<<

Neurochemistry International 140 (2020) 104809

Contents lists available at ScienceDirect

Neurochemistry International

journal homepage: www.elsevier.com/locate/neuint

Review Novel aspects of glutamine synthetase in ammonia homeostasis

Yun Zhou a,*, Tore Eid b, Bjørnar Hassel c, Niels Christian Danbolt a a Neurotransporter Group, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317, Oslo, Norway b Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA c Department of Complex Neurology and Neurohabilitation, Oslo University Hospital, University of Oslo, N-0450, Oslo, Norway

ARTICLE INFO ABSTRACT

Keywords: Elevated blood ammonia (hyperammonemia) is believed to be a major contributor to the neurological sequelae Ammonia following severe liver disease. Ammonia is cleared via two main mechanisms, the urea cycle pathway and the Brain glutamine synthetase reaction. Recent studies of genetically modifiedanimals confirmthe importance of the urea Glutamine synthetase cycle, but also suggest that the glutamine synthetase reaction is more important than previously recognized. Glutamate transporter While the liver clears about two-thirds of the body’s ammonia via the combined action of the urea cycle and Liver Neurotransmitter glutamine synthetase, extrahepatic tissues do not express all the components required for performing a complete Urea cycle urea cycle and therefore depend on the glutamine synthetase reaction for ammonia clearance. The brain is particularly vulnerable to the effects of hyperammonemia, which include impaired extracellular potassium buffering and brain edema. Moreover, the glutamine synthetase reaction is intimately linked to the metabolism of the excitatory and inhibitory neurotransmitters glutamate and gamma aminobutyric acid (GABA), implicating a key role for this enzyme in neurotransmission. This review discusses the emerging roles of glutamine synthetase in brain pathophysiology, particularly aspects related to ammonia homeostasis and hepatic encephalopathy.

experimental portacaval shunting in dogs caused hyperammonemia and encephalopathy (Hahn et al., 1893). The link between hyper­ 1. Introduction ammonemia and encephalopathy has been solidified by a substantial body of evidence from patients (Clemmesen et al., 1999) and animal In mammals, ammonia is a biologically significant intermediate in models of liver disease (for reviews see: Butterworth et al., 1987; several biochemical processes and not merely a waste product from Clemmesen et al., 1999; Bernal et al., 2007; Aldridge et al., 2015). metabolism of nitrogenous compounds. Hyperammonemia (elevated Interestingly, other factors contribute to the encephalopathy because concentrations of ammonia in blood) may be caused by a number of the severity of the encephalopathy does not correlate with the blood conditions, such as liver failure, portocaval shunting, colonization of the ammonia concentration (Shawcross et al., 2011), and because there is gut with urease-producing bacteria, and inborn errors of metabolism not a consistent linear relationship between ammonia levels in the blood (Haberle, 2013). In addition, the brain may be exposed to high con­ and the brain (e.g. Cauli et al., 2008; Zwirner et al., 2010; Kanamori centrations of ammonia during bacterial encephalitis and brain abscess et al., 2002; Dahlberg et al., 2016; Shawcross et al., 2011). One possible formation (Dahlberg et al., 2016). factor is inflammation (Shawcross et al., 2007; for reviews see: Coltart Like several other biochemicals, e.g. glutamate (for reviews see e.g.: et al., 2013; Jayakumar et al., 2015; Tranah et al., 2013; Romero-Gomez Danbolt, 2001), ammonia is toxic if present at excessive concentrations. et al., 2015; Aldridge et al., 2015). Another complicating issue is that the The clinical features of hyperammonemia range in severity and include pathogenesis of ammonia-related encephalopathy is not exclusively personality changes, confusion, seizures, coma, and death (for reviews driven by the liver, but also involves other organs with high ammonia see: Cash et al., 2010; Prakash and Mullen, 2010). Some of the symptoms turnover, such as the skeletal muscle and the kidneys (Aldridge et al., are probably direct consequences of ammonia’s interference with 2015). cellular processes (Bosoi and Rose, 2009), while others are indirect, The scope of the present review is to provide an up to date account of likely reflecting tissue edema (Rovira et al., 2008; Alonso et al., 2014; the mechanisms underlying ammonia metabolism, with particular Larangeira et al., 2018). emphasis on the emerging importance of glutamine synthetase (Glul; EC The toxicity of ammonia was noted more than a century ago when

* Corresponding author. E-mail address: [email protected] (Y. Zhou). https://doi.org/10.1016/j.neuint.2020.104809 Received 29 March 2020; Received in revised form 8 July 2020; Accepted 9 July 2020 Available online 3 August 2020 0197-0186/© 2020 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Abbreviations EAAT2 Excitatory Amino Acid transporter (slc1a2; Pines et al., 1992) ALT alanine aminotransferase (EC 2.6.1.2) FTCD glutamate formiminotransferase (EC 2.1.2.5) AST aspartate aminotransferase (EC 2.6.1.1) GDH glutamate dehydrogenase ASS argininosuccinate synthase (EC 6.3.4.5) GLS2 liver glutaminase (EC 3.5.1.2) ASL argininosuccinate lyase (EC 4.3.2.1) HAL histidine ammonia lyase (EC 4.3.1.3) ARG1 arginase 1 (EC 3.5.3.1) HE hepatic encephalopathy CAS Chemical Abstracts Service registry number OAT ornithine aminotransferase (EC 2.6.1.13) CSF cerebrospinal fluid OCT ornithine transcarbamylase (EC 2.1.3.3) CPS1 carbamoyl-phosphate synthetase 1 (EC 6.3.4.16) MSO methionine sulfoximine (CAS 15985-39-4) CNS central nervous system MTLE mesial temporal lobe epilepsy Cre Cre recombinase (cyclization recombinase) NAGS N-acetylglutamate synthase (EC 2.3.1.1) Glul glutamate ammonia ligase = glutamine synthetase (EC RRID Research Resource Identifiers (https://scicrunch.org/ 6.3.1.2) resources)

6.3.1.2) in ammonia homeostasis and brain function. We will not discuss In contrast, glutamine synthetase and ornithine aminotransferase the pathophysiological sequelae of hyperammonemia, as those have (OAT; EC 2.6.1.13) are both localized exclusively in a rim of 2–3 layers been thoroughly reviewed by others (for review see: Visek, 1968; But­ of perivenous hepatocytes. Further, these hepatocytes also take up terworth et al., 1987; Braissant et al., 2013; Aldridge et al., 2015; Bosoi glutamate and ammonium by, respectively, the glutamate transporter and Rose, 2009)). subtype EAAT2 (slc1a2; see below) and the ammonium transporter Rh B glycoprotein (slc42a2; Haussinger, 1983; Gebhardt et al., 1988; Stoll 2. Zonation of ammonia and glutamate metabolism in the liver et al., 1991; Taylor and Rennie, 1987; Weiner et al., 2003). In the adult liver, glutamate dehydrogenase (GDH; EC 1.4.1.2) is present in high The recent availability of genetically modified animals has made it concentrations around the terminal portal and hepatic veins (thus in easier to uncover the interaction between ammonia metabolism and both periportal and perivenous hepatocytes), but is less abundant in the glutamate metabolism. For instance, urea cycle mutations (e.g. mice intermediate zone (Lamers et al., 1988). The pattern of zonation is lacking CPS1) are associated with severe increases in blood glutamine regulated through β-catenin-dependent and glucagon-dependent mech­ and ammonia levels (Khoja et al., 2019). Additionally, mice lacking anisms (Burke et al., 2009; Cheng et al., 2018). hepatic glutamate dehydrogenase display altered ammonia homeostasis Anterograde and retrograde perfusion experiments, as well as tar­ characterized by increased blood ammonia and reduced conversion to geted lesion experiments, have confirmed the metabolic zonation and urea (Karaca et al., 2018). Loss-of-function mutation in the glutamine suggest that the urea cycle pathway is a low affinity, high capacity transporter SNAT3 (slc38a3) causes several metabolic perturbations in ammonia clearance system that is preferentially localized to the peri­ the liver such as decreased glutamine levels, as well as increased urea portal hepatocytes. In contrast, the glutamine synthetase reaction in the levels (Chan et al., 2016). perivenous hepatocytes is believed to represent a high affinity, low ca­ Although indistinguishable from each other by hematoxylin & eosin pacity ammonia clearance system independent from the urea cycle staining, hepatocytes are functionally different depending on their pathway (Gebhardt and Mecke, 1983; Haussinger, 1983; Gebhardt et al., localization along the porto-central axis of the liver (for review see: 1988). Rappaport, 1958; Gumucio and Miller, 1981; Jungermann and Katz, 1989). For example, the metabolism of nitrogen and glutamate in he­ 3. The importance of the urea cycle in ammonia metabolism patocytes at the entrance of the sinusoids (periportal hepatocytes) is quite different from that of hepatocytes close to the central venules In 1932, Krebs and Henseleit used the surviving tissue slice technique of (perivenous/pericentral hepatocytes, summarized in Fig. 1). This Warburg to show that mammalian liver slices form urea from ammonia pattern first became apparent when the cellular localizations of and carbon dioxide (Krebs, 1942). Their pioneer study not only refuted ammonia-metabolizing enzymes and transporter proteins were deter­ the prevailing hypothesis that ammonium cyanate or related compounds mined by in situ hybridization (Moorman et al., 1994; Berger and were involved in urea formation, but also initiated a new era of Hediger, 2006), by immunohistochemistry (Gebhardt and Mecke, 1983; biochemistry related to urea biosynthesis by definingthe metabolic roles Gaasbeek Janzen et al., 1984; Gaasbeek Janzen et al., 1987; Gebhardt of ornithine, citrulline and arginine. With the efforts from numerous et al., 1988; Gascon-Barre et al., 1989; Kuo et al., 1991; Matsuzawa other biochemists, the chemical reactions involving the five key urea et al., 1997; O’sullivan et al., 1998; Najimi et al., 2014; Hu et al., 2018) cycle enzymes were clarified, i.e. carbamoyl-phosphate synthetase 1 and by microarray analysis (Braeuning et al., 2006). (CPS1), ornithine transcarbamylase (OTC), argininosuccinate synthase The following enzymes are predominantly present in periportal he­ (ASS), argininosuccinate lyase (ASL), and arginase 1 (ARG1). As shown patocytes: carbamoyl-phosphate synthetase 1 (CPS1; EC 6.3.4.16), in Fig. 2, the initial step in the urea cycle is the synthesis of carbamoyl ornithine transcarbamylase (OTC; EC 2.1.3.3), argininosuccinate syn­ phosphate from bicarbonate and ammonia via the mitochondrial thase (ASS; EC 6.3.4.5), argininosuccinate lyase (ASL; EC 4.3.2.1), enzyme carbamoyl phosphate synthetase 1 (CPS1; Krebs, 1942; Cohen arginase 1 (ARG1; EC 3.5.3.1), glutaminase (GLS2; EC 3.5.1.2), alanine and Hayano, 1948; Grisolia and Cohen, 1952). The next step is the aminotransferase (ALT; EC 2.6.1.2), aspartate aminotransferase (AST; formation of citrulline from carbamoyl phosphate and ornithine, cata­ EC 2.6.1.1), and three enzymes involved in histidine catabolism [histi­ lyzed by ornithine transcarbamylase (OTC) which is the only urea cycle dine ammonia lyase (HAL; EC 4.3.1.3), urocanate hydratase (UROC1; EC gene found on the X chromosome (KREBS et al., 1955; Marshall and 4.2.1.49) and glutamate formiminotransferase (FTCD; EC 2.1.2.5)]. Cohen, 1972). This is followed by the cytosolic components of the cycle, These hepatocytes also have the largest uptake capacity for glutamine beginning with the enzyme arginosuccinate synthase (ASS), which cat­ and alanine (Haussinger, 1983; Miyanaka et al., 1998; Gebhardt et al., alyzes the formation of arginosuccinate from citrulline and aspartate 1988; Stoll et al., 1991; Taylor and Rennie, 1987; Baird et al., 2004; (Saheki et al., 1975; Takada et al., 1979; Rochovansky et al., 1977; Botini et al., 2005). Ratner and Pappas, 1949). Arginosuccinate is cleaved via

2 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Fig. 1. Hepatic zonation of ammonia metabolism and glutamate metabolism. Simplified schematic illustration of ammonia removal via the urea cycle and via the glutamine synthetase pathway in the liver. The urea cycle pathway is a low affinity, high capacity ammonia clearance system that is preferen­ tially localized to the periportal hepatocytes, while the glutamine synthetase reaction in the perivenous hepatocytes represent a high affinity, low capacity system for escaping ammonia. Dysfunctions in either urea cycle or glutamine synthesis induces hyper­ ammonemia and causes encephalopathy (Qvarts­ khava et al., 2015; Hakvoort et al., 2017). These processes are intermingled with glutamate meta­ bolism via a differential involvement of enzymes and transporter proteins. Amino acids (AA, except gluta­ mate and aspartate) are taken up and catabolized inside periporal hepatocytes (Haussinger¨ et al., 1992). The two well studied examples are abundant plasma amino acids glutamine (Gln) and alanine, which possibly undergo transport inside the cells by slc38a5 (Taylor and Rennie, 1987). Liver glutaminase (GLS2) hydrolyses glutamine to glutamate and ammonia, and aminotransferases (ATs), e.g. alanine aminotransferase, transfer an amino group to alpha-ketogutarate (α-KG) to produce glutamate. The reaction catalyzed by glutamate dehydrogenase (GDH) is reversible. GDH produces ammonia that provides one nitrogen of the urea molecule. Also, it can furnish mitochondrial glutamate, which supports the formation of aspartate for the urea cycle (Nissim et al., 2003; Karaca et al., 2018). Glutamate partici­ pates in the urea cycle in several ways. First, it plays a regulatory role in the urea cycle because N-acetyl­ glutamate (NAG) is an obligatory activator of car­ bamyl phosphate synthetase 1 (CPS1) and is produced from glutamate and acetyl-coenzyme A. Second, glutamate donates its amino group to form aspartate, which then supplies the second nitrogen of the urea molecule. Third, as described above, the GDH reac­ tion glutamate is a donor of NH3. Perivenous hepa­ tocytes, on the other hand, take up glutamate (Glu) and aspartate (Asp) by the EAAT2 (slc1a2) isoform (Stoll et al 1991; Hu et al 2018). However, these cells apparently can produce glutamine in the absence of exogenous glutamate, and release glutamine probably via slc38a3. Besides alpha-ketoglutarate (α-KG) that is produced by Krebs cycle, ornithine is catabolized in ornithine aminotransferase (OAT)-containing hepa­ tocytes to provide a substrate for glutamine synthesis (Kuo et al 1991; O’sullivan et al 1998). The enzymes or transporter proteins that are predominantly pre­ sent in the periportal zone are indicated in red, while those in perivenous zone are indicated in green, those which are present in both zones are indicated in white and those with little information are indicated in gray.

arginosuccinate lyase (ASL) to arginine and fumarate which enters the transporters that carry ornithine and aspartate across the mitochondrial citric acid cycle (Ratner et al., 1953; Murakami-Murofushi and Ratner, membrane were eventually identified. Ornithine reenters the mito­ 1979). Finally, arginine is cleaved by arginase 1 to urea and ornithine chondria via ORNT1 (slc25a15) in exchange for citrulline (Camacho (Wu and Morris, 1998). Through extensive enzymological studies, it was et al., 1999; Kuno et al., 1990), while aspartate is released to cytosol via also revealed that acetylglutamate, catalyzed by N-acetylglutamate Citrin (slc25a13; Kobayashi et al., 1999). As expected, congenital or synthase (NAGS; EC 2.3.1.1), acts as allosteric activator of carbamoyl acquired deficiencies in any of the above listed enzymes and trans­ phosphate synthetase (Grisolia and Cohen, 1953; Shigesada and Tati­ porters can result in various degrees of hyperammonemia and enceph­ bana, 1971; Hall et al., 1958). Because the urea cycle is carried out in alopathy (summarized in Table 1). The history of the science related to two subcellular compartments (mitochondria and cytosol), mitochon­ the urea cycle and the metabolism of arginine and citrulline has been drial transporters had to be important, and in agreement, the two reviewed in detail elsewhere (Ratner, 1954; Holmes, 1980; Cohen, 1981;

3 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Fig. 2. The urea synthesis in the periportal hepatocytes. Cartoon depicting the formation of urea with the involvement of intermediate metabolites ornithine, citrulline and arginine. The chemical reactions and the involved enzymes/transporters (highlighted by blue) are detailed in the text.

Wu and Morris, 1998). 2019), as well as in enterocytes and immune cells (Stettner et al., 2018). The current non-surgical treatments of urea cycle disorders include Deficiency in these enzymes leads to reduced arginine levels and various approaches aimed at lowering ammonia levels (Tuchman et al., impaired synthesis of nitric oxide with multi-organ dysfunction as the 2008; Haberle et al., 2012a) such as ammonia scavenging agents (e.g. downstream consequence (Erez et al., 2011). sodium phenylacetate and sodium benzoate; Maestri et al., 1991; Enns et al., 2007), a low-protein diet with essential amino acid supplemen­ 4. The importance of liver glutamine synthetase in ammonia tation (Adam et al., 2013), hemodialysis, and arginine or citrulline metabolism supplementation (Brusilow, 1984; Donn and Thoene, 1985; Nagasaka et al., 2006). NAGS deficiencyin humans can be effectively treated with The urea cycle has long been considered to be the primary pathway N-carbamylglutamate (Caldovic et al., 2004; Heibel et al., 2011), and of nitrogen disposal. In the same year when Krebs described the Citrin deficiencyhas been successfully treated with a low-carbohydrate biosynthesis of urea from ammonia and carbon dioxide, he also noted diet with arginine supplementation (Saheki and Song, 1993). Even that the isolated rat liver removed more ammonia from the nutrient though these therapies have significantly improved the overall medium than was accounted for by the formation of urea (Krebs and morbidity and mortality, residual disease sequelae such as develop­ Henseleit, 1932). In 1935 he described the synthesis of glutamine from mental delay and language difficultiesoften persist (Maestri et al., 1991; ammonia and glutamate in the guinea pig kidney, brain, retina and other Kim et al., 2012). Intriguingly, the sequelae of urea cycle disorders tissues (Krebs, 1935). As shown in Table 1, the presence of hyper­ cannot be entirely explained by ammonia toxicity, because patients with glutaminemia in hyperammonemic patients with urea cycle disorders ASL and ORNT1 deficiencies may have neurological impairment in the has been known since the earliest case reports. Speck (1947) and Elliot absence of hyperammonemia (Baruteau et al., 2017; Kim et al., 2012). (1948 ab) independently discovered, respectively, in sheep brain and Moreover, some urea cycle enzymes and transporters are not exclusively pigeon liver, an enzyme system which catalyzes the synthesis of gluta­ expressed in the liver. For instance, ASS, CPS1 and ORNT1 show similar mine by combining glutamate with ammonia (Elliott, 1948; Elliott and patterns of developmental changes in the liver and small intestine Gale, 1948; Speck, 1947). The enzyme was later termed “glutamine (Begum et al., 2002). ARG1 is also found in the brain (Yu et al., 2001) synthetase (GS)" or “glutamate-ammonia ligase (Glul). Meister and and ASL is present in the brain stem (in locus coeruleus; Lerner et al., co-workers used a highly effective stereochemical approach to

4 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Table 1 Phenotypes of urea cycle disorders in human and mouse.

Gene Species Biochemical characteristics Phenotype Ref.

