Minireviews Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Sep 27, 2024; 16(9): 1258-1268
Published online Sep 27, 2024. doi: 10.4254/wjh.v16.i9.1258
Immune remodulation in pediatric inherited metabolic liver diseases
Yi-Chi Wu, Xue-Lin Xiang, Lin-Man Li, Zi-Cheng Lv, Department of Liver Surgery, Renji Hospital (Punan Branch), School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
June-Kong Yong, Yi Zhou, Xi-Cheng Sun, Zi-Jie Zhang, Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
Meng Li, Qiang Xia, Hao Feng, Department of Liver Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, Shanghai Institute of Transplantation, Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200127, China
Huan Tong, Xiao-Ying He, Department of Gynecology, Shanghai First Maternity and Infant Hospital, Shanghai 200012, China
ORCID number: Yi Zhou (0000-0001-8383-7065); Zi-Jie Zhang (0000-0002-2513-3091); Qiang Xia (0000-0002-9199-9723); Hao Feng (0000-0003-4066-8104).
Co-first authors: Yi-Chi Wu and Xue-Lin Xiang.
Co-corresponding authors: Hao Feng and Qiang Xia.
Author contributions: Wu YC, Xiang XL, Yong JK, and Li M conceived, designed, and refined the study protocol; Li LM, Zhang ZJ, Tong H, and He XY were involved in the data collection; Lv ZC, Zhou Y, and Sun XC analyzed the data; Wu YC and Feng H drafted the manuscript. All authors were involved in the critical review of the results and have contributed to, read, and approved the final manuscript. Wu YC and Xiang XL contributed equally to this work as co-first authors. Xia Q and Feng H contributed equally to this work as co-corresponding authors, which accurately reflects the distribution of responsibilities and burdens associated with the time and effort required to complete the review. Both authors have contributed equally to the conception and design of the research. They have collaboratively developed the hypothesis and outlined the experimental approach. Both authors have been involved in the comprehensive review of the existing literature to establish the research context, identify gaps in knowledge, and support the rationale for the study. They have engaged in intellectual discussions and debates that have shaped the direction of the research, leading to the refinement of ideas and the development of innovative approaches. Both co-corresponding authors have been actively involved in drafting the manuscript, ensuring that the writing reflects their collective thoughts, interpretations, and conclusions drawn from the research.
Supported by Shanghai Science and Technology Development Foundation (Outstanding Academic Leader), No. 23XD1423100; and National Natural Science Foundation, No. 82241221 and No. 92059205.
Conflict-of-interest statement: The authors declare that they have no competing interests to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Hao Feng, MD, PhD, Research Scientist, Surgeon, Department of Liver Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, Shanghai Institute of Transplantation, Shanghai Engineering Research Center of Transplantation and Immunology, No. 160 Pujian Road, Shanghai 200127, China. surgeonfeng@live.com
Received: January 28, 2024
Revised: May 28, 2024
Accepted: August 19, 2024
Published online: September 27, 2024
Processing time: 239 Days and 10.4 Hours

Abstract

Inherited metabolic liver diseases arise from genetic mutations that lead to disruptions in liver metabolic pathways and are predominantly observed in pediatric populations. The spectrum of genetic metabolic liver disorders is diverse, encompassing a range of conditions associated with aberrations in iron, copper, carbohydrate, lipid, protein, and amino acid metabolism. Historically, research in the domain of genetic metabolic liver diseases has predominantly concentrated on hepatic parenchymal cell alterations. Nevertheless, emerging studies suggest that inherited metabolic liver diseases exert significant influences on the immune microenvironment, both within the liver and systemically. This review endeavors to encapsulate the immunological features of genetic metabolic liver diseases, aiming to expand the horizons of researchers in this discipline, and to elucidate the underlying pathophysiological mechanisms pertinent to hereditary metabolic liver diseases and to propose innovative therapeutic approaches.

Key Words: Liver transplantation; Inherited metabolic liver diseases; Immune microenvironment; Genetic metabolic liver diseases; Immunotherapy

Core Tip: It remains unclear whether concurrent infections are connected to immunological changes beyond the established metabolic imbalances in inherited metabolic liver diseases. Previous studies often focused on changes in liver parenchyma cells, but recent studies have shown that inherited hepatic metabolic diseases also have a profound impact on the liver’s immune microenvironment.



INTRODUCTION

Hereditary metabolic liver diseases, which are caused by gene mutations leading to liver metabolic disorders, are relatively prevalent among the pediatric population[1]. These conditions can be classified as disorders related to iron, copper, carbohydrate, lipid, protein, and amino acid metabolism based on the metabolites involved. When classified by specific diagnoses, the spectrum of these diseases includes Wilson’s disease (hepatolenticular degeneration), alpha-1 antitrypsin deficiency, urea cycle disorders, propionic acidemia (PA), Dubin-Johnson syndrome, tyrosinemia, and glycogen storage diseases (GSDs), among others. In addition to the well-recognized metabolic disturbances, affected individuals often present with concurrent infections that may be associated with metabolic decompensation. However, the relationship between these concurrent infections and immunological alterations, independent of the established metabolic dysregulations, remains to be elucidated.

Historically, research has concentrated on alterations within hepatic parenchymal cells. Yet, recent studies have demonstrated that inherited hepatic metabolic diseases significantly impact the liver’s immune microenvironment.

In light of these developments, this review aims to encapsulate the characteristics of immune remodeling in genetic liver metabolic diseases, underscoring the importance of a comprehensive analysis of immune remodeling to foster a deeper comprehension of the underlying pathophysiology of these conditions and to propose novel therapeutic strategies informed by these insights (Figure 1).

Figure 1
Figure 1 Immune remodulation in pediatric inherited metabolic liver diseases. GSD1b: Glycogen storage disease type 1b; ROS: Reactive oxygen species; RNS: Reactive nitrogen specie; LT: Liver transplantation; MMA: Methylmalonic acidemia.
NEUTROPHILS AND MACROPHAGES IN GSD AND WILSON DISEASE
GSDs

GSDs represent a group of rare monogenic disorders characterized by impairments in glycogen metabolism, encompassing the synthesis, breakdown (degradation of glycogen into glucose through glucose-6-phosphate [G6P]), and glycolysis (the conversion of glucose to pyruvate), as detailed in Table 1. These intricate, multi-systemic conditions display considerable heterogeneity in their clinical presentations and therapeutic strategies[2,3].

