v
Search
Advanced

Publications > Journals > Journal of Clinical and Translational Hepatology> Article Full Text

  • OPEN ACCESS

Applications of Artificial Intelligence and Smart Devices in Metabolic Dysfunction-associated Steatotic Liver Disease

  • Wenfeng Zhu1,
  • Qi Zheng1,
  • Xinyi Xu1,
  • Xia Yu1,
  • Xianbin Xu1,
  • Huilan Tu1,
  • Yue Yu1,
  • Wubing Ying2,
  • Jiahao Xie2,
  • Guoping Sheng3,* and
  • Jifang Sheng1,* 
 Author information 

Abstract

Metabolic dysfunction-associated steatotic liver disease (MASLD) is now considered to be among the most prevalent chronic liver diseases worldwide. Its comprehensive management encompasses multiple stages, including risk assessment, early detection, stratified intervention, and long-term follow-up. Among these, improving diagnostic accuracy and optimizing individualized therapeutic strategies remain key challenges in both research and clinical practice. In recent years, artificial intelligence and smart devices have developed rapidly and have gradually been applied in the medical field, offering novel tools and pathways for MASLD risk stratification, non-invasive diagnosis, therapeutic evaluation, and patient self-management. This review summarizes the current applications of artificial intelligence and smart devices in MASLD care, highlights their benefits and limitations, and discusses future directions to support precision diagnosis and treatment strategies.

Keywords

Metabolic dysfunction-associated steatotic liver disease, MASLD, Artificial intelligence, Smart device, Diagnosis, Treatment

Introduction

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic liver disease marked by the unregulated accumulation of lipids within the liver, which is closely linked to metabolic dysfunction. Patients generally exhibit at least one cardiovascular metabolic risk factor, such as overweight or obesity, type 2 diabetes, or other metabolic abnormalities.1,2 Epidemiological estimates demonstrate that the global prevalence of MASLD has reached approximately 30% of the adult population.3,4 MASLD is among the most common chronic liver conditions globally, characterized by a gradual progression that starts with simple steatosis and may develop into steatohepatitis, hepatic fibrosis, and eventually lead to cirrhosis or hepatocellular carcinoma.5 Timely identification of individuals at elevated risk and the adoption of early, effective therapeutic strategies are critical to halting MASLD progression and enhancing long-term prognoses. MASLD is now recognized as the predominant form of fatty liver disease globally. Strengthening early detection and intervention efforts in this population plays a pivotal role in preventing progression to advanced liver disease and mitigating the overall healthcare burden.6 The nomenclature of this disease has undergone two significant revisions. In 2020, in light of growing evidence linking obesity and metabolic dysfunction to disease pathogenesis, experts recommended replacing the term nonalcoholic fatty liver disease (NAFLD) with metabolic associated fatty liver disease (MAFLD).7 Subsequently, in 2023, concerns about the potential stigmatization associated with “MAFLD” led major international hepatology associations to advocate for a further revision, officially renaming the disease as MASLD. This updated terminology reflects a more inclusive diagnostic framework, no longer excluding patients with comorbid chronic liver conditions such as viral hepatitis, alcohol use, or drug-induced liver damage.2 Epidemiological studies in U.S. adults have reported the prevalence of NAFLD, MAFLD, and MASLD to be 18.5%, 19.3%, and 20.8%, respectively, demonstrating a clear upward trend.8 Remarkably, approximately 99% of individuals meeting NAFLD diagnostic criteria also fulfill the criteria for MASLD, highlighting a high degree of overlap in their clinical phenotypes. Therefore, findings from previous studies conducted in NAFLD populations remain largely applicable to MASLD research and clinical practice.9–11

Excessive hepatic fat deposition not only causes direct hepatocellular injury but also induces inflammatory responses, fibrosis progression, and apoptosis. In addition, hepatic insulin resistance further exacerbates lipid accumulation and impairs insulin clearance, ultimately creating a vicious cycle of mutual reinforcement.12,13 Clinical evidence indicates that individuals with MASLD face significantly elevated risks of cardiovascular disease, with both prevalence and incidence surpassing those seen in the general population.14 Thus, clinical management of MASLD should emphasize comprehensive control of cardiometabolic comorbidities, including obesity, type 2 diabetes, dyslipidemia, and hypertension, to curb disease advancement and prevent complications.15 Currently, lifestyle intervention remains the foundational strategy for treating MASLD, encompassing scientifically guided weight loss, balanced diet, and regular physical activity. When necessary, these approaches can be combined with pharmacotherapy or other adjunctive treatments. Additionally, early identification and correction of associated metabolic disorders are crucial steps in preventing disease progression.16 In recent years, rapid advancements in artificial intelligence (AI) and smart devices have led to their increasing integration into medical practice. Particularly in MASLD research and clinical practice, AI and smart devices have shown great potential in risk prediction, disease screening, auxiliary diagnosis, and personalized treatment. This review synthesizes recent literature from databases including PubMed, Web of Science, and Google Scholar to summarize the current applications and emerging trends of AI and smart devices in MASLD management, aiming to inform and guide future clinical strategies. Furthermore, this review differs from earlier work that mainly focused on imaging. It provides a broader synthesis that links AI algorithms, smart device applications, and ethical considerations across the entire MASLD management pathway.

Overview of AI and smart devices

AI integrates a multitude of sophisticated algorithms, encompassing machine learning (ML), deep learning, natural language processing (NLP), data mining, and numerous others. ML forms the backbone of this framework. It includes various supervised models such as convolutional neural networks (CNN), decision trees, support vector machines, random forests, and gradient boosting algorithms. In addition, unsupervised, semi-supervised, and reinforcement learning methods are also incorporated (Fig. 1). Deep learning, a major branch of ML, shows particular strength in recognizing complex patterns and handling high-dimensional data.

The collaborative operation model of artificial intelligence technology and smart devices.
Fig. 1  The collaborative operation model of artificial intelligence technology and smart devices.

(Created in BioRender). CNN, convolutional neural networks; DT, decision tree; SVM, support vector machine; RF, random forest; GB, gradient boosting.

In medical research and clinical practice, AI is widely applied to the analysis and modeling of multidimensional data based on clinical indicators, biomarkers, and medical imaging, significantly improving early disease detection capabilities, diagnostic accuracy, and the efficiency of personalized treatment decision-making.17 In disease risk prediction, AI has the capacity to process large-scale patient data to uncover potential risk determinants and estimate the probability of disease development. This facilitates the formulation of individualized preventive strategies tailored to each patient’s specific risk profile. In medical imaging recognition, AI enables rapid and accurate detection of underlying pathological features, improving diagnostic efficiency and significantly reducing the risk of human misjudgment. Additionally, in the field of personalized treatment, AI can provide customized therapeutic recommendations based on patients’ unique clinical and physiological parameters, thereby enhancing treatment outcomes.17,18 The application of these technologies not only significantly reduces the time required for traditional data processing and decision-making but also helps overcome subjective biases inherent in human analysis. This demonstrates important potential for managing chronic diseases, including MASLD. Meanwhile, advancements in smart device technology have also provided new tools for health management. Devices such as smartphones, wearables, and portable medical monitors are now equipped with advanced sensors and multimodal data collection capabilities. They can continuously track critical health indicators, including body weight, waist-to-hip ratio, heart rate, glucose levels, and blood pressure. These devices, combined with AI algorithms for data analysis, can provide users with personalized and dynamic health feedback and intervention recommendations. Figure 2 illustrates the application of AI technology and smart devices in managing MASLD: through continuous monitoring and dynamic feedback to enable real-time health tracking and timely adjustment of intervention strategies based on data trends.19,20

Artificial intelligence and smart devices in clinical management of metabolic dysfunction-associated steatotic liver disease.
Fig. 2  Artificial intelligence and smart devices in clinical management of metabolic dysfunction-associated steatotic liver disease.

(Created in BioRender).

Application of AI in MASLD risk prediction and diagnosis

While liver biopsy remains the gold standard for diagnosing steatotic liver disease, its invasive nature, associated bleeding risks, and low patient acceptance restrict its feasibility for widespread use in population-level screening. In recent years, advances in AI technology have brought new breakthroughs in the early screening of MASLD. By synthesizing a broad spectrum of individual data, including clinical metrics such as body mass index (BMI), glucose and lipid profiles, genetic predispositions, and lifestyle habits, AI can establish multidimensional, personalized risk prediction models, significantly improving the efficiency of identifying high-risk individuals. For example, ML applied to large population cohorts can effectively identify high-risk individuals with potential progression from MASLD to metabolic dysfunction-associated steatohepatitis (MASH), thereby enabling the implementation of personalized, stratified intervention strategies.21 For imaging-based assessments of steatotic liver disease, commonly employed modalities include ultrasound, computed tomography (CT), and magnetic resonance imaging (MRI). Ultrasound is frequently used as a screening method due to its affordability, safety, and non-invasive characteristics. Nevertheless, its diagnostic sensitivity for early-stage or mild steatosis is suboptimal and heavily influenced by operator proficiency. CT can assess the degree of hepatic fat deposition but is susceptible to interference from factors such as iron overload and carries the risk of radiation exposure. MRI, with its high soft tissue resolution and excellent fat quantification capability, is considered the current optimal non-invasive quantitative method, though it is relatively more expensive.22,23 With the continuous advancement of AI, especially ML and deep learning methods, their application in medical imaging analysis has become increasingly widespread. These AI-driven tools can autonomously derive numerous quantitative imaging features and apply sophisticated algorithms or neural networks to support early diagnosis and treatment tracking of fatty liver disease. Multiple studies have introduced AI-based models for imaging diagnosis of hepatic steatosis. Subgroup analyses reveal that whether employing classical ML techniques or more advanced deep learning architectures, these models demonstrate reliable diagnostic performance across different populations, reference standards, imaging techniques, and transfer learning contexts.24

AI-based predictive models using electronic health records (EHR) and laboratory data

AI shows great promise in processing EHR to construct risk assessment models for MASLD. With the shift in disease nomenclature from NAFLD to MASLD, whether some non-invasive tests previously used to predict NAFLD remain applicable to MASLD has become an urgent issue in research and clinical practice. Recent studies have shown that certain non-invasive test tools, such as the fatty liver index, still have strong discriminatory efficacy in identifying MASLD risk, suggesting that they retain application value in the new disease classification framework.25 With the continuous advancement of AI technology, combining non-invasive tests with AI algorithms is anticipated to further streamline MASLD screening workflows—enhancing diagnostic efficiency and reducing the burden on healthcare systems. Currently, commonly used AI methods in EHR-based data analysis include ML, NLP, data mining techniques, and algorithms based on International Classification of Diseases coding. By comprehensively analyzing multidimensional information such as demographic characteristics, lifestyle, physical measurements, and laboratory data contained in EHR, AI models can predict the risk of MASLD and assess its severity. Over time, research priorities have clearly evolved. Early studies mainly compared analyses of methodologies. More recent efforts have shifted toward building sophisticated, multimodal predictive systems. For example, Van Vleck et al.26 reported that NLP outperformed text search and ICD coding in identifying MASLD cases from EHR data. Later, Bonfiglio et al.27 developed a model to predict mortality risk, while Yuan et al.28 created a screening tool for younger individuals based on standard physical examination indicators.

A significant step forward is seen in the N3-MASH model proposed by Zhang et al.29 This framework integrates CXCL10 to reflect inflammation, CK-18 to capture apoptosis, and adjusted BMI as a marker of lipid accumulation. Together, these variables substantially enhance the accuracy of MASH detection. The model marks a transition from dependence on conventional clinical variables to a mechanism-oriented design incorporating novel biomarkers.