CPS1 Human severe hyperammonemia; elevated plasma glutamine; coma; death; protein intolerance; Gelehrter and Snodgrass (1974); reduced citrulline; absence of orotic aciduria developmental retardation; lethargy McReynolds et al. (1981); Wong et al. (1994) Mouse severe hyperammonemia; elevated plasma glutamine and die within 36 h of birth Schofield et al. (1999) (neonatal alanine; reduced citrulline and arginine Khoja et al. (2019) onset) Mouse (late hyperammonemia; encephalopathy; weight loss; death Khoja et al. (2018) onset) elevated plasma glutamine

NAGS Human neontal hyperammonia; elevated plasma glutamine; low can be treated with N-carbamylglutamate Bachmann et al. (1981); Al Kaabi citrulline level; absence of orotic aciduria treatment and El-Hattab (2016) Mouse hyperammonemia; elevated plasma glutamine, glutamate death within 48 h; can be rescued by carbamyl- Senkevitch et al. (2012) and lysine; reduced citrulline, arginine and ornithine l-glutamate and l-citrulline

OCT Human hyperammonemia; hyperglutaminemia coma; convulsions; metabolic acidosis Levin et al. (1969); Kang et al. (1973); Campbell et al. (1973) Mouse elevated plasma ammonia and glutamine; decreased a markedly decreased lifespan; retarded DeMars et al. (1976); citrulline and arginine; orotic aciduria growth; reduced fitness; hepatic fibrosis Batshaw et al. (1995); Wang et al. (2017)

ASS1 Human hyperammonemia; elevated plasma glutamine citrullinemia type I; progressive lethargic; Quinonez and Thoene (1993) premature death; feed poorly; spasticity; mental retardation; seizure; neurological deficits Mouse acute hyperammonemia; elevated plasma citrulline; early neonatal death; severe retardation; Patejunas et al. (1994) decreased arginine alopecia; lethargy; ataxia Perez et al. (2010)

Slc25a13 Human late onset of serious and recurring hyperammonia; often neonatal intrahepatic cholestasis; Saheki and Song (1993); Saheki citrullinemia (moderate compared with CTLN1 patients); neuropsychiatric symptoms; often leading to et al. (2002) decreased plasma glutamine; elevated arginine; an rapid death; reduced carbohydrate intakes increase in plasma threonine-to-serine ratio Mouse elevated ammonia, glutamate and glutamine; higher deficits in ureogenesis and gluconeogenesis; Moriyama et al. (2006) glycerol phosphate shuttle activity fatty liver (strain-dependent); no adult-onset Sinasac et al. (2004) type II citrullinemia

Slc25a13 Mouse hyperammonemia under fed conditions; citrullinemia; decreased survival; growth retardation; fatty Saheki et al. (2007); Saheki et al. and hypoglycemia liver; low-carbohydrate diets including (2012) mGPD increased protein content lead to metabolic correction

ASL Human episodic hyperammonemia; argininosuccinic aciduria; lethargy; poor feeding; tachypnea; seizures, Allan et al. (1958); elevated plasma citrulline, glutamine, alanine and coma; death; cognitive impairments; learning Nagamani et al. (2012) glycine disabilities; trichorrhexis nodosa Mouse hyperammonemia; elevated glutamine, arginosuccinic lethality, reduced body weight Reid Sutton et al. (2003) acid and citrulline; low plasma arginine

ARG1 Human hyperarginemia; hyperammonemia (less frequent) failure to thrive; cognitive dysfunction; Chandra et al. (2019); spasticity; seizures Crombez and Cederbaum (2005); Qureshi et al. (1983) Mouse late-onset hyperammonemia; moderate hyperarginemia; premature death; encephalopahy Iyer et al. (2002) reduced ornithine Deignan et al. (2006) Ballantyne et al. (2016) Kasten et al. (2013)

ORNT1 Human hyperammonaemia; hyperornithinaemia; intellectual disabilities; spastic paraparesis; Camacho et al. (1999); Salvi et al. homocitrullinuria hepatopathy; epilepsy (2001); Tezcan et al. (2012); Kimber et al. (1990); Fecarotta et al. (2006) understand key aspects of Glul function, such as its substrates, catalytic et al., 2010; Boissonnet et al., 2012), suggesting an important role of intermediates, and potential inhibitors (Meister, 1968a, 1968b; Rowe Glul in ammonia metabolism. This notion is further strengthened by the and Meister, 1970; Manning et al., 1969; Wellner and Meister, 1966; report of three human subjects (not related to each other) with partial Cooper et al., 1976). One such inhibitor was methionine sulfoximine congenital Glul deficiency (Haberle et al., 2005; Haberle et al., 2006; (MSO; CAS15985-39-4) which was produced during chemical bleaching Haberle et al., 2012b; reviewed in Spodenkiewicz et al., 2016). The (agenization) of flourwith agene (nitrogen trichloride; CAS 10025-85-1) subjects had increased blood ammonia levels, suffered from severe and causes acute and chronic seizures when ingested by or administered neonatal epileptic encephalopathy, and died young from multi-organ to animals (Campbell et al., 1951; Silver and Johnson, 1947). The use of failure. The severe and systemic manifestations of the Glul deficiency agene as bleaching agent was banned in the USA in 1949. are consistent with the widespread distribution and important roles of The significance of the Glul pathway for ammonia removal became the enzyme. evident when 15N-labeled ammonia was administered to laboratory Investigations of the physiological consequences of Glul have, until animals, resulting in more 15N-labeling of glutamine than of urea (Duda recently, relied heavily on the use of MSO. This approach, however, has and Handler, 1958; Jahoor et al., 1988). Further, systemic administra­ limitations. First, systemically administered MSO inhibits Glul in the tion of MSO reduces glutamine synthesis in multiple organs and leads to entire body, and thus it is unclear whether the neurological symptoms hyperammonemia (Warren and Schenker, 1964; Kant et al., 2014; Blin from systemic MSO are caused by inhibition of liver Glul, brain Glul, or et al., 2002; Rowe and Meister, 1970; Griffith and Meister, 1978; Cloix both. Second, MSO has several known effects other than inhibiting Glul,

5 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Table 2 Phenotypes of conditional glutamine synthetase knockout mice.

Organ/Cell Phenotypes Ref.

Global (all cells from conception) The blastocysts fail to implant and die at ED3.4. He et al. (2007) Brain/most CNS astrocytes (hGFAP-Cre) The mice die soon after birth. They fail to feed and have slightly increased cortical ammonia levels. He et al. (2010a) Cerebral cortex (glutamatergic neurons and The mice are viable, but they have reduced life span, develop spontaneous seizures from 6 weeks of age, Zhou et al. (2019a) glia; Emx1-IRES-Cre) and have decreased locomotive activities with episodes of wild running. Oligodendrocytes (MOGi-Cre) The mice are viable, but neuronal glutamatergic transmission is disrupted in a myelin-independent way. Xin et al. (2019) There are deficits in cocaine-induced locomotor sensitization. Macrophages (CSF1R-CreERT) The mice are viable. But they have an increased capacity to induce T-cell recruitment, a reduced T-cell Palmieri et al. (2017) suppressive potential and an impaired ability to foster endothelial cell branching or cancer cell motility. Endothelial cells (VE-cadherin(PAC)- The mice are viable, but have impaired retinal vessel sprouting during vascular development. Eelen et al. (2018) creERT2 and Pdgfb-creERT2) Liver (Alb-Cre and Alfp-Cre) The mice are viable, but they have systemic hyperammonia, and exhibit behavioral abnormalities Hakvoort et al. (2017); including increased locomotion, impaired fear memory, a slightly reduced life span, reduced exploratory Qvartskhava et al. activity, and delayed habituation to a novel environment. (2015); Chepkova et al. (2017) Muscle (MCK-Cre) The mice are healthy and fertile, but they have a 5-fold higher escaped ammonia when fasting. He et al. (2010b) Kidney (PEPCK-Cre) The mice are viable, but under basal conditions they have increased urinary ammonia excretion. During Lee et al. (2016); Lee metabolic acidosis or during dietary protein restriction, they have impaired adaptive responses in et al. (2017) ammonia excretion. Pancreas (Pdx1-Cre) Pancreatic ductal carcinoma development is supressed. Bott et al. (2019) such as impairing glutathione synthesis by inhibiting ɣ-glutamylcysteine 21Mgn/J, stock no. 003574; RRID: IMSR_JAX:003574)] becomes active synthetase (Bernard-Helary et al., 2002; Shaw and Bains, 2002; Kam and after birth and results in a gradual deletion which completes at the adult Nicoll, 2007). stage (Fig. 3A). To more accurately assess the roles of Glul in health and disease, several transgenic approaches have been undertaken. The firstapproach 5. The excitatory amino acid (glutamate) transporter subtype 2 was to completely delete the Glul gene throughout the body from (EAAT2) is co-expressed with glutamine synthetase in pericentral conception. However, such deletion leads to embryonic demise at em­ hepatocytes, but is not critical for ammonia detoxification bryonic day 3 (E3), suggesting that Glul is crucial for life (He et al., 2007). In a series of subsequent experiments that took advantage of the The pericentral hepatocytes also express EAAT2 (Berger and Cre-LoxP technology (Le and Sauer, 2000), several knockout mouse lines Hediger, 2006; Najimi et al., 2014; Zhou et al., 2014; Hu et al., 2018). with more restricted Glul deletions were independently created by three EAAT2 is the major glutamate transporter in the brain (Haugeto et al., research groups (He et al., 2010a: MGI:4462791, RRID: 1996; Tanaka et al., 1997; Holmseth et al., 2012a), and is the only IMSR_JAX:029827; Qvartskhava et al., 2015: MGI:5750936; Zhou et al., significantly expressed transporter for glutamate and aspartate in he­ 2019a: MGI:6285753) (Table 2). patocytes (Azimifar et al., 2014; Hu et al., 2018). Characterization of mice with liver-selective deletion of Glul The co-expression of Glul with EAAT2 in perivenous hepatocytes elegantly revealed the importance of the enzyme for ammonia homeo­ raises the question of how much blood-derived aspartate and glutamate stasis. The enzyme-deficient mice developed hyperammonemia and contribute to the synthesis of glutamine. Recent investigations using encephalopathy (Qvartskhava et al., 2015; Chepkova et al., 2017). liver-specific EAAT2 knockouts, suggest that EAAT2 cannot supply Further, by giving excess amounts of ammonia to the other enough glutamate to fully exploit the capacity of Glul (Hu et al., 2018). liver-selective Glul knockouts (RRID:IMSR_029827), it was discovered This assumption is based on the facts that the expression of hepatic Glul that Glul in the liver metabolizes approximately 35% of blood ammonia, is orders of magnitude higher than that of EAAT2 (Hu et al., 2018), and the urea cycle metabolizes another 35%, and the rest is degraded else­ that the transport catalyzed by EAAT2 is slow (Otis and Jahr, 1998; Otis where in the body (Hakvoort et al., 2017). While the data agree with and Kavanaugh, 2000; Bergles et al., 2002; Grewer and Rauen, 2005). In prior estimates that 80–87% of blood ammonia is metabolized by the line with this idea, mice lacking hepatic EAAT2 do not display loco­ liver (Aldrete, 1975), the equal contribution of Glul and the urea cycle to motor dysfunction (Hu et al., 2017) like mice lacking hepatic Glul the metabolism was unexpected, as most prior studies had suggested (Qvartskhava et al., 2015). The reason why EAAT2 may not be essential that the urea cycle was the most important. for liver function is that hepatocytes can produce glutamate via other Deletion of Glul from the liver was reported in one study to give pathways such as oxidation of ornithine by ornithine aminotransferase pericentral macrovesicular steatosis (Hakvoort et al., 2017). This would (OAT; O’sullivan et al., 1998) or from α-ketoglutarate (Braeuning et al., be in line with another study showing that hyperammonemia facilitates 2006) (Fig. 1). development of non-alcoholic steatohepatitis (De Chiara et al., 2020). It should be noted, however, that EAAT2 expression in perivenous However, when the liver selective deletion of Glul was replicated by us hepatocytes may be upregulated in disease, e.g. cholestasis (Najimi (Fig. 3D) and by Qvartskhava and coworkers (Qvartskhava et al., 2019), et al., 2014). Further, dietary supplementation with certain amino acids steatosis was not observed. We also did not observe postnatal growth (e.g. aspartate, glutamate, ornithine and arginine) attenuates retardation in our liver-specific glutamine synthetase knockouts carbohydrate-induced hyperammonemia in Citrin/mitochondrial (Fig. 3C) as Hakvoort and colleagues did. Hakvoort and Lamers glycerol-3-phosphate dehydrogenase double knockouts which have an concluded that the differences in steatosis were unlikely due to the impaired malate-aspartate shuttle (Saheki et al., 2019). Cre-line (Alfp-Cre) itself. Therefore, they speculated whether the dif­ ferences could be due to variations in the genetic background of the mice 6. Controversies concerning glutamine as a nitrogen donor for as they had used FVB/N mice while Qvartskhava and we had used urea synthesis C57BL6J mice (Hakvoort and Lamers, 2019). We, however, propose that the difference in steatosis development might be due to the age at which In addition to ammonia-derived nitrogen, other nitrogen donors for Glul is deleted from the liver: the Alfp-Cre construct initiates deletion of urea synthesis have been proposed. In periportal hepatocytes, Glul already at embryonic day E9.5 (Kellendonk et al., 2000; Parviz glutamate-nitrogen enters the urea cycle predominantly as aspartate et al., 2003), whereas the other Cre-line [Alb-Cre (B6.Cg-Tg (Alb-Cre) (Jahoor et al., 1988). But where does the glutamate-nitrogen come

6 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Fig. 3. Glutamine synthetase (GS) deletion that is controlled by the albumin promotor (Alb-Cre) occurs progressively in the liver during postnatal develop­ ment, being nearly complete in the young adult stage (about 7 weeks of old). Panel A. Immunocytochem­ istry analysis of the liver shows the incomplete dele­ tion of the glutamine synthetase protein (red) in the developing liver (P22) and almost complete deletion in mature liver (P55 and P89). Scale bar, 40 μm. Panel B. Immunoblotting analysis of liver-specific GS knockouts (cKO) and wild-type (WT) littermates (58–64 days old) confirm the absence of the gluta­ mine synthetase in the liver and the preservation in the brain. Panel C. The Alb-Cre driven glutamine synthetase knockouts (male, n = 11) have normal postnatal growth as their wildtype littermates do (n = 13) in C57BL6J mice. Panel D. Steatosis is absent in the liver-specific glutamine synthetase knockout fed with regular chows (3 months and 12 months of age; n = 3). The frozen tissue sections are stained for Oil Red O. Livers from high-fat fed mice were used as a positive control (data not shown). Scale bar, 100 μm. The data in the figure has only been published in abstract form (Hu et al., 2017).

et al., 2004). Considering that periportal hepatocytes can take up Table 3 glutamine and also express the liver isoform of glutaminase (which Localization of glutamine synthetase in rodents. catalyzes the conversion of glutamine to glutamate), it follows that the Organ Cellular localization Ref. amide-nitrogen of glutamine may represent a source of urea-nitrogen. Liver perivenous hepatocytes Gebhardt and Mecke (1983); This concept is experimentally supported by observations that addi­ Gaasbeek Janzen et al. tion of glutamine to liver slices results in considerable enhancement of (1987); Hu et al. (2018) urea production (e.g. Bach and Smith, 1956; Nissim et al., 1992). Brain astroglia Martinez-Hernandez et al. However, a major weakness of these studies is a lack of in vivo validation (1977) 15 oligodendrocytes Tansey et al. (1991); Saitoh (Kamin and Handler, 1957). By using N-glutamine, Handler concluded 15 and Araki (2010) that there is negligible formation of urea from N-glutamine in vivo Retina Müller cells Riepe and Norenburg (1977) (Duda and Handler, 1958), but this has been contradicted by another in endothelial cells Eelen et al. (2018) vivo study reporting that glutamine is more important than alanine as a Kidney proximal tubules, intercalated Burch et al. (1978), cells and distal convoluted Verlander et al. (2013) nitrogen donor for urea synthesis (Jahoor et al., 1988). Thus, further tubules investigations are required to fully resolve the role of glutamine in urea Skeletal myocytes Iqbal and Ottaway (1970); synthesis in vivo. muscle He et al. (2010b) Adipose tissue adipocytes van Straaten et al. (2006); 7. Glutamine synthetase in extrahepatic tissues Lie-Venema et al. (1997) Epididymis epithelial cells in the epididymis van Straaten et al. (2006); and testis and Leydig cells Lie-Venema et al. (1997) Glul is strongly expressed in extrahepatic tissues such as the brain, Stomach acid-producing parietal cells van Straaten et al. (2006) the kidneys, the skeletal muscle, the adipose tissue, the male repro­ Gastro- colon Lie-Venema et al. (1997) ductive organs, the pancreas, and the gastro-intestinal tract (see intestinal tract Table 3). These extrahepatic tissues are responsible for approximately Pancreas islets Zhou et al. (2014) one-third of the total ammonia detoxificationcapacity via the glutamine synthetase reaction (Hakvoort et al., 2017). Similar to the liver, the extrahepatic Glul-expressing cells often express glutamate transporters from? Alanine-nitrogen has been suggested because hepatic ammonia (Tables 3 and 4). Interestingly, the pancreas represents an exception, as uptake is accompanied by uptake of alanine in almost equimolar glutamate transporters are not expressed in the islets cells and are only quantities (see Fig. 1; Yang et al., 2000), and because alanine is a major present at very low levels in the exocrine pancreas (Zhou et al., 2014). amino acid extracted by the liver as a substrate for gluconeogenesis Skeletal muscle and kidneys have a particularly high capacity for (Felig et al., 1970). The in vitro work by Brosnan and coworkers confirms ammonia removal (Cruz et al., 2017). Selective deletion of Glul in the the contribution of alanine-nitrogen to urea and to glutamine synthesis, murine skeletal muscle suggests that the maximal capacity of ammonia although the capacity of alanine as a nitrogen source is limited (Brosnan absorption in skeletal muscle is approximately 10% (He et al., 2010b).

7 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Table 4 Localization of excitatory amino acid transporters (EAATs) in rodents.

Organ Subtype Cellular localization Ref.

Liver EAAT2 EAAT2 is the only EAAT-type of transporter in the liver and is selectively Berger and Hediger (2006); expressed in the perivenous hepatocytes Hu et al. (2018) Brain EAAT1 EAAT1 is selective for astroglia. Note that there are some reports claiming Lehre et al. (1995); Ginsberg et al. (1995); Berger and Hediger EAAT1 to be present in neurons, but this has not been supported by later (1998); Berger and Hediger (2000); Hanson et al. (2015) publications. EAAT2 EAAT2 protein is strongly expressed in astroglia Danbolt et al. (1992); Levy et al. (1993); Rothstein et al. (1994); Lehre et al. (1995); Berger and Hediger (1998); Schmitt et al. (1997); Berger and Hediger (2000) EAAT2 mRNA is present in astrocytes and in the majority of neurons in Torp et al. (1994); Schmitt et al. (1996); Torp et al. (1997); Berger multiple CNS regions and Hediger (1998); Berger and Hediger (2000); Berger and Hediger (2001) Torp et al. (1994); EAAT2 is expressed in brain neurons during development, in neurons in Northington et al. (1998); Northington et al. (1999); Martin et al. diseased brain tissue and in cultured neurons (1997); Mennerick et al. (1998); Plachez et al. (2000) Early electron microscopy suggested that nerve terminals, at least in cortex Beart (1976); McLennan (1976); Gundersen et al. (1993) and striatum, are able to take up glutamate though the responsible carrier(s) was not determined. Some of the EAAT2 protein (about 10%) is found in neurons, and is targeted to Chen et al. (2004); the axon-terminals where EAAT2 is the only glutamate transporter. Furness et al. (2008); Zhou et al. (2019b); Petr et al. (2015): Danbolt et al. (2016a) EAAT3 EAAT3 is selective for neurons and present in most if not all neurons, but is Kanai and Hediger (1992); Rothstein et al. (1994); targeted to the cell body and dendrites. Bjoras et al. (1996); Torp et al. (1997); Holmseth et al. (2012a) EAAT4 EAAT4 is expressed in cerebellar Purkinje cells with the highest concentrations Dehnes et al. (1998); Massie et al. (2008) where Zebrin II is, and is also present in scattered neurons in the fore- and midbrain. It is especially enriched in the parts of the dendritic and spine membranes facing astrocytes. Retina EAAT1 EAAT1 is the most abundant of the EAATs in the retina and is expressed by the Lehre et al. (1997); EAAT2 Müller cells. EAAT2 is found in cone photoreceptors and bipolar cells, while Rauen and Kanner (1994); Euler and Wassle (1995); Rauen et al. EAAT5 EAAT5 has been less studied, possibly expressed in Müller cells. (1996); Arriza et al. (1997); Rauen et al. (1999); Rauen (2000); Veruki et al. (2006) Kidney EAAT3 EAAT3 is the only EAAT-type of transporter expressed in the kidney, and is Kanai and Hediger (1992); Hu et al. (2018) found in the proximal tubules Heart EAAT1 Rat hearts contain EAAT3 and EAAT1, but at lower levels than in the brain. Martinov et al. (2014) EAAT3 EAAT2 and EAAT4 were not detected. Skeletal Not reported Muscle Gastro- EAAT3 EAAT3 is the only EAAT-type of transporters in enterocytes, and is found in the Kanai and Hediger (1992); intestinal small intestine and colon with the highest levels in the distal ileum. Hu et al. (2018); Berger and Hediger (2006) tract Exocrine EAAT2 EAAT2 is present at low levels in salivary glands. Berger and Hediger (2006) glands Adipose tissue EAAT1 Several EAATs have been reported in adipocytes, but EAAT1 is the only EAAT Berger and Hediger (2006); Adachi et al. (2007) confirmed by validated in situ hybridization and proteome analysis Pancreas In-depth proteome analysis and characterization of pancreas-specific EAAT2 Zhou et al. (2014) knockout mice show that EAAT2 is not expressed in pancreatic islets as some have reported, but may be expressed at low levels in the exocrine pancreas. In fact, the islets do not express any of the EAATs nor any of the VGLUTs