Table 1 Classification of glycogen storage diseases and their mutation sites.
Condition
Gene(s)
Enzyme(s) or transporter
Inheritance
GSD 0aGYS2Hepatic glycogen synthaseAR
GSD 0bGYS1Muscle glycogen synthaseAR
GSD I, Von Gierke diseaseG6PC1 (GSDIa) and SLC37A4 (GSD Ib)G6Pase (GSD Ia) and G6PT (GSD Ib)AR
GSD II, Pompe diseaseGAAAcid alpha-glucosidaseAR
GSD III (Cori disease; Forbes disease)AGLGlycogen debranching enzymeAR
GSD IV (Andersen disease)GBE1Glycogen branching enzymeAR
GSD V (McArdle disease)PYGMMyophosphorylaseAR
GSD VI (Hers disease)PYGLLiver glycogen phosphorylaseAR
GSD VII (Tarui disease)PFKMMuscle phosphofructokinaseAR
Hepatic GSD IXPHKA2 (GSD IX α2), PHKB (GSD IX β), PHKG2 (GSD IX γ2)Liver phosphorylase kinase α2 (GSD IX α2), liver and muscle phosphorylase kinase β2 (GSD IX β), and phosphorylase kinase γ2 (hepatic and testis isoform) (GSD IX γ2)X-linked (GSD IX α2; females can be affected depending on X inactivation), AR (GSD IX β, and GSD IX γ2)
Muscle GSD IXPHKA1Alpha subunit of muscle phosphorylase kinase (GSD IX α1)X-linked
GSD XPGAM2Muscle phosphoglycerate mutaseAR
GSD XI LDHALactate dehydrogenase AAR
GSD XIIALDOARed blood cell fructose-1,6-bisphosphate aldolase AAR
GSD XIIIENO3Beta-enolaseAR
GSD XVGYG1Glycogenin 1 (muscle isoform)AR
PGM1-CDG (formerly GSD XIV)PGM1Phosphoglucomutase 1AR
FBS (also called GSD XI), Fanconi-Bickel syndromeSLC2A2GLUT2AR
PGK deficiencyPGK1Phosphoglycerate kinaseX-linked

GSD type I (GSD I), also known as von Gierke disease, is a relatively prevalent autosomal recessive disorder within the spectrum of GSDs, with an incidence of approximately 1 in 100000 individuals[2]. GSD I is classified into two subtypes: GSD Ia, which arises from mutations in the G6PC1 gene leading to glucose-6-phosphatase deficiency and represents approximately 80% of cases, and GSD Ib, which results from genetic mutations in the SLC37A4 gene causing a deficiency in glucose-6-phosphate translocase and accounts for the remaining 20%.

Resaz et al[4] employed plasma-derived extracellular vesicles from LS-G6pc-/- mice, a model representative of GSD Ia, to elucidate the regulation of disease-associated microRNA expression. Their target genes were found to be significantly enriched in pathways involved in immune regulation. Cangelosi et al[5] conducted a liver proteomic analysis in LS-G6pc-/- mice, revealing an upregulation of proteins associated with inflammation and immune response. Notably, among these mice, six LS-G6pc-/- mice developed liver adenomas. In those with adenomatous changes, there was an observed increase in proteins involved in tissue inflammation and those influencing macrophage polarization towards the M2 phenotype. These findings suggest that immune regulation plays a pivotal role in the pathogenesis of GSD Ia and is intricately linked to the formation of adenomas and carcinomas. La Rose et al[6] identified a correlation between reduced circulating monocytes and disrupted platelet aggregation in L-G6pc-/- mice under fasting-induced hypoglycemic conditions, potentially relating to the bleeding diathesis observed in GSD Ia.

GSD Ib is characterized by disruptions in glycometabolism, inflammatory bowel disease (IBD), reduced neutrophil count, and impaired neutrophil functionality[7]. Visser et al[7] conducted a comprehensive study encompassing all known individuals with GSD-I born between 1960 and 1995 across 12 European countries. Of the 288 individuals, 57 had GSD Ib, with 54 experiencing neutropenia, defined as an absolute neutrophil count (ANC) below 1 × 109/L. Among these patients, neutropenia was documented in 64% before the age of 1, and in 18% between the ages of 6 and 9. Persistent neutropenia was observed in five patients, while 45 experienced intermittent decreases without discernible periodicity. Neutrophil function investigations in 18 patients with neutropenia revealed abnormal neutrophil function in all cases. Thirty-seven patients reported perioral infections, 27 had perianal infections, and 23 suffered from chronic diarrhea. Of the 20 patients suspected of having IBD, colonoscopic and radiographic examinations were abnormal in all 10 who underwent these procedures. Neutropenia was present in every individual with IBD, as well as in those with perioral and perianal infections[7]. Du et al[8] analyzed three cases of GSD Ib, observing recurrent infections and a decrease in ANC in all patients. Kaczor et al[9] examined 13 pediatric patients with GSD Ib (median age of 5 mo at diagnosis), with 10/13 experiencing severe infections (sepsis and/or pneumonia) as their initial symptoms during the neonatal-infant period, and 4/13 requiring long-term treatment for IBD. Wicker et al[10] conducted a retrospective study of nine French patients with GSD Ib, with an average age of 1.7 years at diagnosis and 3.8 years for the development of IBD. The patients experienced 0.7 acute hospitalizations per year due to infections (0.4 times/year) or digestive system-related symptoms (0.4 times/year). Those with severe clinical manifestations exhibited more pronounced digestive symptoms but also had an earlier onset of neutropenia. During follow-up, there was a declining trend in neutrophil counts in the severe group, with more frequent urgent hospitalizations (median 1.3 times per year), driven by digestive system manifestations (median 0.6 times per year) and infections (median 0.8 times per year). Shimizu et al[11] found that GSD type 1b (GSD1b) recipients are more susceptible to bloodstream infections compared to patients with biliary atresia following liver transplantation (LT), potentially due to lower neutrophil counts. Despite lower tacrolimus levels in the extended post-LT monitoring period for GSD 1b, there were no T-cell-mediated rejection episodes, suggesting that more personalized post-LT immunosuppressive strategies for GSD1b patients may be required. The neutropenia observed in GSD-Ib is attributed to increased neutrophil apoptosis. However, some propose that the blockade of neutrophil maturation within the bone marrow is also a contributing factor to neutropenia. Sim et al[12] utilized the CRISPR/Cas9 system to induce mutations in the G6PT gene, establishing a single-cell-derived G6PT-/- human myeloblastic cell line, HL-60. The G6PT-/- HL-60 cells displayed impaired neutrophil differentiation associated with two mechanisms: (1) Disrupted lipid metabolism leading to delayed metabolic reprogramming; and (2) diminished nuclear transcriptional activity of peroxisome proliferator-activated receptor-gamma, resulting in peroxisome proliferation.