Furthermore, the application of AI in multi-omics research is unveiling new insights into disease mechanisms and biomarker discovery. AI technology has also demonstrated broad application value in metagenomics and metabolomics research on various biological samples, especially in identifying potential key genes and biomarkers for MASLD and its different stages of progression. For detailed research results, please refer to Table 1.21,26,28–34 In genomics and transcriptomics, studies by Conway et al.35 and Park et al.36 have utilized deep learning to identify key genes and signaling pathways, such as the Notch pathway, associated with MASH severity and hepatic fat accumulation. In metabolomics and lipidomics, the work of teams like McGlinchey30 and Chiappini37 has systematically characterized distinct metabolite signatures across disease stages, highlighting the central role of impaired fatty acid metabolism in MASH. Perveen et al.’s study, using decision tree analysis, identified high-density lipoprotein levels as an important predictive marker for the development of hepatic steatosis.31 Additionally, AI-assisted analysis of gut microbiome data highlights the potential of microbial signatures for individualized diagnosis and intervention.38 More importantly, AI’s integrative power is reshaping risk assessment for MASLD. Instead of relying solely on superficial statistical associations, it enables models grounded in biological mechanisms and refined patient subtypes. By combining clinical data with multi-omics information, AI systems can define disease endotypes and guide tailored therapeutic strategies. This progression signals a move toward true precision medicine in metabolic liver disease.

Table 1

EHR and laboratory data

AuthorCountryYearCategoriesMethodsIndicatorsResult
Zhang X et al.29China2025MASHN3-MASHCXCL10, CK-18, BMIAUC: 0.857
Park IG et al.32Korea2024MASLDSVM,RF,CNNGenomic DNAAUC: 0.93
Yuan Y et al.28China2024MASLDNomogram modelAge, Gender, BMI, Waist-to-hip ratio, ALT, LDL-c, HDL-c, UA, and smokingAUC: 0.875
McTeer M et al.21UK et al.2024MASHXGBoostMASLD dataset in EuropeAUC: 0.899
Zhu Q et al.33China2022NASHRF4 lipid moleculesAUC: 0.923
McGlinchey AJ et al.30Sweden et al.2022NAFLDMLClinical dataAUC: F (0–1 vs 2–4) = 0.746, F (0–2 vs 3–4) = 0.778
Kordy K et al.34USA2021NASHRFOral and fecal microbiome, plasma metabolitesAUC: 0.99
Van Vleck TT et al.26USA2019NAFLDNLPEHRSpecificity: 89%, Sensitivity: 93%
Perveen S et al.31Pakistan2018NAFLDDTEHRAUC: 0.73

AI-based diagnostic models using imaging data

AI technology, especially through ML and deep learning models, is playing a growing role in liver imaging, enhancing the accuracy and speed of disease identification and diagnosis. In clinical evaluation, MASLD imaging typically follows a three-step sequence. Ultrasound serves as the first-line screening tool, with AI assisting in automated detection and grading of steatosis. CT often identifies hepatic fat incidentally during unrelated scans, where AI can quantify fat content retrospectively. MRI then provides confirmation and precise quantification, while AI improves workflow efficiency and interpretive accuracy. In recent years, researchers have developed various quantitative fat analysis methods based on ultrasound, CT, and MRI, enabling clinicians to more accurately assess the degree of hepatic lipid accumulation and fibrosis staging. These methods achieve precise quantification of steatosis and fibrosis by automatically extracting key imaging features, providing critical support for early diagnosis, disease staging, and intervention decision-making.39

Ultrasound imaging

Ultrasound examination, as a commonly used imaging modality for diagnosing MASLD, is widely applied in clinical practice. Nonetheless, several limitations persist. Diagnostic accuracy still depends on operator expertise and subjective interpretation. Ultrasound, although sensitive for moderate to advanced steatosis, struggles with mild cases. Subtle steatosis remains difficult to detect, as assessments largely rely on qualitative echogenicity rather than quantitative measures.40 Traditional ultrasound imaging is primarily based on the nonlinear interaction between linearly propagated ultrasonic pulses and tissues, generating harmonic frequencies. Among these, the second harmonic is widely used in clinical image optimization due to its improved signal-to-noise ratio, clear boundaries, and reduced artifacts. However, while qualitative features such as enhanced echoes can be detected when hepatic fat content is elevated, sensitivity remains insufficient for mild fat accumulation. To overcome the limitations of traditional ultrasound, various quantitative ultrasound techniques have been developed in recent years, including elastography, echo signal analysis, and speckle pattern modeling. These methods provide more precise quantitative data on tissue characteristics. For example, ultrasound elastography applies acoustic radiation force pulses via a transducer to measure shear wave velocity within liver tissue, which correlates with tissue stiffness, serving as a non-invasive indicator of fibrosis. Additionally, some algorithms convert the degree of ultrasound attenuation into a controlled attenuation parameter to quantify hepatic fat content. However, these techniques are susceptible to interference from subcutaneous fat thickness and intercostal space width in obese individuals, which can affect the accuracy of the measurements.41 The integration of AI into ultrasound diagnostics has opened new avenues for MASLD detection. By integrating multiple ultrasound imaging features, AI can more accurately assess liver structure and functional status. In recent years, researchers have conducted several exploratory studies in this field, with relevant findings summarized in Table 2.42–61 Various research groups have incorporated deep learning, CNN, and large language models to significantly boost the diagnostic accuracy and efficiency of sonographic imaging. Through algorithm optimization and improvements in image processing techniques, AI has enabled quantitative analysis and grading assessment based on traditional ultrasound, thereby providing a more convenient and reliable approach for MASLD screening and classification. A critical synthesis of recent advancements highlights several distinct, yet complementary, strategies for applying AI to ultrasound-based MASLD diagnosis. These developments move beyond mere automation. They represent a fundamental expansion of ultrasound’s diagnostic capability. The first approach centers on improving diagnostic accuracy directly from standard B-mode images. For instance, Kaffas AE et al.42 showed that a deep learning framework could markedly increase both the sensitivity and the accuracy of MASLD diagnosis using routine ultrasound scans. Their findings suggest that AI can uncover subtle imaging cues overlooked in manual interpretation. A second line of work focuses on the automation and standardization of semi-quantitative metrics. This strategy aims to make results more reproducible and less dependent on operator expertise. The AI system developed by the Santoro group illustrates this point well.43 It automatically calculates the liver-to-kidney ratio, minimizing inter-operator variation and improving consistency across centers. Most notably, a third and transformative direction uses AI to derive quantitative data directly from conventional scans. Models trained on extensive, multi-source datasets that include different scanners and imaging protocols can now estimate liver fat content with high precision. Remarkably, their performance rivals, and in some cases exceeds, that of specialized tools such as FibroScan.62 Collectively, these studies mark a transformative shift for ultrasound in MASLD. AI is not only augmenting conventional practices but also equipping the modality with novel quantitative capabilities, once exclusive to advanced technologies, thereby significantly boosting its clinical utility.

Table 2

Ultrasound

AuthorCountryYearCategoriesMethodsSample sizeReference standardResult
Vianna P et al.44Canada2025Hepatic steatosisCNN250 patientsNonadapted modelsAUC: 0.97
Wu W et al.45China2025MASLDLLM1,542 participantsCAPAUC: 0.831
Kaffas AE et al.42USA2025Hepatic steatosisDL403 patientsMRI-PDFFAUC: S (0 vs 1/2/3) = 0.813, S (0/1 vs 2/3) = 0.959
Drazinos P et al.46Greece2025NAFLDDL112 patientsBiopsyAUC: S (0 vs 1/2/3) = 0.82, S (0/1 vs 2/3) = 0.9156, S (0/1/2 vs 3) = 0.9619
Honarvar M et al.47Canada2024MASLDU-Net architecture139 subjectsMRI or FibroScan CAPSensitivity: 81%, Specificity: 84%
Marques R et al.48Portugal2024Hepatic steatosisNeural network441 patients;205 healthy subjectsUltrasound and elastographyF1 score: ultrasound dataset = 99.5%, elastography dataset = 99.2%, mixed dataset = 98.9%
Liu Y et al.49China2024NAFLDDL710 individualsUltrasound imagesAUC: 0.95
Santoro S et al.43Italy2024Hepatic steatosisAI-based algorithm: HRIA134 patientsMRI-PDFFAUC: 0.98
Kwon H et al.50Korea2024NAFLDAI-QUS35 participantsMRI-PDFFAUC: S (0 vs 1/2/3) = 0.93, S (0/1 vs 2/3) = 0.99, S (0/1/2 vs 3) = 0.98
Vianna P et al.51Canada2023Hepatic steatosisDL199 patientsBiopsyAUC: S (0 vs 1/2/3) = 0.85, S (0/1 vs 2/3) = 0.73, S (0/1/2 vs 3) = 0.67
Yang Y et al.52China2023NAFLDDL928 participantsAbdominal ultrasoundAUC: 0.90
Jeon SK et al.53Korea2023NAFLDCNN173 participantsMRI PDFFAUC: 0.97
Zsombor Z et al.54Hungary2023NAFLDAI-calculated hepatorenal index (AI-HRI)102 patientsMRI-PDFFAUC: Mild steatosis = 0.758, Moderate/severe steatosis = 0.803
Li B et al.55USA et al.2022NAFLDDL3,310 patientsHistologyAUC: Mild steatosis = 0.85, Moderate steatosis = 0.91, Severe steatosis = 0.93
Byra M et al.56USA2022NAFLDCNN135 participantsMRI-PDFFAUC: PDFF≥5% = 0.91, PDFF≥10% = 0.86
Che H et al.57USA2021NAFLDCNN55 subjectsBiopsyAUC: 0.978
Constantinescu EC et al.58Romania2021NAFLDCNN629 grayscale liver imagesB-mode ultrasound and elastographyAUC: 0.93
Han A et al.59Poland2020NAFLDCNN204 participantsMRI-PDFFAUC: 0.98
Cao W et al.60China2020NAFLDDL240 participantsAbdominal ultrasoundAUC: 0.933
Byra M et al.61Poland2018Hepatic steatosisCNN, SVM55 severely obese patientsLiver biopsyAUC: 0.977

CT imaging

CT remains a widely utilized tool in clinical imaging, offering reliable quantification of liver fat, yet its dependence on manual region-of-interest placement makes the process slow, operator-dependent, and unsuitable for large-scale screening. This inherent limitation has positioned full automation as both the central goal and the main advantage of AI in CT-based MASLD evaluation. Recent research efforts have shifted toward automating CT image analysis using deep learning approaches to facilitate MASLD diagnosis. This technology automatically segments the liver region, identifies features of fat deposition, and subsequently quantifies the degree of hepatic steatosis, thereby providing clinicians with supportive diagnostic information. As summarized in Table 3,63–71 the research trajectory clearly shows a shift from proof-of-concept automation to validation in large-scale, real-world populations. Following the early validation by Graffy et al.,63 Martín-Saladich et al.64 advanced the field with a fully automated voxel-level framework. Their system achieved excellent cross-device reproducibility and eliminated manual input. The deep learning model aligned closely with manual readings and captured expected associations between steatosis, age, and BMI, reinforcing the biological credibility of AI-derived quantifications. Further refinements have aimed to improve accuracy and integration into clinical workflows. Studies by Prinz et al.65 and Huo et al.,66 both using CNN architectures, reported consistently high agreement between automated and manual fat quantification across multiple metrics. Collectively, the evidence indicates that AI is no longer merely replicating human measurements. It frequently surpasses them in speed, consistency, and spatial coverage, extending analysis from limited region-of-interest samples to whole-liver assessment. In essence, AI-driven automation is transforming CT from a qualitative or semi-quantitative method into a robust, high-throughput platform for MASLD screening and monitoring. By eliminating the bottlenecks of manual analysis, AI unlocks new potential for opportunistic screening within the vast number of CT scans acquired for unrelated reasons. This shift adds significant value to everyday imaging practice and paves the way for population-level steatosis surveillance.

