logo
AAT Bioquest

Unlocking the Potential of 3D Cell Culture: A Guide to Assay Optimization

Abstract


The exploration of three-dimensional (3D) cell cultures in contemporary biomedical research reveals their pivotal role as a bridge between conventional two-dimensional (2D) models and the intricacies of in vivo systems. It meticulously addresses the hurdles encountered during the transition to 3D cultures, emphasizing the imperative need for nuanced assay optimization strategies. Additionally, the integration of 3D cultures into high throughput screening is examined, alongside contemplations on the promising horizon of organ-on-a-chip and multi-tissue systems in the realm of future possibilities. Despite formidable challenges, 3D cultures offer an avenue to augment research depth and relevance within the domains of cellular research and drug discovery.

 

Introduction


Biomedical research has witnessed a remarkable transformation with the advent of cell culture techniques. The transition from traditional two-dimensional (2D) to three-dimensional (3D) cell culture systems marks a significant milestone in cellular studies. While 2D platforms have served as workhorses in research for decades, their limitations in replicating the complex in vivo environment have become increasingly apparent. The emergence of 3D cell cultures, closely mimicking physiological conditions, promises a more authentic representation of in vivo scenarios. However, this transition is not without its challenges, particularly when it comes to adapting and optimizing assays for 3D culture systems.

 

The Significance of 3D Cultures in Modern Biomedical Research


The shift from traditional 2D cell cultures to advanced 3D cell culture models represents a distinct leap forward in cell biology and tissue engineering. In 2D cultures, cells typically form monolayers on flat surfaces like culture dishes or flasks. While this approach is convenient and suitable for high-throughput screening, its limitations become evident when trying to replicate the intricate three-dimensional structures of tissues and organs. Consequently, 2D cultures fall short in faithfully mimicking in vivo conditions and studying complex, heterogeneous cell interactions.

Conversely, 3D cultures are designed to closely replicate the physiological environment. Cells within these cultures grow in three dimensions, often within a supportive matrix, allowing for realistic cell-cell and cell-matrix interactions. This unique feature makes 3D cultures invaluable for tissue-specific modeling, disease research, and drug testing, offering critical insights into how compounds behave within a tissue-like context. While their advantages are undeniable, it is crucial to acknowledge that 3D cultures can be more challenging to establish and require meticulous consideration of spatial factors. Nevertheless, they provide a more relevant platform for addressing research questions related to tissue development, drug efficacy, and disease mechanisms. Researchers must, therefore, choose between these two culture systems based on their specific research objectives and experimental requirements.
Gel Box Diagram

Comparison of 2D versus 3D cell culture models (figure made in BioRender).

 

Navigating the Challenges of 3D Culture Assay Optimization


The transition from 2D to 3D cell cultures introduces a host of challenges. One particularly noteworthy challenge is the altered dynamics of diffusion within 3D environments. In 2D cultures, the diffusion of nutrients, gases, drugs, and assay reagents is relatively straightforward. However, in 3D cultures, the penetration of these substances becomes inherently more complex, leading to uneven gradients, especially concerning oxygen and essential nutrients. These uneven gradients can significantly impact cellular behavior and, consequently, assay outcomes.

Furthermore, the inherent depth of 3D structures can pose challenges when it comes to imaging clarity. Traditional microscopy techniques, optimized for observing shallow 2D layers, encounter difficulties when visualizing cells situated deeper within 3D structures. This can lead to potential misinterpretations or omissions in data acquisition, underscoring the importance of addressing these imaging challenges in the optimization of assays for 3D cell cultures.

 

Assay Optimization: Tackling the Challenges Head-On


The transition from a traditional 2D experimental setup to a more complex 3D environment represents a significant shift in the field of assay optimization. This shift is not merely a matter of adding an extra dimension but entails a complete reevaluation and redefinition of the entire experimental process. To successfully navigate this transition, tailored approaches to assay optimization become essential.

One critical aspect of this transition involves addressing challenges related to cell viability and proliferation assessment. In traditional 2D systems, commonly used colorimetric assays like the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay are relied upon to measure cell viability. However, in the context of 3D matrices, a new set of challenges arises. The formazan crystals produced during the MTT assay may not solubilize effectively in the dense 3D environment. In response, ATP-based assays, such as the ReadiUse™ Rapid Luminometric ATP Assay Kit, the Cell Meter™ Live Cell ATP Assay kit, or the PhosphoWorks™ ATP Assays have emerged as a more suitable alternative, as they measure cellular ATP levels, offering increased sensitivity and the ability to penetrate deeper into the 3D cultures.