The immune dysregulation in GSD1b extends beyond neutrophil deficiency, implicating both innate and adaptive immune responses. Gehlhaar et al[13] applied mass cytometry (CyTOF) to characterize the peripheral immune profiles in GSD1b patients, revealing a significant reduction in anti-inflammatory macrophages, CD16+ macrophages, and natural killer cells. Additionally, there was a skewed distribution towards central memory and effector memory phenotypes within various T cell subsets. This suggests that under the hypoglycemic conditions associated with GSD1b, immune cell populations may struggle to undergo the necessary metabolic shifts for proper glycolytic activity. The study also noted a general downregulation of CD123, CD14, CCR4, CD24, and CD11b across multiple cell populations, coupled with an upregulation of CXCR3, hinting at impaired immune cell migration as a feature of GSD1b pathology. Jang et al[14] utilized monocytes derived from G6PT-/- mice and human THP-1 monocytes with G6PT deficiency, demonstrating incomplete macrophage differentiation and altered inflammatory cytokine secretion despite increased glucose consumption under inflammatory conditions. Jeon et al[15] employed the CRISPR/Cas9 technique to mutate the G6PT gene in porcine alveolar macrophage 3D4/31 cells, which, lacking G6PT, showed reduced cell growth, bactericidal activity, and antiviral response. These findings of compromised immune functions in GSD1b suggest that future therapeutic interventions may need to target both neutrophils and macrophages.

Certain subtypes of GSDs have been identified to possess significant immunological features. Colonetti et al[16] analyzed plasma samples from treated GSD patients and observed that GSD-Ia/III/IX patients exhibited decreased levels of IL-4, CCL3, CCL22, TNF-β, and VEGF relative to controls. Among the different GSD types, GSD-Ib patients showed elevated G-CSF levels compared to GSD-Ia patients; IL-10 was higher in GSD-Ib than in GSD-Ia, while GSD-III/IX patients had increased CXCL10 levels compared to GSD-Ib individuals. When GSD-I patients were grouped and compared with GSD-III/IX patients, the latter displayed higher CXCL10 and CCL2 levels. Anemia in GSD-I patients correlated with increased IL-4 and CCL3 levels compared to non-anemic patients. Triglyceride levels in GSD-Ia patients without hepatocellular adenoma did not correlate with neutrophil counts or CCL22 levels. Overall, the cytokine profile alterations in GSD patients indicate a disruption in immune homeostasis[17].

Wilson disease

Wilson disease (WD) is a hereditary condition related to copper metabolism, characterized by the abnormal accumulation of copper within the body[18]. The etiology of WD is rooted in mutations affecting the ATP7B gene, which encodes a transmembrane copper-transporting ATPase. These mutations lead to copper accumulation in various organs, including the liver and brain. The progression of clinical symptoms in WD can vary in severity, but liver pathology is a frequently observed outcome. The dysfunction of ATP7B in WD results in excessive copper accumulation in the liver, leading to liver-related pathology[19-21]. Additionally, the excess copper is released into the bloodstream, causing abnormal accumulation in multiple tissues and significantly impacting the brain. This brain accumulation can lead to neurological symptoms and psychiatric disorders[22,23]. WD is most commonly diagnosed in individuals aged 5 to 35 and, despite its rarity, is estimated to affect approximately 1 in 30000 individuals[24]. Recent research has also identified a novel form of cell death dependent on copper, termed “copper-induced apoptosis” or “cuproptosis,” which is distinct from all known cell death pathways. Cuproptosis is characterized by copper binding to acyl-CoA synthetase in the citric acid cycle, leading to protein aggregation, protein toxicity stress, and ultimately cell death[17]. WD is now considered to be closely associated with this process of cuproptosis.

According to recent reports, WD is closely associated with autoimmune disorders. Cases where WD was combined with autoimmune conditions have been found. Acharya et al[25] reported an atypical case involving an 11-year-old with WD developing IgA vasculitis nephritis. This suggests that in WD, there might be a defect in the liver’s management or clearance of IgA/IgA immune complexes, potentially leading to IgA-mediated skin and kidney damage. Pradhan et al[26] reported the case of a 12-year-old girl with WD combined with systemic lupus erythematosus. Ma et al[27] documented the case of an 11-year-old with WD combined with immune thrombocytopenic purpura. Further research is needed to clarify the fundamental pathological mechanisms linking WD with these autoimmune conditions.

The production of autoantibodies is a prevalent characteristic in systemic autoimmune diseases, and both their diversity and abundance serve as crucial diagnostic and prognostic indicators[28,29]. Additionally, autoantibodies might emerge in individuals without any apparent health issues or in conditions induced by alternative mechanisms. Antczak-Kowalska et al[30] found that patients with WD had a higher prevalence of autoantibodies, approximately two times than healthy participants (ANA, ANCA, NSAbs, and ONA). In patients presenting neurological symptoms, the existence of autoantibodies (ANA and ANCA) significantly increased (approximately two times) in contrast to individuals in good health. No significant variations were noted for patients presenting liver symptoms. Jańczyk et al’s study revealed that, among the autoantibodies, only the frequency of ANA was elevated in children with WD compared to the healthy individuals[31]. The presence of autoantibodies did not show a significant association with hepatic steatosis or liver stiffness. Nevertheless, increased liver stiffness (E > 8.2 kPa) was correlated with the production of IgA, IgG, and γ-globulins.

Immunological responses in WD have been noted to exhibit distinct characteristics, with macrophages playing a pivotal role in the development and progression of liver pathologies. In a study by Glavind et al[32], two cohorts comprising 175 individuals with WD were evaluated. The findings indicated that sCD163, a marker of macrophage activation, while not specific to WD, was found at elevated levels in WD patients, with more significant increases in acute cases. Notably, patients with cirrhosis demonstrated higher sCD163 levels compared to those without, which correlated with biochemical indicators of liver damage and hepatic function. These observations suggest that macrophage activation is pronounced in WD and is associated with the hepatic disease phenotype as well as biochemical markers of liver disease. Moreover, sCD163 could potentially serve as an indicator of WD severity.

The dysfunction of the ATP7B protein, as seen in WD, leads to diminished incorporation of copper into ceruloplasmin, thereby reducing the ferroxidase activity essential for intracellular iron efflux. This reduction also impacts the release of copper from hepatocytes into bile, resulting in substantial hepatic copper accumulation and iron deposition. Jończy et al[33] observed a decrease in plasma iron levels in tx-J mice, which carry a missense mutation in the ATP7B gene and serve as a model for WD, alongside iron accumulation in liver cells and hepatic macrophages. Dusek et al[34] reported iron deposition in brain macrophages associated with neurological symptoms in WD. Mi et al[35] employed ATP7B-/- zebrafish and mouse models, as well as ATP7B-knockout HepG2 cell models, to elucidate the role and mechanism of neutrophils in WD. Transcriptome analysis of neutrophils in the liver of ATP7B-/- zebrafish revealed a unique transcriptional signature indicative of N2-type neutrophils. Furthermore, N2-type neutrophils were identified in the liver of mice with ATP7B gene knockout. Interventions targeting transforming growth factor β1, DNA methyltransferases, or signal transducer and activator of transcription 3 in drug-treated ATP7B-knockout mice reduced hepatic N2-type neutrophils, improved liver function, and mitigated hepatic inflammation and fibrosis. The study also found that transforming growth factor β1-mediated epigenetic silencing of Socs3 facilitated N2-type polarization in bone marrow neutrophils. These findings suggest that modulating the activity of N2-type neutrophils could offer an alternative therapeutic approach to enhance liver function in WD.