Table 3

CT

AuthorCountryYearCategoriesMethodsSample sizeReference standardResult
Kim HY et al.67Korea2025Hepatic steatosisDL3,620 subjectsLiver biopsyAUC: 0.868
Vong T et al.68USA2025Hepatic steatosisLLM200 adultsLabeled CT reportsAccuracy: 0.988, Sensitivity: 0.98, Specificity: 1
Martín-Saladich Q et al.64Spain2024MAFLDnn-UNet39 patientsManually assessed by specialistsAUC: 0.94
Yoo J et al.69Korea2024Hepatic steatosisDL362 adultsMRS-PDFFAUC: 0.817
Jeon SK et al.70Korea2024Hepatic steatosisDL252 participantsMRS-PDFFAUC: 0.806
Prinz S et al.65Germany2023Hepatic steatosisCNN197 patientsManual ROIs0.75<AUC<0.87
Pickhardt PJ et al.71USA et al.2021Hepatic steatosisDL1,204 healthy adultsUnenhanced CTAUC: PDFF ≥ 5% = 0.669, PDFF ≥ 10% = 0.854
Graffy PM et al.63Madison2019NAFLDDL9,552 adultsThe manual Hounsfield unit measuresR2: 0.92
Huo Y et al.66USA2019NAFLDCNN246 subjectsAbdominal CT scans with manual liver segmentationPearson correlations = 0.94

MRI imaging

MRI, as a non-invasive technique for hepatic fat quantification, has been widely used in both research and clinical practice. Common MRI-based fat quantification methods include magnetic resonance spectroscopy (MRS), fat-suppressed imaging, water-fat separation techniques, and proton density fat fraction (PDFF). Among these, MRS is considered one of the most precise for non-invasively quantifying intrahepatic fat. It can sensitively detect metabolic changes and pathophysiological states in living tissue and has advantages such as low measurement variability, high correlation with histology, and results that are not affected by liver fibrosis, iron deposition, or dietary factors.72 However, a limitation of MRS is that it can only quantify fat content in a single localized region, making it difficult to comprehensively assess fat distribution throughout the entire liver. In contrast, the PDFF technology can reflect the fat content of the entire organ. After multi-factor correction, its quantitative accuracy is highly consistent with MRS and is regarded as a dependable indicator for assessing steatosis. Both MRI-PDFF and MRS have demonstrated strong concordance with histopathological steatosis grading and offer high diagnostic accuracy for the clinical classification of hepatic fat content.73,74 Rather than revalidating MRI-based fat quantification—which is already well established—current research focuses on workflow optimization, computational efficiency, and improved clinical accessibility. As outlined in Table 4,74–81 recent deep learning advances are systematically resolving major limitations in MRI analysis. A leading direction involves automating labor-intensive steps. Martí-Aguado et al.75 developed a CNN algorithm for automated whole-liver segmentation, achieving fat quantification results with strong histological concordance. Another stream of innovation targets refinement of the PDFF metric through advanced neural networks. With the advancement of AI technology, performance evaluation of metrics such as PDFF has also been optimized. Wang et al.76 used deep learning to infer PDFF values with high precision, reinforcing its reliability as an imaging biomarker. Meneses et al.77 proposed the Variable Echo Times Neural Network, which delivered more accurate PDFF estimation than conventional architectures. Despite these technical gains, clinical translation of MRI-PDFF and MRS remains constrained by high cost, specialized hardware, and long acquisition times. Consequently, these modalities are mostly applied in research contexts or for evaluating high-risk patients rather than general screening. Thus, although AI has greatly advanced the precision, automation, and interpretability of MRI-based fat quantification, these methods remain endpoints within the diagnostic pathway, not tools for population-level screening.

Table 4

MRI

AuthorCountryYearCategoriesMethodsSample sizeReference standardResult
Meneses JP et al.77Chile2025Hepatic steatosisVET-Net188 subjectsMRI-PDFFR2: 0.87–0.98
Li S et al.78China2024NAFLDDL20 subjectsGraph-cut algorithmNo significant difference
Wang K et al.76USA2023NAFLDCNN292 participantsCSE-MRICNN-inferred PDFF showed superior agreement with reference (ICC = 0.99, bias = −0.19%)
Bastati N et al.79Austria2023NAFLDUDC46 patientsHistologyAUC: 0.85
Kim JW et al.74Korea2022NAFLDMRS, MRI-PDFF47 patientsBiopsyAUC: ≥S2 (MRS) = 0.860, (MRI-PDFF) = 0.846, ≥S3 (MRS) = 0.878, (MRI-PDFF) = 0.855
Martí-Aguado D et al.75Spain2022NAFLDDL165 participantsBiopsyAUC: 0.97
Pollack BL et al.80Pittsburgh2021NAFLDCNN149 patientsBiopsyAUC: 0.84
Cho Y et al.81Korea2021NAFLDDL77 samplesManual SegmentationStep 3: Dice Coefficients 0.94 ± 0.01, Bland-Altman bias −0.67%

AI-based analytical models for liver histopathology

Liver biopsy continues to serve as the diagnostic gold standard for MASLD. However, its invasiveness, sampling error, and subjective scoring limit its routine use. In response, AI and digital pathology are redefining histological assessment, offering objective, reproducible, and scalable tissue evaluation. New algorithms can now automatically detect and quantify key histological features from biopsy samples, such as fat accumulation, lobular inflammation, ballooning degeneration, and fibrosis. The results demonstrate high concordance with assessments made by experienced pathologists. A critical review of ongoing research reveals a multi-faceted evolution in AI-assisted liver pathology. This shift is not simply incremental—it reflects a conceptual reorientation toward algorithm-supported diagnostics. AI-driven image analysis systems can perform precise evaluations of digitized histological slides, automatically identifying core pathological changes associated with MASLD. Among these, hepatocyte ballooning is a key morphological marker for diagnosing MASLD. The advancement of digital pathology helps overcome limitations of traditional manual slide reading, such as low annotation efficiency and high subjective variability, by enhancing diagnostic objectivity and consistency through high-resolution image acquisition and algorithmic analysis. The combination of high-resolution slide digitization with algorithmic analysis enhances diagnostic precision and facilitates standardization across centers. Yet, methodological gaps persist. The scarcity of large, annotated, and diverse datasets limits generalizability, especially across staining methods, scanners, and populations. Future research urgently needs to construct a large-scale, standardized image database of ballooning degeneration in liver cells and further optimize algorithm structures (such as introducing the Transformer architecture) to improve recognition accuracy. As generative AI continues to mature, the establishment of standardized performance criteria will be crucial for validating its role in augmenting diagnostic consistency.82 Traditional ML models typically depend on extensive labeled datasets to reach high precision, and their deployment in clinical practice is often hampered by concerns over patient privacy and data sensitivity. To this end, the emerging quantum machine learning (QML), as an interdisciplinary technology that fuses quantum computing and classical ML, shows stronger generalization ability and modeling accuracy. Lusnig et al.83 demonstrated the potential of hybrid quantum–classical neural networks, achieving 97% accuracy in classifying biopsy slides. This approach performed particularly well under data-limited conditions, suggesting that QML may address two persistent challenges: the need for extensive annotation and the protection of data privacy. Beyond static diagnosis, AI is now enabling dynamic disease monitoring. Naoumov et al.84 integrated AI with digital pathology to track histological changes during MASH therapy. Their system provided greater sensitivity in detecting fibrosis regression and enabled more dynamic evaluation of therapeutic efficacy. Together, these advances represent more than automation. They signal a paradigm shift—from subjective interpretation toward reproducible, data-driven quantification—reshaping the epistemological foundation of liver pathology itself.

Treatment of MASLD

The treatment of MASLD primarily includes lifestyle modification and pharmacotherapy (Table 5).85–94 The convergence of AI and smart devices is reshaping these traditional strategies, promoting a shift from standardized care to dynamic, personalized, and data-driven management. In lifestyle management, patients are advised to control weight, limit alcohol intake, improve dietary structure, and adhere to regular exercise. In terms of drug therapy, several drug candidates are currently in clinical trials. In addition, patients with chronic hepatitis B who have concomitant metabolic abnormalities (such as glucose dysregulation, dyslipidemia, or overweight) should also be comprehensively evaluated and actively intervene with their metabolic markers to reduce the risk of MASLD progression. Probiotic therapy has also been recognized as potentially valuable in the management of MASLD. It has been shown that probiotics can play a positive role in improving liver enzyme levels, regulating lipid metabolism, promoting weight control, and alleviating insulin resistance by modulating intestinal flora.95 Bariatric surgery has emerged as an alternative treatment option for patients with MASLD combined with obesity who do not respond well to lifestyle interventions and medications. Chen S et al.96 used a multi-omics approach to analyze the support for the use of sleeve gastrectomy as an effective means of ameliorating hepatic steatosis and inflammation when other weight loss strategies are ineffective.

Table 5

Treatment

AuthorCountryYearCategoriesDrugsMethodsReferenceResult
Harrison SA et al.90USA2025MASHFFAR1/FFAR4 agonistRCTPlaceboResponse rates: 300 mg = 29.3%, placebo = 11.3%
Ozlu Karahan T et al.91Turkey2025MASLDNALLMGuidelinesAccuracy: mean energy = 91.3 ± 11.0%
Zhang L et al.88China2025MAFLDQigui Jiangzhi Formula (QGJZF)AlphaFold-AIpTM scores (range 0–1)pTM scores: PRKAA2 = 0.97, SIRT1 = 0.93
Harrison SA et al.85UK2024NASHResmetiromRCTPlaceboNASH resolution rate: 100 mg = 29.9%, placebo = 9.7%
Wang Y et al.87China2024NASHLiDi FGF21LiDi platformBaselineNAS scores: low-dose = 3.83 ± 0.98, high-dose = 3.33 ± 0.82, baseline = 6.00 ± 0.63
Newsome PN et al.92UK et al.2023NASHAOC3 inhibitorDouble-blind, placebo-controlledPlaceboAOC3 activity relative to baseline: 10 mg = 3.3%, Placebo = 90.4%
Saldarriaga OA et al.89USA2023MASLDNADLKey FindingsUpregulated Targets: CCR2, CCR5, CCL2, CCL5, LGALS3
Yu H et al.93China2023NAFLDAescin (Aes)HepG2 cell models/Aes facilitates Nrf2 nuclear translocation
Sessa L et al.86Italy et al.2023NASH5-hydroxytryptamine 2A receptor (5-HT2AR)SoftMining Platform5HT2AR-CHO-K1 cellsCompetitive binding studies in 5HT2AR-CHO-K1 cells validated the in silico prediction
Lee WY et al.94Korea2022NAFLDFlavonoidsMachine-learning model: AI-DTIHepG2/AML12 cell modelsScreening and functional validation of candidate flavonoids

Lifestyle interventions

Lifestyle-related factors play a pivotal role in the onset and progression of MASLD. Yuan et al.97 demonstrated that intensive lifestyle intervention can reverse hepatic fibrosis in MASH, with AI-assisted imaging revealing the most evident regression in the portal region. However, maintaining adherence to such interventions remains difficult—particularly among socioeconomically disadvantaged groups, who often face higher MASLD risk due to poor dietary habits.98 AI-enabled smart devices provide scalable and individualized solutions for this challenge.

In recent years, with the help of smart devices combined with AI algorithms, clinicians can achieve real-time monitoring of patients’ diet, physical activity, and metabolism-related indicators, and then generate personalized intervention programs. For example, a model based on Gradient Boosting Regression predicts individual postprandial glucose fluctuations and combines gut microbiome data with dietary habits to optimize an individualized dietary structure.99 They are especially valuable for patients with prediabetes or type 2 diabetes and MASLD, where precise glycemic control is essential.

Nutritional strategies are fundamental in the prevention and management of MASLD. Among various dietary patterns, the Mediterranean diet has shown considerable efficacy in lowering hepatic lipid accumulation and improving metabolic health.100 With the support of smart devices and AI, the implementation of medical nutrition therapy is more efficient and precise. Patients can use the image recognition feature to record their daily diet. The system automatically analyzes the nutritional content and guides them to follow a healthy diet structure while dynamically adjusting energy intake based on real-time data. It has been demonstrated that this type of intelligent system is effective in improving liver triglyceride levels and other metabolic parameters.101 This scenario suits motivated individuals who benefit from structured, feedback-driven dietary support.

With advancements in digital health, smart devices now offer capabilities such as remote patient monitoring and online follow-up, allowing healthcare providers to track real-time fluctuations in liver-related biomarkers—including alanine aminotransferase and aspartate transaminase—as well as body weight. This continuous monitoring facilitates timely treatment adjustments based on a patient’s clinical progression. This is equally useful for patients undergoing new drug therapies or those with advanced fibrosis who require close follow-up.

