Clin Mol Hepatol > Volume 31(Suppl); 2025 > Article
Caddeo and Romeo: Precision medicine and nucleotide-based therapeutics to treat steatotic liver disease

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a complex multifactorial disease and becoming the leading cause of liver-related morbidity and mortality. MASLD spans from isolated steatosis to metabolic dysfunction-associated steatohepatitis (MASH), that may progress to cirrhosis and hepatocellular carcinoma (HCC). Genetic, metabolic, and environmental factors strongly contribute to the heterogeneity of MASLD. Lifestyle intervention and weight loss represent a viable treatment for MASLD. Moreover, Resmetirom, a thyroid hormone beta receptor agonist, has recently been approved for MASLD treatment. However, most individuals treated did not respond to this therapeutic, suggesting the need for a more tailored approach to treat MASLD. Oligonucleotide-based therapies, namely small-interfering RNA (siRNA) and antisense oligonucleotide (ASO), have been recently developed to tackle MASLD by reducing the expression of genes influencing MASH progression, such as PNPLA3 and HSD17B13. Here, we review the latest progress made in the synthesis and development of oligonucleotide-based agents targeting genetic determinants of MASH.

INTRODUCTION

Metabolic dysfunction–associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), ranges from simple hepatic steatosis to metabolic dysfunction-associated steatohepatitis (MASH), leading to fibrosis, and ultimately to cirrhosis and hepatocellular carcinoma (HCC) [1]. While the early stage of MASLD can be treated by weight loss, the late stage of MASLD can only be tackled by liver transplant [2]. MASLD coexists with other metabolic derangements, namely obesity and hypertriglyceridemia, the two strongest independent predictors of this disease, along with insulin resistance and type 2 diabetes mellitus (T2DM) [3]. Hence, individuals with MASLD have also an intrinsic risk for developing cardio-metabolic syndrome caused by these risk factors [4]. However, it is not clear why an individual enters a specific clinical trajectory leading to liver-related as opposed to cardio-renal-vascular-related events and vice versa. The presence of different clinical trajectories spanning from liver to extrahepatic complications suggests the existence of different types of MASLD with specific disease-causing mechanisms.
In the context of disease heterogeneity, precision medicine aims to find the most efficacious treatment with no side effects by exploiting individual variability in genes, environment, and lifestyle [5]. In a framework of precision medicine, clinical features, genetic background, and omics forge the molecular understanding of diseases offering the means to effectively treat them. In addition, the application of novel techniques and technologies to ensure specific organ and gene targeting may also be considered a form of precision medicine.
In this review, we start by reasoning on the different strategies to implement precision medicine and continue by examining precision medicine based on inborn genetic variants and the use of nucleotide-based therapeutics to treat MASLD.

USING ACQUIRED GENETIC CHANGES TO IMPLEMENT PRECISION MEDICINE

In the field of oncology, understanding the consequences of acquired DNA changes has been used to implement precision medicine for decades. For example, retinoic acid was demonstrated as an effective treatment for individuals with acute promyelocytic leukaemia for the first time in 1988 [6]. Acute promyelocytic leukaemia is caused by a (15;17) (q24;q21) translocation generating a PML-RARA fusion gene. This fusion gene binds to the RAR/RXR target genes and acts as a potent transcriptional repressor hindering its activation by physiological concentrations of ligands, with the ultimate effect of blocking differentiation of granulocytes. Hence, treatment with all trans retinoic acid (ATRA), a potent derivate of retinoids, overcomes this repression with large and unprecedented beneficial effects on survival for this cancer. As one may anticipate, ATRA is still the backbone treatment for this disease [6].
A precision medicine journey starts with the identification of recurrent genetic changes in individuals with cancer. After their identification, understanding the molecular defect caused by the genetic change is paramount to identify drugs overcoming this disorder. Oncologists forged precision medicine roadmaps with several therapeutic regimes today approved when specific genetic variants/rearrangements arise in determined genes. In the future, one may envisage classification and treatment of cancer not based on the organ/cell type the cancerous cells belong to, but on the mutations the cancerous cells harbour.

USING INBORN GENETIC VARIANTS TO IMPLEMENT PRECISION MEDICINE

Inborn, also known as germline, genetic variants are inherited from each biological parent and are stable over a lifetime. MASLD has a strong genetic component with common genetic variants in the PNPLA3 [7,8] and the TM6SF2 having collectively the largest effect sizes [9-11]. This contrasts other multifactorial diseases, including diabetes and obesity, where common genetic variants have less pronounced effect size in determining these traits. However, a common genetic variant in the transcription factor 7-like 2 (TCF7L2) gene, the strongest genetic risk contributor of T2DM progression, has a sizeable allelic relative risk of ~1.4 that is comparable to PNPLA3 and TM6SF2 variants [12].
Another specific feature of common variants increasing the risk of MASLD is the interaction with excess in body weight, insulin resistance and body fat distribution [13-16]. Due to this interaction, in the absence of metabolic stressors genetic variants have a low and sometimes even negligible effect on increasing the risk of MASLD. However, due to the obesity epidemics, these metabolic stressors become every year more present in the general population.
Among treating physicians in Hepatology among other disciplines, there is always the argument on the lack of clinical value of routinely assessing the presence of genetic variants in individuals with MASLD based on the fact that genetic background cannot be changed. On the other hand, there are those in favour arguing that lifestyle changes due to awareness of the potential risk of MASLD will mitigate the risk itself in virtue of the gene-environment interaction. Indeed, the identification of these variants early in life may encourage individuals to strive for reducing weight excess and being more active. Interestingly, Harris et al. [17] showed that the presence of a specific genotype makes less effective the beneficial effect of exercising in MASLD individuals, and that reduction of serum biomarkers of liver fibro-inflammation after weight loss was more pronounced in carriers of a specific genotype.
A case of interest is when a genetic variant minor allele, as for example the HSD17B13 rs72613567:TA variant, results in a loss of function and associates with a protection against MASLD. This event opens the avenue to the identification of therapeutic targets to treat MASLD. Indeed, targeted downregulation/ablation of specific genes is relatively easy now days due to the application of nucleotide-based therapeutics to pharmacology. Over time, all this is leading to a precision medicine approach by giving tailored-lifestyle counselling and targeting specific genetic variants in individuals with MASLD (Fig. 1).

USING MACHINE LEARNING TO IMPLEMENT PRECISION MEDICINE

The development of precision medicine strategies involves the integration of data deriving from clinical health-related records, computational analyses, artificial intelligence (AI), biobanks, and multi-omics analyses [18]. Indeed, AI, machine learning, and deep learning are used to generate accurate in silico predictive models aiming to implement precision medicine in hepatology [19]. Multiparametric data sets generated by using clinical health-related records, imaging techniques and multi-omics, can be integrated by validated algorithms. These algorithms can predict biological processes, functional associations, chemical interactions of a disease-causing protein, diagnosis, clinical outcomes, or the effect of lifestyle interventions by clustering individuals into more homogenous categories [20].
One of the greatest advantages of AI is the possibility to account for complex and nonlinear relationships among traits in the identification of meaningful patterns to predict disease progression. In 2023, Serra-Burriel et al. [21] generated the Liver Risk Score (LRS) and effectively predicted liver fibrosis at a population level by applying machine learning, namely a recursive feature elimination algorithm, to clinically available data [21]. Indeed, the LRS outperformed the other non-invasive tests in predicting fibrosis resolution in individuals after metabolic surgery [22].
Machine learning can also be used to detect independent predictors of disease that may be clinically useful to identify individuals with MASLD. For example, in this study hypertriglyceridemia is, after body mass index, the second strongest independent predictors of liver triglyceride content, while T2DM, contrarily to the epidemiological evidence, is not [3]. Noteworthily, studies on PNPLA3 rs738409 p.I148M, the strongest genetic determinant of MASLD, are leading to the concept of life-style recommendation tailored on this genetic feature. For instance, Davis et al. [23] showed that overweight Hispanic children carrying the PNPLA3 148M/M variant had lower capacity to hydrolyse hepatic triglycerides and, therefore, increased hepatic triglyceride accumulation as compared to wild type, proposing that the use of a personalized low carbohydrate diet in this group would lead to more beneficial results [23]. Moreover, one year-long lifestyle modification in MASLD subjects led to a three-fold higher reduction of intrahepatic triglyceride content in the PNPLA3 148M/M group compared to wild type carriers, suggesting a higher sensitivity of these individuals to lifestyle modification. Indeed, dietary interventions, based on genetic and clinical data, are pivotal to obtain beneficial therapeutic outcomes for MASLD [24]. In addition, AI can be used for treatment optimization, risk assessment, and cost-effectiveness analyses to reduce costs and measure the impact of interventions [25], as the cost of remedying adverse drug reactions is usually higher than the cost of medication itself [26]. Overall, the application of medical AI models in hepatology has the potential to help clinicians in the medical practice from diagnosis to outcome prediction.