Neutrophil extracellular traps (NETs) are directly implicated in several disease processes and are known to contribute to systemic inflammation, with significant deposition observed in hepatic sinusoids. Cichon et al[36] investigated the influence of copper on NETs formation in the hepatic vasculature during septicemia in a WD mouse model. They found that mutations in the ATP7 gene resulted in reduced NETs release during systemic inflammation, where the excess copper in WD mice directly impaired the capacity to release NETs. This study has broadened our comprehension of how copper metabolism in WD influences NETs release and its subsequent pathological implications.

T CELLS IN UREA CYCLE DISORDERS

In the urea cycle, there are a total of six enzymes involved[37]. The deficiency or defect of any of these enzymes can disrupt the normal functioning of this cycle, leading to the inability to convert the metabolic product of protein, ammonia, into urea, causing its accumulation in the body[38,39]. This results in increased blood ammonia levels, leading to a range of clinical manifestations. This group of genetic metabolic disorders are commonly known as urea cycle disorders. There are several diseases classified as urea cycle disorders, such as argininemia, arginine succinate lyase deficiency, arginine succinate synthase deficiency, carbamoyl phosphate synthetase deficiency, ornithine-δ-transaminase deficiency, and ornithine transcarboxylase deficiency, among others[40].

Hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome, an uncommon autosomal recessive disorder related to the urea cycle resulting from mutations in the OTCD gene, is characterized by metabolic complications and commonly presents acute hyperammonemia and cross-infections associated with metabolic imbalance. Silvera-Ruiz et al[41] described a case of HHH syndrome and detected a reduction in the serum gamma globulin fraction and decreased levels of serum IgG, as well as significant alterations in the phenotype and function of T and B cell immune components. This suggests that this rare metabolic genetic defect might lead to immune impairment in patients and could be associated with the high incidence of cross-infections observed in patients with urea cycle disorders.

Reports on cases of urea cycle disorders further confirm the crucial role of immune regulation in these conditions. The research findings of da Silva et al[42] demonstrate a significant increase in indoleamine 2,3-dioxygenase (IDO) activity (Kyn/Trp) with age. Among the metabolites most strongly associated with elevated IDO, ornithine transcarbamylase (Orn/Arg) exhibits a robust correlation, potentially contributing to the aging of the immune system. These findings provide compelling evidence of metabolite influence on the body’s immune microenvironment, advocating interventions to reduce ornithine transcarbamylase activity.

Awasthi et al’s study revealed that supplementing L-arginine created a favorable environment for parasite growth in mice with malaria[43]. Consequently, the infected mice supplemented with L-arginine died earlier than the control group of infected wild-type mice. Conversely, supplementation of L-citrulline inhibited parasite growth, leading to increased survival time in the mice. Flow CyTOF analysis indicated that supplementing L-arginine increased CTLA-4 in the population of T cells and augmented regulatory T cells (Treg cells), inducing immune suppression. In contrast, supplementation of L-citrulline had no impact on the population of T cells, and the number of Treg cells decreased in P. berghei-infected mice[43]. This suggests that metabolites have a profound influence on the biological behavior of T cells.

NAÏVE HELPER T CELLS IN ORGANIC ACIDEMIAS

Organic acidemia (OA) stems from enzyme deficiencies within the body’s metabolic processes of organic acids, resulting in the accumulation of these acids and their byproducts, leading to a spectrum of disorders[44-46]. In 1966, Tanaka et al[47] identified the first case of isovaleric acidemia (IVA) using gas chromatographic/mass spectrometry technology, and since then, dozens of similar conditions have been progressively identified. Most OAs are inherited as autosomal recessive disorders, with common diseases within this category including methylmalonic acidemia (MMA), PA, and IVA[48].

Organic acids impact clinical manifestations of diseases by influencing the immune system. For instance, high concentrations of methylmalonic acid in the bloodstream can induce immune cells to release cytokines associated with inflammation like TNF-α and IL-6. These cytokines, along with elevated methylmalonic acid, stimulate immune cells to generate reactive oxygen species (ROS) and reactive nitrogen species (RNS). Methylmalonic acid, cytokines, ROS, and RNS present in the serum can penetrate the blood-brain barrier, triggering cognitive impairment[49]. Medina-Torres et al[50] conducted a retrospective study involving 11 patients with PA and MMA, collectively known as propionate inherited metabolic disorders (PIEM), which are OAs. The results indicated that 91% of the patients exhibited immunological abnormalities. With the exception of one patient, all individuals showed decreased absolute counts of lymphocyte subsets, particularly a common reduction in CD4+ T lymphocytes. Furthermore, among the 11 participants, 9 had a low CD4/CD8 ratio. These findings suggest that monitoring immune biomarkers, such as lymphocyte subsets, may be meaningful during the follow-up of patients with PIEM. Altun et al[51] conducted a study involving 33 patients with OA and 32 age- and gender-matched healthy controls to evaluate the immune status of OA patients. Among the 33 patients, 21 (88%) were diagnosed with MMA, while 10 (33%) were identified with PA, and 2 (6.6%) with IVA. Sepsis was present in 18 patients (55%) among the studied group. The numbers of naïve helper T cells and recent thymic emigrants were notably reduced in OA patients (P < 0.001). Several anomalies affecting humoral immunity were also observed, including memory B cells and immunoglobulins. These findings suggest that OA patients might exhibit adaptive immune deficiencies, making them more susceptible to infections. Organic acids can trigger immune-related conditions in diseases. Aydin Köker et al[52] reported a case where hemophagocytic lymphohistiocytosis developed as a consequence of PA. Zhu et al[53] reported the case of a 5.5-year-old girl who simultaneously suffered from MMA, acute lymphoblastic leukemia, and congenital heart disease. However, it is currently unclear how organic acids induce other disorders in the context of the disease.

OTHER INHERITED METABOLIC LIVER DISEASES

Abnormal tyrosine metabolism is a genetic disorder affecting the liver and kidneys due to reduced activity of fumarylacetoacetate hydrolase, the ultimate enzyme in tyrosine breakdown. Succinylacetone in urine or blood is a characteristic feature of this disorder. High tyrosine levels comprise various types; among them, tyrosinemia type I (or hepatorenal tyrosinemia, HT1) exhibits the broadest clinical and pathological manifestations. Nitisinone (NTBC) is a drug used to treat tyrosinemia type I (Figure 2). Wei et al[54] employed NTBC as a means to explore the potential role of the Phe/Tyr degradation pathway in inflammatory responses. They discovered that NTBC exhibited efficacy in alleviating lipopolysaccharide-induced septic shock in mice. Mechanistically, this protective effect was associated with the accumulation of 4-hydroxyphenylpyruvic acid (4-HPP), an intermediate product induced by NTBC treatment in the Phe/Tyr degradation pathway. 4-HPP inhibited the initiation and activation of the NLRP3 inflammasome, thereby reducing the release of IL-1β, as shown in Figure 2. Similar to NTBC, 4-HPP was also effective in alleviating endotoxin shock in mice. These findings suggest that the Tyr degradation pathway might serve as a plausible center for immunoregulation and could be used as a therapeutic approach to mitigate inflammation.