AI chatbots are also gradually playing a role in assisted management, providing not only medication reminders and health education but also basic psychological support that can help improve patient compliance. Related studies show that some AI chatbots currently score high on health-related questions, but clinical application still requires professional medical judgment. With continued algorithm optimization, their clinical practicality is expected to improve further in the future.102 In overburdened healthcare systems, AI chatbots and digital companions can also serve as first-line educational tools, addressing common patient concerns and reinforcing adherence.

AI-powered systems further support individualized care by integrating clinical data, laboratory results, and histopathological findings to formulate tailored therapeutic plans that span lifestyle modifications and pharmacologic options. Mobile health applications and wearable devices are becoming increasingly valuable tools in MASLD care. These tools enable the ongoing collection of behavioral data, including physical activity, dietary intake, and sleep metrics, which supports more precise and dynamic intervention strategies.103 Exercise is another widely accepted intervention with proven benefits for MASLD. Regular physical activity not only contributes to weight reduction and decreased hepatic fat but also elevates high-density lipoprotein cholesterol levels.104 A health management app that combines AI algorithms can help patients set personalized exercise plans and dynamically adjust them based on feedback from wearable sensors. At the same time, these devices can also monitor indicators such as weight and waist circumference in real time and work with AI models to predict the potential effects of weight loss on liver fat improvement. Studies have shown that achieving a weight loss of over 10% can result in substantial improvements in MASH and liver fibrosis severity.105 Closed-loop feedback mechanisms enhance motivation, especially for individuals struggling with weight maintenance, by offering concrete targets and continuous reinforcement.

Drug development

While pharmacological research into MASLD therapies has advanced in recent years, only a limited number of medications have received approval. Drugs currently in late-stage clinical development include incretin-based therapies like glucagon-like peptide-1 and its multi-agonists, metabolic modulators like peroxisome proliferator-activated receptor, fibroblast growth factor 21 (FGF21), and thyroid hormone receptor β (THR-β) agonists, or novel drugs targeting new mechanisms, such as fatty acid synthase inhibitors.106,107 The development of AI technology has significantly accelerated the research and development process of MASLD-related drugs in multiple stages. Through virtual screening, structural prediction, and multi-omics integration analysis, AI has not only promoted new drug discovery but also driven the re-development of existing drugs for new indications. For example, Resmetirom is a highly selective THR-β agonist. A notable breakthrough occurred in 2024 when the U.S. Food and Drug Administration approved Resmetirom as the first drug for treating MASH. AI-assisted analysis was extensively used in the target identification, mechanism elucidation, and clinical study design of this drug. In its pivotal Phase III clinical trial, AI algorithms were used to quantitatively assess changes in liver fibrosis before and after treatment, demonstrating that Resmetirom has definitive efficacy in significantly improving liver fibrosis.85,108,109 In addition, the potential of AI in drug screening and indication expansion continues to be validated. HuX et al. utilized the DiscoveryStudio19 platform, which integrates virtual screening, molecular docking, ADME property prediction, and toxicity assessment, to screen potential farnesoid X receptor agonists, providing a new drug candidate idea for MASH treatment.110 Similarly, Sessa L and colleagues leveraged AI methodologies to validate the therapeutic potential of the 5-hydroxytryptamine 2A receptor, identifying its antagonists as promising candidates for MASH therapy.86 In the field of protein therapy research, Wang Y’s team has developed a new form of bioactive FGF21 based on the “lipidation-dimerization” platform, named LiDi FGF21. This molecule demonstrates superior pharmacological properties, expanding the application prospects of protein-based drugs in the treatment of MASLD.87 AI technology also provides new insights into the modernization of traditional Chinese medicine. Zhang L et al. identified potential target proteins for the traditional Chinese medicine compound QGJZF in MASLD using the SymMap database and predicted the structures of its key proteins using AlphaFold, revealing that it may exert anti-adipogenic and anti-inflammatory effects through the AMPK/SIRT1-TFEB pathway.88 Furthermore, Saldarriaga et al. applied deep learning to multi-dimensional datasets in fatty liver fibrosis, identifying disease-stage-dependent heterogeneity in macrophage populations and highlighting CCR2 and Galectin-3 as potential therapeutic targets in advanced MASLD.89

In summary, AI is fundamentally reshaping MASLD drug discovery by accelerating target identification, optimizing clinical trials, and enhancing predictive profiling.111

Ethical, regulatory, and data security considerations

Integrating AI and smart devices into MASLD care thus holds great promise for improving diagnostic precision and therapeutic outcomes.112 However, its real-world application raises complex ethical, regulatory, and data security concerns (Fig. 3). Therefore, the development and deployment of AI must align with ethically grounded frameworks, consistent with the World Health Organization guidance on the Ethics and Governance of AI for Health. The World Health Organization identifies six guiding principles: protecting autonomy; advancing human welfare and safety; ensuring transparency and intelligibility; reinforcing accountability; promoting equity and inclusiveness; and encouraging responsiveness and sustainability.113

Responsible Artificial intelligence in metabolic dysfunction-associated steatotic liver disease: A framework built on ethics, regulation, and security.
Fig. 3  Responsible Artificial intelligence in metabolic dysfunction-associated steatotic liver disease: A framework built on ethics, regulation, and security.

(Created in BioRender). AI, artificial intelligence; MASLD, metabolic dysfunction-associated steatotic liver disease; HIPAA, health insurance portability and accountability act; GDPR, general data protection regulation; PDPO, personal data (privacy) ordinance; APPI, act on the protection of personal information.

Recent analyses reveal that bias and fairness dominate ethical discourse, followed by concerns about safety, reliability, transparency, accountability, model misuse, and privacy—particularly in relation to large language models.114,115 Concurrently, the evolution of AI operates within established regulatory structures, which define critical operational boundaries and reinforce the protection of personal data and privacy.116 As jurisdictions adapt their own legal standards, an international consensus toward harmonized regulation is increasingly essential.

From a technical standpoint, privacy-preserving techniques, such as federated learning and differential privacy, are vital to building multi-layered safeguards. Only through such a multidisciplinary approach can these innovative tools be deployed safely and equitably, upholding the rights and welfare of individuals in real-world settings.

Ethical AI for MASLD

Algorithmic bias and health disparities

The performance and generalizability of AI models depend critically on the diversity of their training data. When development datasets are disproportionately sourced from limited demographic segments, such as particular ethnic, geographic, or socioeconomic groups, models are prone to substantial performance decline in clinically underrepresented populations. This not only reduces clinical reliability but may also amplify existing health inequities.117 To mitigate such effects, it is essential to systematically construct diverse and inclusive training cohorts and incorporate bias detection and mitigation protocols across the entire model lifecycle. These strategies should consider local context—target demographics, deployment settings, algorithm type, and the specific biases being addressed.118

Transparency and explainability

Many advanced AI systems, particularly deep learning–based systems, function as “black boxes” due to their inherently opaque internal decision logic, which remains largely inaccessible to human interpretation. For healthcare providers to trust AI-based diagnostic aids, such as hepatic steatosis grading, transparency in algorithmic reasoning is essential.119 Explainable AI (XAI) addresses this need by employing both global and local interpretability techniques to uncover salient features contributing to predictive outcomes. This capacity to elucidate model behavior is critical for fostering informed clinical acceptance and enabling the responsible embedding of AI tools into medical practice.120

Accountability and human oversight

Excessive dependence on AI in medical contexts can lead to significant errors in areas such as predictive analytics, system oversight, device selection, and even clinical decision-making. Many AI models are often inadequately trained on specific features, such as dialects and medical colloquialisms; they risk generating inaccurate diagnoses, medication explanations, or treatment recommendations.121 It is therefore essential to reaffirm that AI serves strictly as an adjunctive decision-support mechanism rather than as an autonomous clinical authority. The final judgment and accountability for all diagnostic or therapeutic decisions must rest with qualified healthcare professionals. These practitioners must critically appraise algorithm-generated suggestions and retain the ultimate decision-making power, ensuring consistent human oversight and intervention throughout patient care.

Regulatory frameworks

The development and deployment of medical AI must occur within a robust regulatory framework, with the core objective of safeguarding patient data privacy and security. Several major frameworks currently provide guidance for this evolving landscape.

Health Insurance Portability and Accountability Act (HIPAA)

In the United States, HIPAA stands as a foundational statute safeguarding Protected Health Information.122 Despite its importance, HIPAA’s jurisdiction is incomplete—it excludes data handled by non-covered entities, omits patient-generated content, and overlooks extensive non-health datasets that can indirectly reveal health conditions. To establish comprehensive protection for the health information ecosystem, it is imperative to either expand HIPAA’s applicability or create a separate regulatory regime for currently uncovered health-related data. Proposed legislative pathways include enacting a general health data protection rule supplemented by specific provisions for different data processors, or adopting a unified framework inspired by the EU’s General Data Protection Regulation (GDPR) that applies to all personal data processors while incorporating dedicated rules for health information.123

GDPR

The European Union’s GDPR regulation has become a global benchmark for personal data governance.124 It mandates strict adherence to the principles of lawfulness, fairness, and transparency in all stages of data processing. Under GDPR, information must be collected for clearly defined and legitimate purposes (purpose limitation), and only the minimal data necessary for those purposes may be processed (data minimization). Controllers remain fully accountable for compliance with these standards. In MASLD-related studies involving EU participants, explicit informed consent and transparent data handling practices are indispensable, reflecting the regulation’s emphasis on autonomy and accountability.

Other international frameworks

Comparable data protection systems have been enacted in multiple jurisdictions. Prominent examples encompass the Personal Data (Privacy) Ordinance(PDPO) in Hong Kong125 and Japan’s Act on the Protection of Personal Information(APPI).126 Both are grounded in the shared values of purpose limitation, proportionality, and accountability, offering principled oversight of personal data processing.

Nevertheless, legal compliance alone does not guarantee full protection.127,128 It underscores the necessity for multi-layered privacy-preserving architectures in AI-driven MASLD research.

Data privacy and security strategies

To navigate these complex regulatory terrains and mitigate privacy risks, advanced technical safeguards have been progressively incorporated into AI systems.129

De-identification

De-identification aims to remove or obscure personal identifiers from data. In structured data, methods such as polymorphic encryption have improved resilience against re-identification while maintaining analytical utility. Moreover, large language models are now capable of achieving higher accuracy in automatic de-identification of unstructured text, substantially improving privacy protection for clinical narratives.130

Differential privacy

Differential privacy introduces calibrated random noise into datasets or query results, thereby ensuring that the inclusion or exclusion of any single individual cannot be inferred.131 This approach ensures that the presence or absence of any individual in the dataset cannot be inferred from the analysis results, while still preserving the accuracy of statistical findings at the aggregate level.

Federated learning

Federated learning enables distributed model training across multiple institutions without the exchange of raw data. This paradigm is particularly advantageous for multicenter MASLD studies, allowing algorithmic generalization and cross-population robustness while keeping sensitive data localized within each contributing hospital or research site.132

Synthetic data generation

Synthetic data technology creates artificial datasets that mimic the statistical properties of real-world data but exclude actual patient information.133 Such datasets can be used safely for model testing and algorithm refinement, effectively eliminating privacy concerns inherent in traditional EHR-based development.

Clinical translation of AI in MASLD: Beneficial populations, clinical scenarios, and future pathways

This section explores the transition of AI from research prototypes to real-world applications in MASLD management. It identifies the clinicians and patient populations most likely to benefit and highlights key clinical scenarios where these tools add measurable value. Within this framework, AI emerges not merely as a computational instrument but as a strategic enabler of precision medicine and more equitable healthcare delivery.134

Populations that would benefit most

AI technologies hold considerable promise for improving MASLD management—enhancing diagnostic accuracy, facilitating early intervention, and widening access to care, especially in resource-limited settings.135 They also contribute to objective evaluation metrics in clinical research and therapeutic trials.