USING NUCLEOTIDE-BASED THERAPEUTICS TO IMPLEMENT PRECISION MEDICINE

Nucleotide-based therapeutics, namely small interfering RNA (siRNA) and antisense oligonucleotides (ASO), are techniques capable of selectively ablating a specific gene in a determined disease pathway, thus improving disease phenotypes. A potential drawback of this treatment is the unwanted ablation of off-target genes. However, until now, these treatment modalities have been shown to be well tolerated as they can be administered in very low doses [27,28]. In the pharmacokinetics, the conjugation of these molecules with N-acetylgalactosamine (GalNAc) moieties played a pivotal role. This chemical modification dramatically increases the uptake of the molecule in the hepatocyte due to its binding with the hepatocyte-specific transmembrane asialo-glycoprotein receptor (Ashwell–Morell receptor, ASGPR) that is present specifically on this cell type. This conjugation has allowed unprecedented specificity in targeting the liver and has reduced the drug dose avoiding side effects deriving from the downregulation of the gene in organs other than the liver [29].

FUNDAMENTAL MOLECULAR MECHANISM OF SIRNA

RNA interference (RNAi) is an ancient intracellular defence mechanism through which target mRNAs are degraded before translation as a defence against exogenous genetic material. Small interfering RNAs (also known as short interfering RNA, and silencing RNA, siRNA) have been identified in 1998 by Andrew Fire and Craig Mello [30] in Caenorhabditis elegans granting these basic scientists the 2006 Nobel Prize in Physiology or Medicine award for their discovery.
siRNAs are 20–24-long base pairs double-stranded non-coding RNA molecules composed of a passenger strand (sense strand), used as a drug delivery device, and a guide strand (antisense strand) responsible for triggering RNAi-based gene silencing [31].
siRNA-induced post transcriptional gene silencing starts with the interaction of siRNA with the multidomain type III ribonuclease DICER. DICER, by binding the double-stranded siRNA, cleaves the putative sense strand to generate a mature siRNA that is then loaded onto the RNA-induced silencing complex (RISC) [32]. Alternatively, gene silencing may start with the direct incorporation of siRNA into RISC where its Argonaute-2 component, a doublestranded RNase, degrades the sense strand to form a mature siRNA-RISC complex. The remaining antisense strand, complementary to the mRNA of interest, drives the RISC complex to the mRNA target sequence, thus leading to its cleavage and degradation through the activity of intracellular exonucleases (Fig. 2A). Sequence complementarity between the guide strand and the target mRNA is crucial to reduce unwanted sequence-dependent off-target transcript silencing due to partial homology with the mRNA of interest [33]. Indeed, partial complementary pairing can cause the unpredictable degradation of similar mRNAs, thence inducing the downregulation of untargeted genes.

FUNDAMENTAL ASO MOLECULAR MODE OF ACTION

ASOs are single-stranded nucleic acids (synthetic RNA or DNA sequences) designed to bind the pre- or mature mRNA target through complementary base-pairing. The binding degrades the target transcript and alters the splicing, therefore blocking endogenous or exogenous protein translation [34]. ASOs exert their activity through two mechanisms: a) occupancy-mediated RNA degradation that triggers the cleavage of the target transcript either by Argonaute-2, specific for RNA-RNA-like duplexes, or RNase-H1, that specifically cleaves the RNA– DNA-like heteroduplexes formed by ASOs and the complementary RNA target sequence; and b) occupancy-only mechanism that causes steric interference without any cleavage of the RNA of interest by triggering the alternative splicing of pre-mRNA, and therefore affecting mRNA stability and maturation through the inhibition of the 5′ end CAP formation or the 3′ end polyadenylation, or preventing protein translation by blocking the interaction mRNA-ribosome via steric hindrance (Fig. 2B) [35,36]. Notably, short methylated oligonucleotides modulate gene expression by inducing DNA hypermethylation at specific promoters, thus inhibiting transcription and protein translation for a longer time. Interestingly, DNA methylation is stable even after cell division, leading to a longer-lasting silencing when compared to standard unmethylated oligonucleotides [37].
Depending on the disease molecular mechanism, these therapeutics can downregulate or upregulate the expression of the gene of interest, resulting in lower or higher protein levels. In this regard, ASOS may be used to regulate the methylation of promoter regions of survival motor neuron gene 2 (SMN2) and, therefore, to enhance its gene and protein expression [38]. Indeed, homozygous deletions or mutations in SMN1, a gene highly homologous to SMN2, are responsible for the development of spinal muscular atrophy (SMA) as they lead to the synthesis of a truncated and unstable SMN protein [39,40]. By boosting the transcription of SMN2 and, consequently, the expression of functional SMN protein, mice improved motor function and increased their survival rate [38]. Noteworthily, ASOs activities depend on the intracellular availability of RNase-H1 or Argounate-2. To exert its enzymatic activity, RNAse-H1 needs a very high level of complementarity between the single-strand sequence of ASOs and the target mRNA, thus reducing off-target transcript silencing. Contrarywise, Argonaute-2 does not require the same level of sequence complementarity, and is therefore more likely to result in off target effects [33]. In the last years, several mRNA-targeting ASOs entered clinical trials, such as Mipomersen and Volanesorsen for homozygous familial hypercholesterolaemia and familial chylomicronaemia syndrome, respectively [41,42].

CHEMICAL MODIFICATIONS OF NUCLEOTIDE-BASED THERAPEUTICS TO INCREASE STABILITY

Unmodified oligonucleotides are susceptible to a rapid nuclease-mediated degradation within minutes, they induce the activation of the innate immune system and off-target toxicity due to their intracellular accumulation causing RNAi-dysfunction, and therefore they did not go through in clinical practice [27,43].
However, low stability, specificity, bioavailability, cellular uptake, and the induction of unwanted off-target effects have been mitigated by introducing chemical modifications in three specific sites of the oligonucleotides: phosphate backbone, ribose moiety or nucleobase [27]. These chemical modifications allowed scientists to better understand the gene silencing machinery and to evaluate new modalities aimed to reduce off-target effects and unintended mRNA silencing [34].
Among the phosphates backbone modifications, the phosphorothioate (PS) linkage is one of the most used in oligonucleotides. On the one hand, moderate degree of PS linkage renders oligonucleotides more hydrophobic, resistant to nuclease digestion and easily bound to albumin allowing a longer half-life and higher cellular uptake [44,45]. On the other hand, high degree of PS modification reduces the specificity of the oligonucleotide for its target, thus inducing severe toxic effects [46]. Alternative modifications at the phosphate backbone include 5′-(E)-vinyl phosphonate, phosphorodithioate, methylphosphonate and methoxypropylphosphonate. All these modifications enhance the activity of the oligonucleotide-based drug [27].
Ribose is usually modified by a 2′-O-methyl group, thus improving stability, binding affinity, nuclease resistance and reducing immune reactions [47,48]. Other 2’-sugar-modified oligonucleotides already in clinical trial are characterized by the addition of 2′-methoxyethyl or 2′-fluoro chemical modifications [49].
Nucleobase modifications with uridine analogous and cytidine stabilize the base pair making oligonucleotides more resistant to serum nucleases, and reduce innate immune activation usually triggered by Toll-like receptor-dependent or independent pathways [50].

CHEMICAL MODIFICATIONS OF NUCLEOTIDE-BASED THERAPEUTICS TO ALLOW TARGETED ORGAN DELIVERY

The conjugation of multivalent-GalNAc moieties to the 3’- and 5’-terminals of oligonucleotides allows targeting hepatocyte-specific mRNAs as GalNAc selectively binds the ASGPR, a protein highly expressed in the membrane of hepatocytes while poorly expressed on the surface of other cell types [51]. ASGPR allows the internalization of circulating glycoproteins exposing GalNAc glycans via clathrin-mediated endocytosis [52]. Trivalent-GalNAC conjugation requires the integration of modified ribose and 5’ phosphate resulting in a metabolically stabilized oligonucleotide scaffold with a higher delivery capability [53]. These minor chemical changes and optimized structures have a large impact on the metabolic stability of GalNAc-oligonucleotides conjugates, as they show high stability, specificity, and long-term safety by attenuating the activation of innate immune system, liver-specific uptake, and robust gene silencing of the targeted mRNA in the liver [52]. Importantly, they can be administered subcutaneously monthly or even twice a year, thus potentially increasing compliance [29,54]. For example, Inclisiran, a cholesterol-lowering siRNA conjugated to a triantennary GalNAc targeting proprotein convertase subtilsin/kexin type 9 (PCSK9) mRNA, is currently approved for the treatment of familial hypercholesterolaemia and mixed hyperlipidaemia in adults [55]. Moreover, siRNAs therapies against liver disease are now in clinical studies with some close to being approved as therapies (Table 1).

OLIGONUCLEOTIDE-BASED THERAPEUTICS DELIVERY METHODS

Naked oligonucleotides are hardly uptaken by cells due to their size, high hydrophilicity, and negative charge [46]. Therefore, delivery systems, such as nanoparticles, aptamers, and exosomes, are required to guarantee cellular uptake by energy-dependent endocytosis or pinocytosis [45,56]. These delivery systems are also employed to increase the molecule half-life by avoiding serum RNase A-like nucleases degradation and renal clearance, and to escape immune response [28,29]. Once inside the cell, the delivery system must release the oligonucleotide-based molecule either in the cytoplasm or inside the nucleus where they exert their activity by activating the RISC complex or RNase-H1 [36].

