Liver toxicity testing is a crucial element of preclinical safety assessment, offering early insights into the hepatotoxic potential of drug candidates and chemical substances. As the primary organ involved in xenobiotic metabolism and detoxification, the liver is highly susceptible to compound-induced injury. Identifying hepatotoxicity early in the development pipeline is essential to avoid costly late-stage failures and to ensure the progression of safe therapeutic agents. BOC Sciences combines state-of-the-art technology platforms with deep toxicological expertise to deliver a full spectrum of liver toxicity testing services. Our capabilities include both in vitro and in vivo assessments, mechanistic studies, biomarker profiling, and histopathological evaluations.
Liver toxicity testing, while critical, often presents substantial challenges that can compromise research outcomes. Conventional liver assays may lack sensitivity to detect early hepatocellular injury, leading to delayed toxicity identification and increased development costs. Developing reliable in vitro liver models, such as primary hepatocytes, spheroids, or microfluidic liver-on-a-chip systems, requires high technical expertise and is resource intensive. In vivo studies must address species-specific differences in hepatic metabolism and transporter expression, which often complicate translational relevance. Additionally, interpretation of liver toxicity data relies on integrated evaluation of biochemical parameters (e.g., ALT, AST, bilirubin), histopathology, and molecular markers such as CYP enzymes and apoptosis regulators. These tasks demand cross-functional collaboration between toxicologists, pathologists, and bioinformaticians. Finally, project timelines are frequently strained by the complexity of workflows, data volume, and the need for customized study design and reporting.
We offer a full range of in vitro models (e.g., human hepatocytes, HepaRG, liver organoids) and in vivo systems (rodent and non-rodent) to evaluate hepatotoxic effects under diverse conditions.
We investigate hepatotoxic mechanisms including oxidative stress, mitochondrial dysfunction, lipid accumulation, and cholestasis, supporting mechanism-informed safety evaluation.
All studies are conducted under strict SOPs and quality frameworks to ensure accuracy, consistency, and reproducibility across every stage of testing.
Our platforms enable quantitative measurement of key liver injury markers such as ALT, AST, ALP, bilirubin, GLDH, and novel biomarkers like miR-122 and HMGB1.
BOC Sciences provides comprehensive liver toxicity evaluation services to support drug safety assessment and biomedical research. Leveraging advanced biochemical and pathological techniques, we deliver precise analysis of liver injury mechanisms, functional impairment, and toxicological impacts.
Our panel includes alanine aminotransferase (ALT), aspartate aminotransferase (AST), and lactate dehydrogenase (LDH) measurements, offering sensitive detection of hepatocyte damage, mitochondrial injury, and cell membrane integrity, critical for early identification of acute liver injury.
We assess alkaline phosphatase (ALP), γ-glutamyl transferase (GGT), total bilirubin (TBIL), and direct bilirubin (DBIL) to detect bile excretion impairment and distinguish between hemolytic, hepatocellular, and obstructive jaundice types with high specificity.
Our evaluation of albumin (Alb), prealbumin (PA), prothrombin time (PT), and cholinesterase (CHE) comprehensively reflects hepatic protein synthesis capacity and coagulation status, aiding prognosis and monitoring of liver failure progression.
We quantify malondialdehyde (MDA), glutathione (GSH), superoxide dismutase (SOD), and inflammatory cytokines (TNF-α, IL-6) to reveal oxidative damage and inflammatory responses associated with drug-induced liver injury (DILI).
Our services include liver biopsy with hematoxylin-eosin staining and immunohistochemistry for apoptosis (e.g., caspase-3) and fibrosis markers (e.g., α-SMA), providing detailed tissue-level insights into liver damage and remodeling.
We specialize in assessing liver toxicity related to nanomaterials and natural products by examining hepatic uptake, clearance, oxidative stress, and elucidating specific mechanisms of hepatotoxicity to ensure comprehensive toxicological profiling.
BOC Sciences offers flexible and tailored liver toxicity testing services designed to meet the unique requirements of each client. Our testing portfolio supports a broad range of products, including small molecules, biologics, natural products, nanomaterials, and novel therapeutic modalities. By integrating advanced biochemical assays, histopathological evaluation, and molecular biomarkers, we develop customized testing strategies to comprehensively assess hepatotoxic risks. Our expert team collaborates closely with clients to optimize study design, ensuring precise, reproducible results aligned with project goals. Whether for preclinical safety evaluation or mechanistic research, BOC Sciences delivers bespoke solutions that accelerate drug development and enhance decision-making confidence. We welcome inquiries to discuss your specific needs and co-develop the ideal liver toxicity assessment plan.