Healthcare professionals

For physicians in hepatology, radiology, or primary care, AI-based decision support offers substantial gains. By reducing observer variability and highlighting subtle imaging features, such systems improve diagnostic confidence and consistency. They can also automate repetitive workflows—such as steatosis quantification—and prioritize high-risk cases. For junior clinicians or those outside tertiary centers, these tools function as valuable learning aids and clinical references.136

Populations in resource-limited settings

In underserved areas lacking hepatology expertise, AI-enabled portable ultrasound and automated image analysis can decentralize screening from hospitals to communities. This decentralization promotes earlier diagnosis and intervention. Moreover, digital platforms powered by AI can facilitate remote counseling and adherence tracking, helping sustain long-term care continuity.137

High-risk populations for primary prevention

Individuals with obesity, type 2 diabetes, metabolic syndrome, or related cardiometabolic risk factors form a critical group for preventive strategies. AI-based prediction models integrating routine laboratory and clinical data can identify early hepatic involvement, offering a valuable window for timely lifestyle or pharmacologic intervention before irreversible fibrosis develops.138

Patients enrolled in clinical trials or on pharmacotherapy

In advanced clinical settings, AI-driven digital pathology and imaging analytics provide reproducible quantification of histologic changes, such as steatosis reduction or fibrosis regression.139 These capabilities enhance precision in endpoint measurement, optimize trial efficiency, and support data-driven therapeutic adjustments.

Clinical scenarios of application

AI integration benefits several key clinical workflows, from early detection to long-term management.

Risk stratification and routine screening

When embedded in primary care systems, AI algorithms analyzing EHR and imaging data can flag individuals at risk of MASLD.140 This shift toward proactive, data-informed screening represents a major step away from reactive treatment models.

Lifestyle management and adherence promotion

Mobile health tools and smart wearables can monitor diet, exercise, and metabolic indicators in real time.141 Their feedback mechanisms encourage behavioral adherence and enable personalized guidance through clinician-patient connectivity.

Disease monitoring and follow-up

Remote AI systems leveraging wearable sensors allow continuous disease tracking beyond clinical visits. They can detect early warning signs of progression or noncompliance, reducing reliance on in-person follow-up and improving care accessibility for rural populations.142

Drug response monitoring in drug development

In drug trials, AI-enhanced imaging (such as MRI-PDFF) and computational pathology provide precise, objective measures of treatment efficacy, such as improvements in fibrosis stage or reductions in steatosis burden.143,144 These methods streamline endpoint evaluation, improving the efficiency and robustness of MASLD drug development.

Strengths, challenges, and future research directions

AI is increasingly recognized as a transformative force in healthcare, offering substantial opportunities to enhance diagnostic accuracy and clinical decision-making. However, realizing its full potential in MASLD management requires overcoming substantial technical, ethical, and translational challenges through continuous research and validation.

Strengths: Expanding diagnostic and clinical utility

Evidence increasingly supports the value of AI in refining diagnostic accuracy for hepatic steatosis and fibrosis while reducing interobserver variability. Beyond diagnostics, integration with wearable and behavioral data enables personalized interventions, sustained adherence, and dynamic disease tracking. Continuous monitoring of behavioral, metabolic, and lifestyle parameters enables tailored recommendations—ranging from dietary guidance to activity adjustment—while reinforcing patient adherence. Parallel progress in AI-driven drug discovery and trial optimization further accelerates the identification of novel therapeutic targets and the objective evaluation of drug efficacy. Collectively, these advances expand the reach of precision hepatology and strengthen data-driven care.

Challenges and clinical translation limitations

Despite this progress, routine clinical adoption remains limited. Most AI models rely on retrospective, single-center data, constraining generalizability. Moreover, reference standards differ widely—ranging from histology to MRI or controlled attenuation parameter—hindering comparison. The opaque “black-box” characteristics of many deep learning systems also impede clinician trust and limit regulatory approval. Equally pressing are the gaps in study populations and outcomes. Patients with viral hepatitis, alcohol-related liver disease, or mixed etiologies are frequently underrepresented, and few studies provide robust longitudinal or cost-effectiveness data. Beyond technical challenges, systemic and social barriers also persist. Cost barriers, inadequate digital literacy, and uneven device access exacerbate disparities, especially across rural or low-income communities. Even where devices are available, age, education, and cultural norms may limit engagement. Advanced imaging technologies such as MRI, though powerful, remain concentrated in tertiary centers and entail high costs, reinforcing inequity. Without targeted digital inclusion strategies, these innovations risk amplifying rather than reducing healthcare inequity.

Future research directions

Bridging the divide between laboratory innovation and real-world application is now a central goal for AI in MASLD. Future research must target current gaps—limited external validation, inadequate multimodal integration, and scarce real-world evidence—through a deliberate and iterative translational framework.

First, algorithmic innovation and data fusion are essential. The next generation of models should be interpretable, resilient, and computationally efficient, capable of integrating imaging, clinical, biochemical, and behavioral data. Techniques such as foundation models, transfer learning, and even QML may offer pathways to improved adaptability, particularly in low-data or resource-constrained settings.

Second, systematic validation and consensus-building must follow. Large-scale, multicenter cohorts across diverse populations are vital to ensure external robustness. Standardized annotation protocols, harmonized evaluation metrics, and shared reference datasets would further support reproducibility and comparability across platforms.

Third, ethical and regulatory translation must advance in parallel with technological progress. XAI and privacy-preserving strategies such as federated learning can strengthen transparency and data security, fostering clinical and regulatory acceptance.

Finally, real-world implementation should become an integral part of future studies. Pilot deployments within clinical environments, supported by adaptive policy frameworks, can evaluate usability, workflow compatibility, and economic impact. Continuous learning systems that incorporate patient-generated data and digital biomarkers can, in turn, support dynamic, personalized management. Collectively, these coordinated efforts provide a pragmatic roadmap for transforming AI-driven MASLD research into tangible clinical benefit.

Conclusions

The integration of AI and smart devices is progressively reshaping the comprehensive management framework for MASLD, offering novel perspectives on improving clinical outcomes. By harnessing diverse datasets, including EHR and imaging studies, AI-based predictive tools have greatly enhanced early disease detection, particularly in primary care and for identifying at-risk individuals before symptom onset. In the field of imaging diagnosis, AI-based quantitative analysis technologies for ultrasound, CT, and MRI have effectively overcome the limitations of traditional methods in terms of sensitivity and subjectivity, making reliable assessment more accessible in non-specialist settings. Beyond standard imaging modalities, AI’s analytical power is also being applied to digital pathology. Emerging algorithms, such as QML, have demonstrated high accuracy in liver pathology image recognition, opening up new avenues for non-invasive diagnostic methods. Meanwhile, wearable devices allow real-time tracking of metabolic parameters. When paired with AI algorithms, these systems can generate personalized, adaptive intervention plans, marking a shift from traditional, experience-based lifestyle guidance to a more precise, data-centric management model, which is crucial for long-term patient engagement and adherence. In drug discovery, AI-assisted virtual screening technology has accelerated the discovery and clinical translation of various novel targeted drugs. A typical example is the THR-β agonist Resmetirom, in whose development AI-assisted virtual screening played a pivotal role in candidate selection and optimization, thereby accelerating its path to clinical application and supporting the principles of precision medicine. Despite these impressive advances, translating AI innovations into routine MASLD care continues to face considerable technical and structural obstacles. The opacity of complex models undermines clinical trust, while privacy regulations restrict the multi-center data sharing needed for robust generalization. Overcoming these barriers requires a concerted effort that integrates technical innovation with rigorous ethical and regulatory frameworks. Future priorities should include developing XAI and privacy-preserving computation, alongside establishing standardized validation and regulatory pathways to ensure model safety and equity. In the long run, successful clinical integration will depend on comprehensive multi-center validation across diverse populations and rigorous cost-effectiveness analyses.

With the continuous advancement of technologies such as deep learning and quantum computing, as well as improvements in the performance of mobile sensing devices, the full-cycle management of MASLD will become more intelligent, accurate, and personalized, which will help build a more efficient chronic disease management system.

Declarations

Funding

This study was supported by the National Key Research and Development Program (2024YFA1307101).

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

Conception and design of the study (WZ, GS, JS), drafting of the manuscript, figure preparation, data visualization (WZ), provision of study materials (WZ, QZ, XyX, XY, XbX, HT, YY, WY, JX, GS, JS), and critical revisions of the manuscript (GS, JS). All authors approved the final version and publication of the manuscript.