Figure 2
Figure 2 Mechanism of nitisinone in treating tyrosinemia. 4-HPP: 4-hydroxyphenylpyruvic acid.

Liver genetic metabolic diseases encompass a wide range of conditions, and their pathophysiological mechanisms are somewhat linked to the immune microenvironment. However, most of these conditions are rare diseases with few cases and limited related reports, so a detailed enumeration is not feasible here.

GENE THERAPY FOR INHERITED METABOLIC DISORDERS OF THE LIVER: IMMUNOLOGICAL CHALLENGES

Gene therapy is increasingly recognized as a promising therapeutic strategy for a variety of genetic disorders, particularly for rare metabolic diseases with limited treatment options, such as those affecting liver function. LT currently stands as the only definitive cure for these conditions. The liver’s capacity, representing 10%-15% of the body’s total blood volume, positions it as an optimal site for protein secretion into the circulatory system[55]. Traditionally, viral vectors including lentiviruses, adenoviruses, retroviruses, and adeno-associated viruses (AAVs) have been the mainstays for liver cell targeting. However, over recent decades, an array of non-viral vectors, such as naked DNA, liposomes, and nanocarriers, have risen as effective alternatives for genetic material delivery[56]. Gene therapy approaches can be broadly categorized into two types based on administration methods: Ex vivo and in vivo. Ex vivo gene therapy involves the extraction, culture, amplification, and vector-mediated modification of a patient’s cells, followed by their reintroduction into the patient. In vivo gene therapy, on the other hand, entails the direct administration of therapeutic agents to the patient via various systemic or localized routes. For liver diseases, in vivo gene therapy is often more appropriate given the complexities associated with ex vivo manipulation and amplification of liver cells[57]. A multitude of studies have attested to the efficacy of liver-directed gene therapy. However, the translation of these methods into clinical practice is encumbered by numerous challenges, chief among them being the innate and adaptive immune responses elicited by gene therapy products. While adenoviruses have garnered considerable clinical experience in oncology, their application in the treatment of genetic metabolic liver diseases has been constrained by potential inflammatory reactions. In contrast, AAVs have seen broader application in liver-targeted gene therapy[58].

Crigler-Najjar syndrome, a recessive hereditary disorder of liver metabolism attributable to mutations in the UGT1A1 gene, currently has LT as its sole definitive treatment. D’Antiga et al[59] reported on a phase 1-2 clinical trial of gene therapy for Crigler-Najjar syndrome, wherein patients administered a higher dose of adeno-associated virus serotype 8 vector encoding UGT1A1 (GNT0003) showed a reduction in bilirubin levels and remained free from phototherapy requirements for at least 78 wk post-vector infusion. During follow-up, ALT levels increased in four patients, associated with a T-cell-mediated response to the vector. However, T-cell-mediated immune responses against UGT1A1 and AAV8 were challenging to detect owing to the concurrent use of immunosuppressive drugs. In all five patients, IgG and IgM antibody levels against AAV8 peaked concurrently before returning to baseline levels, with sustained high titers of neutralizing antibodies observed[58]. Hordeaux et al[60] developed the AAVhu68 vector for Pompe disease (GSD II) treatment, a type F adenovirus closely related to AAV9, carrying an engineered human acid alpha-glucosidase (hGAA) with an insulin-like growth factor 2 variant (vIGF2) peptide tag. Eleven cynomolgus monkeys treated with intravenous AAVhu68.vIGF2.hGAA exhibited a spectrum of severe anti-hGAA responses, from absent to severe cell-mediated myocarditis with elevated cardiac troponin I levels, resulting in cardiac toxicity in five monkeys. Eggers et al’s study similarly illustrated that while the adenovirus vector AT845 is efficacious for gene therapy in Pompe disease, high doses can trigger anti-acid alpha-glucosidase (GAA) immune responses, inflammation, and cardiac abnormalities[61]. In contrast, a vector expressing cynomolgus monkey GAA did not induce detectable pathological changes, suggesting that AT845’s toxicity may stem from an anti-GAA heterologous immune response. In a gene therapy trial, Somanathan et al[62] administered arterial delivery using adenovirus type 5 (Ad5) as a vector to a patient with ornithine transcarbamylase deficiency, which regrettably led to fatal systemic inflammation. Further investigation indicated that neutralizing antibodies against Ad5 may enhance transduction and activation of dendritic cells (DCs) by Ad5, potentially post-formation of Ig-Ad5 immune complexes binding to DCs. This finding may shed light on the mechanisms behind systemic inflammatory reactions induced by adenovirus-based gene therapy.

To surmount the immune challenges associated with gene therapy for hepatic genetic metabolic diseases, various strategies have been explored. For instance, the use of viral vectors with lower immunogenicity in their capsids has been investigated, alongside the combination of gene therapy with immunosuppressive treatments. Chandler et al[63] examined the potential of AAV44.9 as a vector for gene therapy in MMA. AAV44.9, initially isolated as a contaminant in the rhesus adenovirus SV15 monkey kidney cell culture system, shares the highest homology with AAVrh.8 and features a novel capsid. In human trials, AAV44.9 has demonstrated low immunogenicity, reducing the risk of neutralizing antibodies and enhancing safety. Tissue distribution in treated MMA mice indicates that AAV44.9 possesses efficient transduction in the liver and heart, also reducing mortality and disease-related metabolic products, positioning it as a promising vector for clinical applications. Choi et al[64] discovered that the combination of bortezomib and mouse-specific CD20 monoclonal antibody as immunosuppressive therapy can effectively mitigate the production of neutralizing antibodies against AAV. This strategy holds potential for the elimination or amelioration of immune responses triggered by neutralizing antibodies.

CONCLUSION

The field of immunology related to genetic metabolic liver diseases is replete with promising prospects. Approaches such as immune modulation, immunocellular therapy, and immune suppression may improve disease progression and symptom severity. Interventions targeting immune responses could mitigate inflammation and immune-related tissue damage, thereby protecting affected liver tissues. Additionally, the deployment of immunocellular therapies, including stem cells or immune-regulatory cells, may foster repair or replacement of damaged liver tissues, offering more enduring therapeutic benefits for patients.

