Abstract
Citrin deficiency (CD) is a recessive, liver disease caused by sequence variants in the SLC25A13 gene encoding a mitochondrial aspartate–glutamate transporter. CD manifests as different age‐dependent phenotypes and affects crucial hepatic metabolic pathways including malate–aspartate‐shuttle, glycolysis, gluconeogenesis, de novo lipogenesis and the tricarboxylic acid and urea cycles. Although the exact pathophysiology of CD remains unclear, impaired use of glucose and fatty acids as energy sources due to NADH shuttle defects and PPARα downregulation, respectively, indicates evident energy deficit in CD hepatocytes. The present review summarizes current trends on available and potential treatments for CD. Baseline recommendation for CD patients is dietary management, often already present as a self‐selected food preference, that includes protein and fat‐rich food, and avoidance of excess carbohydrates. At present, liver transplantation remains the sole curative option for severe CD cases. Our extensive literature review indicated medium‐chain triglycerides (MCT) as the most widely used CD treatment in all age groups. MCT can effectively improve symptoms across disease phenotypes by rapidly supplying energy to the liver, restoring redox balance and inducing lipogenesis. In contrast, sodium pyruvate restored glycolysis and displayed initial preclinical promise, with however limited efficacy in adult CD patients. Ursodeoxycholic acid, nitrogen scavengers and l‐arginine treatments effectively address specific pathophysiological aspects such as cholestasis and hyperammonemia and are commonly administered in combination with other drugs. Finally, future possibilities including restoring redox balance, amino acid supplementation, enhancing bioenergetics, improving ureagenesis and mRNA/DNA‐based gene therapy are also discussed.
Keywords: Citrin deficiency, malate‐aspartate‐shuttle, medium‐chain triglycerides, SLC25A13, ursodeoxycholic acid
1. INTRODUCTION
1.1. General aspects and biochemistry
Citrin deficiency (CD) is a monogenic, autosomal recessive disease caused by mutations in the SLC25A13 (or citrin, OMIM *603859) gene. 1 The disease affects primarily the malate–aspartate‐shuttle in the liver, but consequently also impacts several other pathways including glycolysis, gluconeogenesis, de novo lipogenesis and the tricarboxylic acid (TCA) and urea cycles. CD is a global condition with high prevalence in East Asia including Japan, mainly due to several frequent mutations. 2 , 3 To date, more than 100 different citrin pathogenic mutations have been reported. 2 CD manifests as three different age‐dependent phenotypes: neonatal intrahepatic cholestasis caused by CD (NICCD, OMIM #605814) in newborns or infants; followed by a generally asymptomatic, silent period, or by failure to thrive and dyslipidemia caused by CD (FTTDCD) in children; and finally the most severe form, citrullinemia type 2 (CTLN2, OMIM #603471) in adolescent or adult patients. 4 , 5 , 6
Citrin is an aspartate–glutamate carrier localized at the inner mitochondrial membrane and is mainly expressed in the liver, 7 while the isoform aralar, encoded by the SLC25A12 gene (OMIM *603667), is predominately expressed in skeletal muscle, the brain, kidneys, and the heart. 8 Citrin is a crucial component of the malate–aspartate shuttle that facilitates the transfer of reducing equivalents of NADH molecules from cytoplasm to mitochondria, a vital mechanism for maintaining the redox balance of both compartments (Figure 1). The loss of functional citrin disturbs the NADH/NAD+ ratio, leading to reduced levels of NADH in mitochondria and lack of electron carrier molecules for ATP production. 2 In the cytosol, lack of citrin results in an increased NADH/NAD+ ratio, thereby inhibiting glycolysis and gluconeogenesis. Given the importance of liver gluconeogenesis during fasting, 9 impaired gluconeogenesis can lead to hypoglycemia, which has been observed in NICCD and other CD phenotypes. 3 , 10 , 11 , 12 , 13
FIGURE 1.

Representation of relevant metabolic pathways implicated in CD. Transport steps and biochemical pathways involved in glucose metabolism/TCA cycle (red), malate–aspartate shuttle, amino acid and nucleotide synthesis (black), protein degradation and urea cycle (green), lipid metabolism (purple) are schematically shown. Dashed lines indicate simplified reaction steps. Citrin is colored in green and other mitochondrial carriers, citrate carrier (CIC), carnitine‐acylcarnitine translocase (CACT), mitochondrial pyruvate carrier (MPC), oxoglutarate carrier (OGC), ornithine transporter 1 (ORNT1) and amino acid transporter (AAT) are shown in shades of blue. Enzymes: pyruvate dehydrogenase (PDH), citrate synthase (CS), malate dehydrogenase (MDH), aspartate aminotransferase (AAT), carbamoyl phosphate synthetase 1 (CPS1), ornithine transcarbamylase (OTC), argininosuccinate synthetase (ASS), argininosuccinate lyase (ASL), and arginase 1 (ARG1) are highlighted in red. ATP – adenosine triphosphate, NAD+ − nicotinamide adenine dinucleotide, TCA – tricarboxylic acid, NH3 – ammonia, HCO3− – bicarbonate ion. Created with BioRender.com.
Cytosolic aspartate, transported by citrin in exchange for glutamate, is utilized for the production of amino acids, including asparagine, for de novo nucleotide synthesis and in the urea cycle. 14 , 15 In this latter pathway, aspartate is one of the substrates of argininosuccinate synthetase (ASS), being essential for ammonia detoxification in the form of nontoxic urea. In adolescents and adults with CD, a so far unexplained liver‐specific decrease in ASS without any abnormality in the ASS1 gene or hepatic mRNA levels as well as a reduced cytosolic aspartate level impair ASS functionality, resulting in increased plasma citrulline and ammonia concentrations, 5 , 16 , 17 but this needs further investigations as some patients with adolescent and adult CD (AACD, a term suggested to replace the previous disease name “CTLN2” 18 ) exhibited levels of ASS activity in liver tissue of more than 60% of controls (personal communication Prof. T. Saheki). Hyperammonemia is a main contributor to the most severe complication in these patients, which is the development of brain edema. 19
Furthermore, CD is associated with chronic liver disease ranging from neonatal or infantile onset cholestasis to fatty liver, cirrhosis and in a few patients hepatocellular carcinoma. 3 , 17 , 20 Although the precise molecular mechanism underlying CD‐associated liver disease has yet to be elucidated, recent studies showed downregulation of peroxisome proliferator‐activated receptor alpha (PPARα), a protein that plays a crucial role in regulating lipid metabolism and energy homeostasis gene expression levels, 21 , 22 , 23 as a possible contributing factor. Specifically, PPARα downregulation impairs β‐oxidation, resulting in reduced fatty acid breakdown and hepatic lipid accumulation. 21
1.2. The phenotypes of CD
Newborns and infants with citrin deficiency (NICCD) exhibit various clinical symptoms including low birth weight, jaundice, hepatomegaly, intrahepatic cholestasis, and in severe cases, fatty liver. 3 , 10 , 24 This clinical scenario is accompanied by alterations of the amino acid profile, galactosemia, hypoproteinemia and hypoglycemia, but only rarely hyperammonemia. 25 , 26 In most cases, symptoms resolve by 1 year of age, spontaneously or with dietary adjustments. 10 However, in few cases NICCD progressed into liver failure, requiring liver transplantation 10 , 24 , 27 or even resulting in fatal infantile liver cirrhosis. 6 , 28 , 29
After resolution of NICCD, patients enter an intermediate, usually silent period, with sometimes mild, nonspecific symptoms such as abdominal pain and fatigue, and in more severe cases FTTDCD, characterized by growth impairment, recurrent hypoglycemia, hyperlipidemia, pancreatitis, gastrointestinal symptoms, fatty liver and fatigue. 5 , 6 , 30
Finally, less than 20% of patients progress to the most severe form of the condition when they reach adolescent or adult age. 31 The reason why only a subset of CD patients are affected by AACD as the most severe form is still unclear, but its onset can be triggered by alcohol intake, excessive carbohydrate overload, fatigue, medication, infections and surgery. 21 , 32 , 33 Affected patients may develop a severe liver steatosis and cognitive impairment due to hyperammonemic encephalopathy. 17 , 19 , 32 In some adult patients, the disease rapidly progressed to fatal severe brain edema and acute liver failure, with liver transplantation currently being the only definitive therapy. 34 , 35
A more detailed description of the clinical situation of this multifaceted condition including diagnostic aspects can be found in the preceding article in this Special Issue, dedicated to the “Clinical landscape of citrin deficiency”. 18
2. OVERVIEW OF THE CURRENT TREATMENT LANDSCAPE
Current treatment for CD is characterized by various approaches described across different publications, but there is an absence of standardized treatment protocols and lack of preventive early interventions. The absence of a definitive cure for CD, except liver transplantation, which is limited and challenging, compounds the issue further. In recent years, clinical reports have suggested the benefit of therapies such as medium‐chain triglyceride (MCT) oil, sodium pyruvate, arginine as well as dietary interventions 6 , 32 , 36 , 37 , 38 (Table 1). However, a consolidated resource is lacking, which would synthesize the existing clinical experiences, research findings, and emerging therapies.
TABLE 1.