References

  1. Eslam M, Sanyal AJ, George J, International Consensus Panel. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020;158(7):1999-2014.e1 View Article PubMed/NCBI
  2. Rinella ME, Lazarus JV, Ratziu V, Francque SM, Sanyal AJ, Kanwal F, et al. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. J Hepatol 2023;79(6):1542-1556 View Article PubMed/NCBI
  3. Amini-Salehi E, Letafatkar N, Norouzi N, Joukar F, Habibi A, Javid M, et al. Global Prevalence of Nonalcoholic Fatty Liver Disease: An Updated Review Meta-Analysis comprising a Population of 78 million from 38 Countries. Arch Med Res 2024;55(6):103043 View Article PubMed/NCBI
  4. Miao L, Targher G, Byrne CD, Cao YY, Zheng MH. Current status and future trends of the global burden of MASLD. Trends Endocrinol Metab 2024;35(8):697-707 View Article PubMed/NCBI
  5. Sah AK, Afzal M, Elshaikh RH, Abbas AM, Shalabi MG, Prabhakar PK, et al. Innovative Strategies in the Diagnosis and Treatment of Liver Cirrhosis and Associated Syndromes. Life (Basel) 2025;15(5):779 View Article PubMed/NCBI
  6. Wallace C, Gamkrelidze I, Estes C, Razavi H, Sanyal AJ. Modeling the health and economic impact of pharmacologic therapies for MASLD in the United States. J Hepatol 2025;83(1):21-30 View Article PubMed/NCBI
  7. Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J Hepatol 2020;73(1):202-209 View Article PubMed/NCBI
  8. Song R, Li Z, Zhang Y, Tan J, Chen Z. Comparison of NAFLD, MAFLD and MASLD characteristics and mortality outcomes in United States adults. Liver Int 2024;44(4):1051-1060 View Article PubMed/NCBI
  9. Hagström H, Vessby J, Ekstedt M, Shang Y. 99% of patients with NAFLD meet MASLD criteria and natural history is therefore identical. J Hepatol 2024;80(2):e76-e77 View Article PubMed/NCBI
  10. Younossi ZM, Paik JM, Stepanova M, Ong J, Alqahtani S, Henry L. Clinical profiles and mortality rates are similar for metabolic dysfunction-associated steatotic liver disease and non-alcoholic fatty liver disease. J Hepatol 2024;80(5):694-701 View Article PubMed/NCBI
  11. Song SJ, Lai JC, Wong GL, Wong VW, Yip TC. Can we use old NAFLD data under the new MASLD definition?. J Hepatol 2024;80(2):e54-e56 View Article PubMed/NCBI
  12. Lee WH, Najjar SM, Kahn CR, Hinds TD. Hepatic insulin receptor: new views on the mechanisms of liver disease. Metabolism 2023;145:155607 View Article PubMed/NCBI
  13. Lim S, Kim JW, Targher G. Links between metabolic syndrome and metabolic dysfunction-associated fatty liver disease. Trends Endocrinol Metab 2021;32(7):500-514 View Article PubMed/NCBI
  14. Lin A, Wong ND, Razipour A, McElhinney PA, Commandeur F, Cadet SJ, et al. Metabolic syndrome, fatty liver, and artificial intelligence-based epicardial adipose tissue measures predict long-term risk of cardiac events: a prospective study. Cardiovasc Diabetol 2021;20(1):27 View Article PubMed/NCBI
  15. Zhou XD, Targher G, Byrne CD, Somers V, Kim SU, Chahal CAA, et al. An international multidisciplinary consensus statement on MAFLD and the risk of CVD. Hepatol Int 2023;17(4):773-791 View Article PubMed/NCBI
  16. Eslam M, El-Serag HB, Francque S, Sarin SK, Wei L, Bugianesi E, et al. Metabolic (dysfunction)-associated fatty liver disease in individuals of normal weight. Nat Rev Gastroenterol Hepatol 2022;19(10):638-651 View Article PubMed/NCBI
  17. Dinani AM, Kowdley KV, Noureddin M. Application of Artificial Intelligence for Diagnosis and Risk Stratification in NAFLD and NASH: The State of the Art. Hepatology 2021;74(4):2233-2240 View Article PubMed/NCBI
  18. Li Y, Wang X, Zhang J, Zhang S, Jiao J. Applications of artificial intelligence (AI) in researches on non-alcoholic fatty liver disease(NAFLD) : A systematic review. Rev Endocr Metab Disord 2022;23(3):387-400 View Article PubMed/NCBI
  19. Dunn MA, Kappus MR, Bloomer PM, Duarte-Rojo A, Josbeno DA, Jakicic JM. Wearables, Physical Activity, and Exercise Testing in Liver Disease. Semin Liver Dis 2021;41(2):128-135 View Article PubMed/NCBI
  20. Yeşil F, Çövener Özçelik Ç. Effect of Wearable Technology on Metabolic Control and the Quality of Life in Children and Adolescents with Type 1 Diabetes: A Systematic Review and Meta-Analysis. Balkan Med J 2024;41(4):261-271 View Article PubMed/NCBI
  21. McTeer M, Applegate D, Mesenbrink P, Ratziu V, Schattenberg JM, Bugianesi E, et al. Machine learning approaches to enhance diagnosis and staging of patients with MASLD using routinely available clinical information. PLoS One 2024;19(2):e0299487 View Article PubMed/NCBI
  22. Reinshagen M, Kabisch S, Pfeiffer AFH, Spranger J. Liver Fat Scores for Noninvasive Diagnosis and Monitoring of Nonalcoholic Fatty Liver Disease in Epidemiological and Clinical Studies. J Clin Transl Hepatol 2023;11(5):1212-1227 View Article PubMed/NCBI
  23. Bohte AE, van Werven JR, Bipat S, Stoker J. The diagnostic accuracy of US, CT, MRI and 1H-MRS for the evaluation of hepatic steatosis compared with liver biopsy: a meta-analysis. Eur Radiol 2011;21(1):87-97 View Article PubMed/NCBI
  24. Zhao Q, Lan Y, Yin X, Wang K. Image-based AI diagnostic performance for fatty liver: a systematic review and meta-analysis. BMC Med Imaging 2023;23(1):208 View Article PubMed/NCBI
  25. Crudele L, De Matteis C, Novielli F, Di Buduo E, Petruzzelli S, De Giorgi A, et al. Fatty Liver Index (FLI) is the best score to predict MASLD with 50% lower cut-off value in women than in men. Biol Sex Differ 2024;15(1):43 View Article PubMed/NCBI
  26. Van Vleck TT, Chan L, Coca SG, Craven CK, Do R, Ellis SB, et al. Augmented intelligence with natural language processing applied to electronic health records for identifying patients with non-alcoholic fatty liver disease at risk for disease progression. Int J Med Inform 2019;129:334-341 View Article PubMed/NCBI
  27. Bonfiglio C, Campanella A, Donghia R, Bianco A, Franco I, Curci R, et al. Development and Internal Validation of a Model for Predicting Overall Survival in Subjects with MAFLD: A Cohort Study. J Clin Med 2024;13(4):1181 View Article PubMed/NCBI
  28. Yuan Y, Xu M, Zhang X, Tang X, Zhang Y, Yang X, et al. Development and validation of a nomogram model for predicting the risk of MAFLD in the young population. Sci Rep 2024;14(1):9376 View Article PubMed/NCBI
  29. Zhang X, Zheng MH, Liu D, Lin Y, Song SJ, Chu ES, et al. A blood-based biomarker panel for non-invasive diagnosis of metabolic dysfunction-associated steatohepatitis. Cell Metab 2025;37(1):59-68.e3 View Article PubMed/NCBI
  30. McGlinchey AJ, Govaere O, Geng D, Ratziu V, Allison M, Bousier J, et al. Metabolic signatures across the full spectrum of non-alcoholic fatty liver disease. JHEP Rep 2022;4(5):100477 View Article PubMed/NCBI
  31. Perveen S, Shahbaz M, Keshavjee K, Guergachi A. A Systematic Machine Learning Based Approach for the Diagnosis of Non-Alcoholic Fatty Liver Disease Risk and Progression. Sci Rep 2018;8(1):2112 View Article PubMed/NCBI
  32. Park IG, Yoon SJ, Won SM, Oh KK, Hyun JY, Suk KT, et al. Gut microbiota-based machine-learning signature for the diagnosis of alcohol-associated and metabolic dysfunction-associated steatotic liver disease. Sci Rep 2024;14(1):16122 View Article PubMed/NCBI
  33. Zhu Q, Li H, Ao Z, Xu H, Luo J, Kaurich C, et al. Lipidomic identification of urinary extracellular vesicles for non-alcoholic steatohepatitis diagnosis. J Nanobiotechnology 2022;20(1):349 View Article PubMed/NCBI
  34. Kordy K, Li F, Lee DJ, Kinchen JM, Jew MH, La Rocque ME, et al. Metabolomic Predictors of Non-alcoholic Steatohepatitis and Advanced Fibrosis in Children. Front Microbiol 2021;12:713234 View Article PubMed/NCBI
  35. Conway J, Pouryahya M, Gindin Y, Pan DZ, Carrasco-Zevallos OM, Mountain V, et al. Integration of deep learning-based histopathology and transcriptomics reveals key genes associated with fibrogenesis in patients with advanced NASH. Cell Rep Med 2023;4(4):101016 View Article PubMed/NCBI
  36. Park J, MacLean MT, Lucas AM, Torigian DA, Schneider CV, Cherlin T, et al. Exome-wide association analysis of CT imaging-derived hepatic fat in a medical biobank. Cell Rep Med 2022;3(12):100855 View Article PubMed/NCBI
  37. Chiappini F, Coilly A, Kadar H, Gual P, Tran A, Desterke C, et al. Metabolism dysregulation induces a specific lipid signature of nonalcoholic steatohepatitis in patients. Sci Rep 2017;7:46658 View Article PubMed/NCBI
  38. Ghosh S, Zhao X, Alim M, Brudno M, Bhat M. Artificial intelligence applied to ‘omics data in liver disease: towards a personalised approach for diagnosis, prognosis and treatment. Gut 2025;74(2):295-311 View Article PubMed/NCBI
  39. Cao JS, Lu ZY, Chen MY, Zhang B, Juengpanich S, Hu JH, et al. Artificial intelligence in gastroenterology and hepatology: Status and challenges. World J Gastroenterol 2021;27(16):1664-1690 View Article PubMed/NCBI
  40. Li J, Zou B, Yeo YH, Feng Y, Xie X, Lee DH, et al. Prevalence, incidence, and outcome of non-alcoholic fatty liver disease in Asia, 1999-2019: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2019;4(5):389-398 View Article PubMed/NCBI
  41. Srinivasa Babu A, Wells ML, Teytelboym OM, Mackey JE, Miller FH, Yeh BM, et al. Elastography in Chronic Liver Disease: Modalities, Techniques, Limitations, and Future Directions. Radiographics 2016;36(7):1987-2006 View Article PubMed/NCBI
  42. Kaffas AE, Bhatraju KC, Vo-Phamhi JM, Tiyarattanachai T, Antil N, Negrete LM, et al. Development of a Deep Learning Model for Classification of Hepatic Steatosis from Clinical Standard Ultrasound. Ultrasound Med Biol 2025;51(2):242-249 View Article PubMed/NCBI
  43. Santoro S, Khalil M, Abdallah H, Farella I, Noto A, Dipalo GM, et al. Early and accurate diagnosis of steatotic liver by artificial intelligence (AI)-supported ultrasonography. Eur J Intern Med 2024;125:57-66 View Article PubMed/NCBI
  44. Vianna P, Mehrbod P, Chaudhary M, Eickenberg M, Wolf G, Belilovsky E, et al. Unsupervised Test-Time Adaptation for Hepatic Steatosis Grading Using Ultrasound B-Mode Images. IEEE Trans Ultrason Ferroelectr Freq Control 2025;72(5):601-611 View Article PubMed/NCBI
  45. Wu W, Guo Y, Li Q, Jia C. Exploring the potential of large language models in identifying metabolic dysfunction-associated steatotic liver disease: A comparative study of non-invasive tests and artificial intelligence-generated responses. Liver Int 2025;45(4):e16112 View Article PubMed/NCBI
  46. Drazinos P, Gatos I, Katsakiori PF, Tsantis S, Syrmas E, Spiliopoulos S, et al. Comparison of deep learning schemes in grading non-alcoholic fatty liver disease using B-mode ultrasound hepatorenal window images with liver biopsy as the gold standard. Phys Med 2025;129:104862 View Article PubMed/NCBI
  47. Honarvar M, Lobo J, Schneider C, Wolfe N, Gawrieh S, Loomba R, et al. Deep Learning Based Shear Wave Detection and Segmentation Tool for Use in Point-of-Care for Chronic Liver Disease Assessments. Ultrasound Med Biol 2024;50(12):1812-1820 View Article PubMed/NCBI
  48. Marques R, Santos J, André A, Silva J. Ultrasound Versus Elastography in the Diagnosis of Hepatic Steatosis: Evaluation of Traditional Machine Learning Versus Deep Learning. Sensors (Basel) 2024;24(23):7568 View Article PubMed/NCBI
  49. Liu Y, Yu W, Wang P, Huang Y, Li J, Li P. Deep Learning With Ultrasound Images Enhance the Diagnosis of Nonalcoholic Fatty Liver. Ultrasound Med Biol 2024;50(11):1724-1730 View Article PubMed/NCBI