As we delve deeper into immunological research, a clearer understanding of the interplay between genetic metabolic liver diseases and the immune system will emerge. This knowledge could pave the way for more precise, personalized treatment strategies, thereby enhancing therapeutic outcomes and improving patients’ quality of life. Although this domain remains in its exploratory phase, the relentless advancements in immunology herald a new era of potential treatments for genetic metabolic liver diseases.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A, Grade C, Grade D

Novelty: Grade A, Grade B, Grade C

Creativity or Innovation: Grade A, Grade C, Grade C

Scientific Significance: Grade A, Grade B, Grade C

P-Reviewer: Herrero-Maceda MR; Ullah K S-Editor: Qu XL L-Editor: Wang TQ P-Editor: Yuan YY

References
1.  Ferreira CR, Cassiman D, Blau N. Clinical and biochemical footprints of inherited metabolic diseases. II. Metabolic liver diseases. Mol Genet Metab. 2019;127:117-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
2.  Hannah WB, Derks TGJ, Drumm ML, Grünert SC, Kishnani PS, Vissing J. Glycogen storage diseases. Nat Rev Dis Primers. 2023;9:46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
3.  Ozen H. Glycogen storage diseases: new perspectives. World J Gastroenterol. 2007;13:2541-2553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 161]  [Cited by in F6Publishing: 165]  [Article Influence: 9.7]  [Reference Citation Analysis (1)]
4.  Resaz R, Cangelosi D, Morini M, Segalerba D, Mastracci L, Grillo F, Bosco MC, Bottino C, Colombo I, Eva A. Circulating exosomal microRNAs as potential biomarkers of hepatic injury and inflammation in a murine model of glycogen storage disease type 1a. Dis Model Mech. 2020;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
5.  Cangelosi D, Resaz R, Petretto A, Segalerba D, Ognibene M, Raggi F, Mastracci L, Grillo F, Bosco MC, Varesio L, Sica A, Colombo I, Eva A. A Proteomic Analysis of GSD-1a in Mouse Livers: Evidence for Metabolic Reprogramming, Inflammation, and Macrophage Polarization. J Proteome Res. 2019;18:2965-2978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
6.  La Rose AM, Bazioti V, Hoogerland JA, Svendsen AF, Groenen AG, van Faassen M, Rutten MGS, Kloosterhuis NJ, Dethmers-Ausema B, Nijland JH, Mithieux G, Rajas F, Kuipers F, Lukens MV, Soehnlein O, Oosterveer MH, Westerterp M. Hepatocyte-specific glucose-6-phosphatase deficiency disturbs platelet aggregation and decreases blood monocytes upon fasting-induced hypoglycemia. Mol Metab. 2021;53:101265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
7.  Visser G, Rake JP, Fernandes J, Labrune P, Leonard JV, Moses S, Ullrich K, Smit GP. Neutropenia, neutrophil dysfunction, and inflammatory bowel disease in glycogen storage disease type Ib: results of the European Study on Glycogen Storage Disease type I. J Pediatr. 2000;137:187-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 149]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
8.  Du C, Li Z, Wei H, Zhang M, Hu M, Zhang C, Luo X, Liang Y. Clinical analysis and long-term treatment monitoring of 3 patients with glycogen storage disease type Ib. BMC Med Genomics. 2021;14:81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
9.  Kaczor M, Wesół-Kucharska D, Greczan M, Kierus K, Kałużny Ł, Duś-Żuchowska M, Ehmke Vel Emczyńska-Seliga E, Ciara E, Książyk J, Rokicki D. Clinical characteristics and long-term outcomes of patients with glycogen storage disease type 1b: a retrospective multi-center experience in Poland. Pediatr Endocrinol Diabetes Metab. 2022;28:207-212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
10.  Wicker C, Roda C, Perry A, Arnoux JB, Brassier A, Castelle M, Servais A, Donadieu J, Bouchereau J, Pigneur B, Labrune P, Ruemmele FM, de Lonlay P. Infectious and digestive complications in glycogen storage disease type Ib: Study of a French cohort. Mol Genet Metab Rep. 2020;23:100581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
11.  Shimizu S, Sakamoto S, Yamada M, Fukuda A, Yanagi Y, Uchida H, Mimori K, Shoji K, Funaki T, Miyairi I, Nakano N, Haga C, Yoshioka T, Imadome KI, Horikawa R, Kasahara M. Immunological features and complications in patients with glycogen storage disease 1b after living donor liver transplantation. Pediatr Transplant. 2021;25:e14104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
12.  Sim SW, Jang Y, Park TS, Park BC, Lee YM, Jun HS. Molecular mechanisms of aberrant neutrophil differentiation in glycogen storage disease type Ib. Cell Mol Life Sci. 2022;79:246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
13.  Gehlhaar A, Shouval D, Santiago EG, Ling G, McCourt B, Werner L, Yerushalmi B, Konnikova L. Immune dysregulation in Glycogen Storage Disease 1b - a CyTOF approach. Res Sq. 2023;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
14.  Jang Y, Park TS, Park BC, Lee YM, Heo TH, Jun HS. Aberrant glucose metabolism underlies impaired macrophage differentiation in glycogen storage disease type Ib. FASEB J. 2023;37:e23216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
15.  Jeon EH, Park TS, Jang Y, Hwang E, Kim SJ, Song KD, Weinstein DA, Lee YM, Park BC, Jun HS. Glucose-6-phosphate transporter mediates macrophage proliferation and functions by regulating glycolysis and mitochondrial respiration. Biochem Biophys Res Commun. 2020;524:89-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
16.  Colonetti K, Pinto E Vairo F, Siebert M, Nalin T, Poloni S, Fernando Wurdig Roesch L, Fischinger Moura de Souza C, Cabral Pinheiro F, Vanessa Doederlein Schwartz I. Cytokine profiling in patients with hepatic glycogen storage disease: Are there clues for unsolved aspects? Cytokine. 2023;162:156088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
17.  Chen L, Min J, Wang F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct Target Ther. 2022;7:378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 349]  [Article Influence: 174.5]  [Reference Citation Analysis (0)]
18.  Bandmann O, Weiss KH, Kaler SG. Wilson's disease and other neurological copper disorders. Lancet Neurol. 2015;14:103-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 477]  [Cited by in F6Publishing: 554]  [Article Influence: 61.6]  [Reference Citation Analysis (0)]
19.  Ferenci P. Regional distribution of mutations of the ATP7B gene in patients with Wilson disease: impact on genetic testing. Hum Genet. 2006;120:151-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 155]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
20.  Dzieżyc K, Karliński M, Litwin T, Członkowska A. Compliant treatment with anti-copper agents prevents clinically overt Wilson's disease in pre-symptomatic patients. Eur J Neurol. 2014;21:332-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 61]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