Although this estimate is much lower than what was proposed in the past (50%; Hod et al., 1982; Lockwood et al., 1979), it should be kept in Table 5 mind that skeletal muscle can upregulate Glul activity and therefore Phenotypes of local administration of MSO. increase ammonia metabolism during liver failure (Hod et al., 1982; Region Phenotype Ref. Desjardins et al., 1999; Clemmesen et al., 2000; Chatauret et al., 2006). Skeletal muscle Abolish the arterio- Hod et al. (1982) The kidneys also adapt to the consequences of a failing liver by reducing venous difference of ammonia release into the systemic circulation by slowing down ammonia + ammonia production and by increasing ammonium (NH ) excretion in Trigeminal subnucleus Altered mechanical Chiang et al. (2007); 4 caudalis activation threshold and Tsuboi et al. (2011) the urine (Dejong et al., 1993; Jalan and Kapoor, 2003). responses to graded A full discussion of ammonia metabolism in the kidneys and skeletal mechanical stimuli; muscle, however, is beyond the scope of this review, as these topics are attenuated nocifensive covered well elsewhere (e.g. Mutch and Banister, 1983; Graham and behavior Entorhinal-hippocampal Recurrent seizures Eid et al. (2008); MacLean, 1998; van de Poll et al., 2004; Weiner and Verlander, 2013; area/hippocampus/central Dhaher et al. (2015); Weiner et al., 2015; Olde Damink et al., 2002; Adeva et al., 2012). nucleus of the amygdala Gruenbaum et al. (2015); 8. Glutamine synthetase is essential for brain function Trigeminal motor nucleus Altered jaw-opening Mostafeezur et al. reflex (2014) Ventriculocisternal space Changes in ventilation Hoop et al. (1988) The importance of brain Glul was first uncovered by local adminis­ tration of MSO to the central nervous system (CNS). Depending on the injection site, MSO caused recurrent seizures, altered responses to graded mechanical stimuli, attenuated nocifensive behavior, or altered

8 Y. Zhou et al. Neurochemistry International 140 (2020) 104809 jaw-opening reflex (Table 5) (Eid et al., 2008; Dhaher et al., 2015; EAAT2/slc1a2; Danbolt et al., 1992; Lehre et al., 1995), converts Gruenbaum et al., 2015; Chiang et al., 2007; Tsuboi et al., 2011; Mos­ glutamate into glutamine via the Glul pathway (Martinez-Hernandez tafeezur et al., 2014). The MSO studies are, however, associated with et al., 1977) and releases glutamine into the extracellular space. Neurons some uncertainty because MSO not only inhibits glutamine synthetase, are able to take up glutamine (Schousboe et al., 1979; Dyste et al., 1989) but also decreases tissue glutathione (Shaw and Bains, 2002), increases via glutamine transporters. It is possible that several glutamine trans­ astrocyte glycogen (Bernard-Helary et al., 2002), and excites neurons porters are involved as there are at least 14 different solute carrier via a Glul-independent mechanism (Kam and Nicoll, 2007). Depletion of proteins with the ability to transport glutamine (for reviews see: neuronal glutathione is shown to increase cytosolic glutamate and af­ Mackenzie and Erickson, 2004; Bhutia and Ganapathy, 2016; Danbolt fects excitatory neurotransmission (Sedlak et al., 2019). et al., 2016a; Hellsten et al., 2017). Inside neurons, glutamine can be Several knockout approaches for Glul have recently become avail­ converted back to glutamate by phosphate-activated glutaminase able (as explained above), making highly accurate in vivo studies of the (PAG/GLS1; Kvamme et al., 2001). A full discussion of glutamine and enzyme possible. However, deletion of Glul in the entire CNS resulted in glutamate metabolism in the brain have been thoroughly reviewed by early neonatal death (He et al., 2010a). More restricted deletions would others (e.g. Yudkoff, 2017; Hertz and Zielke, 2004; McKenna and Fer­ thereby be necessary in order to obtain longer survival. Unfortunately, reira, 2016; Bak et al., 2018; Hertz and Chen, 2018; Hertz and Rothman, there are not that many astroglial Cre drivers, and that the commonly 2016). used ones (e.g. hGFAP-Cre) are active in most CNS astrocytes resulting in The glutamate-glutamine cycle model, however, probably over­ too extensive deletion. It was therefore a breakthrough when we (Zhou simplifies a complex reality, and not all available data fit with this et al., 2019a) were able to selectively delete Glul in the cerebral cortex model. (A) Although it is clear that neurons can take up glutamine, it is (i.e. neocortex and hippocampus) using the Emx1_IRES Cre line. These not known how and to what extent glutamine can enter the nerve ter­ mice survive well into adulthood and exhibit numerous pathological minals as no glutamine transporter protein has so far been shown features such as altered locomotive activity, progressive neuro­ immunocytochemically at the electron microscopic level to be present in degeneration, and spontaneous recurrent seizures (Zhou et al., 2019a). axon-terminals in brain tissue (for review see: Mackenzie and Erickson, In contrast, deletion of glutamine synthetase in the liver (Hakvoort et al., 2004; Conti and Melone, 2006; Grewal et al., 2009; Zhou and Danbolt, 2017), the kidneys (Lee et al., 2016), the skeletal muscle (He et al., 2014). Possible candidates comprise SNAT7 (slc38a7) and SNAT8 2010b), the pancreas (Bott et al., 2019) or the adipose tissue (Zhou Y and (slc38a8), but uncertainty remains as the corresponding knockout mice Danbolt NC, personal observations) barely affected mortality and had a are not yet available as negative controls and quantitative information mild phenotype with no evidence of behavioral seizures (Table 2). has yet to be obtained (Hagglund et al., 2011, 2015). Recently, it has been reported that the glutamine transporter SNAT1 (Slc38a1) partici­ 9. Glutamine production and roles of glutamine in pates in the regulation of the GABA levels in GABAergic terminals neurotransmission although the SNAT1 protein itself was not detected at the terminals (Qureshi et al., 2019; Qureshi et al., 2020). There is electrophysiological Glutamine is abundantly present in the central nervous system, and evidence for glutamine uptake at the Calyx of Held synapse (Billups the synthesis is largely catalyzed by astroglial glutamine synthetase et al., 2013). However, this synapse consists of a special giant nerve (Zhou et al., 2019a). The proposed major role is to be a precursor of the terminal which may not be representative for the majority of synapses in neurotransmitters glutamate (excitatory) and ɣ-amino butyric acid the mammalian CNS. The majority of mammalian synapses have bou­ (GABA; inhibitory). For more than half a century, compartmentation of tons that are very small and we know less about them (von Gersdorff and glutamate-glutamine metabolism has been central to how we envision Borst, 2002). (B) Another unexplored observation is that glutamine glutamine as a precursor for neuronal glutamate (Berl et al., 1962; Berl administered to the intact brain is mostly metabolized to CO2 (Zielke et al., 1968; Berl et al., 1970; Martinez-Hernandez et al., 1977; van den et al., 1998), in agreement with an important role for glutamine as a Berg and Garfinkel,1971 ; Bradford et al., 1978; Hamberger et al., 1979). neuronal energy substrate (Bradford et al., 1978; Hamberger et al., As outlined in Fig. 4, neurotransmitter glutamate is released by exocy­ 1979; Zielke et al., 2009). (C) Electrophysiological analyses of isolated tosis of synaptic vesicles from nerve terminals (for review: Sudhof, brain slices suggest that glutamatergic neurotransmission can be sus­ 2014). The surrounding astrocytes rapidly take up the released gluta­ tained in the absence of the cycle (Kam and Nicoll, 2007). (D) The ter­ mate via two astroglial glutamate transporters (EAAT1/slc1a3 and minals can obtain glutamate independently of glutamine as there is

Fig. 4. Overview of the glutamate–glutamine cycle. (1) Glutamate (Glu) is released from vesicles in the axon-terminals into the extracellular fluidfrom where it can (2) bind to glutamate receptors (GLU-R) before it is inactivated by uptake into astrocytes (3) via excitatory amino acid transporters (EAATs). (4) Inside astrocytes it is converted to glutamine (Gln) via the enzyme glutamine synthetase (Glul). (5) Gln is next shuttled from astrocytes to neurons via mechanisms that remain to be definedunequivocally. (6) Once inside the neurons, Gln is converted back to Glu by glutaminase, which is particularly enriched in mitochondria. (7) Finally, Glu is concentrated in synaptic vesicles via vesicular glutamate transporters (VGLUTs), thus completing the cycle. It should ne noted that (8) axonal terminal expressing EAAT2 will partly short circuit the cycle and that (9) there is an export of glutamine from the brain to the blood.

9 Y. Zhou et al. Neurochemistry International 140 (2020) 104809 some de novo synthesis (Hassel and Bråthe, 2000), and as they can take studies. Moreover, the use of Glul knockout tissue as a control for up glutamate directly via EAAT2 because a portion of EAAT2 (5–10%) is antibody specificityhas shown that Glul is expressed in a subpopulation present in axonal terminals (Furness et al., 2008; Danbolt et al., 2016a; of oligodendrocytes that emerges at P21 and is particularly densely Petr et al., 2015; Zhou et al., 2019b; McNair et al., 2019). (E) As stained in caudal regions of the CNS (Xin et al., 2019). The late onset and McKenna and Yudkoff pointed out earlier, the model ignores the facts regional heterogeneity in Glul expression, may explain earlier incon­ that the cycle is not stoichiometric and that the brain avidly exchanges sistent observations. Finally, selective removal of Glul in endothelial many metabolites with the blood (McKenna, 2007; Yudkoff, 2017). In cells impairs retinal vessel spouting during vascular development (Eelen line with this, the supply of glutamine to terminals may not always keep et al., 2018), and deletion of Glul in macrophages in tumor-bearing mice up with glutamate release (Waagepetersen et al., 2005; Kam and Nicoll, promotes tumor vessel pruning, vascular normalization, accumulation 2007; Marx et al., 2015), because there is a glutamine loss due to the of cytotoxic T-cells, and metastasis inhibition (Palmieri et al., 2017). export from the brain to the blood (Cangiano et al., 1983). This export The above findings challenge the concept that Glul in the CNS is process is mediated by several transporters including LAT1 (slc7a5), exclusively expressed in astrocytes and therefore the two-compartment LAT2 (slc7a8) and SNAT3 (slc38a3) which are robustly expressed in (neuron-glia) model of the glutamate-glutamine metabolic cycle, which brain microvessel endothelial cells (Duelli et al., 2000; Dolgodilina is thought to be critical for the synthesis of the major excitatory and et al., 2016; Dolgodilina et al., 2020). Thus, the relative contributions of inhibitory neurotransmitters glutamate and GABA (e.g. van den Berg the various mechanisms are still debated and a complicating factor is and Garfinkel,1971 ; Rothman et al., 1999). While knockout approaches that this may differ between brain regions and in disease. Marx and can be used to gain a more detailed and accurate understanding of the co-workers has recently tried to put available quantitative data together glutamate-glutamine cycle, they are associated with limitations. For (Marx et al., 2015). instance, the insertion of a promotor-Cre construct into a genome sometimes disturbs the expression of other genes (Forni et al., 2006; 10. Glutamine synthetase in the CNS may be expressed in cells Giusti et al., 2014). It is therefore important to assess whether the Cre other than astrocytes promotor interferes with the study. Likewise, when tamoxifen-inducible constructs have been used, possible side-effects of tamoxifen must be It is well established that Glul is expressed in astrocytes (Marti­ carefully controlled for. This is particularly relevant to cancer and nez-Hernandez et al., 1977); however, it is still debated whether the immunological research, as tamoxifen is an anti-cancer drug and ap­ enzyme is expressed in significant quantities in other CNS cells such as pears to have immunomodulatory effects which are independent of the oligodendrocytes, endothelial cells, microglia and neurons. The lack of a estrogen-receptor (Corriden et al., 2015; for review see: Behjati and consensus is partly due to how Glul is quantifiedin vitro and the fact that Frank, 2009). Another reason to be cautious is that gene expression the expression of the enzyme is regulated by cell to cell interactions and profilesof many cell types, including endothelial cells and macrophages, soluble factors that may be very different in vitro versus in vivo (e.g. are significantlyinfluenced by the microenvironment and may therefore Barakat-Walter and Droz, 1990; Cahoy et al., 2008). This point was well differ between locations and functional states (Gordon and Pluddemann, illustrated by Barakat-Walter and Droz (1990) who found that dorsal 2017; Garlanda and Dejana, 1997). Thus, the expression patterns of Glul root ganglion cells grown in vitro express Glul while those grown in vivo in endothelial cells of the brain and in microglia (a macrophage-like cell do not. Another factor contributing to the lack of consensus is the residing in the CNS) need further studies. It will be interesting to learn different results obtained with immunocytochemistry performed in more about the metabolic consequences of deleting glutamine synthe­ different laboratories with different antibodies and labeling protocols tase in endothelial and microglia in vivo. (Martinez-Hernandez et al., 1977; Norenberg and Martinez-Hernandez, 1979; Cammer, 1990; Tansey et al., 1991; D’Amelio et al., 1990; 11. Brain glutamine synthetase-deficiency and liver-induced Yamamoto et al., 1989; Takasaki et al., 2010), suggesting that differ­ hyperammonemia affect the brain in different ways ences in the specificityof immunohistochemical procedures play a role, as this is extensively discussed in prior publications (e.g. Danbolt et al., The classical hyperammonemia syndrome with increased blood 1998; Holmseth et al., 2006; Lorincz and Nusser, 2008; Saper, 2009; ammonia levels leads to astroglial swelling without significantneuronal Holmseth et al., 2012b; Danbolt et al., 2016b). death (Diemer and Laursen, 1977; Laursen and Diemer, 1979; Martin It is important to resolve the questions with regards the cellular et al., 1987; Desjardins et al., 2012; for reviews see: Butterworth et al., expression of Glul because if the enzyme is expressed in other cell types, 1987; Aldridge et al., 2015), and to glutamine accumulation in the brain then this will impact the interpretation of a number of studies of the (Cooper et al., 1979; Ratnakumari et al., 1992; Haussinger et al., 1994; ammonia metabolism. Fortunately, the recent development of knockout Bosman et al., 1990; Rao and Norenberg, 2001) despite models is expected to close key gaps in knowledge and open up for new ammonia-mediated downregulation of astroglial Glul (14–25%, Girard fields of research that may ultimately translate to novel treatments for et al., 1993; Desjardins et al., 1999; Haussinger et al., 2005). ammonia-related disorders. By knocking out Glul in a cell-specific Selective deletion of hepatic Glul causes a similar syndrome char­ manner, the functional consequences can be studied with a high level acterized by hyperammonemia with astroglial swelling, no visible of accuracy and the tissue can also serve as a specificity control for neurodegeneration, and a minor reduction in life span (Qvartskhava immunocytochemical staining of the enzyme. For example, one of the et al., 2015). The mice display altered behavior characterized by Glul knockout approaches (Emx1Cre/Glul-Flox) targets astrocytes, glu­ increased locomotion, reduced exploratory activities and delayed tamatergic neurons, and oligodendrocytes in the cerebral cortex, but habituation to a novel environment as well as impaired fear memory spares cortical GABAergic neurons and cells in other parts of the brain (Qvartskhava et al., 2015; Chepkova et al., 2017). (Zhou et al., 2019a; Gorski et al., 2002). Another approach selectively In contrast, selective deletion of Glul in the whole brain in mice re­ deletes Glul in oligodendrocytes (Xin et al., 2019), endothelial cells sults in a 14-fold decline in cortical glutamine and a 1.6-fold increase in (Eelen et al., 2018) and macrophages (Palmieri et al., 2017). cortical ammonia levels (He et al., 2010a). These mice die about three Surprisingly, mice lacking oligodendrocyte Glul have about 20% days after birth unless they are fed by hand. In contrast, when Glul is reduction in brain glutamate and glutamine levels and exhibit disturbed selectively deleted in the cerebral cortex (Zhou et al., 2019a), there is a glutamatergic neurotransmission, but unaffected myelination (Xin et al., 4-fold reduction in cortical glutamine and the majority of the animals 2019). The finding of normal myelination in vivo is in contrast to an in survive for several months. There is, however, a progressive gliosis and vitro study where glutamine synthetase deficiencyaffects differentiation impaired neurovascular coupling. These animals exhibit overall of oligodendrocytes (Saitoh and Araki, 2010). These results emphasize decreased locomotion with brief episodes of wild running, and many the importance of validating in vitro findings with subsequent in vivo develop spontaneous recurrent seizures and neurodegeneration that