GENETIC THERAPEUTIC TARGETS IN MASLD

Human genetics strongly contributes to the onset and progression of steatotic liver disease (SLD) [57,58]. In the last decades, genome-wide association studies (GWAS) have been used to associate genetic variants in genes involved in lipid metabolism, such as PNPLA3, TM6SF2, MBOAT7, GCKR, HSD17B13, MARC1, APOE and GPAM, with SLD progression or protection (Fig. 3) [58]. Genotyping may also be used to predict liver disease progression. Indeed, the weighted sum of these single nucleotide polymorphisms (SNPs), namely polygenic risk scores (PRS), allows the identification of individuals that will not develop SLD [16,18]. PRS can also be used to re-stratify individuals in fibrosis classes measured by non-invasive predictive tests [15] potentially facilitating early screening, interventions, prioritization of follow up and treatments of high-risk groups. Furthermore, the identification of these genetic variants and the understanding of their fundamental mechanisms have led to several genetically validated therapeutic targets to treat MASLD (Table 1). However, routine genetic screening is available only in tertiary level hospitals.

Patatin-like phospholipase domain-containing protein 3 (PNPLA3)

PNPLA3 is a triacylglycerol lipase that localizes in lipid droplets (LD), endoplasmic reticulum (ER) and Golgi apparatus that hydrolyses triglycerides, phospholipids and retinyl esters, mostly in adipocytes and hepatocytes [59]. PNPLA3 and its protein synthesis are induced during obesity [14], while the PNPLA3 rs738409 variant encodes for a p.I148M substitution that results in a loss of function of the protein enzymatic activity [60]. In addition, the I148M variant compromises protein-mediated retinol release from hepatic stellate cells promoting their activation and thus inflammation, fibrosis and carcinogenesis [61,62].
The PNPLA3 rs738409 polymorphism has been robustly associated with an increased susceptibility to the entire spectrum of hepatic injury development (to MASH and cirrhosis) and increased risk of HCC, by impairing mitochondrial function and antioxidant response, and by hindering lipid and retinol metabolism in hepatocyte and hepatic stellate cells, respectively [63]. Luukkonen et al. [64] showed that PNPLA3 rs738409 carriers have hepatic mitochondrial dysfunction that causes increased hepatic β-oxidation/ketogenesis, impaired de novo lipogenesis and mitochondrial citrate synthase flux that might predispose to the progression from early-stage to late-stage MASLD [64]. In addition, these individuals seem to be protected from cardiovascular disease as they exhibit an antiatherogenic lipid profile, and a better response to lifestyle modifications [24,65-67].
Interestingly, PNPLA3 turnover in hepatocytes is controlled by the bifunctional apoptosis regulator (BFAR) which promotes PNPLA3 degradation by inducing its ubiquitylation [68]. On the contrary, the PNPLA3 I148M mutant isoform eludes ubiquitylation and proteasomal degradation and accumulates in the LDs where it hampers triglyceride breakdown causing impaired very low density lipoprotein (VLDL) secretion [69,70].
Noteworthily, the effect of carrying the 148M mutant protein is larger in women because PNPLA3 expression is regulated by estrogens [71]. From this perspective, a potential PNPLA3 inhibitor may be more effective for the treatment of women with MASLD and carrying the 148M mutant protein.
Pnpla3 silencing by GalNAc-conjugated ASO ameliorated MASLD in human PNPLA3 I148M mutant knock-in mice fed a NASH-inducing diet [72]. Besides, administration of PNPLA3 I148M siRNA in hepatocyte-specific human PNPLA3 I148M knock-in mice re-established PNPLA3 WT phenotype [73]. It is critical to consider that the expression and enzymatic activity of PNPLA3 would not be recovered following the silencing of the I148M variant [58]. To date, several oligonucleotide-based drugs targeting PNPLA3 are in early phase clinical trials to treat SLD (Table 1).

17-beta hydroxysteroid dehydrogenase 13 (HSD17B13)

HSD17B13 is an LD-associated protein expressed only in hepatocytes with a yet-definedfunction. The rs72613567:TA polymorphism encodes for a frameshift resulting in a truncated HSD17B13 protein that is present at a low level in the hepatocytes. The minor allele of this variant associates with lower transaminases and with prevention of MASLD progression that is consistent among several ethnic groups [74-77]. However, the variant protective effect on liver disease without influencing liver steatosis may be elicited through mitigation of inflammation [78,79]. Recently, Ma et al. [80] showed that therapeutic administration of Hsd17b13 ASO induced a dose-dependent inhibition of the hepatic expression of Hsd17b13 and a significant reduction of liver steatosis in a NASH-like hepatic fibrosis mice model, but it did not affect the hepatic fibrosis phenotype [80].
Currently, various oligonucleotide-based drugs targeting HSD17B13 are in early phase clinical trials (Table 1) with some promising findings. Indeed, subcutaneous administration of ARO-HSD was well tolerated and led to a dose-dependent depletion of the HSD17B13 mRNA and protein levels, followed by a reduction of circulating alanine transaminase (ALT) levels in healthy adult volunteers and individuals with MASH [81].

Mitochondrial amidoxime-reducing component 1 (MTARC1)

MTARC1 is a reductase that localizes in the outer membrane of mitochondria and catalyses the reduction of N-oxygenated compounds, thus mediating detoxification reactions under acidic and hypoxic conditions [82,83]. The MTARC1 rs2642438 minor and protective allele encodes for an alanine to threonine substitution at position 165 of the protein (p.A165T) that results in lower intracellular protein levels due to a higher ubiquitin-mediated proteosome degradation [84]. The MTARC1 p.A165T variant does not affect the enzymatic activity of the protein, but its stability [85,86]. This variant is associated with lower hepatic triglyceride content, lobular inflammation, serum ALT, alkaline phosphatase and cholesterol levels, and with protection against MASLD severity and HCC [84,87]. Indeed, MTARC1 deficiency seems to increase intracellular fatty acid oxidation and substrate utilization [88]. Recently, Lewis et al. [89] showed that the administration of GalNAc-siMtarc1 to mice fed the Gubra Amylin NASH diet reduced liver fat content but did not cause phenotypic change in fibrosis despite markers of fibrosis being improved. Importantly, circulating triglyceride levels increased following the GalNAc-siMtarc1 administration [89]. Moreover, Mtarc1 hepatocyte-specific knockdown by GalNAc-Mtarc1 siRNA improved lipid profiles and protected against MASH progression in obese and diet-induced MASH mouse models [90]. To date, the NN6581-4860 siRNA-based drug inhibitor of MTARC1 is currently in a phase 1 clinical trial (Table 1). Interestingly, MTARC1 inhibition may work with the same mechanism of thyroid hormone receptor beta agonist Resmetirom, the first drug approved by the U.S. FDA for treating subjects with noncirrhotic-NASH with moderate to advanced hepatic fibrosis (NCT03900429) [91], namely increase in beta-energy substrate utilization.

Membrane bound O-acyltransferase domain-containing 7 (MBOAT7)

MBOAT7 is a lysophosphatidylinositol acyltransferase (LPIAT1) that localizes in ER, mitochondria-associated membranes and LD [92]. MBOAT7 is a multi-transmembrane protein [93,94] catalysing the acyl-chain remodelling of phospholipids, preferentially transferring polyunsaturated fatty acids (PUFAs), such as arachidonic acid, to phosphatidylinositol [95]. MBOAT7, by finely regulating the availability of acyl-CoA and lysophospholipids, can predispose to the onset and progression of MASLD-driven liver damage [96]. Indeed, the rs641738 C>T variant in the locus containing the MBOAT7 gene has been linked to reduced gene and protein expression, and to increased risk of hepatic steatosis and progressive MASLD, characterized by higher levels of serum ALT, inflammation, fibrosis, and HCC [92,97]. On the other hand, the rs641738 C>T variant has been associated with lower serum triglycerides [98]. Recently, Sharpe et al. [99] showed that hepatic MBOAT7 overexpression in murine NASH models significantly improved hepatic lipid content and serum transaminases levels, but no change in NASH histology was detected [99]. Moreover, hepatocyte-specific MBOAT7 knockout in mice worsened liver steatosis and fibrosis by inducing triglyceride synthesis and the expression of Tafazzin (TAZ), a master regulator of hepatic fibrosis [100,101]. Indeed, Moore et al. [101] proved for the first time that MBOAT7 depletion in hepatocytes induced changes in phospholipids profile by promoting a cholesterol trafficking pathway able to induce the expression and activity of TAZ and Indian hedgehog (IHH), a known TAZ-induced pro-fibrotic factor. Indeed, carriers of the rs641738 C>T variant with MASH have higher TAZ and IHH levels. Conversely, hepatocyte-MBOAT7 restoration in MASH mice reduced the expression of TAZ, thus slowing down fibrosis development [101].
Noteworthily, homozygous carriers of loss of function mutations in MBOAT7 have severe neuronal defects, intellectual disability, autism and epilepsy due to the involvement of MBOAT7 in brain development [102-105].