Summary of clinical studies using MCT or sodium pyruvate as treatments for CD.
| Publications | Patient information | Post‐treatment outcome summary | |
|---|---|---|---|
| Cohort size; phenotype | Age range | ||
| Case reports with MCT therapy | |||
| Hayasaka et al. 43 | N = 4; NICCD | 1.8–4 m.o. |
|
| Hayasaka et al. 42 | N = 5; AACD | 38–62 y.o. |
|
| Otsuka et al. 52 | N = 2; silent period | 7 & 10 y.o. |
|
| Hayasaka et al. 38 | N = 6; AACD | 48–68 y.o. |
|
| Watanabe et al. 51 | N = 2; AACD | 48 & 61 y.o. |
|
| Aoki et al. 40 | N = 3; NICCD | 12–45 d.o. |
|
| Case reports with sodium pyruvate therapy | |||
| Mutoh et al. 37 | N = 1; AACD | 13 y.o. |
|
| Yazaki et al. 34 | N = 2; AACD | 34 & 41 y.o. |
|
| Yazaki et al. 58 | N = 1; AACD | 72 y.o. |
|
| Kogure et al. 44 | N = 1; AACD | 68 y.o. |
|
| Nagasaka et al. 45 | N = 10; silent period | 3–13 y.o. |
|
Abbreviations: α‐KG, alpha‐ketoglutarate; AACD, adolescent and adult citrin deficiency (formerly CTLN2); ACarn, acetylcarnitine; Ala, alanine; ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; Amy, amylase; Arg, arginine; aPTT, activated partial thromboplastin time; ASS, argininosuccinate synthetase; AST, aspartate aminotransferase; BHB, β‐hydroxybutyrate; BUN, blood urea nitrogen; Carn, carnitine; CD, citrin deficiency; ChE, cholinesterase; Citr, citrate; Cit, citrulline; Cr, creatinine; D. bil, direct bilirubin; FBG, fasting blood glucose; Fe, iron; FFA, free fatty acids; FI, fasting insulin: FIB, fibrinogen; FR, fischer ratio; Fum, fumarate; Gln, glutamine; GGT, gamma‐glutamyl transpeptidase; Glu, glutamate; GS, glutamine synthetase; HE, hpatic encephalopathy; Hb, hemoglobin; HbA1c, hemoglobin A1c; HDL‐C, high‐density lipoprotein cholesterol; HYA, hyaluronic acid; ICitr, isocitrate; L/P, lactate to pyruvate ratio; Lac, lactate; LDH, lactate dehydrogenase; Mal, malate; MCT, medium chain triglycerides; Met, methionine; Na+, sodium ions; NaPy, sodium pyruvate; NICCD, neonatal intrahepatic cholestasis caused by citrin deficiency; NH3, ammonia; OGTT, oral glucose tolerance test; PSTI, pancreatic secretory trypsin inhibitor; PLT, platelet count; Pyr, pyruvate; RBC, red blood cell count; Suc, succinate; T/S, threonine to serine ratio; TG, triglycerides; T. bil, total bilirubin; T. chol, total cholesterol; TBA, total bile acid; TCA, tricarboxylic acid; Thr, threonine; TIBC, total iron‐binding capacity; TP, total protein; Tyr, tyrosine; UA, uric acid; WBC, white blood cell count.
In Figure 2, we provide an overview of the current therapies for CD as they are already practiced or just planned or discussed, analyze future and potential treatment options, and highlight existing challenges.
FIGURE 2.

Overview of the current and prospective treatment approaches for CD. Red color indicates pathways and processes implicated in CD manifestation. Current and prospective treatment options and their effects are marked in green and yellow, respectively. In CD, an elevated NADH/NAD+ ratio inhibits carbohydrate metabolism and energy production. For addressing this energy shortage, medium chain triglycerides (MCT) are used to stimulate beta‐oxidation, serving as an alternative source for adenosine triphosphate (ATP) production. Sodium pyruvate (NaPyr) addresses redox imbalance by stimulating gluconeogenesis and acting as an energy reservoir. In CD, urea cycle activity is diminished resulting in ammonium accumulation. To manage hyperammonemia, nitrogen scavengers eliminate excess ammonium, while arginine boosts ammonium detoxification via the urea cycle. Accordingly, l‐aspartate‐l‐ornithine (LOLA) supplementation elevates aspartate levels, stimulating ureagenesis and aspartate‐dependent pathways. Peroxisome proliferator‐activated receptor alpha (PPARα) downregulation in CD impairs β‐oxidation and transport of long‐chain fatty acids across the mitochondrial membrane, for which l‐carnitine supplementation could aid β‐oxidation restoration. Compounds like nicotinamide riboside and KL1333 aim to increase the nicotinamide adenine dinucleotide (NAD) production, restoring redox balance and increasing energy production. Simultaneously, to enhance reduced argininosuccinate synthetase (ASS) content, frequently observed in CD patients, natural occurring molecules including fisetin and spinosyn A can be employed. Finally, emerging genetic methodologies such as DNA and RNA therapy aim to target the underlying genetic defect or induce transient endogenous citrin protein formation, respectively. Created with BioRender.com.
2.1. Dietary management
The dietary management of classical urea cycle disorders (UCD) such as citrullinemia type 1 (ASS deficiency) is characterized by protein restriction and high carbohydrate intake to provide sufficient energy. 47 In contrast, the hallmark of CD patients, even without clinical intervention, is a strong dietary preference for food rich in protein and fat, and low in carbohydrates. 30 , 48 The recommended diet in CD comprises intake of 15%–25% protein, 40%–50% fat, and 30%–40% carbohydrates as excess carbohydrates can lead to manifestation of symptoms and metabolic decompensation. 33 , 49
The reason why CD patients cannot tolerate high amounts of carbohydrates is hypothesized to be due to the accumulation of cytosolic NADH as a result of impairments in the malate–aspartate shuttle. Hereby, redox‐sensitive processes such as glycolysis, gluconeogenesis and ureagenesis are affected, explaining the occurrence of carbohydrate aversion, hypoglycemia, and elevated blood ammonia. 15 , 21 , 50 A linear relationship between plasma ammonia and postprandial glucose levels in CD patients has been shown, and it was hypothesized that intake of carbohydrates may lead to an increase in cytosolic NADH/NAD+ ratio, which may contribute to impaired ureagenesis and hyperammonemia. 5 Unlike carbohydrates, the utilization of protein and fat as energy sources is possible in CD as these metabolites do not contribute to the cytosolic buildup of NADH. 32
Although dietary management has been reported to be effective as a baseline therapy, it still cannot address all the symptoms associated with CD. For instance, in a cohort of 32 CD patients (10 asymptomatic, 22 FTTDCD) who either self‐selected or were prescribed a low carbohydrate, high protein/fat diet, about half exhibited slow growth and patients with FTTDCD also experienced abdominal pain and/or hypoglycemia. 30 Furthermore, the inadequacy of dietary management is exemplified in adult CD patients, who typically show the strongest dietary preferences but still may exhibit severe symptoms such as hyperammonemia, hepatic encephalopathy (HE), low body mass index (BMI), and significant biochemical derangements. 34 , 39 This necessitates alternative or additional therapeutic interventions such as anaplerotic therapies or other medications, which are described in the following sections.
2.2. Medium chain triglycerides (MCT)
An emerging concept that explains the pathophysiology of CD is the chronic energy deficit in the liver due to the impairment in the malate–aspartate shuttle. 32 , 50 Many metabolic processes affected in CD require ATP, and it has been postulated that insufficient ATP levels are a main determinant of the disease. 38 , 50 MCT can rapidly supply energy to the liver, hereby treating the underlying energy shortage, hence MCT was suggested to have the potential to benefit all CD phenotypes. 38 Additionally, it is thought that MCT may promote the malate‐citrate shuttle to improve the hepatic cytosolic NADH/NAD+ ratio. 32 , 50
On this basis, there has been continued interest in MCT as a potential therapy to treat the underlying energy deficit associated with CD. Therefore, herein we review the available literature to provide an overview on the usage and efficacy of MCT to treat CD (Table 2).
TABLE 2.
Summary of biochemical findings in CD patients from clinical reports using MCT as a treatment.
| Patient age | Biochemical findings a | |||
|---|---|---|---|---|
| Improved but not normalized | Normalized | Unchanged | Worsened | |
| Hayasaka et al. 43 (Phenotype: NICCD) | ||||
| 1.8 m.o | Cit, Thr, ALB, T. bil, D. bil, ALP, GGT, ChE, TBA | Met, Tyr, AST, ALT | LDH | |
| 2 m.o | Cit, Thr, Met, Arg, ALB, T. bil, D. bil, AST, ALT, ALP, GGT, ChE, TBA | Tyr | LDH | |
| 2.8 m.o | Cit, Thr, Met, Arg, ALB, T. bil, D. bil, AST, ALP, GGT, ChE, TBA, PSTI | Tyr, ALT | LDH | |
| 4 m.o | Cit, Thr, Met, ALB, T. bil, D. bil, AST, ALT, GGT, ChE, TBA | NH3, Tyr, Arg, LDH, ALP | ||
| Hayasaka et al. 42 (Phenotype: AACD) | ||||
| 38 y.o | NH3, Cit, PSTI | Glu | Gln, FR, ALB, T. bil, AST, ALT, LDH, ALP, ChE, T. chol, TG, BUN, Fe, TIBC | GGT, HYA |
| 41 y.o | Cit, GGT | NH3, T. bil | Glu, Gln, FR, ALB, ALT, LDH, ChE, TG, BUN, Fe, TIBC | AST, ALP, T. chol, HYA |
| 53 y.o | Cit, Glu | NH3, Gln, FR | ALB, T. bil, AST, ALT, LDH, ALP, GGT, ChE, BUN | T. chol, PSTI |
| 53 y.o | Cit, GGT | NH3, FR, ALT, PSTI | Glu, Gln, ALB, T. bil, AST, LDH, ALP, ChE, T.chol, BUN | TG |
| 62 y.o | PSTI, HYA | NH3, Cit, Gln, FR, AST, ALT, ALP, GGT, ChE, TG, Fe, TIBC | Glu, ALB, T. bil, LDH, T. chol, BUN | |
| Otsuka et al. 52 (Phenotype: silent period) | ||||
| 7 y.o | Reference ranges not provided | |||
| 10 y.o | ||||
| Hayasaka et al. 38 (Phenotype: AACD) | ||||
| 48 y.o | Cit, Arg, FR | NH3, ALB, ALT | Gln | |
| 54 y.o | Cit, FR | NH3, Arg, ALT | Gln, ALB, ChE | |
| 60 y.o | Cit, ChE | Arg, FR, ALB | Gln, ALT | NH3 |
| 62 y.o | Cit, FR | NH3, Gln, ALB, ALT, ChE | Arg | |
| 67 y.o | Cit | NH3, Gln, ALB, ChE | FR, ALT | Arg |
| 68 y.o | NH3, Cit, Gln, FR, ALT, ChE | Arg, ALB | ||
| Watanabe et al. 51 (Phenotype: AACD) | ||||
| 48 y.o | Cit, FR | NH3, ALB, AST, ALT, GGT, TG | Gln, ALP, ChE | |
| 61 y.o | Cit, ChE | NH3, Gln, ALB, AST, ALP, GGT | FR, ALT | |
| Aoki et al. 40 (Phenotype: NICCD) | ||||
| 12 d.o | Antithrombin | aPTT, FIB | ||
| 28 d.o | Antithrombin, Factors II, IX | FIB, Factors V, VII, VIII, X, XI | aPTT, Factor XII | |
| 45 d.o | Antithrombin, aPTT | FIB, Factors II, V, VII, VIII, IX, X, XI | Factor XII | |
Abbreviations: α‐KG, alpha‐ketoglutarate; AACD, adolescent and adult citrin deficiency (formerly CTLN2); ACarn, acetylcarnitine; Ala, alanine; ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; Amy, amylase; Arg, arginine; aPTT, activated partial thromboplastin time; ASS, argininosuccinate synthetase; AST, aspartate aminotransferase; BHB, β‐hydroxybutyrate; BUN, blood urea nitrogen; Carn, carnitine; CD, citrin deficiency; ChE, cholinesterase; Citr, citrate; Cit, citrulline; Cr, creatinine; D. bil, direct bilirubin; FBG, fasting blood glucose; Fe, iron; FFA, free fatty acids; FI, fasting insulin: FIB, fibrinogen; FR, fischer ratio; Fum, fumarate; Gln, glutamine; GGT, gamma‐glutamyl transpeptidase; Glu, glutamate; GS, glutamine synthetase; HE, hpatic encephalopathy; Hb, hemoglobin; HbA1c, hemoglobin A1c; HDL‐C, high‐density lipoprotein cholesterol; HYA, hyaluronic acid; ICitr, isocitrate; L/P, lactate to pyruvate ratio; Lac, lactate; LDH, lactate dehydrogenase; Mal, malate; MCT, medium chain triglycerides; Met, methionine; Na+, sodium ions; NaPy, sodium pyruvate; NICCD, neonatal intrahepatic cholestasis caused by citrin deficiency; NH3, ammonia; OGTT, oral glucose tolerance test; PSTI, pancreatic secretory trypsin inhibitor; PLT, platelet count; Pyr, pyruvate; RBC, red blood cell count; Suc, succinate; T/S, threonine to serine ratio; TG, triglycerides; T. bil, total bilirubin; T. chol, total cholesterol; TBA, total bile acid; TCA, tricarboxylic acid; Thr, threonine; TIBC, total iron‐binding capacity; TP, total protein; Tyr, tyrosine; UA, uric acid; WBC, white blood cell count.