10 Y. Zhou et al. Neurochemistry International 140 (2020) 104809 commence around 6 weeks of age. A portion of animals die suddenly and Al Kaabi, E.H., El-Hattab, A.W., 2016. N-acetylglutamate synthase deficiency: novel unexpectedly, leading to an overall reduced life-span (Zhou et al., mutation associated with neonatal presentation and literature review of molecular and phenotypic spectra. Mol. Genet. Metab. Rep. 8, 94–98. https://doi.org/10.1016/ 2019a). j.ymgmr.2016.08.004. The phenotype of the cortical Glul knockout mice are in good Aldrete, J.S., 1975. Quantification of the capacity of the liver to remove ammonia from agreement with the observations that patients with partial loss of the circulation of dogs with portacaval transposition. Surg. Gynecol. Obstet. 141, 399–404. function mutations in the Glul gene had brain atrophy and severe Aldridge, D.R., Tranah, E.J., Shawcross, D.L., 2015. Pathogenesis of hepatic neonatal epileptic encephalopathy (for review see: Spodenkiewicz et al., encephalopathy: role of ammonia and systemic inflammation. J. Clin. Exp. Hepatol. 2016). These humans had insufficient Glul activities, not only in the 5, S7–S20. https://doi.org/10.1016/j.jceh.2014.06.004. Allan, J.D., Cusworth, D.C., Dent, C.E., Wilson, V.K., 1958. A disease, probably brain, but in the entire body including liver, skeletal muscle, kidneys and hereditary characterised by severe mental deficiency and a constant gross skin. They died young from multiple organ failures. Further, the defects abnormality of aminoacid metabolism. Lancet 1, 182–187. https://doi.org/10.1016/ were present already at conception, and in this context it should be s0140-6736(58)90666-4. Alonso, J., Cordoba, J., Rovira, A., 2014. Brain magnetic resonance in hepatic noted that the deleterious effects of hyperammonemia may be more encephalopathy. Semin. Ultrasound CT MR 35, 136–152. https://doi.org/10.1053/j. severe to the developing brain than the mature brain (Braissant et al., sult.2013.09.008. 2013). Arriza, J.L., Eliasof, S., Kavanaugh, M.P., Amara, S.G., 1997. Excitatory amino acid Patients with mesial temporal lobe epilepsy (MTLE) have a defi­ transporter 5, a retinal glutamate transporter coupled to a chloride conductance. Proc. Natl. Acad. Sci. U. S. A. 94, 4155–4160. https://doi.org/10.1073/ ciency in glial Glul in the epileptogenic hippocampus (Eid et al., 2004; pnas.94.8.4155. van der Hel et al., 2005); however, this differs from the above cases as Azimifar, S.B., Nagaraj, N., Cox, J., Mann, M., 2014. Cell-type-resolved quantitative – the loss of Glul in MTLE is limited to small regions of the brain and likely proteomics of murine liver. Cell Metabol. 20, 1076 1087. https://doi.org/10.1016/ j.cmet.2014.11.002. occurs later in life when the brain is fully or near-fully developed. Bach, S.J., Smith, M., 1956. Glutamine; a nitrogen source in urea synthesis. Biochem. J. These distinct signatures may suggest different therapeutic ap­ 64, 417–425. https://doi.org/10.1042/bj0640417. proaches. For instance, pharmacological inhibition of Glul was sug­ Bachmann, C., Krahenbuhl, S., Colombo, J.P., Schubiger, G., Jaggi, K.H., Tonz, O., 1981. N-acetylglutamate synthetase deficiency: a disorder of ammonia detoxication. gested to temporarily relief brain edema by attenuating ammonia- N. Engl. J. Med. 304, 543. https://doi.org/10.1056/NEJM198102263040918. induced astroglial swelling, and by ameliorating some of the reactive Baird, F.E., Beattie, K.J., Hyde, A.R., Ganapathy, V., Rennie, M.J., Taylor, P.M., 2004. astroglial cytoskeletal alterations (Blei et al., 1994; Tanigami et al., Bidirectional substrate fluxes through the system N (SNAT5) glutamine transporter may determine net glutamine fluxin rat liver. J. Physiol. 559, 367–381. https://doi. 2005). This strategy, however, worsen the situation in awake intact org/10.1113/jphysiol.2003.060293. animals (Rangroo Thrane et al., 2013). On the other hand, glutamine Bak, L.K., Walls, A.B., Schousboe, A., Waagepetersen, H.S., 2018. Astrocytic glycogen supplementation to a child with inherited Glul deficiency partially metabolism in the healthy and diseased brain. J. Biol. Chem. 293, 7108–7116. https://doi.org/10.1074/jbc.R117.803239. corrected the glutamine level in the cerebrospinal fluid, improved Ballantyne, L.L., Sin, Y.Y., Al-Dirbashi, O.Y., Li, X., Hurlbut, D.J., Funk, C.D., 2016. Liver- alertness and EEG characteristics (Haberle et al., 2012b). specific knockout of arginase-1 leads to a profound phenotype similar to inducible whole body arginase-1 deficiency. Mol. Genet. Metab. Rep. 9, 54–60. https://doi. org/10.1016/j.ymgmr.2016.10.003. 12. Concluding remarks Barakat-Walter, I., Droz, B., 1990. Glutamine synthetase is expressed by primary sensory neurons from chick embryos in vitro but not in vivo: influence of skeletal muscle – Glutamine synthetase plays a significant role in ammonia meta­ extract. Eur. J. Neurosci. 2, 836 844. https://doi.org/10.1111/j.1460-9568.1990. tb00395.x. bolism, and its dysfunction leads to neurological diseases. An improved Baruteau, J., Jameson, E., Morris, A.A., Chakrapani, A., Santra, S., Vijay, S., Kocadag, H., understanding of the regulation and physiological implications of Beesley, C.E., Grunewald, S., Murphy, E., et al., 2017. Expanding the phenotype in glutamine synthesis in health and disease will likely uncover novel drug argininosuccinic aciduria: need for new therapies. J. Inherit. Metab. Dis. 40, 357–368. https://doi.org/10.1007/s10545-017-0022-x. targets for patients who suffer from hyperammonemia. Batshaw, M.L., Yudkoff, M., McLaughlin, B.A., Gorry, E., Anegawa, N.J., Smith, I.A., Hyman, S.L., Robinson, M.B., 1995. The sparse Fur mouse as a model for gene therapy in ornithine carbamoyltransferase deficiency. Gene Ther. 2, 743–749. Declaration of competing interest Beart, P.M., 1976. The autoradiographic localization of L-(3H) glutamate in synaptosomal preparations. Brain Res. 103, 350–355. https://doi.org/10.1016/ 0006-8993(76)90804-0. The authors declare no competing financial interests. Begum, L., Jalil, M.A., Kobayashi, K., Iijima, M., Li, M.X., Yasuda, T., Horiuchi, M., del Arco, A., Satrustegui, J., Saheki, T., 2002. Expression of three mitochondrial solute carriers, citrin, aralar1 and ornithine transporter, in relation to urea cycle in mice. Acknowledgement Biochim. Biophys. Acta 1574, 283–292. https://doi.org/10.1016/s0167-4781(01) 00376-1. Behjati, S., Frank, M.H., 2009. The effects of tamoxifen on immunity. Curr. Med. Chem. This work was supported by the Norwegian Research Council 16, 3076–3080. https://doi.org/10.2174/092986709788803042. (240844), the Novo Nordisk Fonden (Grant no. NNF14OC0010959 and Berger, U.V., Hediger, M.A., 2001. Differential distribution of the glutamate transporters NNF15OC0016528), funds from the University of Oslo (SERTA and GLT-1 and GLAST in tanycytes of the third ventricle. J. Comp. Neurol. 433, 101–114. UNIFOR-FRIMED and UNIFOR-Anders Jahres Fond and Halvor Høies https://doi.org/10.1002/cne.1128. Berger, U.V., Hediger, M.A., 2000. Distribution of the glutamate transporters GLAST and Fond), the National Institute of Neurological Disorders and Stroke GLT-1 in rat circumventricular organs, meninges, and dorsal root ganglia. J. Comp. (NINDS, Grants no. NS058674, NS070824, NS109062, and NS109734), Neurol. 421, 385–399. https://doi.org/10.1002/(sici)1096-9861(20000605)421: < > and the National Center for Advancing Translational Science (NCATS, 3 385::aid-cne7 3.0.co;2-s. Berger, U.V., Hediger, M.A., 2006. Distribution of the glutamate transporters GLT-1 Grant no. RR024139). The contents of the publication are solely the (SLC1A2) and GLAST (SLC1A3) in peripheral organs. Anat. Embryol. 211, 595–606. responsibility of the authors and do not necessarily represent the official https://doi.org/10.1007/s00429-006-0109-x. view of NINDS or NCATS. Berger, U.V., Hediger, M.A., 1998. Comparative analysis of glutamate transporter expression in rat brain using differential double in situ hybridization. Anat. Embryol. 198, 13–30. https://doi.org/10.1007/s004290050161. References Bergles, D.E., Tzingounis, A.V., Jahr, C.E., 2002. Comparison of coupled and uncoupled currents during glutamate uptake by GLT-1 transporters. J. Neurosci. 22, 10153–10162. https://doi.org/10.1523/JNEUROSCI.22-23-10153.2002. Adachi, J., Kumar, C., Zhang, Y., Mann, M., 2007. In-depth analysis of the adipocyte Berl, S., Nicklas, W.J., Clarke, D.D., 1968. Compartmentation of glutamic acid proteome by mass spectrometry and bioinformatics. Mol. Cell. Proteomics 6, metabolism in brain slices. J. Neurochem. 15, 131–140. https://doi.org/10.1111/ 1257–1273. https://doi.org/10.1074/mcp.M600476-MCP200. j.1471-4159.1968.tb06184.x. Adam, S., Almeida, M.F., Assoun, M., Baruteau, J., Bernabei, S.M., Bigot, S., Berl, S., Nicklas, W.J., Clarke, D.D., 1970. Compartmentation of citric acid cycle Champion, H., Daly, A., Dassy, M., Dawson, S., et al., 2013. Dietary management of metabolism in brain: labelling of glutamate, glutamine, aspartate and gaba by urea cycle disorders: European practice. Mol. Genet. Metabol. 110, 439–445. several radioactive tracer metabolites. J. Neurochem. 17, 1009–1015. https://doi. https://doi.org/10.1016/j.ymgme.2013.09.003. org/10.1111/j.1471-4159.1970.tb02254.x. Adeva, M.M., Souto, G., Blanco, N., Donapetry, C., 2012. Ammonium metabolism in humans. Metabolism 61, 1495–1511. https://doi.org/10.1016/j. metabol.2012.07.007.

11 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Berl, S., Takagaki, G., Clarke, D.D., Waelsch, H., 1962. Metabolic compartments in vivo: Campbell, A.G., Rosenberg, L.E., Snodgrass, P.J., Nuzum, C.T., 1973. Ornithine ammonia and glutamic acid metabolism in brain and liver. J. Biol. Chem. 237, transcarbamylase deficiency: a cause of lethal neonatal hyperammonemia in males. 2562–2569. N. Engl. J. Med. 288, 1–6. https://doi.org/10.1056/NEJM197301042880101. Bernal, W., Hall, C., Karvellas, C.J., Auzinger, G., Sizer, E., Wendon, J., 2007. Arterial Campbell, P.N., Work, T.S., Mellanby, E., 1951. The isolation of a toxic substance from ammonia and clinical risk factors for encephalopathy and intracranial hypertension agenized wheat flour. Biochem. J. 48, 106–113. https://doi.org/10.1042/ in acute liver failure. Hepatology 46, 1844–1852. https://doi.org/10.1002/ bj0480106. hep.21838. Cangiano, C., Cardelli-Cangiano, P., James, J.H., Rossi-Fanelli, F., Patrizi, M.A., Bernard-Helary, K., Ardourel, M.-Y., Hevor, T., Cloix, J.-F., 2002. In vivo and in vitro Brackett, K.A., Strom, R., Fischer, J.E., 1983. Brain microvessels take up large glycogenic effects of methionine sulfoximine are different in two inbred strains of neutral amino acids in exchange for glutamine. Cooperative role of Na+-dependent mice. Brain Res. 929, 147–155. https://doi.org/10.1016/s0006-8993(01)03380-7. and Na+-independent systems. J. Biol. Chem. 258, 8949–8954. Bhutia, Y.D., Ganapathy, V., 2016. Glutamine transporters in mammalian cells and their Cash, W.J., McConville, P., McDermott, E., McCormick, P.A., Callender, M.E., functions in physiology and cancer. Biochim. Biophys. Acta 1863, 2531–2539. McDougall, N.I., 2010. Current concepts in the assessment and treatment of hepatic https://doi.org/10.1016/j.bbamcr.2015.12.017. encephalopathy. QJM 103, 9–16. https://doi.org/10.1093/qjmed/hcp152. Billups, D., Marx, M.-C., Mela, I., Billups, B., 2013. Inducible presynaptic glutamine Cauli, O., Rodrigo, R., Boix, J., Piedrafita, B., Agusti, A., Felipo, V., 2008. Acute liver transport supports glutamatergic transmission at the calyx of Held synapse. failure-induced death of rats is delayed or prevented by blocking NMDA receptors in J. Neurosci. 33, 17429–17434. https://doi.org/10.1523/JNEUROSCI.1466-13.2013. brain. Am. J. Physiol. Gastrointest. Liver Physiol. 295, G503–G511. https://doi.org/ Bjoras, M., Gjesdal, O., Erickson, J.D., Torp, R., Levy, L.M., Ottersen, O.P., Degree, M., 10.1152/ajpgi.00076.2008. Storm-Mathisen, J., Seeberg, E., Danbolt, N.C., 1996. Cloning and expression of a Chan, K., Busque, S.M., Sailer, M., Stoeger, C., Broer, S., Daniel, H., Rubio-Aliaga, I., neuronal rat brain glutamate transporter. Brain Res. Mol. Brain Res. 36, 163–168. Wagner, C.A., 2016. Loss of function mutation of the Slc38a3 glutamine transporter https://doi.org/10.1016/0169-328x(95)00279-2. reveals its critical role for amino acid metabolism in the liver, brain, and kidney. Blei, A.T., Olafsson, S., Therrien, G., Butterworth, R.F., 1994. Ammonia-induced brain Pflügers Archiv 468, 213–227. https://doi.org/10.1007/s00424-015-1742-0. edema and intracranial hypertension in rats after portacaval anastomosis. Chandra, S.R., Christopher, R., Ramanujam, C.N., Harikrishna, G.V., 2019. Hepatology 19, 1437–1444. Hyperargininemia experiences over last 7 Years from a tertiary care center. Blin, M., Crusio, W.E., Hevor, T., Cloix, J.F., 2002. Chronic inhibition of glutamine J. Pediatr. Neurosci. 14, 2–6. https://doi.org/10.4103/jpn.JPN_1_19. synthetase is not associated with impairment of learning and memory in mice. Brain Chatauret, N., Desjardins, P., Zwingmann, C., Rose, C., Rao, K.V., Butterworth, R.F., Res. Bull. 57, 11–15. https://doi.org/10.1016/s0361-9230(01)00631-1. 2006. Direct molecular and spectroscopic evidence for increased ammonia removal Boissonnet, A., Hevor, T., Cloix, J.-F., 2012. Phenotypic differences between fast and capacity of skeletal muscle in acute liver failure. J. Hepatol. 44, 1083–1088. https:// slow methionine sulfoximine-inbred mice: seizures, anxiety, and glutamine doi.org/10.1016/j.jhep.2005.11.048. synthetase. Epilepsy Res. 98, 25–34. https://doi.org/10.1016/j. Chen, W., Mahadomrongkul, V., Berger, U.V., Bassan, M., DeSilva, T., Tanaka, K., eplepsyres.2011.08.012. Irwin, N., Aoki, C., Rosenberg, P.A., 2004. The glutamate transporter GLT1a is Bosman, D.K., Deutz, N.E., De Graaf, A.A., vd Hulst, R.W., Van Eijk, H.M., Bovee, W.M., expressed in excitatory axon terminals of mature hippocampal neurons. J. Neurosci. Maas, M.A., Jorning, G.G., Chamuleau, R.A., 1990. Changes in brain metabolism 24, 1136–1148. https://doi.org/10.1523/JNEUROSCI.1586-03.2004. during hyperammonemia and acute liver failure: results of a comparative 1H-NMR Cheng, X., Kim, S.Y., Okamoto, H., Xin, Y., Yancopoulos, G.D., Murphy, A.J., spectroscopy and biochemical investigation. Hepatology 12, 281–290. https://doi. Gromada, J., 2018. Glucagon contributes to liver zonation. Proc. Natl. Acad. Sci. U. org/10.1002/hep.1840120215. S. A. 115, E4111–E4119. https://doi.org/10.1073/pnas.1721403115. Bosoi, C.R., Rose, C.F., 2009. Identifying the direct effects of ammonia on the brain. Chepkova, A.N., Sergeeva, O.A., Gorg, B., Haas, H.L., Klocker, N., Haussinger, D., 2017. Metab. Brain Dis. 24, 95–102. https://doi.org/10.1007/s11011-008-9112-7. Impaired novelty acquisition and synaptic plasticity in congenital hyperammonemia Botini, F.F., Suzuki-Kemmelmeier, F., Nascimento, E.A., Ide, L.T., Bracht, A., 2005. caused by hepatic glutamine synthetase deficiency. Sci. Rep. 7, 40190. https://doi. Zonation of alanine metabolism in the bivascularly perfused rat liver. https://doi. org/10.1038/srep40190. org/10.1111/j.1478-3231.2005.01093.x, 25, 861-871. Chiang, C.-Y., Wang, J., Xie, Y.-F., Zhang, S., Hu, J.W., Dostrovsky, J.O., Sessle, B.J., Bott, A.J., Shen, J., Tonelli, C., Zhan, L., Sivaram, N., Jiang, Y.-P., Yu, X., Bhatt, V., 2007. Astroglial glutamate-glutamine shuttle is involved in central sensitization of Chiles, E., Zhong, H., et al., 2019. Glutamine anabolism plays a critical role in nociceptive neurons in rat medullary dorsal horn. J. Neurosci. 27, 9068–9076. pancreatic cancer by coupling carbon and nitrogen metabolism. Cell Rep. 29, https://doi.org/10.1523/JNEUROSCI.2260-07.2007. 1287–1298. https://doi.org/10.1016/j.celrep.2019.09.056 e6. Clemmesen, J.O., Kondrup, J., Ott, P., 2000. Splanchnic and leg exchange of amino acids Braeuning, A., Ittrich, C., Kohle, C., Hailfinger,S., Bonin, M., Buchmann, A., Schwarz, M., and ammonia in acute liver failure. Gastroenterology 118, 1131–1139. https://doi. 2006. Differential gene expression in periportal and perivenous mouse hepatocytes. org/10.1016/s0016-5085(00)70366-0. FEBS J. 273, 5051–5061. https://doi.org/10.1111/j.1742-4658.2006.05503.x. Clemmesen, J.O., Larsen, F.S., Kondrup, J., Hansen, B.A., Ott, P., 1999. Cerebral Braissant, O., McLin, V.A., Cudalbu, C., 2013. Ammonia toxicity to the brain. J. Inherit. herniation in patients with acute liver failure is correlated with arterial ammonia Metab. Dis. 36, 595–612. https://doi.org/10.1007/s10545-012-9546-2. concentration. Hepatology 29, 648–653. https://doi.org/10.1002/hep.510290309. Bradford, H.F., Ward, H.K., Thomas, A.J., 1978. Glutamine - a major substrate for nerve Cloix, J.-F., Tahi, Z., Martin, B., Hevor, T., 2010. Selection of two lines of mice based on endings. J Neurochem 30, 1453–1459. https://doi.org/10.1111/j.1471-4159.1978. latency to onset of methionine sulfoximine seizures. Epilepsia 51, 118–128. https:// tb10477.x. doi.org/10.1111/j.1528-1167.2009.02200.x. Brosnan, J.T., Brosnan, M.E., Nissim, I., 2004. A window into cellular metabolism: Cohen, P.P., 1981. The ornithine-urea cycle: biosynthesis and regulation of carbamyl hepatic metabolism of (15)N-labelled substrates. Metab. Eng. 6, 6–11. https://doi. phosphate synthetase I and ornithine transcarbamylase. Curr. Top. Cell. Regul. 18, org/10.1016/j.ymben.2003.10.006. 1–19. https://doi.org/10.1016/b978-0-12-152818-8.50008-6. Brusilow, S.W., 1984. Arginine, an indispensable amino acid for patients with inborn Cohen, P.P., Hayano, M., 1948. The enzymatic steps in urea synthesis. J. Biol. Chem. 172, errors of urea synthesis. J. Clin. Invest. 74, 2144–2148. https://doi.org/10.1172/ 405–415. JCI111640. Coltart, I., Tranah, T.H., Shawcross, D.L., 2013. Inflammation and hepatic Burch, H.B., Choi, S., McCarthy, W.Z., Wong, P.Y., Lowry, O.H., 1978. The location of encephalopathy. Arch. Biochem. Biophys. 536, 189–196. https://doi.org/10.1016/j. glutamine synthetase within the rat and rabbit nephron. Biochem. Biophys. Res. abb.2013.03.016. Commun. 82, 498–505. https://doi.org/10.1016/0006-291x(78)90902-6. Conti, F., Melone, M., 2006. The glutamine commute: lost in the tube? Neurochem. Int. Burke, Z.D., Reed, K.R., Phesse, T.J., Sansom, O.J., Clarke, A.R., Tosh, D., 2009. Liver 48, 459–464. https://doi.org/10.1016/j.neuint.2005.11.016. zonation occurs through a beta-catenin-dependent, c-Myc-independent mechanism. Cooper, A.J., McDonald, J.M., Gelbard, A.S., Gledhill, R.F., Duffy, T.E., 1979. The Gastroenterology 136, 2316–2324. https://doi.org/10.1053/j.gastro.2009.02.063 metabolic fate of 13N-labeled ammonia in rat brain. J. Biol. Chem. 254, 4982–4992. e1-3. Cooper, A.J., Stephani, R.A., Meister, A., 1976. Enzymatic reactions of methionine Butterworth, R.F., Giguere, J.F., Michaud, J., Lavoie, J., Layrargues, G.P., 1987. sulfoximine. Conversion to the corresponding alpha-imino and alpha-keto acids and Ammonia: key factor in the pathogenesis of hepatic encephalopathy. Neurochem. to alpha-ketobutyrate and methane sulfinimide. J. Biol. Chem. 251, 6674–6682. Pathol. 6, 1–12. https://doi.org/10.1007/bf02833598. Corriden, R., Hollands, A., Olson, J., Derieux, J., Lopez, J., Chang, J.T., Gonzalez, D.J., Cahoy, J.D., Emery, B., Kaushal, A., Foo, L.C., Zamanian, J.L., Christopherson, K.S., Nizet, V., 2015. Tamoxifen augments the innate immune function of neutrophils Xing, Y., Lubischer, J.L., Krieg, P.A., Krupenko, S.A., Thompson, W.J., Barres, B.A., through modulation of intracellular ceramide. Nat. Commun. 6, 8369. https://doi. 2008. A transcriptome for astrocytes, neurons, and oligodendrocytes: a new org/10.1038/ncomms9369. resource for understanding brain development and function. J. Neurosci. 28, Crombez, E.A., Cederbaum, S.D., 2005. Hyperargininemia due to liver arginase 264–278. https://doi.org/10.1523/JNEUROSCI.4178-07.2008. deficiency. Mol. Genet. Metabol. 84, 243–251. https://doi.org/10.1016/j. Caldovic, L., Morizono, H., Daikhin, Y., Nissim, I., McCarter, R.J., Yudkoff, M., ymgme.2004.11.004. Tuchman, M., 2004. Restoration of ureagenesis in N-acetylglutamate synthase Cruz, N.F., Dienel, G.A., Patrick, P.A., Cooper, A.J., 2017. Organ distribution of (13)N deficiency by N-carbamylglutamate. J. Pediatr. 145, 552–554. https://doi.org/ following intravenous injection of [(13)N]ammonia into portacaval-shunted rats. 10.1016/j.jpeds.2004.06.047. Neurochem. Res. 42, 1683–1696. https://doi.org/10.1007/s11064-016-2096-5. Camacho, J.A., Obie, C., Biery, B., Goodman, B.K., Hu, C.A., Almashanu, S., Steel, G., D’Amelio, F., Eng, L.F., Gibbs, M.A., 1990. Glutamine synthetase immunoreactivity is Casey, R., Lambert, M., Mitchell, G.A., Valle, D., 1999. Hyperornithinaemia- present in oligodendroglia of various regions of the central nervous system. Glia 3, hyperammonaemia-homocitrullinuria syndrome is caused by mutations in a gene 335–341. https://doi.org/10.1002/glia.440030504. encoding a mitochondrial ornithine transporter. Nat. Genet. 22, 151–158. https:// Dahlberg, D., Ivanovic, J., Hassel, B., 2016. Toxic levels of ammonia in human brain doi.org/10.1038/9658. abscess. J. Neurosurg. 124, 854–860. https://doi.org/10.3171/2015.1.JNS142582. Cammer, W., 1990. Glutamine synthetase in the central nervous system is not confinedto Danbolt, N.C., 2001. Glutamate uptake. Prog. Neurobiol. 65, 1–105. https://doi.org/ astrocytes. J. Neuroimmunol. 26, 173–178. https://doi.org/10.1016/0165-5728(90) 10.1016/s0301-0082(00)00067-8. 90088-5.