Transmembrane 6 superfamily member 2 (TM6SF2)

TM6SF2 is a multi-transmembrane protein that localizes in the ER and LD of hepatocytes and intestinal cells [106,107]. The TM6SF2 rs58542926 (p.E167K) minor allele associates with increased MASLD and fibrosis but reduces risk of cardiovascular events [108,109]. The aminoacidic change results in a protein with reduced stability and, therefore, the loss of function causes liver fat accumulation by retention of lipoproteins [110]. Indeed, carriers of the TM6SF2 rs58542926 genetic variant have MASLD due to increased lipid biosynthesis and defective VLDL assembly and secretion. This induces hepatic lipid entrapment resulting in lower serum lipid levels and reduced risk of myocardial infarction [111,112].
The TM6SF2 protein is stabilized by ER lipid raft proteins (ERLINs) and in turns stabilizes apolipoprotein B by two intraluminal loops [113]. Interestingly, the effect of TM6SF2 E167K on MASLD is weakened by the protective effect of the ERLIN1 rs2862954 (p.I291V) loss of function variant against hepatic fat accumulation [114].

Pleckstrin and sec7 domain-containing 3 (PSD3)

PSD3 is a member of the guanine nucleotide exchange factor for ARF-6 (EAF6) family. The rs71519934 variant in the PSD3 gene, resulting in the p.L186T amino acid substitution, has been associated, although with some inconsistencies, with protection against MASLD development [115]. Moreover, PSD3 downregulation by siRNAs in primary and immortalized hepatocytes in vitro, as well as Psd3 silencing by GalNAc-conjugated ASO in mice fed a NASH-inducing diet, ameliorated MASLD [115].

Cell death-inducing DNA fragmentation factor-like effector b (CIDEB)

CIDEB localizes in hepatic ER and LD [116]. CIDEB is an important regulator of the VLDL pathway as it interacts with apolipoprotein B (APOB) to allow triglyceride-enriched VLDL particles biogenesis, lipidation and maturation [116,117]. On the one hand, Cideb-deficient mice showed smaller hepatic LD, decreased de novo lipogenesis and lower lipid exchange activities [118]. On the other hand, they displayed higher hepatic triglyceride content due to reduced VLDL secretion, and lower levels of circulating free fatty acids and triglycerides [116]. Recently, Chen et al. [119] showed that under ER stress the expression of Cideb is suppressed by C/EBP homologous protein (Chop), thus causing the inhibition of VLDL maturation and transport, and inducing hepatic oxidative stress, triglyceride accumulation and inflammation [119]. Strikingly, in a multistage analysis including more than one million individuals, loss-of-function variants plus missense variants in CIDEB have been associated with lower ALT levels and reduced risk of SLD development [120].

OTHER POTENTIAL GENETIC THERAPEUTIC TARGETS IN MASLD

The apolipoprotein E (APOE) rs429358 [121], glucokinase regulator (GCKR) rs1260326 [122] and the glycerol-3-phosphate acyltransferase, mitochondrial (GPAM) rs2792751 [121] genetic variants contribute to MASLD susceptibility. However, these variants are very pleiotropic with effects on multiple traits that make problematic the manipulation of these genes for therapeutic purposes. For example, the GCKR rs1260326 minor allele increases liver triglyceride content by modulating the handling of glucose and triglycerides in hepatocytes. Indeed, carriers of the rs1260326 minor allele show lower serum glucose and insulin levels, have higher LDL cholesterol and circulating triglycerides and lower intrahepatic triglyceride levels [123,124]. The dissociation between intrahepatic and circulating triglycerides makes manipulation of this gene problematic. The APOE rs429358 variant is strongly associated with an increased risk of Alzheimer disease [125], whilst the same minor allele protects against MASLD, making this gene unattractive to target.
Finally, the GPAM rs2792751 minor allele associates with higher hepatic triglyceride content and transaminases [121]. However, while some data suggest that downregulation of this gene would be effective in reducing liver triglyceride content, to the best of our knowledge, targeting this gene for MASLD is not in any current pharmaceutical pipeline.

CONCLUSIONS

Significant progress has been made in the last decade in designing and synthesizing chemically modified oligonucleotides with important metabolic stability, specificity and efficacy capable of post-transcriptional hepatic gene silencing for clinical purposes. One of the main advantages of nucleotide-based therapies is that the target is not a protein with its own three-dimensional structure, stability and specificity that can move into the cells, but they only need to bind with their antisense strand the right nucleotide sequence of the target mRNA. Moreover, they can only have antagonist effects, they are characterized by a very high selectivity, and their synthesis is easier than other tools, such as antibodies [31]. Another significant advantage of modern oligonucleotides is that they can target “undruggable targets” without off target effects due to the interaction with unspecific mRNAs. To date, oligonucleotides technology is rapidly advancing, and it already represents an encouraging novel method for the treatment of rare disease as well as SLD, with promising studies in late-stage clinical development. Thanks to their efficacy, low toxicity, high specificity, and high tissue distribution, oligonucleotides-based drugs will soon represent a pivotal tool for personalized therapeutic strategies for the treatment of both the early and late stages of hepatic disorders [51].

FOOTNOTES

Authors’ contribution
Drafting of the manuscript: AC; Critical revision and finalization of the manuscript: SR. Final approval of the manuscript: AC, SR.
Conflicts of Interest
The authors have no conflicts to disclose.

Figure 1.
Differences between traditional and precision medicine. (A) Traditional medicine is based on a one-size-fits-all approach that involves the use of only rigid, standardized, already defined methods for clinical care, regardless of the background of each individual. (B) Precision medicine is based on the concept that one drug fits a specific subset of individuals stratified based on their (epi)genetic, clinical, environmental, and ethnic/sex background. Created with BioRender.com.

cmh-2024-0438f1.jpg
Figure 2.
Molecular mechanism of action of nucleotide-based therapeutics. (A) ds-siRNA, once introduced into hepatocytes by endocytosis, interacts with the enzyme DICER or with the RNA-induced silencing complex (RISC) that, by degrading its passenger strand, produces a mature ss-siRNA complementary to the mRNA target. (B) ss-ASO either interacts with RNase-H1 or the RISC complex to bind and cleave the complementary mRNA target sequence, or directly interacts with the mRNA or pre-RNA of interest affecting its stability, maturation or causing alternative splicing, thus preventing its translation to protein. Created with BioRender.com.

cmh-2024-0438f2.jpg
Figure 3.
Representative figure of the main validated hepatic gene targets involved in SLD. APOB100, Apolipoprotein B; ATGL (PNPLA2), Patatin like phospholipase domain containing 2; CD36, CD36 Molecule (CD36 Blood Group); CGI-58, comparative gene identification- 58 (Abhydrolase domain containing 5 (ABHD5), Lysophosphatidic acid acyltransferase); CIDEB, Cell death-inducing DNA fragmentation factor-like effector b; DGTA2, Diacylglycerol O-acyltransferase 2; FFA, free fatty acid; GLUT, Glucose transporter; GPAM, Glycerol-3-phosphate acyltransferase 1, Mitochondrial; HSD17B13, 17-beta hydroxysteroid dehydrogenase 1; LD, lipid droplet; MBOAT7, Membrane bound O-acyltransferase domain-containing 7; MTARC1, Mitochondrial amidoxime-reducing component 1; MTTP, Microsomal triglyceride transfer protein; PLA2, Phospholipase A2; PNPLA3, Patatin-like phospholipase domain-containing protein 3; PSD3, Pleckstrin and sec7 domain-containing 3; TM6SF2, Transmembrane 6 superfamily member 2; VLDL, very low density lipoprotein. Created with BioRender.com.