Based on reference ranges provided in citations.
A total of 151 publications that specifically mention the use of MCT to treat any form of CD were identified of which six manuscripts were included in the below literature review. 38 , 40 , 42 , 43 , 51 , 52 The included studies were all case reports and originated from Japan, with however low homogeneity concerning CD phenotypes, MCT dosage and duration, and outcome measures. Therefore, a meta‐analysis on the identified studies was not possible and instead the data was summarized and described together.
2.2.1. Effects of MCT in NICCD
MCT remains to be the most widely administered supplement for NICCD. A recent nationwide study in Japan showed that 36 out of 40 NICCD patients were treated with MCT. 3 Considering that NICCD patients often present with cholestasis, MCT may be particularly effective as a nutritional supplement compared to other forms of fat since it is easily digested under cholestatic conditions to provide energy to the liver.
A single study in four NICCD infants showed that treatment with lactose‐restricted formula supplemented with MCT normalized most abnormal laboratory findings along with increased body weights and the resolution of cholestasis by the end of the treatment period in all patients (Table S1). 43 Patients 1 to 3 (all siblings), were given MCT‐containing formula, while patient 4 was given a lactose‐free formula mixed with MCT oil due to complications associated with galactosemia. 43
The second included study described three NICCD patients who presented with coagulopathies. One patient received a lactose‐free formula supplemented with MCT oil while the remaining two patients received MCT formulas. It should be noted, however, that details of MCT dosage were not provided. The authors reported that all three patients saw improvements in their coagulation factors after receiving nutritional therapy, with most laboratory values returning to normal ranges. 40
Collectively, these preliminary results suggest that MCT treatment can significantly improve and normalize most laboratory parameters and resolve clinical complications associated with NICCD.
2.2.2. Effects of MCT in the silent period
MCT treatment was examined in two siblings in the silent period who frequently experienced hypoglycemic attacks and complained of fatigue. After 4 months of MCT treatment, fatigue and fasting blood glucose levels in both siblings improved and no hypoglycemia was noted after an oral glucose tolerance test (OGTT). 52 One patient (brother) exhibited elevations in postprandial blood ammonia levels that were normalized after MCT therapy. Postprandial lactate‐to‐pyruvate (L/P) ratios of both siblings were elevated before treatment but normalized after MCT therapy. 52 Taken together, these findings suggest that MCT may also have potential in improving clinical symptoms associated with the silent period or FTTDCD, but it should be noted that there are no detailed data on the long‐term use of MCT in these CD phenotypes.
2.2.3. Effects of MCT in adolescent and adult CD
In a cohort of 11 adult CD patients, nine patients who initially presented with HE, hyperammonemia, and disturbed consciousness recovered from encephalopathy after receiving MCT therapy, with five patients regaining consciousness within 1 to 3 days. 38 , 42 , 51 Of this cohort, five were also reported to be free of encephalopathic relapse, one chose to undergo liver transplantation, 38 and one experienced a relapse in HE although this was attributed to work‐related stress 38 (Table S2). Most biochemical parameters, including transaminases, improved in all 11 patients after MCT treatment. Ammonia levels were normalized in nine out of 11 patients after treatment, but were not normalized in the remaining two patients, who had considerably higher baseline ammonia levels. While plasma citrulline levels decreased in all 11 patients following MCT treatment, they did not fully normalize in most patients (9/11) but remained elevated, possibly due to a decrease in the amount of ASS protein and enzymatic activity. 38
Fatty liver, commonly observed in CD, is postulated to be a consequence of hepatic PPARα downregulation. 17 , 53 , 54 MCT supplementation in animals has indicated both beneficial and harmful outcomes on liver steatosis. 55 , 56 Yet, successful resolution of fatty liver was observed in 2 CD patients under long‐term MCT treatment. 42 The authors postulate that MCT can promote lipogenesis through the malate‐citrate shuttle, leading to PPARα upregulation. 50
2.2.4. Discussion and conclusions concerning MCT as treatment for CD
To the best of our knowledge, this work represents the first literature review of the potential efficacy of MCT with the present results suggesting improvements in clinical symptoms in all CD phenotypes. However, it is important to note that the above reports are based on a relatively small sample size without controls, and some biochemical parameters did not significantly improve following MCT treatment. It is also unclear whether MCT alone is effective in treating or even preventing onset of pathology in adolescent and adult CD patients. While optimal doses for MCT remain to be established for each phenotype, a recent publication has provided suggested dosages based on expert opinion. 41 Given the biochemical rationale of MCT and its potential efficacy for CD, further studies are urgently needed to systematically investigate its use in CD and to identify any potential long‐term side effects. Finally, the obvious inability of MCT to cure all metabolic abnormalities in CD highlights the need to consider additional modalities such as those discussed in the following sections.
2.3. Sodium pyruvate
Sodium pyruvate (NaPy) has been suggested as a treatment for CD due to its potential ability to alleviate redox imbalance by stimulating gluconeogenesis and to serve as an energy source. 37 , 57 Initial preclinical studies on the citrin‐KO mouse model demonstrated that liver perfusion of NaPy was effective in improving hepatic L/P ratios in a dose dependent manner and enhanced ureagenesis from exogenously administered ammonia but without reducing elevated plasma citrulline levels. 57
The first clinical study followed 2 years later when NaPy was administered to a 13‐year‐old patient who presented with low BMI, fatigue, and postprandial lethargy without hyperammonemia. Laboratory findings revealed significantly elevated plasma citrulline, pancreatic secretory trypsin inhibitor (PSTI), HDL‐cholesterol, and Thr/Ser ratio. NaPy therapy was initiated at 177 mg/kg/day with arginine and the patient was monitored periodically for up to 3 years. 37 Improvements in BMI, food intake, general fatigue, and postprandial lethargy were noted within 6 months of therapy, however, NaPy had limited efficacy at improving biochemical parameters. PSTI levels were normalized in the patient, but abnormal plasma amino acids (Cit, Arg, Gln, Thr/Ser ratio), and HDL‐cholesterol levels persisted. The authors concluded that NaPy therapy was effective in the patient. 37
A further survey of the current literature on the specific use of NaPy in CD identified four case reports in adult CD patients, one nonrandomized study in a cohort of 10 children in the silent period (Table 3) and no studies of NaPy use in NICCD patients. 34 , 37 , 44 , 45 , 58
TABLE 3.
Summary of biochemical findings in CD patients from clinical reports using sodium pyruvate as a treatment.
| Patient age (years) | NaPy dose (g/day)/duration (days) | Biochemical findings a | |||
|---|---|---|---|---|---|
| Improved but not normalized | Normalized | Unchanged | Worsened | ||
| Mutoh et al. 37 (Phenotype: AACD) | |||||
| 13 | 6/1095 | T/S | TG, PSTI | T. bil, AST, ALT, Amy, T. chol, HDL‐C, BUN, UA, NH3, TP, Lac, Pyr, Na+, Cit, Ala, FR | Arg, Gln |
| Yazaki et al. 34 (Phenotype: AACD) | |||||
| 34 | 3–15/77 | Exact values not provided (results in graph format) | |||
| 41 | 3/23 | ||||
| Yazaki et al. 58 (Phenotype: AACD) | |||||
| 72 | 3–9/121 | ALB, Arg, FR | RBC, Hb, TP, T. chol, ALP, GGT, Amy, PSTI | WBC, PLT, Cr, TG, ALT, LDH, T. bil, prothrombin time, activated partial prothromboplasmin time, NH3 | BUN, AST, Cit |
| Kogure et al. 44 (Phenotype: AACD) | |||||
| 68 | 4.5–8/1825 | Cit | NH3, PSTI | ||
| Nagasaka et al. 45 (Phenotype: silent period) b | |||||
| 3–13 | 300 c /91–182 | Carn | Citr, ICitr, ɑ‐KG, ACarn, BHB, FFA, FBG, HbA1c, Lac, Pyr, L/P | Suc, Fum, Mal, FI | |
Abbreviations: α‐KG, alpha‐ketoglutarate; AACD, adolescent and adult citrin deficiency (formerly CTLN2); ACarn, acetylcarnitine; Ala, alanine; ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; Amy, amylase; Arg, arginine; aPTT, activated partial thromboplastin time; ASS, argininosuccinate synthetase; AST, aspartate aminotransferase; BHB, β‐hydroxybutyrate; BUN, blood urea nitrogen; Carn, carnitine; CD, citrin deficiency; ChE, cholinesterase; Citr, citrate; Cit, citrulline; Cr, creatinine; D. bil, direct bilirubin; FBG, fasting blood glucose; Fe, iron; FFA, free fatty acids; FI, fasting insulin: FIB, fibrinogen; FR, fischer ratio; Fum, fumarate; Gln, glutamine; GGT, gamma‐glutamyl transpeptidase; Glu, glutamate; GS, glutamine synthetase; HE, hpatic encephalopathy; Hb, hemoglobin; HbA1c, hemoglobin A1c; HDL‐C, high‐density lipoprotein cholesterol; HYA, hyaluronic acid; ICitr, isocitrate; L/P, lactate to pyruvate ratio; Lac, lactate; LDH, lactate dehydrogenase; Mal, malate; MCT, medium chain triglycerides; Met, methionine; Na+, sodium ions; NaPy, sodium pyruvate; NICCD, neonatal intrahepatic cholestasis caused by citrin deficiency; NH3, ammonia; OGTT, oral glucose tolerance test; PSTI, pancreatic secretory trypsin inhibitor; PLT, platelet count; Pyr, pyruvate; RBC, red blood cell count; Suc, succinate; T/S, threonine to serine ratio; TG, triglycerides; T. bil, total bilirubin; T. chol, total cholesterol; TBA, total bile acid; TCA, tricarboxylic acid; Thr, threonine; TIBC, total iron‐binding capacity; TP, total protein; Tyr, tyrosine; UA, uric acid; WBC, white blood cell count.