12 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Danbolt, N.C., Furness, D.N., Zhou, Y., 2016a. Neuronal vs glial glutamate uptake: Elliott, W.H., 1948. Adenosinetriphosphate in glutamine synthesis. Nature 161, 128. resolving the conundrum. Neurochem. Int. 98, 29–45. https://doi.org/10.1016/j. Elliott, W.H., Gale, E.F., 1948. Glutamine-synthesizing system of Staphylococcus aureus; neuint.2016.05.009. its inhibition by crystal violet and methionine-sulphoxide. Nature 161, 129. https:// Danbolt, N.C., Lehre, K.P., Dehnes, Y., Chaudhry, F.A., Levy, L.M., 1998. Localization of doi.org/10.1038/161129a0. transporters using transporter-specific antibodies. Methods Enzymol. 296, 388–407. Enns, G.M., Berry, S.A., Berry, G.T., Rhead, W.J., Brusilow, S.W., Hamosh, A., 2007. https://doi.org/10.1016/s0076-6879(98)96028-1. Survival after treatment with phenylacetate and benzoate for urea-cycle disorders. Danbolt, N.C., Storm-Mathisen, J., Kanner, B.I., 1992. An [Na+ + K+]coupled L- N. Engl. J. Med. 356, 2282–2292. https://doi.org/10.1056/NEJMoa066596. glutamate transporter purified from rat brain is located in glial cell processes. Erez, A., Nagamani, S.C., Shchelochkov, O.A., Premkumar, M.H., Campeau, P.M., Neuroscience 51, 295–310. https://doi.org/10.1016/0306-4522(92)90316-t. Chen, Y., Garg, H.K., Li, L., Mian, A., Bertin, T.K., et al., 2011. Requirement of Danbolt, N.C., Zhou, Y., Furness, D.N., Holmseth, S., 2016b. Strategies for argininosuccinate lyase for systemic nitric oxide production. Nat. Med. 17, immunohistochemical protein localization using antibodies: what did we learn from 1619–1626. https://doi.org/10.1038/nm.2544. neurotransmitter transporters in glial cells and neurons. Glia 64, 2045–2064. Euler, T., Wassle, H., 1995. Immunocytochemical identification of cone bipolar cells in https://doi.org/10.1002/glia.23027. the rat retina. J. Comp. Neurol. 361, 461–478. https://doi.org/10.1002/ De Chiara, F., Thomsen, K.L., Habtesion, A., Jones, H., Davies, N., Gracia-Sancho, J., cne.903610310. Manicardi, N., Hall, A., Andreola, F., Paish, H.L., Reed, L.H., , A.A., Leslie, J., Fecarotta, S., Parenti, G., Vajro, P., Zuppaldi, A., Della Casa, R., Carbone, M.T., Oakley, F., Rombouts, K., Mookerjee, R.P., Mann, J., Jalan, R., 2020. Ammonia Correra, A., Torre, G., Riva, S., Dionisi-Vici, C., Santorelli, F.M., Andria, G., 2006. scavenging prevents progression of fibrosis in experimental nonalcoholic fatty liver HHH syndrome (hyperornithinaemia, hyperammonaemia, homocitrullinuria), with disease. Hepatology 71, 874–892. https://doi.org/10.1002/hep.30890. fulminant hepatitis-like presentation. J. Inherit. Metab. Dis. 29, 186–189. https:// Dehnes, Y., Chaudhry, F.A., Ullensvang, K., Lehre, K.P., Storm-Mathisen, J., Danbolt, N. doi.org/10.1007/s10545-006-0120-7. C., 1998. The glutamate transporter EAAT4 in rat cerebellar Purkinje cells: a Felig, P., Pozefsky, T., Marliss, E., Cahill, G.F., 1970. Alanine: key role in glutamate-gated chloride channel concentrated near the synapse in parts of the gluconeogenesis. Science 167, 1003–1004. https://doi.org/10.1126/ dendritic membrane facing astroglia. J. Neurosci. 18, 3606–3619. https://doi.org/ science.167.3920.1003. 10.1523/JNEUROSCI.18-10-03606.1998. Forni, P.E., Scuoppo, C., Imayoshi, I., Taulli, R., Dastru, W., Sala, V., Betz, U.A., Deignan, J.L., Livesay, J.C., Yoo, P.K., Goodman, S.I., O’Brien, W.E., Iyer, R.K., Muzzi, P., Martinuzzi, D., Vercelli, A.E., Kageyama, R., Ponzetto, C., 2006. High Cederbaum, S.D., Grody, W.W., 2006. Ornithine deficiency in the arginase double levels of Cre expression in neuronal progenitors cause defects in brain development knockout mouse. Mol. Genet. Metabol. 89, 87–96. https://doi.org/10.1016/j. leading to microencephaly and hydrocephaly. J. Neurosci. 26, 9593–9602. https:// ymgme.2006.04.007. doi.org/10.1523/JNEUROSCI.2815-06.2006. Dejong, C.H., Deutz, N.E., Soeters, P.B., 1993. Metabolic adaptation of the kidney to Furness, D.N., Dehnes, Y., Akhtar, A.Q., Rossi, D.J., Hamann, M., Grutle, N.J., hyperammonemia during chronic liver insufficiency in the rat. Hepatology 18, Gundersen, V., Holmseth, S., Lehre, K.P., Ullensvang, K., Wojewodzic, M., Zhou, Y., 890–902. https://doi.org/10.1002/hep.1840180422. Attwell, D., Danbolt, N.C., 2008. A quantitative assessment of glutamate uptake into DeMars, R., LeVan, S.L., Trend, B.L., Russell, L.B., 1976. Abnormal ornithine hippocampal synaptic terminals and astrocytes: new insights into a neuronal role for carbamoyltransferase in mice having the sparse-Fur mutation. Proc. Natl. Acad. Sci. excitatory amino acid transporter 2 (EAAT2). Neuroscience 157, 80–94. https://doi. U. S. A. 73, 1693–1697. https://doi.org/10.1073/pnas.73.5.1693. org/10.1016/j.neuroscience.2008.08.043. Desjardins, P., Du, T., Jiang, W., Peng, L., Butterworth, R.F., 2012. Pathogenesis of Gaasbeek Janzen, J.W., Gebhardt, R., ten Voorde, G.H., Lamers, W.H., Charles, R., hepatic encephalopathy and brain edema in acute liver failure: role of glutamine Moorman, A.F., 1987. Heterogeneous distribution of glutamine synthetase during rat redefined. Neurochem. Int. 60, 690–696. https://doi.org/10.1016/j. liver development. J. Histochem. Cytochem. 35, 49–54. https://doi.org/10.1177/ neuint.2012.02.001. 35.1.2878950. Desjardins, P., Rao, K.V., Michalak, A., Rose, C., Butterworth, R.F., 1999. Effect of Gaasbeek Janzen, J.W., Lamers, W.H., Moorman, A.F., de Graaf, A., Los, J.A., Charles, R., portacaval anastomosis on glutamine synthetase protein and gene expression in 1984. Immunohistochemical localization of carbamoyl-phosphate synthetase brain, liver and skeletal muscle. Metab. Brain Dis. 14, 273–280. https://doi.org/ (ammonia) in adult rat liver; evidence for a heterogeneous distribution. 10.1023/a:1020741226752. J. Histochem. Cytochem. 32, 557–564. https://doi.org/10.1177/32.6.6373912. Dhaher, R., Wang, H., Gruenbaum, S.E., Tu, N., Lee, T.-S.W., Zaveri, H.P., Eid, T., 2015. Garlanda, C., Dejana, E., 1997. Heterogeneity of endothelial cells. Specific markers. Effects of site-specific infusions of methionine sulfoximine on the temporal Arterioscler. Thromb. Vasc. Biol. 17, 1193–1202. https://doi.org/10.1161/01. progression of seizures in a rat model of mesial temporal lobe epilepsy. Epilepsy Res. atv.17.7.1193. 115, 45–54. https://doi.org/10.1016/j.eplepsyres.2015.05.005. Gascon-Barre, M., Benbrahim, N., Tremblay, C., 1989. Hepatic zonation of drug Diemer, N.H., Laursen, H., 1977. Glial cell reactions in rats with hyperammoniemia metabolizing enzymes. Studies on hepatocytes isolated from the periportal or induced by urease or porto-caval anastomosis. Acta Neurol. Scand. 55, 425–442. perivenous region of the liver acinus. Can. J. Physiol. Pharmacol. 67, 1015–1022. https://doi.org/10.1111/j.1600-0404.1977.tb07623.x. https://doi.org/10.1139/y89-160. Dolgodilina, E., Camargo, S.M., Roth, E., Herzog, B., Nunes, V., Palacin, M., Verrey, F., Gebhardt, R., Burger, H.J., Heini, H., Schreiber, K.L., Mecke, D., 1988. Alterations of 2020. Choroid plexus LAT2 and SNAT3 as partners in CSF amino acid homeostasis hepatic enzyme levels and of the acinar distribution of glutamine synthetase in maintenance. Fluids Barriers CNS 17, 17. https://doi.org/10.1186/s12987-020- response to experimental liver injury in the rat. Hepatology 8, 822–830. https://doi. 0178-x. org/10.1002/hep.1840080421. Dolgodilina, E., Imobersteg, S., Laczko, E., Welt, T., Verrey, F., Makrides, V., 2016. Brain Gebhardt, R., Mecke, D., 1983. Heterogeneous distribution of glutamine synthetase interstitial fluidglutamine homeostasis is controlled by blood-brain barrier SLC7A5/ among rat liver parenchymal cells in situ and in primary culture. EMBO J. 2, LAT1 amino acid transporter. J. Cerebr. Blood Flow Metabol. 36, 1929–1941. 567–570. https://doi.org/10.1177/0271678X15609331. Gelehrter, T.D., Snodgrass, P.J., 1974. Lethal neonatal deficiencyof carbamyl phosphate Donn, S.M., Thoene, J.G., 1985. Prospective prevention of neonatal hyperammonaemia synthetase. N. Engl. J. Med. 290, 430–433. https://doi.org/10.1056/ in argininosuccinic acidura by arginine therapy. J. Inherit. Metab. Dis. 8, 18–20. NEJM197402212900804. https://doi.org/10.1007/BF01805478. Ginsberg, S.D., Martin, L.J., Rothstein, J.D., 1995. Regional deafferentation down- Duda, G.D., Handler, P., 1958. Kinetics of ammonia metabolism in vivo. J. Biol. Chem. regulates subtypes of glutamate transporter proteins. J. Neurochem. 65, 2800–2803. 232, 303–314. https://doi.org/10.1046/j.1471-4159.1995.65062800.x. Duelli, R., Enerson, B.E., Gerhart, D.Z., Drewes, L.R., 2000. Expression of large amino Girard, G., Giguere, J.F., Butterworth, R.F., 1993. Region-selective reductions in acid transporter LAT1 in rat brain endothelium. J. Cerebr. Blood Flow Metabol. 20, activities of glutamine synthetase in rat brain following portacaval anastomosis. 1557–1562. https://doi.org/10.1097/00004647-200011000-00005. Metab. Brain Dis. 8, 207–215. https://doi.org/10.1007/bf01001062. Dyste, G.N., Menezes, A.H., VanGilder, J.C., 1989. Symptomatic Chiari malformations. Giusti, S.A., Vercelli, C.A., Vogl, A.M., Kolarz, A.W., Pino, N.S., Deussing, J.M., An analysis of presentation, management, and long-term outcome. J. Neurosurg. 71, Refojo, D., 2014. Behavioral phenotyping of Nestin-Cre mice: implications for 159–168. https://doi.org/10.3171/jns.1989.71.2.0159. genetic mouse models of psychiatric disorders. J. Psychiatr. Res. 55, 87–95. https:// Eelen, G., Dubois, C., Cantelmo, A.R., Goveia, J., Bruning, U., DeRan, M., Jarugumilli, G., doi.org/10.1016/j.jpsychires.2014.04.002. van Rijssel, J., Saladino, G., Comitani, F., et al., 2018. Role of glutamine synthetase Gordon, S., Pluddemann, A., 2017. Tissue macrophages: heterogeneity and functions. in angiogenesis beyond glutamine synthesis. Nature 561, 63–69. https://doi.org/ BMC Biol. 15, 53. https://doi.org/10.1186/s12915-017-0392-4. 10.1038/s41586-018-0466-7. Gorski, J.A., Talley, T., Qiu, M., Puelles, L., Rubenstein, J.L., Jones, K.R., 2002. Cortical Eid, T., Ghosh, A., Wang, Y., Beckstrom, H., Zaveri, H.P., Lee, T.-S.W., Lai, J.C., excitatory neurons and glia, but not GABAergic neurons, are produced in the Emx1- Malthankar-Phatak, G.H., de Lanerolle, N.C., 2008. Recurrent seizures and brain expressing lineage. J. Neurosci. 22, 6309–6314. https://doi.org/10.1523/ pathology after inhibition of glutamine synthetase in the hippocampus in rats. Brain JNEUROSCI.22-15-06309.2002. 131, 2061–2070. https://doi.org/10.1093/brain/awn133. Graham, T.E., MacLean, D.A., 1998. Ammonia and amino acid metabolism in skeletal Eid, T., Thomas, M.J., Spencer, D.D., Runden-Pran, E., Lai, J.C., Malthankar, G.V., Kim, J. muscle: human, rodent and canine models. Med. Sci. Sports Exerc. 30, 34–46. H., Danbolt, N.C., Ottersen, O.P., de Lanerolle, N.C., 2004. Loss of glutamine https://doi.org/10.1097/00005768-199801000-00006. synthetase in the human epileptogenic hippocampus: possible mechanism for raised Grewal, S., Defamie, N., Zhang, X., De Gois, S., Shawki, A., Mackenzie, B., Chen, C., extracellular glutamate in mesial temporal lobe epilepsy. Lancet 363, 28–37. https:// Varoqui, H., Erickson, J.D., 2009. SNAT2 amino acid transporter is regulated by doi.org/10.1016/s0140-6736(03)15166-5. amino acids of the SLC6 gamma-aminobutyric acid transporter subfamily in