BOC Sciences offers an extensive portfolio of hepatotoxicity assays to support early safety evaluation, mechanism elucidation, and drug screening applications. Leveraging validated cellular models and advanced analytical platforms, our hepatotoxicity testing services encompass the following 12 robust methods:
This assay evaluates membrane integrity by quantifying lactate dehydrogenase (LDH) released from damaged cells into the culture medium. It is a cost-effective and sensitive method for assessing cytotoxicity. However, medium components such as fetal bovine serum (FBS) may interfere with the assay and should be carefully controlled.
PI is a membrane-impermeable dye that selectively stains non-viable cells. It is widely used for high-throughput screening (HTS) of cytotoxic agents. Dye concentration optimization is essential to minimize false-positive results.
These assays indirectly measure cellular metabolic activity via mitochondrial enzyme reduction. WST-1 and XTT provide improved water solubility for real-time assessment. Use of electron-coupling agents such as phenazine methosulfate is recommended to enhance sensitivity.
Using fluorescent probes like JC-1, this assay detects mitochondrial depolarization, an early indicator of oxidative stress and apoptosis. It enables sensitive monitoring of mitochondrial function under drug treatment.
This method evaluates lipid peroxidation by detecting malondialdehyde (MDA) via thiobarbituric acid-reactive substances (TBARS). While rapid and easy to implement, specificity limitations may lead to overestimation of MDA levels.
We assess cellular oxidative defense mechanisms through the quantification of antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GPx). These assays help characterize redox imbalance induced by hepatotoxic agents.
This assay evaluates the inhibition of bile acid efflux transporters (BSEP/MRP2), serving as a high-throughput-compatible tool for detecting cholestasis risk. However, test compound interference should be considered during assay interpretation.
5(6)-Carboxy-2',7'-dichlorofluorescein diacetate (CDFDA) is used to monitor MRP2-mediated excretion of fluorescent metabolites, enabling the assessment of bile canalicular damage and transporter function.
Using sandwich-cultured human hepatocytes, this assay measures urea production ratios (DICI values) to distinguish between cholestatic and non-cholestatic hepatotoxicity. It provides a mechanistically relevant model for cholestasis screening.
A specific stain for intracellular neutral lipids such as triglycerides, Oil Red O is applied to both 2D and 3D hepatocyte/organoid models. While effective for steatosis detection, it does not differentiate lipid subtypes.
By tracking fluorescently labeled fatty acids or quantifying apolipoprotein B100 (APOB100), this assay evaluates dysregulated lipid metabolism associated with hepatic steatosis and lipid export impairment.
This assay assesses hepatic fibrosis via collagen quantification. Sirius Red staining binds specifically to collagen fibers for imaging-based analysis, while the hydroxyproline assay offers sensitive colorimetric detection after acid hydrolysis. Together, they provide reliable evaluation of liver fibrogenesis.
BOC Sciences has established a comprehensive hepatotoxicity testing platform encompassing both in vitro and in vivo models. Leveraging standardized protocols and advanced technical expertise, we provide reliable and efficient support for liver toxicity assessment and mechanistic research throughout early-stage drug development.