Based on reference ranges provided in citations.
Reference ranges not provided. Data are compared against control values obtained from healthy volunteers.
Units in mg/kg/day.
Reports on the effectiveness of NaPy in adult CD patients provided mixed results. In two patients, NaPy treatment in conjunction with dietary management was not effective in reducing the frequency of encephalopathic attacks nor did it improve hyperammonemia. Eventually, both patients underwent successful liver transplantation. 34 In another report, a 73‐year‐old CD patient was treated with NaPy for 4 months and while PSTI and Fischer ratios were normalized, the majority of laboratory findings were not improved or even worsened. 58 Long‐term NaPy treatment in another adult CD patient normalized blood ammonia levels and there was no recurrence of encephalopathy although plasma citrulline and PSTI levels remained elevated. 44
A nonrandomized clinical study investigating the effects of NaPy therapy on TCA metabolites in 10 CD children showed no corrections in metabolite profiles including plasma L/P ratios after 3 to 6 months of therapy. 45 As the clinical symptoms of the patients were not detailed, it is unknown whether NaPy led to any functional improvements.
Although studies are limited, the collective findings suggest limited efficacy of NaPy in CD. Most biochemical results were not significantly improved by NaPy and recurrent encephalopathic attacks remained. 34 , 58 While initial preclinical findings were promising, 57 the limited efficacy seen in clinical studies could be due to poor oral bioavailability of NaPy. Because in mouse studies NaPy was administered through liver perfusion techniques, 57 the contributions of the human GI tract, where NaPy is potentially decarboxylated, were not accounted for. 46 In healthy adults, orally ingested NaPy even at a relatively high dose of 25 g failed to increase whole blood or plasma pyruvate concentrations, which may be due to rapid intestinal decarboxylation of NaPy, likely through fermentation by gut bacteria, 46 and/or uptake and metabolism by skeletal muscle. 59 The development of new formulations of pyruvate with enhanced oral bioavailability may be helpful in this regard.
Although both MCT and NaPy aim to address the same fundamental defects associated with the condition, current findings from limited studies suggest that MCT may be more effective. Another important practical consideration worth mentioning is that NaPy is not an approved drug, while MCT is a widely available nutraceutical and therefore easily accessible.
2.4. Ursodeoxycholic acid (UDCA)
Cholestasis is a commonly observed symptom associated with NICCD 10 and while the cause of this is not fully understood, it has been suggested that the impaired energy production caused by CD leads to a reduction in the production and secretion of bile acids. 33 UDCA is a mainstay treatment for cholestatic liver diseases including cholestasis associated with NICCD by improving bile acid biosynthesis and bile flow. 3 , 10 , 60 , 61 UDCA has also been reported to have properties that may protect hepatocytes against bile acid‐induced cytotoxicity. 62 Although mainly indicated for NICCD, the use of UDCA in post‐NICCD patients has also been reported in the literature, 3 , 52 , 63 but there is a lack of systematic studies into the use of UDCA in CD.
2.5. Nitrogen scavengers
Nitrogen scavengers such as benzoate, phenylacetate, and phenylbutyrate are first line therapies in the management of hyperammonemia in UCDs. 47 This class of drugs bypass the urea cycle to provide an alternative pathway for nitrogen excretion by conjugating with nitrogen bearing amino acids such as glycine or glutamine and are later excreted in urine. 3 , 47 , 64 , 65 Nitrogen scavengers are most often used for adult CD patients to manage elevated ammonia levels, 3 , 66 but there are no systematic studies in CD available.
2.6. l‐arginine
l‐arginine is an intermediary metabolite of the urea cycle used to clinically manage hyperammonemia in UCDs (except for arginase 1 deficiency) and is often administered together with nitrogen scavengers. 65 , 67 l‐arginine lowers ammonia levels by enhancing ureagenesis. 68 Its use and effectiveness in CD were first reported in a 37‐year‐old patient where arginine granules were effective in lowering and maintaining normal blood ammonia levels. When arginine therapy was temporarily discontinued in this patient, ammonia levels increased significantly, and the patient fell into a coma. 36 However, in contrast to most UCDs (except for arginase 1 deficiency), plasma arginine levels in CD are often elevated, questioning the role of arginine supplementation in this condition. There are no systematic studies available, but a thorough biochemical study would be of interest to understand the mechanism of action of arginine therapy in CD.
2.7. Liver transplantation
At present, liver transplantation remains the only curative treatment for CD and is typically performed in adult patients with severe symptoms who are unresponsive to other treatments, 15 , 19 , 69 and rarely in NICCD patients. 24 , 27 Post‐transplantation, laboratory results and clinical symptoms of patients were reported to normalize. Interestingly, the unique food preferences of patients also disappeared after liver transplantation and they no longer require diet restrictions. 34 , 69 , 70 , 71 These drastic improvements also highlight that CD primarily affects the liver. Despite these positive outcomes, significant drawbacks concerning liver transplantation remain such as lifelong immunosuppression and associated risks, high costs, and shortage of suitable donors.
2.8. General discussion on the current treatment landscape
The present overview demonstrates that research on existing therapies for CD remains at a relatively nascent stage. In order to advance the establishment and development of effective therapies for CD, several outstanding issues need to be addressed first.
Efficiently characterizing and organizing large patient cohorts with CD is vital for expediting therapy evaluation and development, as it facilitates insights into the disease's history, streamlines patient recruitment, and reduces study costs.
Furthermore, identification of CD‐specific biomarkers and establishment of informative functional tests in patients are needed to reliably assess the efficacy of therapies in clinical trials. Current biochemical parameters such as plasma amino acids, liver function markers, and ammonia levels are nonspecific for CD and may fluctuate greatly depending on the time of sampling. A potential functional test for CD is to examine the ureagenesis function of patients across phenotypes, as this may be a good indicator of disease severity and progression and whether patients are at risk of developing the adult form of the disease. Recently, new methods have been developed that use low concentrations of stable isotope tracer coupled with more robust analytical methods that may be considered. 72
Finally, there is a need to develop suitable preclinical models of CD to facilitate preclinical studies on potential therapies. To date, only two animal models exist for CD, the citrin‐KO mouse that fails to display features of human CD, and the citrin/mitochondrial glycerol‐3‐phosphate dehydrogenase (mGPD) double‐KO mouse that demonstrates several features of the disease. 73 , 74 Also, mice express much higher hepatic levels of the citrin protein isoform, aralar, which may functionally compensate for the loss of citrin. 75 The development of other animal models, such as rat models, may be considered as rats are genetically and metabolically closer to humans compared to mice. 76 Cellular models of CD are limited and include a citrin‐KO cellular model in the HepG2 hepatoblastoma cell line 77 as well as hepatocyte‐like cells derived from CD‐induced pluripotent stem cells. 22 However, it has been shown that HepG2 cells lack the expression of several urea cycle enzymes, resulting in limitations for studies of the urea cycle, 78 , 79 while iPSC‐derived hepatocytes are generally immature and more closely resemble fetal rather than adult hepatocytes. 80
3. FUTURE THERAPEUTIC OUTLOOK
3.1. Restoring the redox balance
In vivo and in vitro models of CD demonstrate a disrupted NADH/NAD+ ratio as a result of impaired malate–aspartate shuttle activity. 73 , 74 , 77 This imbalance causes impairment of glycolysis and gluconeogenesis in the cytosol, while the shortage of NADH in mitochondria impairs the function of the TCA cycle and hence ATP production.
Redox balance can be restored by increasing the amount of NAD+ in the cytosol, either by providing a precursor molecule such as nicotinamide riboside (NR) 77 or by pharmacologically stimulating NAD+ synthesis. 81 The latter approach has been pursued using KL1333, a compound able to modulate NAD(P)H:quinone oxidoreductase towards enhanced NADH to NAD+ conversion. 81 KL1333 treatment was able to restore NAD+ and ATP levels and improve mitochondrial biogenesis of skin fibroblasts from a patient with mitochondrial encephalomyopathy, lactic acidosis and stroke‐like episodes. 81
Another study focused on HepG2 citrin‐knockout (KO) cells for assessing the potential effect of NR treatment. These cells exhibit several characteristics of CD, including increased ammonia accumulation, higher cytosolic NADH/NAD+ ratio and impaired glycolysis. 77 HepG2 citrin‐KO cells supplemented with NR successfully reduced the cytosolic NADH/NAD+ ratio, enhanced glycolysis and fatty acid oxidation, but did not improve ammonia detoxification, 77 highlighting the need for combination treatment for additionally targeting ammonia‐related CD manifestations. Taken together, these results highlight the importance and ability of NADH/NAD+ rebalancing approaches in establishing new treatments for CD.
3.2. Aspartate and other amino acids supplementation
The most serious consequence of adult CD is a condition resembling HE, 44 and it is speculated that in addition to the energy deficit in liver cells, insufficient cytosolic aspartate levels hamper urea cycle functionality. Therefore, restoration of aspartate availability could rationally aid ureagenesis function.
Amino acid supplementation (as well as MCT and NaPy) for restoring urea cycle function has been tested on citrin/mGPD double‐KO mice with induced hyperammonemia. 73 , 82 Combinations of either ornithine and alanine or ornithine and aspartate were equally effective in decreasing blood ammonia to levels similar to control animals. Furthermore, both treatment combinations increased hepatic aspartate and decreased hepatic citrulline concentrations by ASS protein activation. 82
For ameliorating HE and hyperammonemia in patients not affected by CD, supplementation with L‐ornithine‐L‐aspartate (LOLA) has been proposed and tested in a double‐blind randomized controlled trial as well as a randomized clinical trial. 83 , 84 , 85 Oral administration of LOLA to patients with stable, overt, chronic encephalopathy was beneficial at improving the extent and preventing further episodes of HE and in reducing blood ammonia concentrations. 83 , 84
Overall, LOLA and other amino acids supplementation show some promise in mitigating ammonia‐induced brain edema and have already undergone clinical trials for different liver conditions, 86 but further investigations of LOLA and other amino acids on broader aspects of CD are required.