cmh-2024-0438f3.jpg
Table 1.
A summary of nucleotide-based therapeutics (siRNA or ASO) targeting the main genetic therapeutic targets in MASLD currently undergoing clinical trials
Target Compound Developer Type Clinical trial ID Clinical stage Target population Status
PNPLA3 AZD2693 Astrazeneca ASO NCT05809934 Phase 2b Non-cirrhotic non-NASH carriers of the PNPLA3 rs738409 I148M allele Recruiting
D7830C00004
2022-001629-65
PNPLA3 AZD2693 Astrazeneca ASO NCT04483947 D7830C00002 Phase 1 Non-cirrhotic NASH carriers of the PNPLA3 rs738409 I148M allele Completed
PNPLA3 AZD2693 Astrazeneca ASO NCT05107336 Phase 1 Healthy participants Completed
D7830C00006
PNPLA3 AZD2693 Astrazeneca ASO NCT04142424 Phase 1 Subjects of non-childbearing potential in overweight, but otherwise healthy subjects, and healthy chinese and japanese subjects Completed
D7830C00001
PNPLA3 AZD2693 Astrazeneca ASO NCT05919069 Phase 1 Individuals with hepatic impairment Recruiting
D7830C00008
PNPLA3 LY3849891 Eli Lilly and Company siRNA NCT05395481 Phase 1 NAFLD subjects carriers of the PNPLA3 I148M allele Recruiting
18214
J3W-MC-GZOA
PNPLA3 ALN-PNP Regeneron Pharma- ceuticals siRNA NCT05648214 Phase 1 NAFLD subjects carriers of a specific variant of the PNPLA3 gene Recruiting
ALN-PNP-HV-2227
PNPLA3 ALN-PNP Regeneron Pharma- ceuticals siRNA NCT06024408 Phase 1 Participants with NAFLD and a PNPLA3 genetic risk Factor Not yet recruiting
ALN-PNP-NASH-2255
PNPLA3 AMG 609 Amgen siRNA NCT04857606 Phase 1 MASLD and MASH individuals Completed
20200001
PNPLA3 JNJ-75220795 Janssen Research & Development, LLC siRNA NCT04844450 Phase 1 Participants with certain genetic predis-position to NAFLD Completed
ARO-PNPLA3 CR108997
75220795NAS1001
HSD17B13 ARO-HSD Arrowhead Pharma- ceuticals Inc. siRNA NCT04202354 Phase 1/2 Healthy volunteers and individuals with NASH or suspected NASH Completed
AROHSD1001
HSD17B13 AZD7503 AstraZeneca ASO NCT05560607 Phase 1 NAFLD or NASH individuals Completed
D9230C00003
HSD17B13 AZD7503 AstraZeneca ASO NCT06093542 Phase 1 Healthy Japanese participants Completed
D9230C00005
HSD17B13 AZD7503 AstraZeneca ASO NCT05864391 Phase 1 Individuals with suspected Non-cirrhotic NASH Recruiting
D9230C00002
HSD17B13 ALN-HSD Regeneron Pharmaceuticals siRNA NCT05519475 ALN-HSDNASH-2130 Phase 2 NASH participants with genetic risk factors Not yet recruiting
HSD17B13 ALN-HSD Alnylam Pharmaceuticals siRNA NCT04565717 Phase 1 Adult subjects with NASH Terminated
HSD17B13 GSK4532990 GlaxoSmithKline siRNA NCT06104319 Phase 2 Adult participants with NASH or suspected NASH Recruiting
218675
HSD17B13 GSK4532990 GlaxoSmithKline siRNA NCT05583344 Phase 2 b Adults with NASH Recruiting
218672
2022-002538-14
2023-507503-62-00
HSD17B13 VSA006 Visirna Therapeutics siRNA NCT06322628 Phase 2 Chinese NASH individuals Not yet recruiting
HK Limited VSA006-2001
MTARC1 NN6581-4860 Novo Nordisk A/S siRNA NCT05599945 Phase 1 Healthy adults Recruiting
NN6581-4860

siRNA, small interfering RNA; ASO, antisense oligonucleotide; MASLD, Metabolic dysfunction–associated steatotic liver disease; PNPLA3, Patatin-like phospholipase domain-containing protein 3; HSD17B13, 17-beta hydroxysteroid dehydrogenase 13; MTARC1, Mitochondrial amidoxime-reducing component 1; NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis.

Abbreviations

AI
artificial intelligence
ALT
alanine transaminase
ASGPR
asialo-glycoprotein receptor
ASO
antisense oligonucleotide
ATRA
all trans retinoic acid
BFAR
bifunctional apoptosis regulator
CIDEB
Cell death-inducing DNA fragmentation factor-like effector b
ER
endoplasmic reticulum
ERLIN
ER lipid raft proteins
GalNAc
N-acetylgalactosamine
GAN
Gubra Amylin NASH diet
GWAS
genome-wide association study
HCC
hepatocellular carcinoma
HSD17B13
17-beta hydroxysteroid dehydrogenase 13
IHH
Indian hedgehog
LD
lipid droplet
LPIAT1
lysophosphatidylinositol acyltransferase 1
LRS
liver risk score
MASH
metabolic dysfunction-associated steatohepatitis
MASLD
Metabolic dysfunction–associated steatotic liver disease
MBOAT7
Membrane bound O-acyltransferase domain-containing 7
MTARC1
Mitochondrial amidoxime-reducing component 1
NAFLD
non-alcoholic fatty liver disease
PCSK9
proprotein convertase subtilsin/kexin type 9
PNPLA3
Patatin-like phospholipase domain-containing protein 3
PS
phosphorothioate
PSD3
Pleckstrin and sec7 domain-containing 3
PUFA
polyunsaturated fatty acid
RISC
RNA-induced silencing complex
RNAi
RNA interference
siRNA
small interfering RNA
SLD
steatotic liver disease
SNP
single nucleotide polymorphism
TAZ
Transcriptional Coactivator With PDZ-Binding Motif
T2DM
type 2 diabetes mellitus
TM6SF2
Transmembrane 6 superfamily member 2
TCF7L2
transcription factor 7-like 2
VLDL
very low density lipoprotein