  50. Kwon H, Kim MG, Oh S, Kim Y, Jung G, Lee HJ, et al. Application of Quantitative Ultrasonography and Artificial Intelligence for Assessing Severity of Fatty Liver: A Pilot Study. Diagnostics (Basel) 2024;14(12):1237 View Article PubMed/NCBI
  51. Vianna P, Calce SI, Boustros P, Larocque-Rigney C, Patry-Beaudoin L, Luo YH, et al. Comparison of Radiologists and Deep Learning for US Grading of Hepatic Steatosis. Radiology 2023;309(1):e230659 View Article PubMed/NCBI
  52. Yang Y, Liu J, Sun C, Shi Y, Hsing JC, Kamya A, et al. Nonalcoholic fatty liver disease (NAFLD) detection and deep learning in a Chinese community-based population. Eur Radiol 2023;33(8):5894-5906 View Article PubMed/NCBI
  53. Jeon SK, Lee JM, Joo I, Yoon JH, Lee G. Two-dimensional Convolutional Neural Network Using Quantitative US for Noninvasive Assessment of Hepatic Steatosis in NAFLD. Radiology 2023;307(1):e221510 View Article PubMed/NCBI
  54. Zsombor Z, Rónaszéki AD, Csongrády B, Stollmayer R, Budai BK, Folhoffer A, et al. Evaluation of Artificial Intelligence-Calculated Hepatorenal Index for Diagnosing Mild and Moderate Hepatic Steatosis in Non-Alcoholic Fatty Liver Disease. Medicina (Kaunas) 2023;59(3):469 View Article PubMed/NCBI
  55. Li B, Tai DI, Yan K, Chen YC, Chen CJ, Huang SF, et al. Accurate and generalizable quantitative scoring of liver steatosis from ultrasound images via scalable deep learning. World J Gastroenterol 2022;28(22):2494-2508 View Article PubMed/NCBI
  56. Byra M, Han A, Boehringer AS, Zhang YN, O’Brien WD, Erdman JW, et al. Liver Fat Assessment in Multiview Sonography Using Transfer Learning With Convolutional Neural Networks. J Ultrasound Med 2022;41(1):175-184 View Article PubMed/NCBI
  57. Che H, Brown LG, Foran DJ, Nosher JL, Hacihaliloglu I. Liver disease classification from ultrasound using multi-scale CNN. Int J Comput Assist Radiol Surg 2021;16(9):1537-1548 View Article PubMed/NCBI
  58. Constantinescu EC, Udriștoiu AL, Udriștoiu ȘC, Iacob AV, Gruionu LG, Gruionu G, et al. Transfer learning with pre-trained deep convolutional neural networks for the automatic assessment of liver steatosis in ultrasound images. Med Ultrason 2021;23(2):135-139 View Article PubMed/NCBI
  59. Han A, Byra M, Heba E, Andre MP, Erdman JW, Loomba R, et al. Noninvasive Diagnosis of Nonalcoholic Fatty Liver Disease and Quantification of Liver Fat with Radiofrequency Ultrasound Data Using One-dimensional Convolutional Neural Networks. Radiology 2020;295(2):342-350 View Article PubMed/NCBI
  60. Cao W, An X, Cong L, Lyu C, Zhou Q, Guo R. Application of Deep Learning in Quantitative Analysis of 2-Dimensional Ultrasound Imaging of Nonalcoholic Fatty Liver Disease. J Ultrasound Med 2020;39(1):51-59 View Article PubMed/NCBI
  61. Byra M, Styczynski G, Szmigielski C, Kalinowski P, Michałowski Ł, Paluszkiewicz R, et al. Transfer learning with deep convolutional neural network for liver steatosis assessment in ultrasound images. Int J Comput Assist Radiol Surg 2018;13(12):1895-1903 View Article PubMed/NCBI
  62. Yin H, Fan Y, Yu J, Xiong B, Zhou B, Sun Y, et al. Quantitative US fat fraction for noninvasive assessment of hepatic steatosis in suspected metabolic-associated fatty liver disease. Insights Imaging 2024;15(1):159 View Article PubMed/NCBI
  63. Graffy PM, Sandfort V, Summers RM, Pickhardt PJ. Automated Liver Fat Quantification at Nonenhanced Abdominal CT for Population-based Steatosis Assessment. Radiology 2019;293(2):334-342 View Article PubMed/NCBI
  64. Martín-Saladich Q, Pericàs JM, Ciudin A, Ramirez-Serra C, Escobar M, Rivera-Esteban J, et al. Metabolic-associated fatty liver voxel-based quantification on CT images using a contrast adapted automatic tool. Med Image Anal 2024;95:103185 View Article PubMed/NCBI
  65. Prinz S, Murray JM, Strack C, Nattenmüller J, Pomykala KL, Schlemmer HP, et al. Novel measures for the diagnosis of hepatic steatosis using contrast-enhanced computer tomography images. Eur J Radiol 2023;160:110708 View Article PubMed/NCBI
  66. Huo Y, Terry JG, Wang J, Nair S, Lasko TA, Freedman BI, et al. Fully automatic liver attenuation estimation combing CNN segmentation and morphological operations. Med Phys 2019;46(8):3508-3519 View Article PubMed/NCBI
  67. Kim HY, Lee KJ, Lee SS, Choi SJ, Kim DH, Heo S, et al. Diagnosis of moderate-to-severe hepatic steatosis using deep learning-based automated attenuation measurements on contrast-enhanced CT. Abdom Radiol (NY) 2025;50(9):4139-4147 View Article PubMed/NCBI
  68. Vong T, Rizer N, Jain V, Thompson VL, Dredze M, Klein EY, et al. Automated identification of incidental hepatic steatosis on Emergency Department imaging using large language models. Hepatol Commun 2025;9(3):e0638 View Article PubMed/NCBI
  69. Yoo J, Joo I, Jeon SK, Park J, Yoon SH. Utilizing fully-automated 3D organ segmentation for hepatic steatosis assessment with CT attenuation-based parameters. Eur Radiol 2024;34(9):6205-6213 View Article PubMed/NCBI
  70. Jeon SK, Joo I, Park J, Yoo J. Automated hepatic steatosis assessment on dual-energy CT-derived virtual non-contrast images through fully-automated 3D organ segmentation. Radiol Med 2024;129(7):967-976 View Article PubMed/NCBI
  71. Pickhardt PJ, Blake GM, Graffy PM, Sandfort V, Elton DC, Perez AA, et al. Liver Steatosis Categorization on Contrast-Enhanced CT Using a Fully Automated Deep Learning Volumetric Segmentation Tool: Evaluation in 1204 Healthy Adults Using Unenhanced CT as a Reference Standard. AJR Am J Roentgenol 2021;217(2):359-367 View Article PubMed/NCBI
  72. Stern C, Castera L. Non-invasive diagnosis of hepatic steatosis. Hepatol Int 2017;11(1):70-78 View Article PubMed/NCBI
  73. Gottfriedova H, Dezortova M, Sedivy P, Pajuelo D, Burian M, Sticova E, et al. Comparison of ultrasound to MR and histological methods for liver fat quantification. Eur J Radiol 2025;183:111931 View Article PubMed/NCBI
  74. Kim JW, Lee CH, Yang Z, Kim BH, Lee YS, Kim KA. The spectrum of magnetic resonance imaging proton density fat fraction (MRI-PDFF), magnetic resonance spectroscopy (MRS), and two different histopathologic methods (artificial intelligence vs. pathologist) in quantifying hepatic steatosis. Quant Imaging Med Surg 2022;12(11):5251-5262 View Article PubMed/NCBI
  75. Martí-Aguado D, Jiménez-Pastor A, Alberich-Bayarri Á, Rodríguez-Ortega A, Alfaro-Cervello C, Mestre-Alagarda C, et al. Automated Whole-Liver MRI Segmentation to Assess Steatosis and Iron Quantification in Chronic Liver Disease. Radiology 2022;302(2):345-354 View Article PubMed/NCBI
  76. Wang K, Cunha GM, Hasenstab K, Henderson WC, Middleton MS, Cole SA, et al. Deep Learning for Inference of Hepatic Proton Density Fat Fraction From T1-Weighted In-Phase and Opposed-Phase MRI: Retrospective Analysis of Population-Based Trial Data. AJR Am J Roentgenol 2023;221(5):620-631 View Article PubMed/NCBI
  77. Meneses JP, Qadir A, Surendran N, Arrieta C, Tejos C, Andia ME, et al. Unbiased and reproducible liver MRI-PDFF estimation using a scan protocol-informed deep learning method. Eur Radiol 2025;35(5):2843-2854 View Article PubMed/NCBI
  78. Li S, Wang Z, Ding Z, She H, Du YP. Accelerated four-dimensional free-breathing whole-liver water-fat magnetic resonance imaging with deep dictionary learning and chemical shift modeling. Quant Imaging Med Surg 2024;14(4):2884-2903 View Article PubMed/NCBI
  79. Bastati N, Perkonigg M, Sobotka D, Poetter-Lang S, Fragner R, Beer A, et al. Correlation of histologic, imaging, and artificial intelligence features in NAFLD patients, derived from Gd-EOB-DTPA-enhanced MRI: a proof-of-concept study. Eur Radiol 2023;33(11):7729-7743 View Article PubMed/NCBI
  80. Pollack BL, Batmanghelich K, Cai SS, Gordon E, Wallace S, Catania R, et al. Deep Learning Prediction of Voxel-Level Liver Stiffness in Patients with Nonalcoholic Fatty Liver Disease. Radiol Artif Intell 2021;3(6):e200274 View Article PubMed/NCBI
  81. Cho Y, Kim MJ, Park BJ, Sim KC, Keu YS, Han YE, et al. Active Learning for Efficient Segmentation of Liver with Convolutional Neural Network-Corrected Labeling in Magnetic Resonance Imaging-Derived Proton Density Fat Fraction. J Digit Imaging 2021;34(5):1225-1236 View Article PubMed/NCBI
  82. Zheng TL, Sha JC, Deng Q, Geng S, Xiao SY, Yang WJ, et al. Object detection: A novel AI technology for the diagnosis of hepatocyte ballooning. Liver Int 2024;44(2):330-343 View Article PubMed/NCBI
  83. Lusnig L, Sagingalieva A, Surmach M, Protasevich T, Michiu O, McLoughlin J, et al. Hybrid Quantum Image Classification and Federated Learning for Hepatic Steatosis Diagnosis. Diagnostics (Basel) 2024;14(5):558 View Article PubMed/NCBI
  84. Naoumov NV, Brees D, Loeffler J, Chng E, Ren Y, Lopez P, et al. Digital pathology with artificial intelligence analyses provides greater insights into treatment-induced fibrosis regression in NASH. J Hepatol 2022;77(5):1399-1409 View Article PubMed/NCBI
  85. Harrison SA, Bedossa P, Guy CD, Schattenberg JM, Loomba R, Taub R, et al. A Phase 3, Randomized, Controlled Trial of Resmetirom in NASH with Liver Fibrosis. N Engl J Med 2024;390(6):497-509 View Article PubMed/NCBI
  86. Sessa L, Concilio S, Fominaya J, Eletto D, Piotto S, Busquets X. A new serotonin 2A receptor antagonist with potential benefits in Non-Alcoholic Fatty Liver Disease. Life Sci 2023;314:121315 View Article PubMed/NCBI
  87. Wang Y, Shen L, Wang C, Dong Y, Hua H, Xu J, et al. Lipidation-dimerization platform unlocks treatment potential of fibroblast growth factor 21 for non-alcoholic steatohepatitis. J Control Release 2024;376:1130-1142 View Article PubMed/NCBI
  88. Zhang L, Zheng Y, Shao M, Chen A, Liu M, Sun W, et al. AlphaFold-based AI docking reveals AMPK/SIRT1-TFEB pathway modulation by traditional Chinese medicine in metabolic-associated fatty liver disease. Pharmacol Res 2025;212:107617 View Article PubMed/NCBI
  89. Saldarriaga OA, Wanninger TG, Arroyave E, Gosnell J, Krishnan S, Oneka M, et al. Heterogeneity in intrahepatic macrophage populations and druggable target expression in patients with steatotic liver disease-related fibrosis. JHEP Rep 2024;6(1):100958 View Article PubMed/NCBI
  90. Harrison SA, Alkhouri N, Ortiz-Lasanta G, Rudraraju M, Tai D, Wack K, et al. A phase IIb randomised-controlled trial of the FFAR1/FFAR4 agonist icosabutate in MASH. J Hepatol 2025;83(2):293-303 View Article PubMed/NCBI
  91. Ozlu Karahan T, Kenger EB, Yilmaz Y. Artificial Intelligence-Based Diets: A Role in the Nutritional Treatment of Metabolic Dysfunction-Associated Steatotic Liver Disease?. J Hum Nutr Diet 2025;38(2):e70033 View Article PubMed/NCBI
  92. Newsome PN, Sanyal AJ, Neff G, Schattenberg JM, Ratziu V, Ertle J, et al. A randomised Phase IIa trial of amine oxidase copper-containing 3 (AOC3) inhibitor BI 1467335 in adults with non-alcoholic steatohepatitis. Nat Commun 2023;14(1):7151 View Article PubMed/NCBI
  93. Yu H, Yan S, Jin M, Wei Y, Zhao L, Cheng J, et al. Aescin can alleviate NAFLD through Keap1-Nrf2 by activating antioxidant and autophagy. Phytomedicine 2023;113:154746 View Article PubMed/NCBI
  94. Lee WY, Lee CY, Lee JS, Kim CE. Identifying Candidate Flavonoids for Non-Alcoholic Fatty Liver Disease by Network-Based Strategy. Front Pharmacol 2022;13:892559 View Article PubMed/NCBI
  95. Huang Y, Wang X, Zhang L, Zheng K, Xiong J, Li J, et al. Effect of Probiotics Therapy on Nonalcoholic Fatty Liver Disease. Comput Math Methods Med 2022;2022:7888076 View Article PubMed/NCBI