3.3. Enhancing bioenergetics
Contributing to the energy deficit in CD, 32 citrin deficient hepatocytes struggle to utilize glucose due to impaired glycolysis or fatty acids due to reduced β‐oxidation rates attributed to PPARα downregulation. Additionally, it has been proposed that fatty liver observed in some cases of CD is the result of decreased β‐oxidation without the corresponding decrease in lipogenesis. 77 Therefore, stimulating β‐oxidation using e.g. L‐carnitine could increase mitochondrial NADH and ATP supply as well as reduce the buildup of excess hepatic fatty acids. L‐carnitine is a natural constituent of human cells 87 and plays an important role in fatty acid metabolism. 88 It acts as an obligatory cofactor for fatty acid oxidation and facilitates long‐chain fatty acid transport across the mitochondrial membranes, thereby promoting β‐oxidation and ATP generation. 88 , 89 Supplementation of L‐carnitine in the context of pulmonary arterial hypertension led to augmentation of fatty acid oxidation and lipolysis, and reduction of lipid accumulation in vitro. 90 Nevertheless, future clinical studies to rectify bioenergetic deficits in CD with compounds such as L‐carnitine would be needed for improving our understanding of the disease and its management.
3.4. Enhancing argininosuccinate synthetase and other urea cycle enzymes
Hyperammonemic episodes in CD are generally attributed to the lack of aspartate and, at least in the majority but not all of the respective patients, decreased ASS activity. 11 , 17 , 91 While cytosolic aspartate deficiency is a direct consequence of a defective citrin protein, the reason for decreased ASS function remains unclear, and requires additional studies for a correct and complete understanding of the origin of hyperammonemia in CD. Several FDA‐approved small molecules are shown to upregulate ASS expression or enhance its activity, namely fisetin and spinosyn A. 92 Specifically for fisetin, which is a flavonoid molecule commonly found in fruits and vegetables, it has been proposed to upregulate both ASS and argininosuccinate lyase (ASL) protein expression in various cancer cell lines and in vivo in mice enterocytes. 92 Furthermore, one study has demonstrated that spinosyn A, component of the commonly‐used pesticide spinosad with low toxicity in mammals, 93 , 94 , 95 and its derivative can specifically bind to ASS protein and dose‐dependently enhance its catalytic activity. 95 Therefore, possible analogous effects mediated by fisetin or spinosyn A could potentially restore urea cycle function in CD hepatocytes both in vitro and in vivo. While these molecules have not yet been tested in relevant CD models, they present an intriguing possibility for alleviating hyperammonemia in CD patients.
3.5. Gene therapy
Gene therapy has been shown to represent a valuable and promising approach for treating various diseases including inherited metabolic liver disorders such as Wilson's disease and UCDs, e.g. ornithine transcarbamylase (OTC) and ASS deficiency. 96 , 97 , 98 By directly targeting the underlying genetic abnormality, gene therapy has the potential to provide long‐lasting or even permanent solutions to conditions without previous successful treatment options. Gene therapy holds tremendous potential for treating metabolic liver diseases including CD, since it may be performed in a carefully planned approach during the silent period, hereby preventing progression of liver disease and the onset of adolescent or adult CD. Below, we describe established or currently planned approaches towards this goal.
3.6. mRNA‐based gene therapy approaches
mRNA‐based therapies have emerged as a recent and innovative approach in gene therapy that showed promising results in the treatment of inborn errors of liver metabolism, including UCDs. 99 , 100 , 101 , 102 , 103 mRNA molecules, commonly delivered intravenously via lipid nanoparticles are largely taken up by the hepatocytes, where they use the cell's own translational and transport machinery, bypassing the need for exogenous enzyme production. 99 , 104 Furthermore, mRNA therapy does not integrate into the genome, mitigating the risk of insertional mutagenesis, however, due to their short lifespan in cells, regular mRNA infusions are required. 100 , 101
Notably, a breakthrough in mRNA therapy for CD was achieved using codon‐optimized mRNA encoding the human citrin protein, which was encapsulated in lipid nanoparticles that were administered intravenously to citrin‐KO and double KO (ctrn/mGPD‐dKO) mice. 73 , 74 , 105 The goal of this study was to increase hepatic citrin levels and restore metabolic balance. Since the majority of disease‐causing citrin mutations lead to a complete absence of citrin protein, 2 , 106 even a partial restoration of citrin protein can result in clinical improvement. This was in fact achieved by restoration of citrin levels to 2% to 5% of wild‐type within 24 h post‐injection in the citrin‐KO mice, when plasma citrulline and ammonia were effectively reduced to near physiological levels. 105
Further validation on the safety of this approach and its effect on the different symptoms of CD such as on fatty acid accumulation or on ASS protein levels and activity are yet to be determined but mRNA therapy holds promise for alleviating the disease.
3.7. DNA‐based gene therapy approaches
In DNA gene therapy, a functional copy of the defective gene is introduced into the patient's cells, allowing for the correction or modification of genetic defects at the DNA level. Correction of UCDs via liver‐directed gene therapy has been reported in mouse models of OTC, 107 ASS1, 97 ASL, 108 CPS1 109 and ARG1 deficiency. 110 While gene therapy faces challenges related to immune responses triggered by the recognition of an exogenous sequence as a foreign entity, an approach that provides cells with a similar sequence to the endogenous one may help circumvent these immune responses. In the case of CD, aralar, which shares approximately 80% identity with citrin and exhibits similar transport properties, emerges as a suitable candidate for gene therapy due to its expression in various cell types, including certain liver cells, namely Kupffer cells. 111
Recently, the possibility of introducing an exogenous aralar sequence in the hepatocytes of citrin‐KO mice was explored as a potential therapeutic strategy. 74 The introduction of an exogenous aralar sequence substantially restored the function of the malate–aspartate shuttle in CD liver mitochondria and balanced NADH/NAD+ levels in hepatocytes isolated from citrin‐KO mice. 75 Furthermore, it has been reported that citrin is around 8 times more abundant than aralar in mice liver while in human liver citrin is 400 times more abundant, 75 suggesting that aralar therapy could be clinically beneficial. It is important to note that overexpression of aspartate/glutamate carriers (AGCs) has been connected with different cancer types, including hepatocellular carcinoma, 112 , 113 therefore, providing exogenous AGC to patients has to be finely tuned. It is also unclear whether there will be potential immunological responses against wild‐type citrin protein given citrin's intracellular localization in the inner mitochondrial membrane and the fact that its isoform aralar is being endogenously expressed. 75 Hence, gene editing of the defective citrin gene should also be attempted. While the effect of exogenous citrin or aralar expression on hallmarks of CD such as hyperammonemia and fatty liver is yet to be determined, gene therapy holds potential as the future of CD treatment.
4. CONCLUSIONS
Since the discovery of SLC25A13 as the causative gene for CD 25 years ago, advancement in the understanding of CD and developments in dietary and therapeutic interventions have enhanced the quality of life in affected patients. Among these therapies, the present review highlights MCT as the most promising therapeutic candidate for all CD phenotypes given its biochemical rationale. However, only few additional effective therapeutic options exist for CD and there is still no definite and curative treatment besides liver transplantation. Large opportunities remain in the research and development of new effective therapies, from small molecules that can alleviate the impact of the condition to a potential cure such as gene therapies to express a functional copy of the citrin protein. There is also the need to sufficiently characterize and organize large patient cohorts for natural history studies and clinical trials, to establish effective biomarkers and functional tests, as well as to generate suitable preclinical models in order to advance the development of treatments for CD.
AUTHOR CONTRIBUTIONS
Toni Vuković and Li Eon Kuek contributed equally to this manuscript.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflicts of interest.
ETHICS STATEMENT
This study is a review paper, which does not require approval by any Institutional Review Board (IRB).
Supporting information
Table S1. Clinical data of NICCD patients before and after MCT treatment. 43
Table S2. Clinical data of CD patients with AACDa before and after MCT treatment.
ACKNOWLEDGMENTS
The authors acknowledge the role of Citrin Foundation to achieve the goals described in the preceding concluding remarks. Work on urea cycle disorders at the University Children's Hospital Zurich is supported by the Swiss National Science Foundation (grants 320030_207965 and CRSII‐222794 to JH), the University Research Priority Program ITINERARE and by Citrin Foundation (Grant Reference number: RG22005). Open access funding provided by Universitat Zurich.