REFERENCES

1. Rinella ME, Lazarus JV, Ratziu V, Francque SM, Sanyal AJ, Kanwal F, et al.; NAFLD Nomenclature consensus group. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. J Hepatol 2023;79:1542-1556.
pmid
2. Müller PC, Kabacam G, Vibert E, Germani G, Petrowsky H. Current status of liver transplantation in Europe. Int J Surg 2020;82S:22-29.
pmid
3. Tavaglione F, Marafioti G, Romeo S, Jamialahmadi O. Machine learning reveals the contribution of lipoproteins to liver triglyceride content and inflammation. J Clin Endocrinol Metab 2024;dgae371.
crossref pdf
4. Wang X, Zhang L, Dong B. Molecular mechanisms in MASLD/MASH-related HCC. Hepatology 2024 Feb 13;doi: 10.1097/HEP.0000000000000786.
crossref
5. Lonardo A. Precision medicine in nonalcoholic fatty liver disease. J Gastroenterol Hepatol 2022;37:1175-1178.
crossref pmid pdf
6. Sanz MA, Fenaux P, Tallman MS, Estey EH, Löwenberg B, Naoe T, et al. Management of acute promyelocytic leukemia: updated recommendations from an expert panel of the European LeukemiaNet. Blood 2019;133:1630-1643.
crossref pmid pmc pdf
7. Sookoian S, Pirola CJ. Meta-analysis of the influence of I148M variant of patatin-like phospholipase domain containing 3 gene (PNPLA3) on the susceptibility and histological severity of nonalcoholic fatty liver disease. Hepatology 2011;53:1883-1894.
crossref pmid pdf
8. Sookoian S, Castaño GO, Burgueño AL, Gianotti TF, Rosselli MS, Pirola CJ. A nonsynonymous gene variant in the adiponutrin gene is associated with nonalcoholic fatty liver disease severity. J Lipid Res 2009;50:2111-2116.
crossref pmid pmc
9. Trépo E, Valenti L. Update on NAFLD genetics: from new variants to the clinic. J Hepatol 2020;72:1196-1209.
crossref pmid
10. Pelusi S, Baselli G, Pietrelli A, Dongiovanni P, Donati B, McCain MV, et al. Rare pathogenic variants predispose to hepatocellular carcinoma in nonalcoholic fatty liver disease. Sci Rep 2019;9:3682.
crossref pmid pmc pdf
11. Anstee QM, Darlay R, Cockell S, Meroni M, Govaere O, Tiniakos D, et al.; EPoS Consortium Investigators. Genomewide association study of non-alcoholic fatty liver and steatohepatitis in a histologically characterised cohort. J Hepatol 2020;73:505-515. J Hepatol 2020;73:505-515 Erratum in: J Hepatol 2021;74:1274-1275. Erratum in: J Hepatol 2023;78:1085-1086.
pmid
12. Grant SF, Thorleifsson G, Reynisdottir I, Benediktsson R, Manolescu A, Sainz J, et al. Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 2006;38:320-323.
crossref pmid pdf
13. Barata L, Feitosa MF, Bielak LF, Halligan B, Baldridge AS, Guo X, et al. Insulin resistance exacerbates genetic predisposition to nonalcoholic fatty liver disease in individuals without diabetes. Hepatol Commun 2019;3:894-907.
crossref pmid pmc pdf
14. Stender S, Kozlitina J, Nordestgaard BG, Tybjærg-Hansen A, Hobbs HH, Cohen JC. Adiposity amplifies the genetic risk of fatty liver disease conferred by multiple loci. Nat Genet 2017;49:842-847.
crossref pmid pmc pdf
15. De Vincentis A, Tavaglione F, Jamialahmadi O, Picardi A, Antonelli Incalzi R, Valenti L, et al. A polygenic risk score to refine risk stratification and prediction for severe liver disease by clinical fibrosis scores. Clin Gastroenterol Hepatol 2022;20:658-673.
crossref pmid
16. Bianco C, Jamialahmadi O, Pelusi S, Baselli G, Dongiovanni P, Zanoni I, et al. Non-invasive stratification of hepatocellular carcinoma risk in non-alcoholic fatty liver using polygenic risk scores. J Hepatol 2021;74:775-782.
crossref pmid pmc
17. Harris SJ, Smith N, Hummer B, Schreibman IR, Faust AJ, Geyer NR, et al. Exercise training improves serum biomarkers of liver fibroinflammation in patients with metabolic dysfunction-associated steatohepatitis. Liver Int 2024;44:532-540.
crossref pmid pmc
18. Sookoian S, Pirola CJ. Precision medicine in nonalcoholic fatty liver disease: new therapeutic insights from genetics and systems biology. Clin Mol Hepatol 2020;26:461-475.
crossref pmid pmc pdf
19. Su TH, Wu CH, Kao JH. Artificial intelligence in precision medicine in hepatology. J Gastroenterol Hepatol 2021;36:569-580.
crossref pmid pdf
20. Vora LK, Gholap AD, Jetha K, Thakur RRS, Solanki HK, Chavda VP. Artificial intelligence in pharmaceutical technology and drug delivery design. Pharmaceutics 2023;15:1916.
crossref pmid pmc
21. Serra-Burriel M, Juanola A, Serra-Burriel F, Thiele M, Graupera I, Pose E, et al.; LiverScreen Consortium Investigators. Development, validation, and prognostic evaluation of a risk score for long-term liver-related outcomes in the general population: a multicohort study. Lancet 2023;402:988-996 Erratum in: Lancet 2023;402:964.
pmid
22. Raverdy V, Tavaglione F, Chatelain E, Caiazzo R, Saponaro C, Lassailly G, et al. Performance of non-invasive tests for liver fibrosis resolution after bariatric surgery. Metabolism 2024;153:155790.
crossref pmid
23. Davis JN, Lê KA, Walker RW, Vikman S, Spruijt-Metz D, Weigensberg MJ, et al. Increased hepatic fat in overweight Hispanic youth influenced by interaction between genetic variation in PNPLA3 and high dietary carbohydrate and sugar consumption. Am J Clin Nutr 2010;92:1522-1527.
crossref pmid pmc
24. Shen J, Wong GL, Chan HL, Chan RS, Chan HY, Chu WC, et al. PNPLA3 gene polymorphism and response to lifestyle modification in patients with nonalcoholic fatty liver disease. J Gastroenterol Hepatol 2015;30:139-146.
pmid
25. Lai-Hung Wong G, Sung JJ. The emerging role of big data in gastroenterology and hepatology. J Gastroenterol Hepatol 2019;34:307-308.
crossref pmid pdf
26. Lorbek G, Urlep Ž, Rozman D. Pharmacogenomic and personalized approaches to tackle nonalcoholic fatty liver disease. Pharmacogenomics 2016;17:1273-1288.
crossref pmid
27. Hu B, Zhong L, Weng Y, Peng L, Huang Y, Zhao Y, et al. Therapeutic siRNA: state of the art. Signal Transduct Target Ther 2020;5:101.
crossref pmid pmc pdf
28. Setten RL, Rossi JJ, Han SP. The current state and future directions of RNAi-based therapeutics. Nat Rev Drug Discov 2019;18:421-446 Erratum in: Nat Rev Drug Discov 2020;19:291. Erratum in: Nat Rev Drug Discov 2020;19:290.
crossref pmid pdf
29. Zhang J, Chen B, Gan C, Sun H, Zhang J, Feng L. A comprehensive review of small interfering RNAs (siRNAs): mechanism, therapeutic targets, and delivery strategies for cancer therapy. Int J Nanomedicine 2023;18:7605-7635.
crossref pmid pmc pdf
30. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998;391:806-811.
crossref pmid pdf
31. Alshaer W, Zureigat H, Al Karaki A, Al-Kadash A, Gharaibeh L, Hatmal MM, et al. siRNA: mechanism of action, challenges, and therapeutic approaches. Eur J Pharmacol 2021;905:174178 Erratum in: Eur J Pharmacol 2022;916:174741.
crossref pmid
32. Lee YY, Kim H, Kim VN. Sequence determinant of small RNA production by DICER. Nature 2023;615:323-330.
crossref pmid pdf
33. Jackson AL, Burchard J, Schelter J, Chau BN, Cleary M, Lim L, et al. Widespread siRNA “off-target” transcript silencing mediated by seed region sequence complementarity. RNA 2006;12:1179-1187.
crossref pmid pmc
34. Bajan S, Hutvagner G. RNA-based therapeutics: from antisense oligonucleotides to miRNAs. Cells 2020;9:137.
crossref pmid pmc
35. Crooke ST, Liang XH, Crooke RM, Baker BF, Geary RS. Antisense drug discovery and development technology considered in a pharmacological context. Biochem Pharmacol 2021;189:114196.
crossref pmid
36. Gheibi-Hayat SM, Jamialahmadi K. Antisense oligonucleotide (AS-ODN) technology: principle, mechanism and challenges. Biotechnol Appl Biochem 2021;68:1086-1094.
crossref pmid pdf
37. Hoffman AR, Hu JF. Directing DNA methylation to inhibit gene expression. Cell Mol Neurobiol 2006;26:425-438.
crossref pmid pmc pdf
38. Wang J, Bai J, OuYang S, Wang H, Jin Y, Peng X, et al. Antisense oligonucleotides targeting the SMN2 promoter region enhance SMN2 expression in spinal muscular atrophy cell lines and mouse model. Hum Mol Genet 2022;31:1635-1650.
crossref pmid pdf
39. Monani UR, Lorson CL, Parsons DW, Prior TW, Androphy EJ, Burghes AH, et al. A single nucleotide difference that alters splicing patterns distinguishes the SMA gene SMN1 from the copy gene SMN2. Hum Mol Genet 1999;8:1177-1183.
crossref pmid
40. Lefebvre S, Bürglen L, Reboullet S, Clermont O, Burlet P, Viollet L, et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 1995;80:155-165.
crossref pmid
41. Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, et al. Volanesorsen and triglyceride levels in familial chylomicronemia syndrome. N Engl J Med 2019;381:531-542.
crossref pmid
42. Hair P, Cameron F, McKeage K. Mipomersen sodium: first global approval. Drugs 2013;73:487-493.
crossref pmid pdf
43. Layzer JM, McCaffrey AP, Tanner AK, Huang Z, Kay MA, Sullenger BA. In vivo activity of nuclease-resistant siRNAs. RNA 2004;10:766-771.
crossref pmid pmc
44. Eckstein F. Phosphorothioates, essential components of therapeutic oligonucleotides. Nucleic Acid Ther 2014;24:374-387.