13 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

neocortical neurons and may play no role in delivering glutamine for glutamatergic Haussinger, D., Gorg, B., Reinehr, R., Schliess, F., 2005. Protein tyrosine nitration in transmission. J. Biol. Chem. 284, 11224–11236. https://doi.org/10.1074/jbc. hyperammonemia and hepatic encephalopathy. Metab. Brain Dis. 20, 285–294. M806470200. https://doi.org/10.1007/s11011-005-7908-2. Grewer, C., Rauen, T., 2005. Electrogenic glutamate transporters in the CNS: molecular Haussinger, D., Laubenberger, J., vom Dahl, S., Ernst, T., Bayer, S., Langer, M., mechanism, pre-steady-state kinetics, and their impact on synaptic signaling. Gerok, W., Hennig, J., 1994. Proton magnetic resonance spectroscopy studies on J. Membr. Biol. 203, 1–20. https://doi.org/10.1007/s00232-004-0731-6. human brain myo-inositol in hypo-osmolarity and hepatic encephalopathy. Griffith, O.W., Meister, A., 1978. Differential inhibition of glutamine and gamma- Gastroenterology 107, 1475–1480. https://doi.org/10.1016/0016-5085(94)90552- glutamylcysteine synthetases by alpha-alkyl analogs of methionine sulfoximine that 5. induce convulsions. J. Biol. Chem. 253, 2333–2338. Haussinger,¨ D., Lamers, W.H., Moorman, A.F., 1992. Hepatocyte heterogeneity in the Grisolia, S., Cohen, P.P., 1952. The catalytic role of carbamyl glutamate in citrulline metabolism of amino acids and ammonia. Enzyme 46 (1–3), 72–93. https://doi.org/ biosynthesis. J. Biol. Chem. 198, 561–571. 10.1159/000468779. Grisolia, S., Cohen, P.P., 1953. Catalytic role of of glutamate derivatives in citrulline He, Y., Hakvoort, T.B., Kohler, S.E., Vermeulen, J.L., de Waart, D.R., de Theije, C., ten biosynthesis. J. Biol. Chem. 204, 753–757. Have, G.A., van Eijk, H.M., Kunne, C., Labruyere, W.T., Houten, S.M., Sokolovic, M., Gruenbaum, S.E., Wang, H., Zaveri, H.P., Tang, A.B., Lee, T.-S.W., Eid, T., Dhaher, R., Ruijter, J.M., Deutz, N.E., Lamers, W.H., 2010b. Glutamine synthetase in muscle is 2015. Inhibition of glutamine synthetase in the central nucleus of the amygdala required for glutamine production during fasting and extrahepatic ammonia induces anhedonic behavior and recurrent seizures in a rat model of mesial temporal detoxification. J. Biol. Chem. 285, 9516–9524. https://doi.org/10.1074/jbc. lobe epilepsy. Epilepsy Behav. 51, 96–103. https://doi.org/10.1016/j. M109.092429. yebeh.2015.07.015. He, Y., Hakvoort, T.B., Vermeulen, J.L., Labruyere, W.T., De Waart, D.R., Van Der Gumucio, J.J., Miller, D.L., 1981. Functional implications of liver cell heterogeneity. Hel, W.S., Ruijter, J.M., Uylings, H.B., Lamers, W.H., 2010a. Glutamine synthetase Gastroenterology 80, 393–403. deficiencyin murine astrocytes results in neonatal death. Glia 58, 741–754. https:// Gundersen, V., Danbolt, N.C., Ottersen, O.P., Storm-Mathisen, J., 1993. Demonstration of doi.org/10.1002/glia.20960. glutamate/aspartate uptake activity in nerve endings by use of antibodies He, Y., Hakvoort, T.B., Vermeulen, J.L., Lamers, W.H., Van Roon, M.A., 2007. Glutamine recognizing exogenous D-aspartate. Neuroscience 57, 97–111. https://doi.org/ synthetase is essential in early mouse embryogenesis. Dev. Dynam. 236, 1865–1875. 10.1016/0306-4522(93)90114-u. https://doi.org/10.1002/dvdy.21185. Haberle, J., 2013. Clinical and biochemical aspects of primary and secondary Heibel, S.K., Ah Mew, N., Caldovic, L., Daikhin, Y., Yudkoff, M., Tuchman, M., 2011. N- hyperammonemic disorders. Arch. Biochem. Biophys. 536, 101–108. https://doi. carbamylglutamate enhancement of ureagenesis leads to discovery of a novel org/10.1016/j.abb.2013.04.009. deleterious mutation in a newly definedenhancer of the NAGS gene and to effective Haberle, J., Boddaert, N., Burlina, A., Chakrapani, A., Dixon, M., Huemer, M., Karall, D., therapy. Hum. Mutat. 32, 1153–1160. https://doi.org/10.1002/humu.21553. Martinelli, D., Crespo, P.S., Santer, R., Servais, A., Valayannopoulos, V., Lindner, M., Hertz, L., Chen, Y., 2018. Glycogenolysis, an astrocyte-specific reaction, is essential for Rubio, V., Dionisi-Vici, C., 2012a. Suggested guidelines for the diagnosis and both astrocytic and neuronal activities involved in learning. Neuroscience 370, management of urea cycle disorders. Orphanet J. Rare Dis. 7, 32. https://doi.org/ 27–36. https://doi.org/10.1016/j.neuroscience.2017.06.025. 10.1186/1750-1172-7-32. Hertz, L., Rothman, D.L., 2016. Glucose, lactate, beta-hydroxybutyrate, acetate, GABA, Haberle, J., Gorg, B., Rutsch, F., Schmidt, E., Toutain, A., Benoist, J.-F., Gelot, A., Suc, A.- and succinate as substrates for synthesis of glutamate and GABA in the glutamine- L., Hohne, W., Schliess, F., Haussinger, D., Koch, H.G., 2005. Congenital glutamine glutamate/GABA cycle. In: Schousboe, A., Sonnewald, U. (Eds.), The Glutamate/ deficiency with glutamine synthetase mutations. N. Engl. J. Med. 353, 1926–1933. GABA-Glutamine Cycle - Amino Acid Neurotransmitter Homeostasis. https://doi.org/10.1056/NEJMoa050456. Hertz, L., Zielke, H.R., 2004. Astrocytic control of glutamatergic activity: astrocytes as Haberle, J., Gorg, B., Toutain, A., Rutsch, F., Benoist, J.-F., Gelot, A., Suc, A.-L., Koch, H. stars of the show. Trends Neurosci. 27, 735–743. https://doi.org/10.1016/j. G., Schliess, F., Haussinger, D., 2006. Inborn error of amino acid synthesis: human tins.2004.10.008. glutamine synthetase deficiency. J. Inherit. Metab. Dis. 29, 352–358. https://doi. Hellsten, S.V., Hagglund, M.G., Eriksson, M.M., Fredriksson, R., 2017. The neuronal and org/10.1007/s10545-006-0256-5. astrocytic protein SLC38A10 transports glutamine, glutamate, and aspartate, Haberle, J., Shahbeck, N., Ibrahim, K., Schmitt, B., Scheer, I., O’Gorman, R., suggesting a role in neurotransmission. FEBS Open Bio 7, 730–746. https://doi.org/ Chaudhry, F.A., Ben-Omran, T., 2012b. Glutamine supplementation in a child with 10.1002/2211-5463.12219. inherited GS deficiency improves the clinical status and partially corrects the Hod, G., Chaouat, M., Haskel, Y., Lernau, O.Z., Nissan, S., Mayer, M., 1982. Ammonia peripheral and central amino acid imbalance. Orphanet J. Rare Dis. 7, 48. https:// uptake by skeletal muscle in the hyperammonaemic rat. Eur. J. Clin. Invest. 12, doi.org/10.1186/1750-1172-7-48. 445–450. https://doi.org/10.1111/j.1365-2362.1982.tb02222.x. Hagglund, M.G., Hellsten, S.V., Bagchi, S., Philippot, G., Lofqvist, E., Nilsson, V.C., Holmes, F.L., 1980. Hans Krebs and the discovery of the ornithine cycle. Fed. Proc. 39, Almkvist, I., Karlsson, E., Sreedharan, S., Tafreshiha, A., Fredriksson, R., 2015. 216–225. Transport of L-glutamine, L-alanine, L-arginine and L-histidine by the neuron- Holmseth, S., Dehnes, Y., Huang, Y.H., Follin-Arbelet, V.V., Grutle, N.J., Mylonakou, M. specific Slc38a8 (SNAT8) in CNS. J. Mol. Biol. 427, 1495–1512. https://doi.org/ N., Plachez, C., Zhou, Y., Furness, D.N., Bergles, D.E., Lehre, K.P., Danbolt, N.C., 10.1016/j.jmb.2014.10.016. 2012a. The density of EAAC1 (EAAT3) glutamate transporters expressed by neurons Hagglund, M.G., Sreedharan, S., Nilsson, V.C., Shaik, J.H., Almkvist, I.M., Backlin, S., in the mammalian CNS. J. Neurosci. 32, 6000–6013. https://doi.org/10.1523/ Wrange, O., Fredriksson, R., 2011. Identification of SLC38A7 (SNAT7) protein as a JNEUROSCI.5347-11.2012. glutamine transporter expressed in neurons. J. Biol. Chem. 286, 20500–20511. Holmseth, S., Lehre, K.P., Danbolt, N.C., 2006. Specificity controls for https://doi.org/10.1074/jbc.M110.162404. immunocytochemistry. Anat. Embryol. 211, 257–266. https://doi.org/10.1007/ Hahn, M., Massen, O., Nenki, M., Pavlov, J., 1893. Die Ecksche Fistel zwischen der s00429-005-0077-6. unteren Hohlvene und der Pfortader und ihre Folger für den Organismus. Ach. Exp. Holmseth, S., Zhou, Y., Follin-Arbelet, V.V., Lehre, K.P., Bergles, D.E., Danbolt, N.C., Pathol. Pharmakol. 32, 161–270. https://doi.org/10.1007/BF01995065. 2012b. Specificity controls for immunocytochemistry: the antigen pre-adsorption Hakvoort, T.B., He, Y., Kulik, W., Vermeulen, J.L., Duijst, S., Ruijter, J.M., Runge, J.H., test can lead to inaccurate assessment of antibody specificity. J. Histochem. Deutz, N.E., Koehler, S.E., Lamers, W.H., 2017. Pivotal role of glutamine synthetase Cytochem. 60, 174–187. https://doi.org/10.1369/0022155411434828. in ammonia detoxification. Hepatology 65, 281–293. https://doi.org/10.1002/ Hoop, B., Systrom, D.M., Shih, V.E., Kazemi, H., 1988. Central respiratory effects of hep.28852. glutamine synthesis inhibition in dogs. J. Appl. Physiol. 65, 1099–1109. https://doi. Hakvoort, T.B., Lamers, W.H., 2019. Reply. Hepatology 69, 922–923. https://doi.org/ org/10.1152/jappl.1988.65.3.1099. 10.1002/hep.29728. Hu, Q.-X., Eid, T., Danbolt, N.C., Zhou, Y., 2017. Phenotypic differences between brain- Hall, L.M., Metzenberg, R.L., Cohen, P.P., 1958. Isolation and characterization of a and liver-specific glutamine synthetase knockout mice. J. Neurochem. 142, 220. naturally occurring cofactor of carbamyl phosphate biosynthesis. J. Biol. Chem. 230, Hu, Q.X., Ottestad-Hansen, S., Holmseth, S., Hassel, B., Danbolt, N.C., Zhou, Y., 2018. 1013–1021. Expression of glutamate transporters in mouse liver, kidney, and intestine. Hamberger, A.C., Chiang, G.H., Nylen, E.S., Scheff, S.W., Cotman, C.W., 1979. Glutamate J. Histochem. Cytochem. 66, 189–202. https://doi.org/10.1369/ as a CNS transmitter. I. Evaluation of glucose and glutamine as precursors for the 0022155417749828. synthesis of preferentially released glutamate. Brain Res 168, 513–530. https://doi. Iqbal, K., Ottaway, J.H., 1970. Glutamine synthetase in muscle and kidney. Biochem. J. org/10.1016/0006-8993(79)90306-8. 119, 145–156. https://doi.org/10.1042/bj1190145. Hanson, E., Armbruster, M., Cantu, D., Andresen, L., Taylor, A., Danbolt, N.C., Dulla, C. Iyer, R.K., Yoo, P.K., Kern, R.M., Rozengurt, N., Tsoa, R., O’Brien, W.E., Yu, H., G., 2015. Astrocytic glutamate uptake is slow and does not limit neuronal NMDA Grody, W.W., Cederbaum, S.D., 2002. Mouse model for human arginase deficiency. receptor activation in the neonatal neocortex. Glia 63, 1784–1796. https://doi.org/ Mol. Cell Biol. 22, 4491–4498. https://doi.org/10.1128/mcb.22.13.4491- 10.1002/glia.22844. 4498.2002. Hassel, B., Bråthe, A., 2000. Neuronal pyruvate carboxylation supports formation of Jahoor, F., Jackson, A.A., Golden, M.H., 1988. In vivo metabolism of nitrogen precursors transmitter glutamate. J. Neurosci. 20, 1342–1347. https://doi.org/10.1523/ for urea synthesis in the postprandial rat. Ann. Nutr. Metab. 32, 240–244. https:// JNEUROSCI.20-04-01342.2000. doi.org/10.1159/000177447. Haugeto, Ø., Ullensvang, K., Levy, L.M., Chaudhry, F.A., Honore, T., Nielsen, M., Jalan, R., Kapoor, D., 2003. Enhanced renal ammonia excretion following volume Lehre, K.P., Danbolt, N.C., 1996. Brain glutamate transporter proteins form expansion in patients with well compensated cirrhosis of the liver. Gut 52, homomultimers. J. Biol. Chem. 271, 27715–27722. https://doi.org/10.1074/ 1041–1045. https://doi.org/10.1136/gut.52.7.1041. jbc.271.44.27715. Jayakumar, A.R., Rama Rao, K.V., Norenberg, M.D., 2015. Neuroinflammationin hepatic Haussinger, D., 1983. Hepatocyte heterogeneity in glutamine and ammonia metabolism encephalopathy: mechanistic aspects. J. Clin. Exp. Hepatol. 5, S21–S28. https://doi. and the role of an intercellular glutamine cycle during ureogenesis in perfused rat org/10.1016/j.jceh.2014.07.006. liver. Eur. J. Biochem. 133, 269–275. https://doi.org/10.1111/j.1432-1033.1983. Jungermann, K., Katz, N., 1989. Functional specialization of different hepatocyte tb07458.x. populations. Physiol. Rev. 69, 708–764. https://doi.org/10.1152/ physrev.1989.69.3.708.

14 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Kam, K., Nicoll, R., 2007. Excitatory synaptic transmission persists independently of the Le, Y., Sauer, B., 2000. Conditional gene knockout using cre recombinase. Methods Mol. glutamate-glutamine cycle. J. Neurosci. 27, 9192–9200. https://doi.org/10.1523/ Biol. 136, 477–485. https://doi.org/10.1385/1-59259-065-9:477. JNEUROSCI.1198-07.2007. Lee, H.-W., Osis, G., Handlogten, M.E., Lamers, W.H., Chaudhry, F.A., Verlander, J.W., Kamin, H., Handler, P., 1957. Amino acid and protein metabolism. Annu. Rev. Biochem. Weiner, I.D., 2016. Proximal tubule-specific glutamine synthetase deletion alters 26, 419–490. https://doi.org/10.1146/annurev.bi.26.070157.002223. basal and acidosis-stimulated ammonia metabolism. Am. J. Physiol. Ren. Physiol. Kanai, Y., Hediger, M.A., 1992. Primary structure and functional characterization of a 310, F1229–F1242. https://doi.org/10.1152/ajprenal.00547.2015. high-affinityglutamate transporter. Nature 360, 467–471. https://doi.org/10.1038/ Lee, H.-W., Osis, G., Handlogten, M.E., Verlander, J.W., Weiner, I.D., 2017. Proximal 360467a0. tubule glutamine synthetase expression is necessary for the normal response to Kanamori, K., Ross, B.D., Kondrat, R.W., 2002. Glial uptake of neurotransmitter dietary protein restriction. Am. J. Physiol. Ren. Physiol. 313, F116–F125. https:// glutamate from the extracellular fluid studied in vivo by microdialysis and (13)C doi.org/10.1152/ajprenal.00048.2017. NMR. J. Neurochem. 83, 682–695. https://doi.org/10.1046/j.1471- Lehre, K.P., Davanger, S., Danbolt, N.C., 1997. Localization of the glutamate transporter 4159.2002.01161.x. protein GLAST in rat retina. Brain Res. 744, 129–137. https://doi.org/10.1016/ Kang, E.S., Snodgrass, P.J., Gerald, P.S., 1973. Ornithine transcarbamylase deficiency in s0006-8993(96)01022-0. the newborn infant. J. Pediatr. 82, 642–649. https://doi.org/10.1016/s0022-3476 Lehre, K.P., Levy, L.M., Ottersen, O.P., Storm-Mathisen, J., Danbolt, N.C., 1995. (73)80590-6. Differential expression of two glial glutamate transporters in the rat brain: Kant, D., Tripathi, S.1., Qureshi, M.F., Tripathi, S.2., Pandey, S., Singh, G., Kumar, T., quantitative and immunocytochemical observations. J. Neurosci. 15, 1835–1853. Mir, F.A., Jha, S.K., 2014. The effect of glial glutamine synthetase inhibition on https://doi.org/10.1523/JNEUROSCI.15-03-01835.1995. recognition and temporal memories in the rat. Neurosci. Lett. 560, 98–102. https:// Lerner, S., Anderzhanova, E., Verbitsky, S., Eilam, R., Kuperman, Y., Tsoory, M., doi.org/10.1016/j.neulet.2013.12.033. Kuznetsov, Y., Brandis, A., Mehlman, T., Mazkereth, R., McCarter, R., Segal, M., Karaca, M., Martin-Levilain, J., Grimaldi, M., Li, L., Dizin, E., Emre, Y., Maechler, P., Nagamani, S.C., Chen, A., Erez, A., 2019. ASL metabolically regulates tyrosine 2018. Liver glutamate dehydrogenase controls whole-body energy partitioning hydroxylase in the nucleus locus coeruleus. Cell Rep. 29, 2144–2153. https://doi. through amino acid-derived gluconeogenesis and ammonia homeostasis. Diabetes org/10.1016/j.celrep.2019.10.043 e7. 67, 1949–1961. https://doi.org/10.2337/db17-1561. Levin, B., Oberholzer, V.G., Sinclair, L., 1969. Biochemical investigations of Kasten, J., Hu, C., Bhargava, R., Park, H., Tai, D., Byrne, J.A., Marescau, B., De Deyn, P. hyperammonaemia. Lancet 2, 170–174. https://doi.org/10.1016/s0140-6736(69) P., Schlichting, L., Grody, W.W., Cederbaum, S.D., Lipshutz, G.S., 2013. Lethal 91419-6. phenotype in conditional late-onset arginase 1 deficiency in the mouse. Mol. Genet. Levy, L.M., Lehre, K.P., Rolstad, B., Danbolt, N.C., 1993. A monoclonal antibody raised Metabol. 110, 222–230. https://doi.org/10.1016/j.ymgme.2013.06.020. against an [Na(+)+K+]coupled L-glutamate transporter purified from rat brain Kellendonk, C., Opherk, C., Anlag, K., Schutz, G., Tronche, F., 2000. Hepatocyte-specific confirms glial cell localization. FEBS Lett. 317, 79–84. https://doi.org/10.1016/ expression of Cre recombinase. Genesis 26, 151–153. https://doi.org/10.1002/(sici) 0014-5793(93)81495-l. 1526-968x(200002)26:2<151::aid-gene17>3.0.co;2-e. Lie-Venema, H., de Boer, P.A., Moorman, A.F., Lamers, W.H., 1997. Organ-specific Khoja, S., Nitzahn, M., Hermann, K., Truong, B., Borzone, R., Willis, B., Rudd, M., activity of the 5’ regulatory region of the glutamine synthetase gene in developing Palmer, D.J., Ng, P., Brunetti-Pierri, N., Lipshutz, G.S., 2018. Conditional disruption mice. Eur. J. Biochem. 248, 644–659. https://doi.org/10.1111/j.1432- of hepatic carbamoyl phosphate synthetase 1 in mice results in hyperammonemia 1033.1997.00644.x. without orotic aciduria and can be corrected by liver-directed gene therapy. Mol. Lockwood, A.H., McDonald, J.M., Reiman, R.E., Gelbard, A.S., Laughlin, J.S., Duffy, T.E., Genet. Metabol. 124, 243–253. https://doi.org/10.1016/j.ymgme.2018.04.001. Plum, F., 1979. The dynamics of ammonia metabolism in man. Effects of liver Khoja, S., Nitzahn, M., Truong, B., Lambert, J., Willis, B., Allegri, G., Rufenacht, V., disease and hyperammonemia. J. Clin. Invest. 63, 449–460. https://doi.org/ Haberle, J., Lipshutz, G.S., 2019. A constitutive knockout of murine carbamoyl 10.1172/JCI109322. phosphate synthetase 1 results in death with marked hyperglutaminemia and Lorincz, A., Nusser, Z., 2008. Specificity of immunoreactions: the importance of testing hyperammonemia. J. Inherit. Metab. Dis. 42, 1044–1053. https://doi.org/10.1002/ specificity in each method. J. Neurosci. 28, 9083–9086. https://doi.org/10.1523/ jimd.12048. JNEUROSCI.2494-08.2008. Kim, S.Z., Song, W.J., Nyhan, W.L., Ficicioglu, C., Mandell, R., Shih, V.E., 2012. Long- Mackenzie, B., Erickson, J.D., 2004. Sodium-coupled neutral amino acid (System N/A) term follow-up of four patients affected by HHH syndrome. Clin. Chim. Acta 413, transporters of the SLC38 gene family. Pflügers Archiv 447, 784–795. https://doi. 1151–1155. https://doi.org/10.1016/j.cca.2012.03.015. org/10.1007/s00424-003-1117-9. Kimber, I., Cumberbatch, M., Humphreys, M., Hopkins, S.J., 1990. Contact Maestri, N.E., Hauser, E.R., Bartholomew, D., Brusilow, S.W., 1991. Prospective hypersensitivity induces plasma interleukin 6. Int. Arch. Allergy Appl. Immunol. 92, treatment of urea cycle disorders. J. Pediatr. 119, 923–928. https://doi.org/ 97–99. https://doi.org/10.1159/000235232. 10.1016/s0022-3476(05)83044-6. Kobayashi, K., Sinasac, D.S., Iijima, M., Boright, A.P., Begum, L., Lee, J.R., Yasuda, T., Manning, J.M., Moore, S., Rowe, W.B., Meister, A., 1969. Identification of L-methionine Ikeda, S., Hirano, R., Terazono, H., Crackower, M.A., Kondo, I., Tsui, L.C., Scherer, S. S-sulfoximine as the diastereoisomer of L-methionine SR-sulfoximine that inhibits W., Saheki, T., 1999. The gene mutated in adult-onset type II citrullinaemia encodes glutamine synthetase. Biochemistry 8, 2681–2685. https://doi.org/10.1021/ a putative mitochondrial carrier protein. Nat. Genet. 22, 159–163. https://doi.org/ bi00834a066. 10.1038/9667. Marshall, M., Cohen, P.P., 1972. Ornithine transcarbamylase from Streptococcus faecalis Krebs, H.A., 1935. Metabolism of amino acids. IV. Synthesis of glutamine from glutamic and bovine liver. I. Isolation and subunit structure. J. Biol. Chem. 247, 1641–1653. acid and ammonia, and the enzymatic hydrolysis of glutamine in animal tissue. Martin, H., Voss, K., Hufnagl, P., Wack, R., Wassilew, G., 1987. Morphometric and Biochem. J. 29, 1951–1969. densitometric investigations of protoplasmic astrocytes and neurons in human Krebs, H.A., 1942. Urea formation in mammalian liver. Biochem. J. 36, 758–767. hepatic encephalopathy. Exp. Pathol. 32, 241–250. https://doi.org/10.1016/s0232- https://doi.org/10.1042/bj0360758. 1513(87)80035-x. Krebs, H.A., Eggleston, L.V., Knivett, V.A., 1955. Arsenolysis and phosphorolysis of Martin, L.J., Brambrink, A.M., Lehmann, C., Portera-Cailliau, C., Koehler, R., citrulline in mammalian liver. Biochem. J. 59, 185–193. https://doi.org/10.1042/ Rothstein, J., Traystman, R.J., 1997. Hypoxia-ischemia causes abnormalities in bj0590185. glutamate transporters and death of astroglia and neurons in newborn striatum. Ann. Krebs, H.A., Henseleit, K., 1932. Untersuchungen uber die Harnstoffbildung im Neurol. 42, 335–348. https://doi.org/10.1002/ana.410420310. Tierkorper.¨ Biol. Chem. 210 (1–2), 33. https://doi.org/10.1515/bchm2.1932.210.1- Martinez-Hernandez, A., Bell, K.P., Norenberg, M.D., 1977. Glutamine synthetase: glial 2.33. localization in brain. Science 195, 1356–1358. https://doi.org/10.1126/ Kuno, T., Miyazaki, S., Inoue, I., Saheki, T., 1990. Hyperammonemia caused by impaired science.14400. mitochondrial ornithine transport in a patient with partial quantitative deficiencyof Martinov, V., Dehnes, Y., Holmseth, S., Shimamoto, K., Danbolt, N.C., Valen, G., 2014. ornithine carbamoyltransferase. Clin. Biochem. 23, 143–147. https://doi.org/ A novel glutamate transporter blocker, LL-TBOA, attenuates ischaemic injury in the 10.1016/0009-9120(90)80027-g. isolated, perfused rat heart despite low transporter levels. Eur. J. Cardio. Thorac. Kuo, F.C., Hwu, W.L., Valle, D., Darnell, J.E., 1991. Colocalization in pericentral Surg. 45, 710–716. https://doi.org/10.1093/ejcts/ezt487. hepatocytes in adult mice and similarity in developmental expression pattern of Marx, M.-C., Billups, D., Billups, B., 2015. Maintaining the presynaptic glutamate supply ornithine aminotransferase and glutamine synthetase mRNA. Proc. Natl. Acad. Sci. for excitatory neurotransmission. J. Neurosci. Res. 93, 1031–1044. https://doi.org/ U. S. A. 88, 9468–9472. https://doi.org/10.1073/pnas.88.21.9468. 10.1002/jnr.23561. Kvamme, E., Torgner, I.A., Roberg, B., 2001. Kinetics and localization of brain phosphate Massie, A., Cnops, L., Smolders, I., McCullumsmith, R., Kooijman, R., Kwak, S., activated glutaminase. J. Neurosci. Res. 66, 951–958. https://doi.org/10.1002/ Arckens, L., Michotte, Y., 2008. High-affinity Na(+)/K(+)-dependent glutamate jnr.10041. transporter EAAT4 is expressed throughout the rat fore- and midbrain. J. Comp. Lamers, W.H., Janzen, J.W., Moorman, A.F., Charles, R., Knecht, E., Martinez-Ramon, A., Neurol. 511, 155–172. https://doi.org/10.1002/cne.21823. Hernandez-Yago, J., Grisolia, S., 1988. Immunohistochemical localization of Matsuzawa, T., Kobayashi, T., Ogawa, H., Kasahara, M., 1997. Microheterogeneity and glutamate dehydrogenase in rat liver: plasticity of distribution during development intrahepatic localization of human and rat liver cytosolic alanine aminotransferase. and with hormone treatment. J. Histochem. Cytochem. 36, 41–47. https://doi.org/ Biochim. Biophys. Acta 1340, 115–122. https://doi.org/10.1016/s0167-4838(97) 10.1177/36.1.3335769. 00033-2. Larangeira, A.S., Tanita, M.T., Dias, M.A., Filho, O.F., Delfino, V.D., Cardoso, L.T., McKenna, M.C., 2007. The glutamate-glutamine cycle is not stoichiometric: fates of Grion, C.M., 2018. Analysis of cerebral blood flow and intracranial hypertension in glutamate in brain. J. Neurosci. Res. 85, 3347–3358. https://doi.org/10.1002/ critical patients with non-hepatic hyperammonemia. Metab. Brain Dis. 33, jnr.21444. 1335–1342. https://doi.org/10.1007/s11011-018-0245-z. McKenna, M.C., Ferreira, G.C., 2016. Enzyme complexes important for the glutamate- Laursen, H., Diemer, N.H., 1979. Morphometric studies of rat glial cell ultrastructure glutamine cycle. Adv. Neurobiol. 13, 59–98. https://doi.org/10.1007/978-3-319- after urease-induced hyperammonaemia. Neuropathol. Appl. Neurobiol. 5, 345–362. 45096-4_4. https://doi.org/10.1111/j.1365-2990.1979.tb00634.x. McLennan, H., 1976. The autoradiographic localization of L-[3h]glutamate in rat brain tissue. Brain Res. 115, 139–144.