Vuković T, Kuek LE, Yu B, Makris G, Häberle J. The therapeutic landscape of citrin deficiency. J Inherit Metab Dis. 2024;47(6):1157‐1174. doi: 10.1002/jimd.12768
REFERENCES
- 1. Kobayashi K, Sinasac DS, Iijima M, et al. The Gene Mutated in Adult‐Onset Type II Citrullinaemia Encodes a Putative Mitochondrial Carrier Protein. 1999. http://www.genet.sickkids.on.ca/ [DOI] [PubMed]
- 2. Tavoulari S, Lacabanne D, Thangaratnarajah C, Kunji ERS. Pathogenic variants of the mitochondrial aspartate/glutamate carrier causing citrin deficiency. Trends Endocrinol Metab. 2022;33(8):539‐553. doi: 10.1016/j.tem.2022.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Kido J, Häberle J, Sugawara K, et al. Clinical manifestation and long‐term outcome of citrin deficiency: report from a nationwide study in Japan. J Inherit Metab Dis. 2022;45(3):431‐444. doi: 10.1002/jimd.12483 [DOI] [PubMed] [Google Scholar]
- 4. Saheki T, Kobayashi K. Mitochondrial aspartate glutamate carrier (citrin) deficiency as the cause of adult‐onset type II citrullinemia (CTLN2) and idiopathic neonatal hepatitis (NICCD). J Hum Genet. 2002;47(7):333‐341. doi: 10.1007/s100380200046 [DOI] [PubMed] [Google Scholar]
- 5. Saheki T, Inoue K, Tushima A, Mutoh K, Kobayashi K. Citrin deficiency and current treatment concepts. Mol Genet Metab. 2010;100:S59‐S64. doi: 10.1016/j.ymgme.2010.02.014 [DOI] [PubMed] [Google Scholar]
- 6. Song YZ. Genotypic and phenotypic features of citrin deficiency: five‐year experience in a Chinese pediatric center. Int J Mol Med. 2011;28:33‐40. doi: 10.3892/ijmm.2011.653 [DOI] [PubMed] [Google Scholar]
- 7. Palmieri L, Pardo B, Lasorsa FM, et al. Citrin and aralar1 are Ca2+−stimulated aspartate/glutamate transporters in mitochondria. EMBO J. 2001;20(18):5060‐5069. doi: 10.1093/emboj/20.18.5060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. del Arco A, Satrústegui J. Molecular cloning of Aralar, a new member of the mitochondrial carrier superfamily that binds calcium and is present in human muscle and brain. J Biol Chem. 1998;273(36):23327‐23334. doi: 10.1074/jbc.273.36.23327 [DOI] [PubMed] [Google Scholar]
- 9. Zhang X, Yang S, Chen J, Su Z. Unraveling the regulation of hepatic gluconeogenesis. Front Endocrinol (Lausanne). 2019;9:9. doi: 10.3389/fendo.2018.00802 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Ohura T, Kobayashi K, Tazawa Y, et al. Clinical pictures of 75 patients with neonatal intrahepatic cholestasis caused by citrin deficiency (NICCD). J Inherit Metab Dis. 2007;30(2):139‐144. doi: 10.1007/s10545-007-0506-1 [DOI] [PubMed] [Google Scholar]
- 11. Dimmock D, Maranda B, Dionisi‐Vici C, et al. Citrin deficiency, a perplexing global disorder. Mol Genet Metab. 2009;96(1):44‐49. doi: 10.1016/j.ymgme.2008.10.007 [DOI] [PubMed] [Google Scholar]
- 12. Treepongkaruna S, Jitraruch S, Kodcharin P, et al. Neonatal intrahepatic cholestasis caused by citrin deficiency: prevalence and SLC25A13mutations among thai infants. BMC Gastroenterol. 2012;12(1):141. doi: 10.1186/1471-230X-12-141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Arai‐Ichinoi N, Kikuchi A, Wada Y, Sakamoto O, Kure S. Hypoglycemic attacks and growth failure are the most common manifestations of citrin deficiency after 1 year of age. J Inherit Metab Dis. 2021;44(4):838‐846. doi: 10.1002/jimd.12390 [DOI] [PubMed] [Google Scholar]
- 14. Sullivan LB, Gui DY, Hosios AM, Bush LN, Freinkman E, Vander Heiden MG. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell. 2015;162(3):552‐563. doi: 10.1016/j.cell.2015.07.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Saheki T, Moriyama M, Funahashi A, Kuroda E. AGC2 (Citrin) deficiency—from recognition of the disease till construction of therapeutic procedures. Biomol Ther. 2020;10(8):1100. doi: 10.3390/biom10081100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Kobayashi K, Shaheen N, Kumashiro R, et al. A search for the primary abnormality in adult‐onset type II citrullinemia. Am J Hum Genet. 1993;53(5):1024‐1030. [PMC free article] [PubMed] [Google Scholar]
- 17. Komatsu M, Yazaki M, Tanaka N, et al. Citrin deficiency as a cause of chronic liver disorder mimicking non‐alcoholic fatty liver disease. J Hepatol. 2008;49(5):810‐820. doi: 10.1016/j.jhep.2008.05.016 [DOI] [PubMed] [Google Scholar]
- 18. Kido J, Makris G, Santra S, Häberle J. Clinical landscape of citrin deficiency: a global perspective on a multifaceted condition. J Inherit Metab Dis Published online March 19, 2024. doi: 10.1002/jimd.12722 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Ikeda S. Type II (adult onset) citrullinaemia: clinical pictures and the therapeutic effect of liver transplantation. J Neurol Neurosurg Psychiatry. 2001;71(5):663‐670. doi: 10.1136/jnnp.71.5.663 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Ohura T, Kobayashi K, Abukawa D, et al. A novel inborn error of metabolism detected by elevated methionine and/or galactose in newborn screening: neonatal intrahepatic cholestasis caused by citrin deficiency. Eur J Pediatr. 2003;162(5):317‐322. doi: 10.1007/s00431-003-1171-5 [DOI] [PubMed] [Google Scholar]
- 21. Komatsu M, Tanaka N, Kimura T, Yazaki M. Citrin deficiency: clinical and nutritional features. Nutrients. 2023;15(10):2284. doi: 10.3390/nu15102284 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Kim Y, Choi JY, Lee SH, Lee BH, Yoo HW, Han YM. Malfunction in mitochondrial β‐oxidation contributes to lipid accumulation in hepatocyte‐like cells derived from Citrin deficiency‐induced pluripotent stem cells. Stem Cells Dev. 2016;25(8):636‐647. doi: 10.1089/scd.2015.0342 [DOI] [PubMed] [Google Scholar]
- 23. Tanaka N, Moriya K, Kiyosawa K, Koike K, Gonzalez FJ, Aoyama T. PPARα activation is essential for HCV core protein–induced hepatic steatosis and hepatocellular carcinoma in mice. J Clin Investig. 2008;118(2):683‐694. doi: 10.1172/JCI33594 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Tamamori A, Okano Y, Ozaki H, et al. Neonatal intrahepatic cholestasis caused by citrin deficiency: severe hepatic dysfunction in an infant requiring liver transplantation. Eur J Pediatr. 2002;161(11):609‐613. doi: 10.1007/s00431-002-1045-2 [DOI] [PubMed] [Google Scholar]
- 25. Tazawa Y, Kobayashi K, Abukawa D, et al. Clinical heterogeneity of neonatal intrahepatic cholestasis caused by citrin deficiency: case reports from 16 patients. Mol Genet Metab. 2004;83(3):213‐219. doi: 10.1016/j.ymgme.2004.06.018 [DOI] [PubMed] [Google Scholar]
- 26. Tamamori A, Fujimoto A, Okano Y, et al. Effects of Citrin deficiency in the perinatal period: feasibility of newborn mass screening for Citrin deficiency. Pediatr Res. 2004;56(4):608‐614. doi: 10.1203/01.PDR.0000139713.64264.BC [DOI] [PubMed] [Google Scholar]
- 27. Shigeta T, Kasahara M, Kimura T, et al. Liver transplantation for an infant with neonatal intrahepatic cholestasis caused by citrin deficiency using heterozygote living donor. Pediatr Transplant. 2010;14(7):E86‐E88. doi: 10.1111/j.1399-3046.2009.01172.x [DOI] [PubMed] [Google Scholar]
- 28. Chew HB, Ngu LH, Zabedah MY, et al. Neonatal intrahepatic cholestasis associated with citrin deficiency (NICCD): a case series of 11 Malaysian patients. J Inherit Metab Dis. 2010;33(S3):489‐495. doi: 10.1007/s10545-010-9248-6 [DOI] [PubMed] [Google Scholar]
- 29. Zhang MH, Gong JY, Wang JS. Citrin deficiency presenting as acute liver failure in an eight‐month‐old infant. World J Gastroenterol. 2015;21(23):7331‐7334. doi: 10.3748/wjg.v21.i23.7331 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Pinto A, Ashmore C, Batzios S, et al. Dietary management, clinical status and outcome of patients with Citrin deficiency in the UK. Nutrients. 2020;12(11):3313. doi: 10.3390/nu12113313 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Kobayashi K. Screening of nine SLC25A13 mutations: their frequency in patients with citrin deficiency and high carrier rates in Asian populations. Mol Genet Metab. 2003;80(3):356‐359. doi: 10.1016/S1096-7192(03)00140-9 [DOI] [PubMed] [Google Scholar]
- 32. Hayasaka K, Numakura C. Adult‐onset type II citrullinemia: current insights and therapy. Appl Clin Genet. 2018;11:163‐170. doi: 10.2147/TACG.S162084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Okano Y, Ohura T, Sakamoto O, Inui A. Current treatment for citrin deficiency during NICCD and adaptation/compensation stages: strategy to prevent CTLN2. Mol Genet Metab. 2019;127(3):175‐183. doi: 10.1016/j.ymgme.2019.06.004 [DOI] [PubMed] [Google Scholar]
- 34. Yazaki M, Hineno A, Matsushima A, et al. First two cases of adult‐onset type II citrullinemia successfully treated by deceased‐donor liver transplantation in Japan. Hepatol Res. 2012;42(9):934‐939. doi: 10.1111/j.1872-034X.2012.00992.x [DOI] [PubMed] [Google Scholar]
- 35. Chen P, Gao X, Chen B, Zhang Y. Adult‐onset citrullinaemia type II with liver cirrhosis: a rare cause of hyperammonaemia. Open Med. 2021;16(1):455‐458. doi: 10.1515/med-2021-0235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Imamura Y. Effectiveness of carbohydrate‐restricted diet and arginine granules therapy for adult‐onset type II citrullinemia: a case report of siblings showing homozygous SLC25A13 mutation with and without the disease. Hepatol Res. 2003;26(1):68‐72. doi: 10.1016/S1386-6346(02)00331-5 [DOI] [PubMed] [Google Scholar]
- 37. Mutoh K, Kurokawa K, Kobayashi K, Saheki T. Treatment of a citrin‐deficient patient at the early stage of adult‐onset type II citrullinaemia with arginine and sodium pyruvate. J Inherit Metab Dis. 2008;31(S2):343‐347. doi: 10.1007/s10545-008-0914-x [DOI] [PubMed] [Google Scholar]