crossref pmid
45. Koller E, Vincent TM, Chappell A, De S, Manoharan M, Bennett CF. Mechanisms of single-stranded phosphorothioate modified antisense oligonucleotide accumulation in hepatocytes. Nucleic Acids Res 2011;39:4795-4807.
crossref pmid pmc
46. Ku SH, Jo SD, Lee YK, Kim K, Kim SH. Chemical and structural modifications of RNAi therapeutics. Adv Drug Deliv Rev 2016;104:16-28.
crossref pmid
47. Monia BP, Johnston JF, Sasmor H, Cummins LL. Nuclease resistance and antisense activity of modified oligonucleotides targeted to Ha-ras. J Biol Chem 1996;271:14533-14540.
pmid
48. Robbins M, Judge A, Liang L, McClintock K, Yaworski E, MacLachlan I. 2’-O-methyl-modified RNAs act as TLR7 antagonists. Mol Ther 2007;15:1663-1669.
crossref pmid
49. Khvorova A, Watts JK. The chemical evolution of oligonucleotide therapies of clinical utility. Nat Biotechnol 2017;35:238-248.
crossref pmid pmc pdf
50. Judge A, MacLachlan I. Overcoming the innate immune response to small interfering RNA. Hum Gene Ther 2008;19:111-124.
crossref pmid
51. Lu K, Fan Q, Zou X. Antisense oligonucleotide is a promising intervention for liver diseases. Front Pharmacol 2022;13:1061842.
crossref pmid pmc
52. Nair JK, Willoughby JL, Chan A, Charisse K, Alam MR, Wang Q, et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc 2014;136:16958-16961.
crossref pmid
53. Prakash TP, Kinberger GA, Murray HM, Chappell A, Riney S, Graham MJ, et al. Synergistic effect of phosphorothioate, 5’-vinylphosphonate and GalNAc modifications for enhancing activity of synthetic siRNA. Bioorg Med Chem Lett 2016;26:2817-2820.
crossref pmid
54. Foster DJ, Brown CR, Shaikh S, Trapp C, Schlegel MK, Qian K, et al. Advanced siRNA designs further improve in vivo performance of GalNAc-siRNA conjugates. Mol Ther 2018;26:708-717.
crossref pmid pmc
55. Lamb YN. Inclisiran: first approval. Drugs 2021;81:389-395 Erratum in: Drugs 2021;81:1129.
crossref pmid pmc pdf
56. Karaki S, Benizri S, Mejías R, Baylot V, Branger N, Nguyen T, et al. Lipid-oligonucleotide conjugates improve cellular uptake and efficiency of TCTP-antisense in castration-resistant prostate cancer. J Control Release 2017;258:1-9.
crossref pmid
57. Huang G, Wallace DF, Powell EE, Rahman T, Clark PJ, Subramaniam VN. Gene variants implicated in steatotic liver disease: opportunities for diagnostics and therapeutics. Biomedicines 2023;11:2809.
crossref pmid pmc
58. Lindén D, Romeo S. Therapeutic opportunities for the treatment of NASH with genetically validated targets. J Hepatol 2023;79:1056-1064.
crossref pmid
59. Sherman DJ, Liu L, Mamrosh JL, Xie J, Ferbas J, Lomenick B, et al. The fatty liver disease-causing protein PNPLA3-I148M alters lipid droplet-Golgi dynamics. Proc Natl Acad Sci U S A 2024;121:e2318619121.
crossref pmid pmc
60. Pingitore P, Pirazzi C, Mancina RM, Motta BM, Indiveri C, Pujia A, et al. Recombinant PNPLA3 protein shows triglyceride hydrolase activity and its I148M mutation results in loss of function. Biochim Biophys Acta 2014;1841:574-580.
crossref pmid
61. Pirazzi C, Valenti L, Motta BM, Pingitore P, Hedfalk K, Mancina RM, et al. PNPLA3 has retinyl-palmitate lipase activity in human hepatic stellate cells. Hum Mol Genet 2014;23:4077-4085.
crossref pmid pmc
62. Pingitore P, Dongiovanni P, Motta BM, Meroni M, Lepore SM, Mancina RM, et al. PNPLA3 overexpression results in reduction of proteins predisposing to fibrosis. Hum Mol Genet 2016;25:5212-5222.
crossref pmid pmc
63. Caon E, Martins M, Hodgetts H, Blanken L, Vilia MG, Levi A, et al. Exploring the impact of the PNPLA3 I148M variant on primary human hepatic stellate cells using 3D extracellular matrix models. J Hepatol 2024;80:941-956.
crossref pmid
64. Luukkonen PK, Porthan K, Ahlholm N, Rosqvist F, Dufour S, Zhang XM, et al. The PNPLA3 I148M variant increases ketogenesis and decreases hepatic de novo lipogenesis and mitochondrial function in humans. Cell Metab 2023;35:1887-1896 e5.
crossref pmid
65. Luukkonen PK, Qadri S, Lehtimäki TE, Juuti A, Sammalkorpi H, Penttilä AK, et al. The PNPLA3-I148M variant confers an antiatherogenic lipid profile in insulin-resistant patients. J Clin Endocrinol Metab 2021;106:e300-e315.
crossref pmid pdf
66. Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, Pennacchio LA, et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet 2008;40:1461-1465.
crossref pmid pmc pdf
67. Santos RD, Valenti L, Romeo S. Does nonalcoholic fatty liver disease cause cardiovascular disease? Current knowledge and gaps. Atherosclerosis 2019;282:110-120.
crossref pmid
68. Das A, Cheng H, Wang Y, Kinch LN, Liang G, Hong S, et al. The ubiquitin E3 ligase BFAR promotes degradation of PNPLA3. Proc Natl Acad Sci U S A 2024;121:e2312291121.
crossref pmid pmc
69. Sookoian S, Pirola CJ, Valenti L, Davidson NO. Genetic pathways in nonalcoholic fatty liver disease: insights from systems biology. Hepatology 2020;72:330-346.
crossref pmid pmc pdf
70. BasuRay S, Smagris E, Cohen JC, Hobbs HH. The PNPLA3 variant associated with fatty liver disease (I148M) accumulates on lipid droplets by evading ubiquitylation. Hepatology 2017;66:1111-1124.
crossref pmid pmc pdf
71. Cherubini A, Ostadreza M, Jamialahmadi O, Pelusi S, Rrapaj E, Casirati E, et al. Interaction between estrogen receptor-α and PNPLA3 p.I148M variant drives fatty liver disease susceptibility in women. Nat Med 2023;29:2643-2655 Erratum in: Nat Med 2024;30:1212.
pmid pmc
72. Lindén D, Ahnmark A, Pingitore P, Ciociola E, Ahlstedt I, Andréasson AC, et al. Pnpla3 silencing with antisense oligonucleotides ameliorates nonalcoholic steatohepatitis and fibrosis in Pnpla3 I148M knock-in mice. Mol Metab 2019;22:49-61.
crossref pmid pmc
73. Banini BA, Kumar DP, Cazanave S, Seneshaw M, Mirshahi F, Santhekadur PK, et al. Identification of a metabolic, transcriptomic, and molecular signature of patatin-like phospholipase domain containing 3-mediated acceleration of steatohepatitis. Hepatology 2021;73:1290-1306.
crossref pmid pmc pdf
74. Kubiliun MJ, Cohen JC, Hobbs HH, Kozlitina J. Contribution of a genetic risk score to ethnic differences in fatty liver disease. Liver Int 2022;42:2227-2236.
crossref pmid pmc pdf
75. Kallwitz E, Tayo BO, Kuniholm MH, Daviglus M, Zeng D, Isasi CR, et al. Association of HSD17B13 rs72613567:TA with non-alcoholic fatty liver disease in Hispanics/Latinos. Liver Int 2020;40:889-893.
crossref pmid pdf
76. Stender S, Romeo S. HSD17B13 as a promising therapeutic target against chronic liver disease. Liver Int 2020;40:756-757.
crossref pmid pdf
77. Raja AM, Ciociola E, Ahmad IN, Dar FS, Naqvi SMS, Moaeen-Ud-Din M, et al. Genetic susceptibility to chronic liver disease in individuals from Pakistan. Int J Mol Sci 2020;21:3558.
crossref pmid pmc
78. Abul-Husn NS, Cheng X, Li AH, Xin Y, Schurmann C, Stevis P, et al. A protein-truncating HSD17B13 variant and protection from chronic liver disease. N Engl J Med 2018;378:1096-1106.
pmid pmc
79. Pirola CJ, Garaycoechea M, Flichman D, Arrese M, San Martino J, Gazzi C, et al. Splice variant rs72613567 prevents worst histologic outcomes in patients with nonalcoholic fatty liver disease. J Lipid Res 2019;60:176-185.
crossref pmid pmc
80. Ma Y, Cai H, Smith J, Chu CH, Mercer SE, Boehm S, et al. Evaluation of antisense oligonucleotide therapy targeting Hsd17b13 in a fibrosis mice model. J Lipid Res 2024;65:100514.
crossref pmid pmc
81. Mak LY, Gane E, Schwabe C, Yoon KT, Heo J, Scott R, et al. A phase I/II study of ARO-HSD, an RNA interference therapeutic, for the treatment of non-alcoholic steatohepatitis. J Hepatol 2023;78:684-692.
crossref pmid
82. Klein JM, Busch JD, Potting C, Baker MJ, Langer T, Schwarz G. The mitochondrial amidoxime-reducing component (mARC1) is a novel signal-anchored protein of the outer mitochondrial membrane. J Biol Chem 2012;287:42795-42803.
crossref pmid pmc
83. Kubitza C, Bittner F, Ginsel C, Havemeyer A, Clement B, Scheidig AJ. Crystal structure of human mARC1 reveals its exceptional position among eukaryotic molybdenum enzymes. Proc Natl Acad Sci U S A 2018;115:11958-11963.
crossref pmid pmc
84. Dutta T, Sasidharan K, Ciociola E, Pennisi G, Noto FR, Kovooru L, et al. Mitochondrial amidoxime-reducing component 1 p.Ala165Thr increases protein degradation mediated by the proteasome. Liver Int 2024;44:1219-1232.
pmid
85. Ott G, Reichmann D, Boerger C, Cascorbi I, Bittner F, Mendel RR, et al. Functional characterization of protein variants encoded by nonsynonymous single nucleotide polymorphisms in MARC1 and MARC2 in healthy Caucasians. Drug Metab Dispos 2014;42:718-725.
crossref pmid
86. Wu M, Tie M, Hu L, Yang Y, Chen Y, Ferguson D, et al. Fatty liver disease protective MTARC1 p.A165T variant reduces the protein stability of MTARC1. Biochem Biophys Res Commun 2024;702:149655.
crossref pmid pmc
87. Emdin CA, Haas ME, Khera AV, Aragam K, Chaffin M, Klarin D, et al. A missense variant in Mitochondrial Amidoxime Reducing Component 1 gene and protection against liver disease. PLoS Genet 2020;16:e1008629 Erratum in: PLoS Genet 2021;17:e1009503.