21.  European Association for Study of Liver. EASL Clinical Practice Guidelines: Wilson's disease. J Hepatol. 2012;56:671-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 717]  [Cited by in F6Publishing: 700]  [Article Influence: 58.3]  [Reference Citation Analysis (1)]
22.  Roberts EA, Schilsky ML; American Association for Study of Liver Diseases (AASLD). Diagnosis and treatment of Wilson disease: an update. Hepatology. 2008;47:2089-2111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 884]  [Cited by in F6Publishing: 769]  [Article Influence: 48.1]  [Reference Citation Analysis (0)]
23.  Socha P, Janczyk W, Dhawan A, Baumann U, D'Antiga L, Tanner S, Iorio R, Vajro P, Houwen R, Fischler B, Dezsofi A, Hadzic N, Hierro L, Jahnel J, McLin V, Nobili V, Smets F, Verkade HJ, Debray D. Wilson's Disease in Children: A Position Paper by the Hepatology Committee of the European Society for Paediatric Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr. 2018;66:334-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 127]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
24.  Członkowska A, Litwin T, Dusek P, Ferenci P, Lutsenko S, Medici V, Rybakowski JK, Weiss KH, Schilsky ML. Wilson disease. Nat Rev Dis Primers. 2018;4:21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 386]  [Cited by in F6Publishing: 417]  [Article Influence: 69.5]  [Reference Citation Analysis (0)]
25.  Acharya R, Zeng X, Clapp WL, Upadhyay K. IgA vasculitis with nephritis in cirrhotic Wilson disease: Is there an association? Clin Nephrol Case Stud. 2020;8:80-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
26.  Pradhan S, Krishnamurthy S, Jagadisan B, Rajesh NG, Kaliaperumal S, Ramasamy S, Subramanian N. Wilson disease and lupus nephritis: is it coincidence or a true association? Paediatr Int Child Health. 2019;39:146-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
27.  Ma TJ, Sun GL, Yao F, Yang ZL. Wilson disease associated with immune thrombocytopenia: A case report and review of the literature. World J Clin Cases. 2019;7:2630-2636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
28.  Sheldon J. Laboratory testing in autoimmune rheumatic diseases. Best Pract Res Clin Rheumatol. 2004;18:249-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 26]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
29.  Lyons R, Narain S, Nichols C, Satoh M, Reeves WH. Effective use of autoantibody tests in the diagnosis of systemic autoimmune disease. Ann N Y Acad Sci. 2005;1050:217-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 49]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
30.  Antczak-Kowalska M, Członkowska A, Eyileten C, Palejko A, Cudna A, Wolska M, Piechal A, Litwin T. Autoantibodies in Wilson disease: Impact on clinical course. JIMD Rep. 2022;63:508-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Reference Citation Analysis (0)]
31.  Jańczyk W, Bierła JB, Trojanowska I, Wierzbicka-Rucińska A, Cukrowska B, Socha P. Prevalence and Significance of Autoantibody Seropositivity in Children with Wilson's Disease. Diagnostics (Basel). 2023;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
32.  Glavind E, Gotthardt DN, Pfeiffenberger J, Sandahl TD, Bashlekova T, Willemoe GL, Hasselby JP, Weiss KH, Møller HJ, Vilstrup H, Lee WM, Schilsky ML, Ott P, Grønbæk H. The macrophage activation marker soluble CD163 is elevated and associated with liver disease phenotype in patients with Wilson's disease. Orphanet J Rare Dis. 2020;15:173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
33.  Jończy A, Lipiński P, Ogórek M, Starzyński RR, Krzysztofik D, Bednarz A, Krzeptowski W, Szudzik M, Haberkiewicz O, Miłoń A, Grzmil P, Lenartowicz M. Functional iron deficiency in toxic milk mutant mice (tx-J) despite high hepatic ferroportin: a critical role of decreased GPI-ceruloplasmin expression in liver macrophages. Metallomics. 2019;11:1079-1092.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
34.  Dusek P, Hofer T, Alexander J, Roos PM, Aaseth JO. Cerebral Iron Deposition in Neurodegeneration. Biomolecules. 2022;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 46]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
35.  Mi X, Song Y, Deng C, Yan J, Li Z, Li Y, Zheng J, Yang W, Gong L, Shi J. Stimulation of Liver Fibrosis by N2 Neutrophils in Wilson's Disease. Cell Mol Gastroenterol Hepatol. 2023;16:657-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
36.  Cichon I, Ortmann W, Bednarz A, Lenartowicz M, Kolaczkowska E. Reduced Neutrophil Extracellular Trap (NET) Formation During Systemic Inflammation in Mice With Menkes Disease and Wilson Disease: Copper Requirement for NET Release. Front Immunol. 2019;10:3021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
37.  Häberle J, Burlina A, Chakrapani A, Dixon M, Karall D, Lindner M, Mandel H, Martinelli D, Pintos-Morell G, Santer R, Skouma A, Servais A, Tal G, Rubio V, Huemer M, Dionisi-Vici C. Suggested guidelines for the diagnosis and management of urea cycle disorders: First revision. J Inherit Metab Dis. 2019;42:1192-1230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 249]  [Article Influence: 49.8]  [Reference Citation Analysis (0)]
38.  Posset R, Gropman AL, Nagamani SCS, Burrage LC, Bedoyan JK, Wong D, Berry GT, Baumgartner MR, Yudkoff M, Zielonka M, Hoffmann GF, Burgard P, Schulze A, McCandless SE, Garcia-Cazorla A, Seminara J, Garbade SF, Kölker S; Urea Cycle Disorders Consortium and the European Registry and Network for Intoxication Type Metabolic Diseases Consortia Study Group. Impact of Diagnosis and Therapy on Cognitive Function in Urea Cycle Disorders. Ann Neurol. 2019;86:116-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 36]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
39.  Ah Mew N, Krivitzky L, McCarter R, Batshaw M, Tuchman M; Urea Cycle Disorders Consortium of the Rare Diseases Clinical Research Network. Clinical outcomes of neonatal onset proximal versus distal urea cycle disorders do not differ. J Pediatr. 2013;162:324-9.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
40.  Matsumoto S, Häberle J, Kido J, Mitsubuchi H, Endo F, Nakamura K. Urea cycle disorders-update. J Hum Genet. 2019;64:833-847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 107]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
41.  Silvera-Ruiz SM, Gemperle C, Peano N, Olivero V, Becerra A, Häberle J, Gruppi A, Larovere LE, Motrich RD. Immune Alterations in a Patient With Hyperornithinemia-Hyperammonemia-Homocitrullinuria Syndrome: A Case Report. Front Immunol. 2022;13:861516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
42.  da Silva IDCG, Marchioni DML, Carioca AAF, Bueno V, Colleoni GWB. May critical molecular cross-talk between indoleamine 2,3-dioxygenase (IDO) and arginase during human aging be targets for immunosenescence control? Immun Ageing. 2021;18:33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