  96. Chen S, Zeng Q, Cai X, Xue J, Yin G, Song P, et al. Multiomics analyses decipher intricate changes in the cellular and metabolic landscape of steatotic livers upon dietary restriction and sleeve gastrectomy. Int J Biol Sci 2024;20(11):4438-4457 View Article PubMed/NCBI
  97. Yuan HY, Tong XF, Ren YY, Li YY, Wang XL, Chen LL, et al. AI-based digital pathology provides newer insights into lifestyle intervention-induced fibrosis regression in MASLD: An exploratory study. Liver Int 2024;44(10):2572-2582 View Article PubMed/NCBI
  98. Golovaty I, Tien PC, Price JC, Sheira L, Seligman H, Weiser SD. Food Insecurity May Be an Independent Risk Factor Associated with Nonalcoholic Fatty Liver Disease among Low-Income Adults in the United States. J Nutr 2020;150(1):91-98 View Article PubMed/NCBI
  99. Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A, et al. Personalized Nutrition by Prediction of Glycemic Responses. Cell 2015;163(5):1079-1094 View Article PubMed/NCBI
  100. Metro D, Buda M, Manasseri L, Corallo F, Cardile D, Lo Buono V, et al. Role of Nutrition in the Etiopathogenesis and Prevention of Nonalcoholic Fatty Liver Disease (NAFLD) in a Group of Obese Adults. Medicina (Kaunas) 2023;59(3):638 View Article PubMed/NCBI
  101. Simancas-Racines D, Annunziata G, Verde L, Fascì-Spurio F, Reytor-González C, Muscogiuri G, et al. Nutritional Strategies for Battling Obesity-Linked Liver Disease: the Role of Medical Nutritional Therapy in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Management. Curr Obes Rep 2025;14(1):7 View Article PubMed/NCBI
  102. Pugliese N, Wai-Sun Wong V, Schattenberg JM, Romero-Gomez M, Sebastiani G, Aghemo A, et al. Accuracy, Reliability, and Comprehensibility of ChatGPT-Generated Medical Responses for Patients With Nonalcoholic Fatty Liver Disease. Clin Gastroenterol Hepatol 2024;22(4):886-889.e5 View Article PubMed/NCBI
  103. Su TH, Wu CH, Kao JH. Artificial intelligence in precision medicine in hepatology. J Gastroenterol Hepatol 2021;36(3):569-580 View Article PubMed/NCBI
  104. Whyte MB, Shojaee-Moradie F, Sharaf SE, Cuthbertson DJ, Kemp GJ, Barrett M, et al. HDL-apoA-I kinetics in response to 16 wk of exercise training in men with nonalcoholic fatty liver disease. Am J Physiol Endocrinol Metab 2020;318(6):E839-E847 View Article PubMed/NCBI
  105. Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, Torres-Gonzalez A, Gra-Oramas B, Gonzalez-Fabian L, et al. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology 2015;149(2):367-378.e5 View Article PubMed/NCBI
  106. Newsome PN, Loomba R. Therapeutic horizons in metabolic dysfunction-associated steatohepatitis. J Clin Invest 2025;135(13):e186425 View Article PubMed/NCBI
  107. Hu Y, Sun C, Chen Y, Liu YD, Fan JG. Pipeline of New Drug Treatment for Non-alcoholic Fatty Liver Disease/Metabolic Dysfunction-associated Steatotic Liver Disease. J Clin Transl Hepatol 2024;12(9):802-814 View Article PubMed/NCBI
  108. Sanyal AJ, Brunt EM, Kleiner DE, Kowdley KV, Chalasani N, Lavine JE, et al. Endpoints and clinical trial design for nonalcoholic steatohepatitis. Hepatology 2011;54(1):344-353 View Article PubMed/NCBI
  109. Harrison SA, Taub R, Neff GW, Lucas KJ, Labriola D, Moussa SE, et al. Resmetirom for nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled phase 3 trial. Nat Med 2023;29(11):2919-2928 View Article PubMed/NCBI
  110. Hu X, Ge J, Wen Y. Computational study of novel natural agonists targeting farnesoid X receptor. Sci Rep 2024;14(1):17597 View Article PubMed/NCBI
  111. Ndikuryayo F, Gong XY, Hao GF, Yang WC. How Artificial Intelligence Assists in Overcoming Drug Resistance?. Med Res Rev 2025;45(6):1679-1699 View Article PubMed/NCBI
  112. Oleribe OO, Taylor-Robinson AW, Agala VR, Sobande OO, Izurieta R, Taylor-Robinson SD. Global Adoption, Promotion, Impact, and Deployment of AI in Patient Care, Health Care Delivery, Management, and Health Care Systems Leadership: Cross-Sectional Survey. J Med Internet Res 2025;27:e70805 View Article PubMed/NCBI
  113. World Health Organization. 2021;Ethics and Governance of Artificial Intelligence for Health: WHO Guidance. [Online]. Available from: https://www.who.int/publications/i/item/9789240029200
  114. Fei Du R, Lloret Carbonell E, Huang J, Liu S, Wang X, Shen D, et al. Ethics of Foundation Models in Computational Pathology: Overview of Contemporary Issues and Future Implications. IEEE Trans Med Imaging 2025;44(10):4098-4115 View Article PubMed/NCBI
  115. Fareed M, Fatima M, Uddin J, Ahmed A, Sattar MA. A systematic review of ethical considerations of large language models in healthcare and medicine. Front Digit Health 2025;7:1653631 View Article PubMed/NCBI
  116. Jonnagaddala J, Wong ZS. Privacy preserving strategies for electronic health records in the era of large language models. NPJ Digit Med 2025;8(1):34 View Article PubMed/NCBI
  117. Obermeyer Z, Powers B, Vogeli C, Mullainathan S. Dissecting racial bias in an algorithm used to manage the health of populations. Science 2019;366(6464):447-453 View Article PubMed/NCBI
  118. Cary MP, Zink A, Wei S, Olson A, Yan M, Senior R, et al. Mitigating Racial And Ethnic Bias And Advancing Health Equity In Clinical Algorithms: A Scoping Review. Health Aff (Millwood) 2023;42(10):1359-1368 View Article PubMed/NCBI
  119. Salahuddin Z, Woodruff HC, Chatterjee A, Lambin P. Transparency of deep neural networks for medical image analysis: A review of interpretability methods. Comput Biol Med 2022;140:105111 View Article PubMed/NCBI
  120. Karako K, Tang W. Applications of and issues with machine learning in medicine: Bridging the gap with explainable AI. Biosci Trends 2025;18(6):497-504 View Article PubMed/NCBI
  121. Rizzo M. AI in Neurology: Everything, Everywhere, all at Once PART 2: Speech, Sentience, Scruples, and Service. Ann Neurol 2025;98(3):431-447 View Article PubMed/NCBI
  122. Department of Health and Human Services. Health Insurance Portability and Accountability Act of 1996. New York, NY: Department of Health and Human Services; 1996
  123. Cohen IG, Mello MM. HIPAA and Protecting Health Information in the 21st Century. JAMA 2018;320(3):231-232 View Article PubMed/NCBI
  124. Voigt P, Von Dem Bussche A. The EU general data protection regulation (GDPR): A practical guide. Cham, Switzerland: Springer Nature, 2024. Available from:https://link.springer.com/book/10.1007/978-3-031-62328-8
  125. Personal Data (Privacy) Ordinance, Chapter 486 of the Laws of Hong Kong (PDPO). Available from: http://www.hklii.org.hk/hk/legis/ord/486/index.html
  126. Act on the protection of personal information, Act No. 57 of 2003. Available from: https://www. japaneselawtranslation.go.jp/en/laws/view/4241/en
  127. Sweeney L, Yoo JS, Perovich L, Boronow KE, Brown P, Brody JG. Re-identification Risks in HIPAA Safe Harbor Data: A study of data from one environmental health study. Technol Sci 2017;2017:2017082801 PubMed/NCBI
  128. Gadotti A, Rocher L, Houssiau F, Creţu AM, de Montjoye YA. Anonymization: The imperfect science of using data while preserving privacy. Sci Adv 2024;10(29):eadn7053 View Article PubMed/NCBI
  129. Meskó B, Topol EJ. The imperative for regulatory oversight of large language models (or generative AI) in healthcare. NPJ Digit Med 2023;6(1):120 View Article PubMed/NCBI
  130. Jonnagaddala J, Dai H-J, Chen C-T. Large language models for automatic deidentification of electronic health record notes. Singapore: Springer Nature Singapore; 2025. Available from:https://link.springer.com/book/10.1007/978-981-97-7966-6
  131. Zhang S, Li X. Differential privacy medical data publishing method based on attribute correlation. Sci Rep 2022;12(1):15725 View Article PubMed/NCBI
  132. Peng L, Luo G, Zhou S, Chen J, Xu Z, Sun J, et al. An in-depth evaluation of federated learning on biomedical natural language processing for information extraction. NPJ Digit Med 2024;7(1):127 View Article PubMed/NCBI
  133. Chen A, Jonnagaddala J, Nekkantti C, Liaw ST. Generation of Surrogates for De-Identification of Electronic Health Records. Stud Health Technol Inform 2019;264:70-73 View Article PubMed/NCBI
  134. Topol EJ. High-performance medicine: the convergence of human and artificial intelligence. Nat Med 2019;25(1):44-56 View Article PubMed/NCBI
  135. Dunn N, Verma N, Dunn W. Artificial Intelligence for Predictive Diagnostics, Prognosis, and Decision Support in MASLD, Hepatocellular Carcinoma, and Digital Pathology. J Clin Exp Hepatol 2026;16(1):103184 View Article PubMed/NCBI
  136. Shrestha UK. Emerging role of artificial intelligence in gastroenterology and hepatology. World J Gastroenterol 2025;31(39):111495 View Article PubMed/NCBI
  137. Bharadwaj HR, Dahiya DS, Dalal P, Fuad M, Raza HA, Ibrahim M, et al. Artificial Intelligence in Population-Level Gastroenterology and Hepatology: A Comprehensive Review of Public Health Applications and Quantitative Impact. Dig Dis Sci 2025 View Article PubMed/NCBI
  138. Nivethitha V, Daniel RA, Debnath A, Dwarakanathan V, Jeer G, Kavipriya G. Diagnostic accuracy of artificial intelligence models for imaging detection of hepatic steatosis through systematic review and meta analysis. Sci Rep 2025;15(1):34408 View Article PubMed/NCBI
  139. Ratziu V, Hompesch M, Petitjean M, Serdjebi C, Iyer JS, Parwani AV, et al. Artificial intelligence-assisted digital pathology for non-alcoholic steatohepatitis: current status and future directions. J Hepatol 2024;80(2):335-351 View Article PubMed/NCBI
  140. Zhu G, Song Y, Lu Z, Yi Q, Xu R, Xie Y, et al. Machine learning models for predicting metabolic dysfunction-associated steatotic liver disease prevalence using basic demographic and clinical characteristics. J Transl Med 2025;23(1):381 View Article PubMed/NCBI
  141. Kim HJ, Lee KH, Lee JH, Youk H, Lee HY. The Effect of a Mobile and Wearable Device Intervention on Increased Physical Activity to Prevent Metabolic Syndrome: Observational Study. JMIR Mhealth Uhealth 2022;10(2):e34059 View Article PubMed/NCBI
  142. Mahajan A, Heydari K, Powell D. Wearable AI to enhance patient safety and clinical decision-making. NPJ Digit Med 2025;8(1):176 View Article PubMed/NCBI
  143. Pulaski H, Harrison SA, Mehta SS, Sanyal AJ, Vitali MC, Manigat LC, et al. Clinical validation of an AI-based pathology tool for scoring of metabolic dysfunction-associated steatohepatitis. Nat Med 2025;31(1):315-322 View Article PubMed/NCBI
  144. Caussy C, Reeder SB, Sirlin CB, Loomba R. Noninvasive, Quantitative Assessment of Liver Fat by MRI-PDFF as an Endpoint in NASH Trials. Hepatology 2018;68(2):763-772 View Article PubMed/NCBI

About this Article

Cite this article
Zhu W, Zheng Q, Xu X, Yu X, Xu X, Tu H, et al. Applications of Artificial Intelligence and Smart Devices in Metabolic Dysfunction-associated Steatotic Liver Disease. J Clin Transl Hepatol. Published online: Dec 11, 2025. doi: 10.14218/JCTH.2025.00406.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
August 9, 2025 November 11, 2025 November 20, 2025 December 11, 2025
DOI http://dx.doi.org/10.14218/JCTH.2025.00406
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Applications of Artificial Intelligence and Smart Devices in Metabolic Dysfunction-associated Steatotic Liver Disease

Wenfeng Zhu, Qi Zheng, Xinyi Xu, Xia Yu, Xianbin Xu, Huilan Tu, Yue Yu, Wubing Ying, Jiahao Xie, Guoping Sheng, Jifang Sheng
  • Reset Zoom
  • Download TIFF