Model Type | Description |
Primary Human Hepatocytes | Retain complete liver cell functions and metabolic enzyme activities; considered the gold standard for in vitro hepatotoxicity testing, widely used in drug metabolism and toxicity evaluation. |
HepaRG Cell Line | Exhibits stable hepatic functions and expression of various metabolic enzymes, suitable for long-term culture and high-throughput screening. |
3D Liver Spheroids | Constructed via three-dimensional culture techniques to mimic the liver microenvironment, enhancing cell-cell interactions and improving physiological relevance for hepatotoxicity prediction. |
Co-culture Models | Co-culture of hepatocytes with Kupffer cells, stellate cells, and other non-parenchymal cells to simulate the complex liver microenvironment, providing a more comprehensive hepatotoxicity response. |
iPSC-derived Hepatocytes | Derived from induced pluripotent stem cells using differentiation protocols; exhibit hepatic characteristics, suitable for personalized hepatotoxicity assessment and mechanistic studies. |
Model Type | Description |
Rodent Liver Toxicity Models | Administration of compounds via oral, intravenous, or intraperitoneal routes in mice or rats to evaluate liver toxicity in whole-organism contexts, including serum biochemistry and histopathology analysis. |
Transgenic Liver Toxicity Models | Genetically engineered mice with knockout or knock-in of specific metabolic enzymes or signaling pathways, enabling in-depth mechanistic studies of liver toxicity. |
Liver Perfusion Models | Ex vivo perfusion of isolated livers to dynamically monitor hepatic metabolism and toxic responses, combined with histological and biochemical assessments. |
BOC Sciences' hepatotoxicity testing platform is equipped with state-of-the-art instruments designed to deliver comprehensive liver toxicity assessment, mechanistic studies, and biomarker analysis. Our advanced instrumentation ensures high-throughput, sensitive, and reproducible testing, supporting robust data generation for research and development projects in the biochemical and pharmaceutical fields.
Engage in in-depth communication with clients to fully understand their research objectives, compound characteristics (e.g., chemical structure, metabolic profile), and specific hepatotoxicity concerns such as mitochondrial dysfunction, bile acid transport inhibition, or oxidative stress. This ensures a focused and fit-for-purpose testing strategy.
Design a customized hepatotoxicity testing plan based on compound class and toxicological endpoints. Select appropriate in vitro hepatic models (e.g., primary human hepatocytes, HepaRG cells, 3D liver spheroids) and define measurable indicators (e.g., ATP content, ALT/AST release, ROS levels). Establish concentration ranges, exposure durations, and control groups to ensure scientific rigor and reproducibility.
Establish biologically relevant liver model systems with functional expression of metabolic enzymes (e.g., CYP450 isoforms) and hepatic transporters. Where appropriate, integrate 3D cultures or co-culture systems with non-parenchymal liver cells to enhance physiological relevance. Validate system performance to ensure assay reliability and sensitivity.
Conduct systematic toxicity testing under optimized laboratory conditions. Apply multiple assays to evaluate compound-induced effects on cell viability, mitochondrial membrane potential, oxidative stress levels, and bile salt export. Utilize high-content imaging and multiplex technologies to generate multidimensional mechanistic insights.
Collect quantitative data across all assays and perform comprehensive statistical analysis. Derive key toxicological parameters such as IC50, inhibition rates, and time-dependent effects. Apply structure–toxicity relationship (STR) analysis and integrate results for mechanistic interpretation and prioritization of compounds.
Deliver a structured, data-rich report encompassing methodology, graphical and tabular data presentations, hepatotoxicity profiles, and mechanistic evaluations. The report is tailored for internal research teams to support lead optimization and early decision-making.
The most common markers for liver toxicity include elevated levels of enzymes such as alanine aminotransferase (ALT) and aspartate aminotransferase (AST). These enzymes are released into the bloodstream when liver cells are damaged. Additional markers include alkaline phosphatase (ALP), bilirubin, and gamma-glutamyl transferase (GGT). In vitro hepatotoxicity testing often measures these biomarkers alongside cellular viability and mitochondrial function to assess liver injury.
Hepatotoxicity is tested in vitro using various liver cell models such as primary human hepatocytes, HepaRG cells, and 3D liver spheroids. Assays typically measure endpoints like cell viability, enzyme leakage (ALT/AST), mitochondrial membrane potential, oxidative stress, and bile acid transporter function. These models help predict compound-induced liver injury early in the drug development process.
Mitochondrial dysfunction plays a critical role in liver toxicity because mitochondria are essential for energy production and cellular metabolism. Damage to mitochondria can lead to decreased ATP production, increased reactive oxygen species (ROS), and activation of cell death pathways, which contribute to hepatocyte injury. Therefore, assessing mitochondrial health is a key parameter in comprehensive hepatotoxicity evaluation.
Primary human hepatocytes remain the gold standard for in vitro liver toxicity testing due to their intact metabolic enzyme expression and physiological relevance. Advanced models like HepaRG cells and 3D liver spheroids also provide improved prediction by maintaining liver-specific functions and cell-cell interactions. The choice of model depends on the testing objective, with 3D systems increasingly used for long-term and mechanistic studies.