43.  Awasthi V, Chauhan R, Das J. Administration of L-citrulline prevents Plasmodium growth by inhibiting/ modulating T-regulatory cells during malaria pathogenesis. J Vector Borne Dis. 2022;59:45-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
44.  Raina R, Bedoyan JK, Lichter-Konecki U, Jouvet P, Picca S, Mew NA, Machado MC, Chakraborty R, Vemuganti M, Grewal MK, Bunchman T, Sethi SK, Krishnappa V, McCulloch M, Alhasan K, Bagga A, Basu RK, Schaefer F, Filler G, Warady BA. Consensus guidelines for management of hyperammonaemia in paediatric patients receiving continuous kidney replacement therapy. Nat Rev Nephrol. 2020;16:471-482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 43]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
45.  Chen T, Gao Y, Zhang S, Wang Y, Sui C, Yang L. Methylmalonic acidemia: Neurodevelopment and neuroimaging. Front Neurosci. 2023;17:1110942.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 10]  [Reference Citation Analysis (0)]
46.  Pillai NR, Stroup BM, Poliner A, Rossetti L, Rawls B, Shayota BJ, Soler-Alfonso C, Tunuguntala HP, Goss J, Craigen W, Scaglia F, Sutton VR, Himes RW, Burrage LC. Liver transplantation in propionic and methylmalonic acidemia: A single center study with literature review. Mol Genet Metab. 2019;128:431-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 31]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
47.  Tanaka K, Budd MA, Efron ML, Isselbacher KJ. Isovaleric acidemia: a new genetic defect of leucine metabolism. Proc Natl Acad Sci U S A. 1966;56:236-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 220]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
48.  Reddy N, Calloni SF, Vernon HJ, Boltshauser E, Huisman TAGM, Soares BP. Neuroimaging Findings of Organic Acidemias and Aminoacidopathies. Radiographics. 2018;38:912-931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
49.  Li Q, Jin H, Liu Y, Rong Y, Yang T, Nie X, Song W. Determination of Cytokines and Oxidative Stress Biomarkers in Cognitive Impairment Induced by Methylmalonic Acidemia. Neuroimmunomodulation. 2021;28:178-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
50.  Medina-Torres EA, Vela-Amieva M, Galindo-Campos L, Ibarra-González I, Espinosa-Padilla S, Guillén-López S, López-Mejía L, Fernández-Lainez C. Abnormalities in subsets of B and T cells in Mexican patients with inborn errors of propionate metabolism: observations from a single-center case series. Allergol Immunopathol (Madr). 2021;49:101-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
51.  Altun I, Kiykim A, Zubarioglu T, Burtecene N, Hopurcuoglu D, Topcu B, Cansever MS, Kiykim E, Cokugras HC, Aktuglu Zeybek AC. Altered immune response in organic acidemia. Pediatr Int. 2022;64:e15082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
52.  Aydin Köker S, Yeşilbaş O, Köker A, Şevketoğlu E. Propionic acidemia: an extremely rare cause of hemophagocytic lymphohistiocytosis in an infant. Arch Argent Pediatr. 2020;118:e174-e177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
53.  Zhu J, Wan S, Zhao X, Zhu B, Lv Y, Jiang H. Acute Lymphoblastic Leukemia in Combined Methylmalonic Acidemia and Homocysteinemia (cblC Type): A Case Report and Literature Review. Front Genet. 2022;13:856552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
54.  Wei Y, Liu M, Han J, Huang H, Xu S, Zhang S, Jing Q, Wang H, Bu H, Kou Y, Liu Z, Zheng K, Wang Y. A tyrosine catabolic intermediate 4-hydroxyphenylpyruate attenuates murine endotoxic shock by blocking NLRP3 inflammasome activation. Int Immunopharmacol. 2022;111:109098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
55.  Maestro S, Weber ND, Zabaleta N, Aldabe R, Gonzalez-Aseguinolaza G. Novel vectors and approaches for gene therapy in liver diseases. JHEP Rep. 2021;3:100300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 45]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
56.  Blaese RM, Culver KW, Miller AD, Carter CS, Fleisher T, Clerici M, Shearer G, Chang L, Chiang Y, Tolstoshev P, Greenblatt JJ, Rosenberg SA, Klein H, Berger M, Mullen CA, Ramsey WJ, Muul L, Morgan RA, Anderson WF. T lymphocyte-directed gene therapy for ADA- SCID: initial trial results after 4 years. Science. 1995;270:475-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1074]  [Cited by in F6Publishing: 893]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
57.  Bordignon C, Notarangelo LD, Nobili N, Ferrari G, Casorati G, Panina P, Mazzolari E, Maggioni D, Rossi C, Servida P, Ugazio AG, Mavilio F. Gene therapy in peripheral blood lymphocytes and bone marrow for ADA- immunodeficient patients. Science. 1995;270:470-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 594]  [Cited by in F6Publishing: 606]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
58.  Bulaklak K, Gersbach CA. The once and future gene therapy. Nat Commun. 2020;11:5820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 131]  [Article Influence: 32.8]  [Reference Citation Analysis (0)]
59.  D'Antiga L, Beuers U, Ronzitti G, Brunetti-Pierri N, Baumann U, Di Giorgio A, Aronson S, Hubert A, Romano R, Junge N, Bosma P, Bortolussi G, Muro AF, Soumoudronga RF, Veron P, Collaud F, Knuchel-Legendre N, Labrune P, Mingozzi F. Gene Therapy in Patients with the Crigler-Najjar Syndrome. N Engl J Med. 2023;389:620-631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
60.  Hordeaux J, Ramezani A, Tuske S, Mehta N, Song C, Lynch A, Lupino K, Chichester JA, Buza EL, Dyer C, Yu H, Bell P, Weimer JM, Do H, Wilson JM. Immune transgene-dependent myocarditis in macaques after systemic administration of adeno-associated virus expressing human acid alpha-glucosidase. Front Immunol. 2023;14:1094279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Reference Citation Analysis (0)]
61.  Eggers M, Vannoy CH, Huang J, Purushothaman P, Brassard J, Fonck C, Meng H, Prom MJ, Lawlor MW, Cunningham J, Sadhu C, Mavilio F. Muscle-directed gene therapy corrects Pompe disease and uncovers species-specific GAA immunogenicity. EMBO Mol Med. 2022;14:e13968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
62.  Somanathan S, Calcedo R, Wilson JM. Adenovirus-Antibody Complexes Contributed to Lethal Systemic Inflammation in a Gene Therapy Trial. Mol Ther. 2020;28:784-793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
63.  Chandler RJ, Di Pasquale G, Sloan JL, McCoy S, Hubbard BT, Kilts TM, Manoli I, Chiorini JA, Venditti CP. Systemic gene therapy for methylmalonic acidemia using the novel adeno-associated viral vector 44.9. Mol Ther Methods Clin Dev. 2022;27:61-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
64.  Choi SJ, Yi JS, Lim JA, Tedder TF, Koeberl DD, Jeck W, Desai AK, Rosenberg A, Sun B, Kishnani PS. Successful AAV8 readministration: Suppression of capsid-specific neutralizing antibodies by a combination treatment of bortezomib and CD20 mAb in a mouse model of Pompe disease. J Gene Med. 2023;25:e3509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]