- 38. Hayasaka K, Numakura C, Yamakawa M, et al. Medium‐chain triglycerides supplement therapy with a low‐carbohydrate formula can supply energy and enhance ammonia detoxification in the hepatocytes of patients with adult–onset type II citrullinemia. J Inherit Metab Dis. 2018;41(5):777‐784. doi: 10.1007/s10545-018-0176-1 [DOI] [PubMed] [Google Scholar]
- 39. Nakamura M, Yazaki M, Kobayashi Y, et al. The characteristics of food intake in patients with Type II Citrullinemia. J Nutr Sci Vitaminol (Tokyo). 2011;57(3):239‐245. doi: 10.3177/jnsv.57.239 [DOI] [PubMed] [Google Scholar]
- 40. Aoki H, Ogiwara K, Hasegawa M, Nogami K. Hemostatic rebalance in neonatal intrahepatic cholestasis with citrin deficiency. Pediatr Int. 2022;64(1):e14741. doi: 10.1111/ped.14741 [DOI] [PubMed] [Google Scholar]
- 41. Hayasaka K. Pathogenesis and management of citrin deficiency. Intern Med. 2023;2595‐23. doi: 10.2169/internalmedicine.2595-23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Hayasaka K, Numakura C, Toyota K, et al. Medium‐chain triglyceride supplementation under a low‐carbohydrate formula is a promising therapy for adult‐onset type II citrullinemia. Mol Genet Metab Rep. 2014;1:42‐50. doi: 10.1016/j.ymgmr.2013.12.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Hayasaka K, Numakura C, Toyota K, Kimura T. Treatment with lactose (galactose)‐restricted and medium‐chain triglyceride‐supplemented formula for neonatal intrahepatic cholestasis caused by citrin deficiency. JIMD Rep. 2011;2:37‐44. doi: 10.1007/8904_2011_42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Kogure T, Kondo Y, Kakazu E, et al. Three cases of adult‐onset type II citrullinemia treated with different therapies: efficacy of sodium pyruvate and low‐carbohydrate diet. Hepatol Res. 2014;44(6):707‐712. doi: 10.1111/hepr.12170 [DOI] [PubMed] [Google Scholar]
- 45. Nagasaka H, Komatsu H, Inui A, et al. Circulating tricarboxylic acid cycle metabolite levels in citrin‐deficient children with metabolic adaptation, with and without sodium pyruvate treatment. Mol Genet Metab. 2017;120(3):207‐212. doi: 10.1016/j.ymgme.2016.12.011 [DOI] [PubMed] [Google Scholar]
- 46. Oliphant K, Allen‐Vercoe E. Macronutrient metabolism by the human gut microbiome: major fermentation by‐products and their impact on host health. Microbiome. 2019;7(1):91. doi: 10.1186/s40168-019-0704-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Häberle J, Burlina A, Chakrapani A, et al. Suggested guidelines for the diagnosis and management of urea cycle disorders: first revision. J Inherit Metab Dis. 2019;42(6):1192‐1230. doi: 10.1002/jimd.12100 [DOI] [PubMed] [Google Scholar]
- 48. Fukushima K, Yazaki M, Nakamura M, et al. Conventional diet therapy for Hyperammonemia is risky in the treatment of hepatic encephalopathy associated with Citrin deficiency. Intern Med. 2010;49(3):243‐247. doi: 10.2169/internalmedicine.49.2712 [DOI] [PubMed] [Google Scholar]
- 49. Okano Y, Okamoto M, Yazaki M, et al. Analysis of daily energy, protein, fat, and carbohydrate intake in citrin‐deficient patients: towards prevention of adult‐onset type II citrullinemia. Mol Genet Metab. 2021;133(1):63‐70. doi: 10.1016/j.ymgme.2021.03.004 [DOI] [PubMed] [Google Scholar]
- 50. Hayasaka K. Metabolic basis and treatment of citrin deficiency. J Inherit Metab Dis. 2021;44(1):110‐117. doi: 10.1002/jimd.12294 [DOI] [PubMed] [Google Scholar]
- 51. Watanabe Y, Numakura C, Tahara T, et al. Diabetes mellitus exacerbates citrin deficiency via glucose toxicity. Diabetes Res Clin Pract. 2020;164:108159. doi: 10.1016/j.diabres.2020.108159 [DOI] [PubMed] [Google Scholar]
- 52. Otsuka H, Sasai H, Abdelkreem E, et al. Effectiveness of medium‐chain triglyceride oil therapy in two Japanese Citrin‐deficient siblings: evaluation using Oral glucose tolerance tests. Tohoku J Exp Med. 2016;240(4):323‐328. doi: 10.1620/tjem.240.323 [DOI] [PubMed] [Google Scholar]
- 53. Komatsu M, Kimura T, Yazaki M, et al. Steatogenesis in adult‐onset type II citrullinemia is associated with down‐regulation of PPARα. Biochim Biophys Acta. 2015;1852(3):473‐481. doi: 10.1016/j.bbadis.2014.12.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Song YZ, Yazaki M, Saheki T. Citrin deficiency. Human Pathobiochemistry. Springer; 2019:3‐14. doi: 10.1007/978-981-13-2977-7_1 [DOI] [Google Scholar]
- 55. Guimarães J, Bargut TCL, Mandarim‐de‐Lacerda CA, Aguila MB. Medium‐chain triglyceride reinforce the hepatic damage caused by fructose intake in mice. Prostaglandins Leukot Essent Fatty Acids. 2019;140:64‐71. doi: 10.1016/j.plefa.2018.11.005 [DOI] [PubMed] [Google Scholar]
- 56. Ronis MJJ, Baumgardner JN, Sharma N, et al. Medium chain triglycerides dose‐dependently prevent liver pathology in a rat model of non‐alcoholic fatty liver disease. Exp Biol Med. 2013;238(2):151‐162. doi: 10.1258/ebm.2012.012303 [DOI] [PubMed] [Google Scholar]
- 57. Moriyama M, Li MX, Kobayashi K, et al. Pyruvate ameliorates the defect in ureogenesis from ammonia in citrin‐deficient mice. J Hepatol. 2006;44(5):930‐938. doi: 10.1016/j.jhep.2005.09.018 [DOI] [PubMed] [Google Scholar]
- 58. Yazaki M, Kinoshita M, Ogawa S, et al. A 73‐year‐old patient with adult‐onset type II citrullinemia successfully treated by sodium pyruvate and arginine. Clin Neurol Neurosurg. 2013;115(8):1542‐1545. doi: 10.1016/j.clineuro.2012.12.027 [DOI] [PubMed] [Google Scholar]
- 59. Morrison MA, Spriet LL, Dyck DJ. Pyruvate ingestion for 7 days does not improve aerobic performance in well‐trained individuals. J Appl Physiol. 2000;89(2):549‐556. doi: 10.1152/jappl.2000.89.2.549 [DOI] [PubMed] [Google Scholar]
- 60. Huang L, Li S, Chen J, et al. Efficacy and safety of ursodeoxycholic acid in children with cholestasis: a systematic review and meta‐analysis. PLoS One. 2023;18(1):e0280691. doi: 10.1371/journal.pone.0280691 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Wang JY, Wu SD. Ursodeoxycholic acid for the treatment of liver diseases. Liver Pathophysiology. Elsevier; 2017:767‐779. doi: 10.1016/B978-0-12-804274-8.00055-2 [DOI] [Google Scholar]
- 62. Goossens JF, Bailly C. Ursodeoxycholic acid and cancer: from chemoprevention to chemotherapy. Pharmacol Ther. 2019;203:107396. doi: 10.1016/j.pharmthera.2019.107396 [DOI] [PubMed] [Google Scholar]
- 63. Tanaka N, Yazaki M, Kobayashi K. A lean man with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2007;5(3):A32. doi: 10.1016/j.cgh.2006.06.014 [DOI] [PubMed] [Google Scholar]
- 64. de Las HJ, Aldámiz‐Echevarría L, Martínez‐Chantar ML, Delgado TC. An update on the use of benzoate, phenylacetate and phenylbutyrate ammonia scavengers for interrogating and modifying liver nitrogen metabolism and its implications in urea cycle disorders and liver disease. Expert Opin Drug Metab Toxicol. 2017;13(4):439‐448. doi: 10.1080/17425255.2017.1262843 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Batshaw ML, MacArthur RB, Tuchman M. Alternative pathway therapy for urea cycle disorders: twenty years later. J Pediatr. 2001;138(1):S46‐S55. doi: 10.1067/mpd.2001.111836 [DOI] [PubMed] [Google Scholar]
- 66. Saheki T, Kobayashi K, Iijima M, et al. Adult‐onset type II citrullinemia and idiopathic neonatal hepatitis caused by citrin deficiency: involvement of the aspartate glutamate carrier for urea synthesis and maintenance of the urea cycle. Mol Genet Metab. 2004;81:20‐26. doi: 10.1016/j.ymgme.2004.01.006 [DOI] [PubMed] [Google Scholar]
- 67. Matsumoto S, Häberle J, Kido J, Mitsubuchi H, Endo F, Nakamura K. Urea cycle disorders—update. J Hum Genet. 2019;64(9):833‐847. doi: 10.1038/s10038-019-0614-4 [DOI] [PubMed] [Google Scholar]
- 68. Barbul A. Arginine: biochemistry, physiology, and therapeutic implications. J Parenter Enter Nutr. 1986;10(2):227‐238. doi: 10.1177/0148607186010002227 [DOI] [PubMed] [Google Scholar]
- 69. Yazaki M, Ikeda S, Takei Y, et al. Complete neurological recovery of an adult patient with type ii citrullinemia after living related partial liver transplantation. Transplantation. 1996;62(11):1679‐1681. doi: 10.1097/00007890-199612150-00027 [DOI] [PubMed] [Google Scholar]
- 70. Noto D, Takahashi K, Hamaguchi T, et al. A case of adult onset type II citrullinemia with portal‐systemic shunt. J Neurol Sci. 2009;281(1–2):127‐129. doi: 10.1016/j.jns.2009.02.003 [DOI] [PubMed] [Google Scholar]
- 71. Kawata A, Suda M, Tanabe H. Adult‐onset type II citrullinemia: clinical pictures before and after liver transplantation. Intern Med. 1997;36(6):408‐412. doi: 10.2169/internalmedicine.36.408 [DOI] [PubMed] [Google Scholar]
- 72. Allegri G, Deplazes S, Grisch‐Chan HM, et al. A simple dried blood spot‐method for in vivo measurement of ureagenesis by gas chromatography–mass spectrometry using stable isotopes. Clin Chim Acta. 2017;464:236‐243. doi: 10.1016/j.cca.2016.11.038 [DOI] [PubMed] [Google Scholar]
- 73. Saheki T, Iijima M, Li MX, et al. Citrin/mitochondrial glycerol‐3‐phosphate dehydrogenase double knock‐out mice recapitulate features of human citrin deficiency. J Biol Chem. 2007;282(34):25041‐25052. doi: 10.1074/jbc.M702031200 [DOI] [PubMed] [Google Scholar]
- 74. Sinasac DS, Moriyama M, Jalil MA, et al. Slc25a13 ‐knockout mice harbor metabolic deficits but fail to display hallmarks of Adult‐onset Type II Citrullinemia. Mol Cell Biol. 2004;24(2):527‐536. doi: 10.1128/MCB.24.2.527-536.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. González‐Moreno L, Santamaría‐Cano A, Paradela A, et al. Exogenous aralar/slc25a12 can replace citrin/slc25a13 as malate aspartate shuttle component in liver. Mol Genet Metab Rep. 2023;35:100967. doi: 10.1016/j.ymgmr.2023.100967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Neff EP. Rats on the rise. Lab Anim (NY). 2021;50(8):205‐208. doi: 10.1038/s41684-021-00812-0 [DOI] [PubMed] [Google Scholar]