crossref pmid pmc
88. Jones AK, Bajrami B, Campbell MK, Erzurumluoglu AM, Guo Q, Chen H, et al. mARC1 in MASLD: modulation of lipid accumulation in human hepatocytes and adipocytes. Hepatol Commun 2024;8:e0365.
crossref pmid pmc
89. Lewis LC, Chen L, Hameed LS, Kitchen RR, Maroteau C, Nagarajan SR, et al. Hepatocyte mARC1 promotes fatty liver disease. JHEP Rep 2023;5:100693.
crossref pmid pmc
90. Guo Y, Gao Z, LaGory EL, Kristin LW, Gupte J, Gong Y, et al. Liver-specific mitochondrial amidoxime-reducing component 1 (Mtarc1) knockdown protects the liver from diet-induced MASH in multiple mouse models. Hepatol Commun 2024;8:e0419.
crossref pmid pmc
91. Harrison SA, Bedossa P, Guy CD, Schattenberg JM, Loomba R, Taub R, et al.; MAESTRO-NASH Investigators. A phase 3, randomized, controlled trial of resmetirom in NASH with liver fibrosis. N Engl J Med 2024;390:497-509.
pmid
92. Mancina RM, Dongiovanni P, Petta S, Pingitore P, Meroni M, Rametta R, et al. The MBOAT7-TMC4 variant rs641738 increases risk of nonalcoholic fatty liver disease in individuals of European descent. Gastroenterology 2016;150:1219-1230 e6.
crossref pmid pmc
93. Caddeo A, Jamialahmadi O, Solinas G, Pujia A, Mancina RM, Pingitore P, et al. MBOAT7 is anchored to endomembranes by six transmembrane domains. J Struct Biol 2019;206:349-360.
crossref pmid
94. Wang K, Lee CW, Sui X, Kim S, Wang S, Higgs AB, et al. The structure of phosphatidylinositol remodeling MBOAT7 reveals its catalytic mechanism and enables inhibitor identification. Nat Commun 2023;14:3533.
crossref pmid pmc pdf
95. Caddeo A, Hedfalk K, Romeo S, Pingitore P. LPIAT1/MBOAT7 contains a catalytic dyad transferring polyunsaturated fatty acids to lysophosphatidylinositol. Biochim Biophys Acta Mol Cell Biol Lipids 2021;1866:158891.
crossref pmid
96. Caddeo A, Spagnuolo R, Maurotti S. MBOAT7 in liver and extrahepatic diseases. Liver Int 2023;43:2351-2364.
pmid
97. Donati B, Dongiovanni P, Romeo S, Meroni M, McCain M, Miele L, et al. MBOAT7 rs641738 variant and hepatocellular carcinoma in non-cirrhotic individuals. Sci Rep 2017;7:4492.
crossref pmid pmc pdf
98. Teo K, Abeysekera KWM, Adams L, Aigner E, Anstee QM, Banales JM, et al. rs641738C>T near MBOAT7 is associated with liver fat, ALT and fibrosis in NAFLD: a meta-analysis. J Hepatol 2021;74:20-30.
pmid pmc
99. Sharpe MC, Pyles KD, Hallcox T, Kamm DR, Piechowski M, Fisk B, et al. Enhancing hepatic MBOAT7 expression in mice with nonalcoholic steatohepatitis. Gastro Hep Adv 2023;2:558-572.
crossref pmid pmc
100. Tanaka Y, Shimanaka Y, Caddeo A, Kubo T, Mao Y, Kubota T, et al. LPIAT1/MBOAT7 depletion increases triglyceride synthesis fueled by high phosphatidylinositol turnover. Gut 2021;70:180-193.
crossref pmid pmc
101. Moore MP, Wang X, Kennelly JP, Shi H, Ishino Y, Kano K, et al. Low MBOAT7 expression, a genetic risk for MASH, promotes a profibrotic pathway involving hepatocyte TAZ upregulation. Hepatology 2025;81:576-590.
crossref pmid
102. Heidari E, Caddeo A, Zarabadi K, Masoudi M, Tavasoli AR, Romeo S, et al. Identification of novel loss of function variants in MBOAT7 resulting in intellectual disability. Genomics 2020;112:4072-4077.
crossref pmid
103. Lee J, Shamim A, Park J, Jang JH, Kim JH, Kwon JY, et al. Functional and structural changes in the membrane-bound O-acyltransferase family member 7 (MBOAT7) protein: the pathomechanism of a novel MBOAT7 variant in patients with intellectual disability. Front Neurol 2022;13:836954.
crossref pmid pmc
104. Li H, Qi Z, Xie L, Hao C, Li W. The first Chinese intellectual developmental disorder, autosomal recessive 57 patient with two novel MBOAT7 variants. Mol Genet Genomic Med 2024;12:e2391.
crossref pmid pmc
105. Nazmina G, Khan A, Jiang J, Miao Z, Khan SN, Khan MI, et al. Exome sequencing identifies homozygous variants in MBOAT7 associated with neurodevelopmental disorder. Clin Genet 2024;105:423-429.
crossref pmid
106. O’Hare EA, Yang R, Yerges-Armstrong LM, Sreenivasan U, McFarland R, Leitch CC, et al. TM6SF2 rs58542926 impacts lipid processing in liver and small intestine. Hepatology 2017;65:1526-1542.
crossref pmid pmc pdf
107. Mahdessian H, Taxiarchis A, Popov S, Silveira A, Franco-Cereceda A, Hamsten A, et al. TM6SF2 is a regulator of liver fat metabolism influencing triglyceride secretion and hepatic lipid droplet content. Proc Natl Acad Sci U S A 2014;111:8913-8918.
crossref pmid pmc
108. Kozlitina J, Smagris E, Stender S, Nordestgaard BG, Zhou HH, Tybjærg-Hansen A, et al. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nat Genet 2014;46:352-356.
crossref pmid pmc pdf
109. Longo M, Meroni M, Paolini E, Erconi V, Carli F, Fortunato F, et al. TM6SF2/PNPLA3/MBOAT7 loss-of-function genetic variants impact on NAFLD development and progression both in patients and in in vitro models. Cell Mol Gastroenterol Hepatol 2022;13:759-788.
crossref pmid
110. Luo F, Smagris E, Martin SA, Vale G, McDonald JG, Fletcher JA, et al. Hepatic TM6SF2 is required for lipidation of VLDL in a pre-Golgi compartment in mice and rats. Cell Mol Gastroenterol Hepatol 2022;13:879-899.
crossref pmid pmc
111. Dongiovanni P, Petta S, Maglio C, Fracanzani AL, Pipitone R, Mozzi E, et al. Transmembrane 6 superfamily member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease. Hepatology 2015;61:506-514.
crossref pmid pdf
112. Prill S, Caddeo A, Baselli G, Jamialahmadi O, Dongiovanni P, Rametta R, et al. The TM6SF2 E167K genetic variant induces lipid biosynthesis and reduces apolipoprotein B secretion in human hepatic 3D spheroids. Sci Rep 2019;9:11585.
crossref pmid pmc pdf
113. Li BT, Sun M, Li YF, Wang JQ, Zhou ZM, Song BL, et al. Disruption of the ERLIN-TM6SF2-APOB complex destabilizes APOB and contributes to non-alcoholic fatty liver disease. PLoS Genet 2020;16:e1008955.
crossref pmid pmc
114. Rendel MD, Vitali C, Creasy KT, Zhang D, Scorletti E, Huang H, et al. The common p.Ile291Val variant of ERLIN1 enhances TM6SF2 function and is associated with protection against MASLD. Med 2024;5:963-980 e5.
crossref pmid
115. Mancina RM, Sasidharan K, Lindblom A, Wei Y, Ciociola E, Jamialahmadi O, et al. PSD3 downregulation confers protection against fatty liver disease. Nat Metab 2022;4:60-75.
crossref pmid pmc pdf
116. Ye J, Li JZ, Liu Y, Li X, Yang T, Ma X, et al. Cideb, an ER-and lipid droplet-associated protein, mediates VLDL lipidation and maturation by interacting with apolipoprotein B. Cell Metab 2009;9:177-190.
crossref pmid
117. Tiwari S, Siddiqi S, Siddiqi SA. CideB protein is required for the biogenesis of very low density lipoprotein (VLDL) transport vesicle. J Biol Chem 2013;288:5157-5165.
crossref pmid pmc
118. Xu W, Wu L, Yu M, Chen FJ, Arshad M, Xia X, et al. Differential roles of cell death-inducing DNA fragmentation factor-αlike effector (CIDE) proteins in promoting lipid droplet fusion and growth in subpopulations of hepatocytes. J Biol Chem 2016;291:4282-4293.
crossref pmid pmc
119. Chen Q, Fang W, Shen Y, Xu D, Chen Q, Cui K, et al. Suppression of cideb under endoplasmic reticulum stress exacerbated hepatic inflammation by inducing hepatic steatosis and oxidative stress. Free Radic Biol Med 2022;185:67-75.
crossref pmid
120. Verweij N, Haas ME, Nielsen JB, Sosina OA, Kim M, Akbari P, et al. Germline mutations in CIDEB and protection against liver disease. N Engl J Med 2022;387:332-344.
crossref pmid
121. Jamialahmadi O, Mancina RM, Ciociola E, Tavaglione F, Luukkonen PK, Baselli G, et al. Exome-wide association study on alanine aminotransferase identifies sequence variants in the GPAM and APOE associated with fatty liver disease. Gastroenterology 2021;160:1634-1646.e7.
crossref pmid
122. Speliotes EK, Yerges-Armstrong LM, Wu J, Hernaez R, Kim LJ, et al.; GOLD Consortium. Genome-wide association analysis identifies variants associated with nonalcoholic fatty liver disease that have distinct effects on metabolic traits. PLoS Genet 2011;7:e1001324.
crossref pmid pmc
123. Orho-Melander M, Melander O, Guiducci C, Perez-Martinez P, Corella D, Roos C, et al. Common missense variant in the glucokinase regulatory protein gene is associated with increased plasma triglyceride and C-reactive protein but lower fasting glucose concentrations. Diabetes 2008;57:3112-3121.
crossref pmid pmc pdf
124. Dongiovanni P, Stender S, Pietrelli A, Mancina RM, Cespiati A, Petta S, et al. Causal relationship of hepatic fat with liver damage and insulin resistance in nonalcoholic fatty liver. J Intern Med 2018;283:356-370.
crossref pmid pmc pdf
125. Yassine HN, Finch CE. APOE alleles and diet in brain aging and Alzheimer’s disease. Front Aging Neurosci 2020;12:150.
crossref pmid pmc

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: cmh_journal@ijpnc.com
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 713
TOTAL : 2629744
Close layer