15 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

McNair, L.F., Andersen, J.V., Aldana, B.I., Hohnholt, M.C., Nissen, J.D., Sun, Y., Parviz, F., Matullo, C., Garrison, W.D., Savatski, L., Adamson, J.W., Ning, G., Kaestner, K. Fischer, K.D., Sonnewald, U., Nyberg, N., Webster, S.C., et al., 2019. Deletion of H., Rossi, J.M., Zaret, K.S., Duncan, S.A., 2003. Hepatocyte nuclear factor 4alpha neuronal GLT-1 in mice reveals its role in synaptic glutamate homeostasis and controls the development of a hepatic epithelium and liver morphogenesis. Nat. mitochondrial function. J. Neurosci. 39, 4847–4863. https://doi.org/10.1523/ Genet. 34, 292–296. https://doi.org/10.1038/ng1175. JNEUROSCI.0894-18.2019. Patejunas, G., Bradley, A., Beaudet, A.L., O’Brien, W.E., 1994. Generation of a mouse McReynolds, J.W., Crowley, B., Mahoney, M.J., Rosenberg, L.E., 1981. Autosomal model for citrullinemia by targeted disruption of the argininosuccinate synthetase recessive inheritance of human mitochondrial carbamyl phosphate synthetase gene. Somat. Cell Mol. Genet. 20, 55–60. https://doi.org/10.1007/BF02257486. deficiency. Am. J. Hum. Genet. 33, 345–353. Perez, C.J., Jaubert, J., Guenet, J.-L., Barnhart, K.F., Ross-Inta, C.M., Quintanilla, V.C., Meister, A., 1968a. Stereochemical mapping of the active site of glutamine synthetase. Aubin, I., Brandon, J.L., Otto, N.W., DiGiovanni, J., Gimenez-Conti, I., Giulivi, C., Fed. Proc. 27, 100–109. Kusewitt, D.F., Conti, C.J., Benavides, F., 2010. Two hypomorphic alleles of mouse Meister, A., 1968b. The specificity of glutamine synthetase and its relationship to Ass1 as a new animal model of citrullinemia type I and other hyperammonemic substrate conformation at the active site. Adv. Enzymol. Relat. Area Mol. Biol. 31, syndromes. Am. J. Pathol. 177, 1958–1968. https://doi.org/10.2353/ 183–218. https://doi.org/10.1002/9780470122761.ch5. ajpath.2010.100118. Mennerick, S., Dhond, R.P., Benz, A., Xu, W., Rothstein, J.D., Danbolt, N.C., Isenberg, K. Petr, G.T., Sun, Y., Frederick, N.M., Zhou, Y., Dhamne, S.C., Hameed, M.Q., Miranda, C., E., Zorumski, C.F., 1998. Neuronal expression of the glutamate transporter GLT-1 in Bedoya, E.A., Fischer, K.D., Armsen, W., Wang, J., Danbolt, N.C., Rotenberg, A., hippocampal microcultures. J. Neurosci. 18, 4490–4499. https://doi.org/10.1523/ Aoki, C.J., Rosenberg, P.A., 2015. Conditional deletion of the glutamate transporter JNEUROSCI.18-12-04490.1998. GLT-1 reveals that astrocytic GLT-1 protects against fatal epilepsy while neuronal Miyanaka, K., Gotoh, T., Nagasaki, A., Takeya, M., Ozaki, M., Iwase, K., Takiguchi, M., GLT-1 contributes significantlyto glutamate uptake into synaptosomes. J. Neurosci. Iyama, K.I., Tomita, K., Mori, M., 1998. Immunohistochemical localization of 35, 5187–5201. https://doi.org/10.1523/JNEUROSCI.4255-14.2015. arginase II and other enzymes of arginine metabolism in rat kidney and liver. Pines, G., Danbolt, N.C., Bjørås, M., Zhang, Y., Bendahan, A., Eide, L., Koepsell, H., Histochem. J. 30, 741–751. https://doi.org/10.1023/a:1003468726969. Storm-Mathisen, J., Seeberg, E., Kanner, B.I., 1992. Cloning and expression of a rat Moorman, A.F., de Boer, P.A., Watford, M., Dingemanse, M.A., Lamers, W.H., 1994. brain L- glutamate transporter. Nature 360, 464–467. https://doi.org/10.1038/ Hepatic glutaminase mRNA is confinedto part of the urea cycle domain in the adult 360464a0. rodent liver lobule. FEBS Lett. 356, 76–80. https://doi.org/10.1016/0014-5793(94) Plachez, C., Danbolt, N.C., Recasens, M., 2000. Transient expression of the glial 01230-x. glutamate transporters GLAST and GLT in hippocampal neurons in primary culture. Moriyama, M., Li, M.X., Kobayashi, K., Sinasac, D.S., Kannan, Y., Iijima, M., J. Neurosci. Res. 59, 587–593. https://doi.org/10.1002/(SICI)1097-4547 Horiuchi, M., Tsui, L.-C., Tanaka, M., Nakamura, Y., Saheki, T., 2006. Pyruvate (20000301)59:5<587::AID-JNR1>3.0.CO;2-L. ameliorates the defect in ureogenesis from ammonia in citrin-deficient mice. Prakash, R., Mullen, K.D., 2010. Mechanisms, diagnosis and management of hepatic J. Hepatol. 44, 930–938. https://doi.org/10.1016/j.jhep.2005.09.018. encephalopathy. Nat. Rev. Gastroenterol. Hepatol. 7, 515–525. https://doi.org/ Mostafeezur, R.M., Shinoda, M., Unno, S., Zakir, H.M., Takatsuji, H., Takahashi, K., 10.1038/nrgastro.2010.116. Yamada, Y., Yamamura, K., Iwata, K., Kitagawa, J., 2014. Involvement of astroglial Quinonez, S.C., Thoene, J.G., 1993. Citrullinemia type I. glutamate-glutamine shuttle in modulation of the jaw-opening reflex following Qureshi, I.A., Letarte, J., Ouellet, R., Larochelle, J., Lemieux, B., 1983. A new French- infraorbital nerve injury. Eur. J. Neurosci. 39, 2050–2059. https://doi.org/10.1111/ Canadian family affected by hyperargininaemia. J. Inherit. Metab. Dis. 6, 179–182. ejn.12562. https://doi.org/10.1007/BF02310878. Murakami-Murofushi, K., Ratner, S., 1979. Argininosuccinase from bovine brain: Qureshi, T., Sorensen, C., Berghuis, P., Jensen, V., Dobszay, M.B., Farkas, T., Dalen, K.T., isolation and comparison of catalytic, physical, and chemical properties with the Guo, C., Hassel, B., Utheim, T.P., Hvalby, O., Hafting, T., Harkany, T., Fyhn, M., enzymes from liver and kidney. Anal. Biochem. 95, 139–155. https://doi.org/ Chaudhry, F.A., 2019. The Glutamine Transporter Slc38a1 Regulates GABAergic 10.1016/0003-2697(79)90197-0. Neurotransmission and Synaptic Plasticity. Cereb Cortex 29, 5166–5179. https:// Mutch, B.J., Banister, E.W., 1983. Ammonia metabolism in exercise and fatigue: a doi.org/10.1093/cercor/bhz055. review. Med. Sci. Sports Exerc. 15, 41–50. Qureshi, T., Bjorkmo, M., Nordengen, K., Gundersen, V., Utheim, T.P., Watne, L.O., Nagamani, S.C., Erez, A., Lee, B., 2012. Argininosuccinate lyase deficiency. Genet. Med. Storm-Mathisen, J., Hassel, B., Chaudhry, F.A., 2020. Slc38a1 Conveys Astroglia- 14, 501–507. https://doi.org/10.1038/gim.2011.1. Derived Glutamine into GABAergic Interneurons for Neurotransmitter GABA Nagasaka, H., Yorifuji, T., Murayama, K., Kubota, M., Kurokawa, K., Murakami, T., Synthesis. Cells 9. https://doi.org/10.3390/cells9071686. Kanazawa, M., Takatani, T., Ogawa, A., Ogawa, E., Yamamoto, S., Adachi, M., Qvartskhava, N., Jin, C.J., Gorg, B., Haussinger, D., 2019. Comment on Hakvoort et al. Kobayashi, K., Takayanagi, M., 2006. Effects of arginine treatment on nutrition, Hepatology 69, 921–922. https://doi.org/10.1002/hep.29727. growth and urea cycle function in seven Japanese boys with late-onset ornithine Qvartskhava, N., Lang, P.A., Gorg, B., Pozdeev, V.I., Ortiz, M.P., Lang, K.S., Bidmon, H.J., transcarbamylase deficiency. Eur. J. Pediatr. 165, 618–624. https://doi.org/ Lang, E., Leibrock, C.B., Herebian, D., Bode, J.G., Lang, F., Haussinger, D., 2015. 10.1007/s00431-006-0143-y. Hyperammonemia in gene-targeted mice lacking functional hepatic glutamine Najimi, M., Stephenne, X., Sempoux, C., Sokal, E., 2014. Regulation of hepatic EAAT-2 synthetase. Proc. Natl. Acad. Sci. U. S. A. 112, 5521–5526. https://doi.org/10.1073/ glutamate transporter expression in human liver cholestasis. World J. Gastroenterol. pnas.1423968112. 20, 1554–1564. https://doi.org/10.3748/wjg.v20.i6.1554. Rangroo Thrane, V., Thrane, A.S., Wang, F., Cotrina, M.L., Smith, N.A., Chen, M., Xu, Q., Nissim, I., Cattano, C., Nissim, I., Yudkoff, M., 1992. Relative role of the glutaminase, Kang, N., Fujita, T., Nagelhus, E.A., Nedergaard, M., 2013. Ammonia triggers glutamate dehydrogenase, and AMP-deaminase pathways in hepatic ureagenesis: neuronal disinhibition and seizures by impairing astrocyte potassium buffering. Nat. studies with 15N. Arch. Biochem. Biophys. 292, 393–401. https://doi.org/10.1016/ Med. 19, 1643–1648. https://doi.org/10.1038/nm.3400. 0003-9861(92)90008-k. Rao, K.V., Norenberg, M.D., 2001. Cerebral energy metabolism in hepatic Nissim, I., Horyn, O., Luhovyy, B., Lazarow, A., Daikhin, Y., Nissim, I., Yudkoff, M., encephalopathy and hyperammonemia. Metab. Brain Dis. 16, 67–78. https://doi. 2003. Role of the glutamate dehydrogenase reaction in furnishing aspartate nitrogen org/10.1023/a:1011666612822. for urea synthesis: studies in perfused rat liver with 15N. Biochem J. 376 (Pt 1), Rappaport, A.M., 1958. The structural and functional unit in the human liver (liver 179–188. https://doi.org/10.1042/BJ20030997. acinus). Anat. Rec. 130, 673–689. https://doi.org/10.1002/ar.1091300405. Norenberg, M.D., Martinez-Hernandez, A., 1979. Fine structural localization of Ratnakumari, L., Qureshi, I.A., Butterworth, R.F., 1992. Effects of congenital glutamine synthetase in astrocytes of rat brain. Brain Res. 161, 303–310. https://doi. hyperammonemia on the cerebral and hepatic levels of the intermediates of energy org/10.1016/0006-8993(79)90071-4. metabolism in spf mice. Biochem. Biophys. Res. Commun. 184, 746–751. https:// Northington, F.J., Traystman, R.J., Koehler, R.C., Martin, L.J., 1999. GLT1, glial doi.org/10.1016/0006-291x(92)90653-3. glutamate transporter, is transiently expressed in neurons and develops astrocyte Ratner, S., 1954. Urea synthesis and metabolism of arginine and citrulline. Adv. specificity only after midgestation in the ovine fetal brain. J. Neurobiol. 39, Enzymol. Relat. Subj. Biochem. 15, 319–387. https://doi.org/10.1002/ 515–526. 9780470122600.ch8. Northington, F.J., Traystman, R.J., Koehler, R.C., Rothstein, J.D., Martin, L.J., 1998. Ratner, S., Anslow, W.P., Petrack, B., 1953. Biosynthesis of urea. VI. Enzymatic cleavage Regional and cellular expression of glial (GLT1) and neuronal (EAAC1) glutamate of argininosuccinic acid to arginine and fumaric acid. J. Biol. Chem. 204, 115–125. transporter proteins in ovine fetal brain. Neuroscience 85, 1183–1194. https://doi. Ratner, S., Pappas, A., 1949. Biosynthesis of urea; arginine synthesis from citrulline in org/10.1016/s0306-4522(97)00673-8. liver homogenates. J. Biol. Chem. 179, 1199–1212. O’sullivan, D., Brosnan, J.T., Brosnan, M.E., 1998. Hepatic zonation of the catabolism of Rauen, T., 2000. Diversity of glutamate transporter expression and function in the arginine and ornithine in the perfused rat liver. Biochem. J. 330 (Pt 2), 627–632. mammalian retina. Amino Acids 19, 53–62. https://doi.org/10.1007/ https://doi.org/10.1042/bj3300627. s007260070033. Olde Damink, S.W., Deutz, N.E., Dejong, C.H., Soeters, P.B., Jalan, R., 2002. Interorgan Rauen, T., Fischer, F., Wiessner, M., 1999. Glia-neuron interaction by high-affinity ammonia metabolism in liver failure. Neurochem. Int. 41, 177–188. https://doi.org/ glutamate transporters in neurotransmission. Adv. Exp. Med. Biol. 468, 81–95. 10.1016/s0197-0186(02)00040-2. https://doi.org/10.1007/978-1-4615-4685-6_7. Otis, T.S., Jahr, C.E., 1998. Anion currents and predicted glutamate flux through a Rauen, T., Kanner, B.I., 1994. Localization of the glutamate transporter GLT-1 in rat and neuronal glutamate transporter. J. Neurosci. 18, 7099–7110. macaque monkey retinae. Neurosci. Lett. 169, 137–140. https://doi.org/10.1016/ Otis, T.S., Kavanaugh, M.P., 2000. Isolation of current components and partial reaction 0304-3940(94)90375-1. cycles in the glial glutamate transporter EAAT2. J. Neurosci. 20, 2749–2757. Rauen, T., Rothstein, J.D., Wassle, H., 1996. Differential expression of three glutamate Palmieri, E.M., Menga, A., Martin-Perez, R., Quinto, A., Riera-Domingo, C., De Tullio, G., transporter subtypes in the rat retina. Cell Tissue Res. 286, 325–336. https://doi. Hooper, D.C., Lamers, W.H., Ghesquiere, B., McVicar, D.W., Guarini, A., org/10.1007/s004410050702. Mazzone, M., Castegna, A., 2017. Pharmacologic or genetic targeting of glutamine Reid Sutton, V., Pan, Y., Davis, E.C., Craigen, W.J., 2003. A mouse model of synthetase skews macrophages toward an M1-like phenotype and inhibits tumor argininosuccinic aciduria: biochemical characterization. Mol. Genet. Metabol. 78, metastasis. Cell Rep. 20, 1654–1666. https://doi.org/10.1016/j.celrep.2017.07.054. 11–16. https://doi.org/10.1016/s1096-7192(02)00206-8.