- 77. Yau WW, Bin CG, Zhou J, et al. Nicotinamide riboside rescues dysregulated glycolysis and fatty acid β‐oxidation in a human hepatic cell model of citrin deficiency. Hum Mol Genet. 2023;32(11):1922‐1931. doi: 10.1093/hmg/ddad018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Mavri‐Damelin D, Eaton S, Damelin LH, Rees M, Hodgson HJF, Selden C. Ornithine transcarbamylase and arginase I deficiency are responsible for diminished urea cycle function in the human hepatoblastoma cell line HepG2. Int J Biochem Cell Biol. 2007;39(3):555‐564. doi: 10.1016/j.biocel.2006.10.007 [DOI] [PubMed] [Google Scholar]
- 79. Makris G, Veit L, Rüfenacht V, et al. Expression and function of the urea cycle in widely‐used hepatic cellular models. J Inherit Metab Dis. 2024. doi: 10.1002/jimd.12701 [DOI] [PubMed] [Google Scholar]
- 80. Graffmann N, Scherer B, Adjaye J. In vitro differentiation of pluripotent stem cells into hepatocyte like cells – basic principles and current progress. Stem Cell Res. 2022;61:102763. doi: 10.1016/j.scr.2022.102763 [DOI] [PubMed] [Google Scholar]
- 81. Seo KS, Kim JH, Min KN, et al. KL1333, a novel NAD+ modulator, improves energy metabolism and mitochondrial dysfunction in MELAS fibroblasts. Front Neurol. 2018;9:9. doi: 10.3389/fneur.2018.00552 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Saheki T, Moriyama M, Kuroda E, et al. Pivotal role of inter‐organ aspartate metabolism for treatment of mitochondrial aspartate‐glutamate carrier 2 (citrin) deficiency, based on the mouse model. Sci Rep. 2019;9(1):4179. doi: 10.1038/s41598-019-39627-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Jain A, Sharma BC, Mahajan B, et al. L‐ornithine L‐aspartate in acute treatment of severe hepatic encephalopathy: a double‐blind randomized controlled trial. Hepatology. 2022;75(5):1194‐1203. doi: 10.1002/hep.32255 [DOI] [PubMed] [Google Scholar]
- 84. Alvares‐da‐Silva MR, de Araujo A, Vicenzi JR, et al. Oral l‐ornithine‐l‐aspartate in minimal hepatic encephalopathy: a randomized, double‐blind, placebo‐controlled trial. Hepatol Res. 2014;44(9):956‐963. doi: 10.1111/hepr.12235 [DOI] [PubMed] [Google Scholar]
- 85. Bai M, He C, Yin Z, et al. Randomised clinical trial: L− ornithine− L− aspartate reduces significantly the increase of venous ammonia concentration after TIPSS. Aliment Pharmacol Ther. 2014;40(1):63‐71. doi: 10.1111/apt.12795 [DOI] [PubMed] [Google Scholar]
- 86. Butterworth RF, Kircheis G, Hilger N, McPhail MJW. Efficacy of l‐ornithine l‐aspartate for the treatment of hepatic encephalopathy and Hyperammonemia in cirrhosis: systematic review and meta‐analysis of randomized controlled trials. J Clin Exp Hepatol. 2018;8(3):301‐313. doi: 10.1016/j.jceh.2018.05.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Rebouche CJ, Paulson DJ. Carnitine metabolism and function in humans. 1986. www.annualreviews.org [DOI] [PubMed]
- 88. Herrera MD, Rodríguez‐Rodríguez R, Justo ML, de Sotomayor MA, Herrera MD. Critical update for the clinical use of L‐carnitine analogs in cardiometabolic disorders. Vasc Health Risk Manag. 2011;7:169‐176. doi: 10.2147/VHRM.S14356 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Longo N, di San A, Filippo C, Pasquali M. Disorders of carnitine transport and the carnitine cycle. Am J Med Genet C Semin Med Genet. 2006;142C(2):77‐85. doi: 10.1002/ajmg.c.30087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Agrawal V, Hemnes AR, Shelburne NJ, et al. L− Carnitine therapy improves right heart dysfunction through Cpt1‐dependent fatty acid oxidation. Pulm Circ. 2022;12(3):e12107. doi: 10.1002/pul2.12107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Tanaka T, Nagao M, Tsutsumi H. Application of mutation analysis for the previously uncertain cases of Adult‐onset Type II Citrullinemia(CTLN2) and their clinical profiles. Tohoku J Exp Med. 2002;198(2):89‐97. doi: 10.1620/tjem.198.89 [DOI] [PubMed] [Google Scholar]
- 92. Stettner N, Rosen C, Bernshtein B, et al. Induction of nitric‐oxide metabolism in enterocytes alleviates colitis and inflammation‐associated colon cancer. Cell Rep. 2018;23(7):1962‐1976. doi: 10.1016/j.celrep.2018.04.053 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Kirst HA. The spinosyn family of insecticides: realizing the potential of natural products research. J Antibiot (Tokyo). 2010;63(3):101‐111. doi: 10.1038/ja.2010.5 [DOI] [PubMed] [Google Scholar]
- 94. Sparks TC, Dripps JE, Watson GB, Paroonagian D. Resistance and cross‐resistance to the spinosyns – a review and analysis. Pestic Biochem Physiol. 2012;102(1):1‐10. doi: 10.1016/j.pestbp.2011.11.004 [DOI] [Google Scholar]
- 95. Zou Z, Hu X, Luo T, et al. Naturally‐occurring spinosyn a and its derivatives function as argininosuccinate synthase activator and tumor inhibitor. Nat Commun. 2021;12(1):2263. doi: 10.1038/s41467-021-22235-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Murillo O, Luqui DM, Gazquez C, et al. Long‐term metabolic correction of Wilson's disease in a murine model by gene therapy. J Hepatol. 2016;64(2):419‐426. doi: 10.1016/j.jhep.2015.09.014 [DOI] [PubMed] [Google Scholar]
- 97. Chandler RJ, Tarasenko TN, Cusmano‐Ozog K, et al. Liver‐directed adeno‐associated virus serotype 8 gene transfer rescues a lethal murine model of citrullinemia type 1. Gene Ther. 2013;20(12):1188‐1191. doi: 10.1038/gt.2013.53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Cunningham SC, Spinoulas A, Carpenter KH, Wilcken B, Kuchel PW, Alexander IE. AAV2/8‐mediated correction of OTC deficiency is robust in Adult but not neonatal Spfash mice. Mol Ther. 2009;17(8):1340‐1346. doi: 10.1038/mt.2009.88 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. An D, Schneller JL, Frassetto A, et al. Systemic messenger RNA therapy as a treatment for Methylmalonic Acidemia. Cell Rep. 2017;21(12):3548‐3558. doi: 10.1016/j.celrep.2017.11.081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Trepotec Z, Lichtenegger E, Plank C, Aneja MK, Rudolph C. Delivery of mRNA therapeutics for the treatment of hepatic diseases. Mol Ther. 2019;27(4):794‐802. doi: 10.1016/j.ymthe.2018.12.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Prieve MG, Harvie P, Monahan SD, et al. Targeted mRNA therapy for ornithine Transcarbamylase deficiency. Mol Ther. 2018;26(3):801‐813. doi: 10.1016/j.ymthe.2017.12.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Asrani KH, Cheng L, Cheng CJ, Subramanian RR. Arginase I mRNA therapy – a novel approach to rescue arginase 1 enzyme deficiency. RNA Biol. 2018;15(7):914‐922. doi: 10.1080/15476286.2018.1475178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Herrera Sanchez MB, Previdi S, Bruno S, et al. Extracellular vesicles from human liver stem cells restore argininosuccinate synthase deficiency. Stem Cell Res Ther. 2017;8(1):176. doi: 10.1186/s13287-017-0628-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Soria LR, Ah Mew N, Brunetti‐Pierri N. Progress and challenges in development of new therapies for urea cycle disorders. Hum Mol Genet. 2019;28(R1):R42‐R48. doi: 10.1093/hmg/ddz140 [DOI] [PubMed] [Google Scholar]
- 105. Cao J, An D, Galduroz M, et al. mRNA therapy improves metabolic and behavioral abnormalities in a murine model of Citrin deficiency. Mol Ther. 2019;27(7):1242‐1251. doi: 10.1016/j.ymthe.2019.04.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Lin WX, Zeng HS, Zhang ZH, et al. Molecular diagnosis of pediatric patients with citrin deficiency in China: SLC25A13 mutation spectrum and the geographic distribution. Sci Rep. 2016;6(1):29732. doi: 10.1038/srep29732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Wang L, Bell P, Morizono H, et al. AAV gene therapy corrects OTC deficiency and prevents liver fibrosis in aged OTC‐knock out heterozygous mice. Mol Genet Metab. 2017;120(4):299‐305. doi: 10.1016/j.ymgme.2017.02.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Ashley SN, Nordin JML, Buza EL, Greig JA, Wilson JM. Adeno‐associated viral gene therapy corrects a mouse model of argininosuccinic aciduria. Mol Genet Metab. 2018;125(3):241‐250. doi: 10.1016/j.ymgme.2018.08.013 [DOI] [PubMed] [Google Scholar]
- 109. Nitzahn M, Allegri G, Khoja S, et al. Split AAV‐mediated gene therapy restores ureagenesis in a murine model of carbamoyl phosphate Synthetase 1 deficiency. Mol Ther. 2020;28(7):1717‐1730. doi: 10.1016/j.ymthe.2020.04.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Cantero G, Liu XB, Mervis RF, et al. Rescue of the functional alterations of motor cortical circuits in arginase deficiency by neonatal gene therapy. J Neurosci. 2016;36(25):6680‐6690. doi: 10.1523/JNEUROSCI.0897-16.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. del Arco A, Morcillo J, Martínez‐Morales JR, et al. Expression of the aspartate/glutamate mitochondrial carriers aralar1 and citrin during development and in adult rat tissues. Eur J Biochem. 2002;269(13):3313‐3320. doi: 10.1046/j.1432-1033.2002.03018.x [DOI] [PubMed] [Google Scholar]
- 112. Infantino V, Dituri F, Convertini P, et al. Epigenetic upregulation and functional role of the mitochondrial aspartate/glutamate carrier isoform 1 in hepatocellular carcinoma. Biochim Biophys Acta Mol basis Dis. 2019;1865(1):38‐47. doi: 10.1016/j.bbadis.2018.10.018 [DOI] [PubMed] [Google Scholar]
- 113. Mention K, Joncquel Chevalier Curt M, Dessein AF, Douillard C, Dobbelaere D, Vamecq J. Citrin deficiency: does the reactivation of liver aralar‐1 come into play and promote HCC development? Biochimie. 2021;190:20‐23. doi: 10.1016/j.biochi.2021.06.018 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Table S1. Clinical data of NICCD patients before and after MCT treatment. 43
Table S2. Clinical data of CD patients with AACDa before and after MCT treatment.