16 Y. Zhou et al. Neurochemistry International 140 (2020) 104809

Riepe, R.E., Norenburg, M.D., 1977. Muller cell localisation of glutamine synthetase in Shawcross, D.L., Sharifi, Y., Canavan, J.B., Yeoman, A.D., Abeles, R.D., Taylor, N.J., rat retina. Nature 268, 654–655. https://doi.org/10.1038/268654a0. Auzinger, G., Bernal, W., Wendon, J.A., 2011. Infection and systemic inflammation, Rochovansky, O., Kodowaki, H., Ratner, S., 1977. Biosynthesis of urea. Molecular and not ammonia, are associated with Grade 3/4 hepatic encephalopathy, but not regulatory properties of crystalline argininosuccinate synthetase. J. Biol. Chem. 252, mortality in cirrhosis. J. Hepatol. 54, 640–649. https://doi.org/10.1016/j. 5287–5294. jhep.2010.07.045. Romero-Gomez, M., Montagnese, S., Jalan, R., 2015. Hepatic encephalopathy in patients Shawcross, D.L., Wright, G., Olde Damink, S.W., Jalan, R., 2007. Role of ammonia and with acute decompensation of cirrhosis and acute-on-chronic liver failure. inflammation in minimal hepatic encephalopathy. Metab. Brain Dis. 22, 125–138. J. Hepatol. 62, 437–447. https://doi.org/10.1016/j.jhep.2014.09.005. https://doi.org/10.1007/s11011-006-9042-1. Rothman, D.L., Sibson, N.R., Hyder, F., Shen, J., Behar, K.L., Shulman, R.G., 1999. In Shigesada, K., Tatibana, M., 1971. Role of acetylglutamate in ureotelism. I. Occurrence vivo nuclear magnetic resonance spectroscopy studies of the relationship between and biosynthesis of acetylglutamate in mouse and rat tissues. J. Biol. Chem. 246, the glutamate-glutamine neurotransmitter cycle and functional neuroenergetics. 5588–5595. Philos. Trans. R. Soc. Lond. B Biol. Sci. 354, 1165–1177. https://doi.org/10.1098/ Silver, M.L., Johnson, R.E., 1947. White bread and epilepsy in animals. J. Am. Med. rstb.1999.0472. Assoc. 135, 757–760. https://doi.org/10.1001/jama.1947.02890120011005. Rothstein, J.D., Martin, L., Levey, A.I., Dykes-Hoberg, M., Jin, L., Wu, D., Nash, N., Sinasac, D.S., Moriyama, M., Jalil, M.A., Begum, L., Li, M.X., Iijima, M., Horiuchi, M., Kuncl, R.W., 1994. Localization of neuronal and glial glutamate transporters. Neuron Robinson, B.H., Kobayashi, K., Saheki, T., Tsui, L.-C., 2004. Slc25a13-knockout mice 13, 713–725. https://doi.org/10.1016/0896-6273(94)90038-8. harbor metabolic deficits but fail to display hallmarks of adult-onset type II Rovira, A., Alonso, J., Cordoba, J., 2008. MR imaging findings in hepatic citrullinemia. Mol. Cell Biol. 24, 527–536. https://doi.org/10.1128/mcb.24.2.527- encephalopathy. AJNR Am. J. Neuroradiol. 29, 1612–1621. https://doi.org/ 536.2004. 10.3174/ajnr.A1139. Speck, J.F., 1947. The enzymic synthesis of glutamine. J. Biol. Chem. 168, 403. Rowe, W.B., Meister, A., 1970. Identification of L-methionine-S-sulfoximine as the Spodenkiewicz, M., Diez-Fernandez, C., Rufenacht, V., Gemperle-Britschgi, C., convulsant isomer of methionine sulfoximine. Proc. Natl. Acad. Sci. U. S. A. 66, Haberle, J., 2016. Minireview on glutamine synthetase deficiency, an ultra-rare 500–506. https://doi.org/10.1073/pnas.66.2.500. inborn error of amino acid biosynthesis. Biology 5. https://doi.org/10.3390/ Saheki, T., Iijima, M., Li, M.X., Kobayashi, K., Horiuchi, M., Ushikai, M., Okumura, F., biology5040040. Meng, X.J., Inoue, I., Tajima, A., Moriyama, M., Eto, K., Kadowaki, T., Sinasac, D.S., Stettner, N., Rosen, C., Bernshtein, B., Gur-Cohen, S., Frug, J., Silberman, A., Sarver, A., Tsui, L.-C., Tsuji, M., Okano, A., Kobayashi, T., 2007. Citrin/mitochondrial glycerol- Carmel-Neiderman, N.N., Eilam, R., Biton, I., et al., 2018. Induction of nitric-oxide 3-phosphate dehydrogenase double knock-out mice recapitulate features of human metabolism in enterocytes alleviates colitis and inflammation-associated colon citrin deficiency. J. Biol. Chem. 282, 25041–25052. https://doi.org/10.1074/jbc. cancer. Cell Rep. 23, 1962–1976. https://doi.org/10.1016/j.celrep.2018.04.053. M702031200. Stoll, B., McNelly, S., Buscher, H.P., Haussinger, D., 1991. Functional hepatocyte Saheki, T., Inoue, K., Ono, H., Katsura, N., Yokogawa, M., Yoshidumi, Y., Furuie, S., heterogeneity in glutamate, aspartate and alpha-ketoglutarate uptake: a Kuroda, E., Ushikai, M., Asakawa, A., Inui, A., Eto, K., Kadowaki, T., Sinasac, D.S., histoautoradiographical study. Hepatology 13, 247–253. Yamamura, K.-I., Kobayashi, K., 2012. Effects of supplementation on food intake, Sudhof, T.C., 2014. The molecular machinery of neurotransmitter release (Nobel body weight and hepatic metabolites in the citrin/mitochondrial glycerol-3- lecture). Angew Chem. Int. Ed. Engl. 53, 12696–12717. https://doi.org/10.1002/ phosphate dehydrogenase double-knockout mouse model of human citrin deficiency. anie.201406359. Mol. Genet. Metabol. 107, 322–329. https://doi.org/10.1016/j. Takada, S., Saheki, T., Igarashi, Y., Katsunuma, T., 1979. Studies on rat liver ymgme.2012.07.021. argininosuccinate synthetase. Inhibition by various amino acids. J. Biochem. 85, Saheki, T., Kobayashi, K., Iijima, M., Nishi, I., Yasuda, T., Yamaguchi, N., Gao, H.Z., 1309–1314. Jalil, M.A., Begum, L., Li, M.X., 2002. Pathogenesis and pathophysiology of citrin (A Takasaki, C., Yamasaki, M., Uchigashima, M., Konno, K., Yanagawa, Y., Watanabe, M., mitochondrial aspartate glutamate carrier) deficiency. Metab. Brain Dis. 17, 2010. Cytochemical and cytological properties of perineuronal oligodendrocytes in 335–346. https://doi.org/10.1023/a:1021961919148. the mouse cortex. Eur. J. Neurosci. 32, 1326–1336. https://doi.org/10.1111/j.1460- Saheki, T., Kusumi, T., Takada, S., Katsunuma, T., Katunuma, N., 1975. Crystallization 9568.2010.07377.x. and some properties of argininosuccinate synthase from rat liver. FEBS Lett. 58, Tanaka, K., Watase, K., Manabe, T., Yamada, K., Watanabe, M., Takahashi, K., Iwama, H., 314–317. https://doi.org/10.1016/0014-5793(75)80287-0. Nishikawa, T., Ichihara, N., Kikuchi, T., Okuyama, S., Kawashima, N., Hori, S., Saheki, T., Moriyama, M., Kuroda, E., Funahashi, A., Yasuda, I., Setogawa, Y., Gao, Q., Takimoto, M., Wada, K., 1997. Epilepsy and exacerbation of brain injury in mice Ushikai, M., Furuie, S., Yamamura, K.-I., Takano, K., Nakamura, Y., Eto, K., lacking the glutamate transporter GLT-1. Science 276, 1699–1702. https://doi.org/ Kadowaki, T., Sinasac, D.S., Furukawa, T., Horiuchi, M., Tai, Y.H., 2019. Pivotal role 10.1126/science.276.5319.1699. of inter-organ aspartate metabolism for treatment of mitochondrial aspartate- Tanigami, H., Rebel, A., Martin, L.J., Chen, T.-Y., Brusilow, S.W., Traystman, R.J., glutamate carrier 2 (citrin) deficiency,based on the mouse model. Sci. Rep. 9, 4179. Koehler, R.C., 2005. Effect of glutamine synthetase inhibition on astrocyte swelling https://doi.org/10.1038/s41598-019-39627-y. and altered astroglial protein expression during hyperammonemia in rats. Saheki, T., Song, Y.-Z., 1993. Citrin deficiency.In: Adam, M.P., Ardinger, H.H., Pagon, R. Neuroscience 131, 437–449. https://doi.org/10.1016/j.neuroscience.2004.10.045. A., Wallace, S.E., Bean, L.J., Stephens, K., Amemiya, A. (Eds.), GeneReviews® Tansey, F.A., Farooq, M., Cammer, W., 1991. Glutamine synthetase in oligodendrocytes [. University of Washington, Seattle, Seattle (WA). and astrocytes: new biochemical and immunocytochemical evidence. J. Neurochem. Saitoh, F., Araki, T., 2010. Proteasomal degradation of glutamine synthetase regulates 56, 266–272. https://doi.org/10.1111/j.1471-4159.1991.tb02591.x. schwann cell differentiation. J. Neurosci. 30, 1204–1212. https://doi.org/10.1523/ Taylor, P.M., Rennie, M.J., 1987. Perivenous localisation of Na-dependent glutamate JNEUROSCI.3591-09.2010. transport in perfused rat liver. FEBS Lett. 221, 370–374. https://doi.org/10.1016/ Salvi, S., Santorelli, F.M., Bertini, E., Boldrini, R., Meli, C., Donati, A., Burlina, A.B., 0014-5793(87)80958-4. Rizzo, C., Di Capua, M., Fariello, G., Dionisi-Vici, C., 2001. Clinical and molecular Tezcan, K., Louie, K.T., Qu, Y., Velasquez, J., Zaldivar, F., Rioseco-Camacho, N., findings in hyperornithinemia-hyperammonemia-homocitrullinuria syndrome. Camacho, J.A., 2012. Adult-onset presentation of a hyperornithinemia- Neurology 57, 911–914. https://doi.org/10.1212/wnl.57.5.911. hyperammonemia-homocitrullinuria patient without prior history of neurological Saper, C.B., 2009. A guide to the perplexed on the specificityof antibodies. J. Histochem. complications. JIMD Rep. 3, 97–102. https://doi.org/10.1007/8904_2011_71. Cytochem. 57, 1–5. https://doi.org/10.1369/jhc.2008.952770. Torp, R., Danbolt, N.C., Babaie, E., Bjoras, M., Seeberg, E., Storm-Mathisen, J., Schmitt, A., Asan, E., Puschel, B., Jons, T., Kugler, P., 1996. Expression of the glutamate Ottersen, O.P., 1994. Differential expression of two glial glutamate transporters in transporter GLT1 in neural cells of the rat central nervous system: non-radioactive in the rat brain: an in situ hybridization study. Eur. J. Neurosci. 6, 936–942. https:// situ hybridization and comparative immunocytochemistry. Neuroscience 71, doi.org/10.1111/j.1460-9568.1994.tb00587.x. 989–1004. https://doi.org/10.1016/0306-4522(95)00477-7. Torp, R., Hoover, F., Danbolt, N.C., Storm-Mathisen, J., Ottersen, O.P., 1997. Differential Schmitt, A., Asan, E., Puschel, B., Kugler, P., 1997. Cellular and regional distribution of distribution of the glutamate transporters GLT1 and rEAAC1 in rat cerebral cortex the glutamate transporter GLAST in the CNS of rats: nonradioactive in situ and thalamus: an in situ hybridization analysis. Anat. Embryol. 195, 317–326. hybridization and comparative immunocytochemistry. J. Neurosci. 17, 1–10. https://doi.org/10.1007/s004290050051. https://doi.org/10.1523/JNEUROSCI.17-01-00001.1997. Tranah, T.H., Vijay, G.K., Ryan, J.M., Shawcross, D.L., 2013. Systemic inflammationand Schofield,J.P., Cox, T.M., Caskey, C.T., Wakamiya, M., 1999. Mice deficientin the urea- ammonia in hepatic encephalopathy. Metab. Brain Dis. 28, 1–5. https://doi.org/ cycle enzyme, carbamoyl phosphate synthetase I, die during the early neonatal 10.1007/s11011-012-9370-2. period from hyperammonemia. Hepatology 29, 181–185. https://doi.org/10.1002/ Tsuboi, Y., Iwata, K., Dostrovsky, J.O., Chiang, C.Y., Sessle, B.J., Hu, J.W., 2011. hep.510290112. Modulation of astroglial glutamine synthetase activity affects nociceptive behaviour Schousboe, A., Hertz, L., Svenneby, G., Kvamme, E., 1979. Phosphate activated and central sensitization of medullary dorsal horn nociceptive neurons in a rat model glutaminase activity and glutamine uptake in primary cultures of astrocytes. of chronic pulpitis. Eur. J. Neurosci. 34, 292–302. https://doi.org/10.1111/j.1460- J. Neurochem. 32, 943–950. https://doi.org/10.1111/j.1471-4159.1979.tb04579.x. 9568.2011.07747.x. Sedlak, T.W., Paul, B.D., Parker, G.M., Hester, L.D., Snowman, A.M., Taniguchi, Y., Tuchman, M., Lee, B., Lichter-Konecki, U., Summar, M.L., Yudkoff, M., Cederbaum, S.D., Kamiya, A., Snyder, S.H., Sawa, A., 2019. The glutathione cycle shapes synaptic Kerr, D.S., Diaz, G.A., Seashore, M.R., Lee, H.-S., McCarter, R.J., Krischer, J.P., glutamate activity. Proc. Natl. Acad. Sci. U. S. A. 116, 2701–2706. https://doi.org/ Batshaw, M.L., 2008. Cross-sectional multicenter study of patients with urea cycle 10.1073/pnas.1817885116. disorders in the United States. Mol. Genet. Metabol. 94, 397–402. https://doi.org/ Senkevitch, E., Cabrera-Luque, J., Morizono, H., Caldovic, L., Tuchman, M., 2012. 10.1016/j.ymgme.2008.05.004. A novel biochemically salvageable animal model of hyperammonemia devoid of N- van de Poll, M.C., Soeters, P.B., Deutz, N.E., Fearon, K.C., Dejong, C.H., 2004. Renal acetylglutamate synthase. Mol. Genet. Metabol. 106, 160–168. https://doi.org/ metabolism of amino acids: its role in interorgan amino acid exchange. Am. J. Clin. 10.1016/j.ymgme.2012.03.004. Nutr. 79, 185–197. https://doi.org/10.1093/ajcn/79.2.185. Shaw, C.A., Bains, J.S., 2002. Synergistic versus antagonistic actions of glutamate and van den Berg, C.J., Garfinkel, D., 1971. A simulation study of brain compartments. glutathione: the role of excitotoxicity and oxidative stress in neuronal disease. Cell. Metabolism of glutamate and related substances in mouse brain. Biochem. J. 123, Mol. Biol. 48, 127–136. 211–218. https://doi.org/10.1042/bj1230211.

17 Y. Zhou et al. Neurochemistry International 140 (2020) 104809 van der Hel, W.S., Notenboom, R.G., Bos, I.W., van Rijen, P.C., van Veelen, C.W., de Wu, G.Y., Morris, S.M., 1998. Arginine metabolism: nitric oxide and beyond. Biochem. J. Graan, P.N., 2005. Reduced glutamine synthetase in hippocampal areas with neuron 336, 1–17. https://doi.org/10.1042/bj3360001. loss in temporal lobe epilepsy. Neurology 64, 326–333. https://doi.org/10.1212/01. Xin, W., Mironova, Y.A., Shen, H., Marino, R.A., Waisman, A., Lamers, W.H., Bergles, D. WNL.0000149636.44660.99. E., Bonci, A., 2019. Oligodendrocytes support neuronal glutamatergic transmission van Straaten, H.W., He, Y., van Duist, M.M., Labruyere, W.T., Vermeulen, J.L., van via expression of glutamine synthetase. Cell Rep. 27, 2262–2271. https://doi.org/ Dijk, P.J., Ruijter, J.M., Lamers, W.H., Hakvoort, T.B., 2006. Cellular concentrations 10.1016/j.celrep.2019.04.094 e5. of glutamine synthetase in murine organs. Biochem. Cell. Biol. 84, 215–231. https:// Yamamoto, T., Iwasaki, Y., Sato, Y., Yamamoto, H., Konno, H., 1989. Astrocytic doi.org/10.1139/o05-170. pathology of methionine sulfoximine-induced encephalopathy. Acta Neuropathol. Verlander, J.W., Chu, D., Lee, H.-W., Handlogten, M.E., Weiner, I.D., 2013. Expression of 77, 357–368. https://doi.org/10.1007/bf00687370. glutamine synthetase in the mouse kidney: localization in multiple epithelial cell Yang, D., Hazey, J.W., David, F., Singh, J., Rivchum, R., Streem, J.M., Halperin, M.L., types and differential regulation by hypokalemia. Am. J. Physiol. Ren. Physiol. 305, Brunengraber, H., 2000. Integrative physiology of splanchnic glutamine and F701–F713. https://doi.org/10.1152/ajprenal.00030.2013. ammonium metabolism. Am. J. Physiol. Endocrinol. Metab. 278, E469–E476. Veruki, M.L., Morkve, S.H., Hartveit, E., 2006. Activation of a presynaptic glutamate https://doi.org/10.1152/ajpendo.2000.278.3.E469. transporter regulates synaptic transmission through electrical signaling. Nat. Yu, H., Iyer, R.K., Kern, R.M., Rodriguez, W.I., Grody, W.W., Cederbaum, S.D., 2001. Neurosci. 9, 1388–1396. https://doi.org/10.1038/nn1793. Expression of arginase isozymes in mouse brain. J. Neurosci. Res. 66, 406–422. Visek, W.J., 1968. Some aspects of ammonia toxicity in animal cells. J. Dairy Sci. 51, https://doi.org/10.1002/jnr.1233. 286–295. https://doi.org/10.3168/jds.S0022-0302(68)86976-0. Yudkoff, M., 2017. Interactions in the metabolism of glutamate and the branched-chain von Gersdorff, H., Borst, J.G., 2002. Short-term plasticity at the calyx of Held. Nat. Rev. amino acids and ketoacids in the CNS. Neurochem. Res. 42, 10–18. https://doi.org/ Neurosci. 3, 53–64. https://doi.org/10.1038/nrn705. 10.1007/s11064-016-2057-z. Waagepetersen, H.S., Qu, H., Sonnewald, U., Shimamoto, K., Schousboe, A., 2005. Role Zhou, Y., Danbolt, N.C., 2014. Glutamate as a neurotransmitter in the healthy brain. of glutamine and neuronal glutamate uptake in glutamate homeostasis and synthesis J. Neural. Transm. 121, 799–817. https://doi.org/10.1007/s00702-014-1180-8. during vesicular release in cultured glutamatergic neurons. Neurochem. Int. 47, Zhou, Y., Dhaher, R., Parent, M., Hu, Q.-X., Hassel, B., Yee, S.-P., Hyder, F., 92–102. https://doi.org/10.1016/j.neuint.2005.04.012. Gruenbaum, S.E., Eid, T., Danbolt, N.C., 2019a. Selective deletion of glutamine Wang, L., Bell, P., Morizono, H., He, Z., Pumbo, E., Yu, H., White, J., Batshaw, M.L., synthetase in the mouse cerebral cortex induces glial dysfunction and vascular Wilson, J.M., 2017. AAV gene therapy corrects OTC deficiency and prevents liver impairment that precede epilepsy and neurodegeneration. Neurochem. Int. 123, fibrosis in aged OTC-knock out heterozygous mice. Mol. Genet. Metabol. 120, 22–33. https://doi.org/10.1016/j.neuint.2018.07.009. 299–305. https://doi.org/10.1016/j.ymgme.2017.02.011. Zhou, Y., Hassel, B., Eid, T., Danbolt, N.C., 2019b. Axon-terminals expressing EAAT2 Warren, K.S., Schenker, S., 1964. Effect OF an inhibitor OF glutamine synthesis (GLT-1; Slc1a2) are common in the forebrain and not limited to the hippocampus. (methionine sulfoximine) ON ammonia toxicity and metabolism. J. Lab. Clin. Med. Neurochem. Int. 123, 101–113. https://doi.org/10.1016/j.neuint.2018.03.006. 64, 442–449. Zhou, Y., Waanders, L.F., Holmseth, S., Guo, C., Berger, U.V., Li, Y., Lehre, A.-C., Weiner, I.D., Miller, R.T., Verlander, J.W., 2003. Localization of the ammonium Lehre, K.P., Danbolt, N.C., 2014. Proteome analysis and conditional deletion of the transporters, Rh B glycoprotein and Rh C glycoprotein, in the mouse liver. EAAT2 glutamate transporter provide evidence against a role of EAAT2 in pancreatic Gastroenterology 124, 1432–1440. https://doi.org/10.1016/s0016-5085(03)00277- insulin secretion in mice. J. Biol. Chem. 289, 1329–1344. https://doi.org/10.1074/ 4. jbc.M113.529065. Weiner, I.D., Mitch, W.E., Sands, J.M., 2015. Urea and ammonia metabolism and the Zielke, H.R., Collins jr., R.M., Baab, P.J., Huang, Y., Zielke, C.L., Tildon, J.T., 1998. control of renal nitrogen excretion. Clin. J. Am. Soc. Nephrol. 10, 1444–1458. Compartmentation of [14C]glutamate and [14C]glutamine oxidative metabolism in https://doi.org/10.2215/CJN.10311013. the rat hippocampus as determined by microdialysis. J Neurochem 71, 1315–1320. Weiner, I.D., Verlander, J.W., 2013. Renal ammonia metabolism and transport. Comp. https://doi.org/10.1046/j.1471-4159.1998.71031315.x. Physiol. 3, 201–220. https://doi.org/10.1002/cphy.c120010. Zielke, H.R., Zielke, C.L., Baab, P.J., 2009. Direct measurement of oxidative metabolism Wellner, V.P., Meister, A., 1966. Binding of adenosine triphosphate and adenosine in the living brain by microdialysis: a review. J Neurochem 109 (Suppl 1), 24–29. diphosphate by glutamine synthetase. Biochemistry 5, 872–879. https://doi.org/ https://doi.org/10.1111/j.1471-4159.2009.05941.x. 10.1021/bi00867a010. Zwirner, K., Thiel, C., Thiel, K., Morgalla, M.H., Konigsrainer, A., Schenk, M., 2010. Wong, L.J., Craigen, W.J., O’Brien, W.E., 1994. Postpartum coma and death due to Extracellular brain ammonia levels in association with arterial ammonia, carbamoyl-phosphate synthetase I deficiency. Ann. Intern. Med. 120, 216–217. intracranial pressure and the use of albumin dialysis devices in pigs with acute liver https://doi.org/10.7326/0003-4819-120-3-199402010-00007. failure. Metab. Brain Dis. 25, 407–412. https://doi.org/10.1007/s11011-010-9222- x.

18