Table 1. Comparison of MTT and ATP-based assays commonly used in cell viability and proliferation studies.
AspectMTT Assay (2D)MTT Assay (3D)ATP-based Assay (2D)ATP-based Assay (3D)
PrincipleMeasures mitochondrial activity (metabolic activity)Measures mitochondrial activity (metabolic activity)Measures ATP production (cell viability)Measures ATP production (cell viability)
Advantages
  • Widely used, well-established
  • Cost-effective
  • Simple protocol
  • Compatible with various cell types
  • Measures metabolic changes
  • Applicable to some 3D cultures
  • Suitable for simple 3D models
  • Can capture metabolic changes
  • Provides insights into 3D-specific effects
  • Non-destructive to some 3D cultures
  • Direct measurement of cell viability
  • Sensitive and rapid
  • Compatible with various cell types
  • Measures ATP production in real-time
  • Detects changes in real-time, indicating viability
  • Direct measurement of cell viability
  • Suitable for some 3D models
  • Minimal interference from dead cells
  • Suitable for high-throughput screening
  • Provides insights into 3D-specific effects
Disadvantages
  • Indirect measurement of viability
  • May yield false positives in some cases
  • Requires solubilization step for quantification
  • Requires solubilization step for quantification
  • Limited applicability to complex 3D models
  • May require additional assays for accuracy
  • May underestimate viability in certain cases
  • Difficulties in uniform compound exposure
  • Requires ATP extraction or lysis
  • Limited sensitivity in low ATP conditions
  • More expensive reagents
  • May not distinguish between viable and stressed cells
  • Requires ATP extraction or lysis
  • Sensitivity to environmental factors
  • More expensive reagents
  • May not distinguish between viable and stressed cells


In drug studies and other research contexts, tracking cell apoptosis (programmed cell death) is of paramount importance. While conventional 2D assays for apoptosis often rely on colorimetric outputs, these may not provide optimal results within the intricate 3D matrix. Instead, assays utilizing fluorescence or luminescence have proven to be superior, delivering enhanced clarity and sensitivity, making them the preferred choice for 3D settings.

To further enhance our understanding of cellular behavior within 3D structures, researchers have turned to advanced imaging techniques. Among these techniques, confocal and multiphoton microscopy stand out due to their capacity to capture sequential z-stack images. When these images are reconstructed, they offer comprehensive 3D views, enabling researchers to gain deeper insights into cellular organization and behavior within the complex 3D environment.

Additionally, the choice of matrix material for 3D cultures plays a pivotal role in shaping cellular behavior. Whether using natural matrices like collagen or Matrigel or synthetic alternatives, the matrix composition significantly influences assay outcomes. Therefore, it is imperative to carefully fine-tune matrix components to strike the right balance, providing adequate support for cellular activities while avoiding unintended interference with assay results. This consideration underscores the multifaceted nature of assay optimization in the transition from 2D to 3D experimental setups, where researchers must address not only assay methods but also the intricacies of the matrix environment itself.

 

Scaling Up: 3D Cultures in High Throughput Screening


The realm of pharmaceuticals recognizes the immense promise held by 3D cultures, particularly in the context of drug screening. The inclusion of 3D models within high throughput screening (HTS) frameworks, however, necessitates a rigorous commitment to standardization. Even slight variations in critical parameters, such as spheroid size or seeding density, can profoundly influence experimental outcomes. To navigate this terrain effectively, automation, particularly in the domains of imaging and data interpretation, must exhibit a high degree of proficiency in accommodating the unique intricacies inherent to 3D cultures.

To harness the potential of 3D cultures in pharmaceutical research, it is vital to establish standardized protocols. These protocols should encompass the formation of spheroids, the selection of suitable cell lines, and the optimization of culture conditions, including media composition, pH levels, and oxygen concentrations. Given that fluctuations in these parameters can significantly skew results, precision in control is paramount.

Automation plays a pivotal role in ensuring the reliability and consistency of 3D culture experiments. Liquid-handling robots, for instance, can meticulously control seeding density and spheroid size. Moreover, the imaging of 3D structures demands advanced technologies like confocal or multi-photon microscopy, and automating this process enhances efficiency in handling a substantial number of samples.

The challenge of interpreting data arising from 3D cultures is further accentuated by their three-dimensional nature. Specialized software and algorithms are imperative to accurately quantify parameters such as spheroid size, shape, and viability. Integrating this unique dataset into HTS workflows necessitates the development of databases and computational tools tailored to the specific demands of 3D assays, encompassing both imaging and molecular profiling data.

Incorporating high-content screening (HCS) systems is particularly beneficial when dealing with 3D cultures, as they furnish a comprehensive array of information extending beyond conventional viability assays. By marrying automated imaging with advanced data analysis, HCS extracts intricate details about cellular morphology and protein localization, among others.

Sustaining the integrity of 3D cultures over time mandates rigorous quality control measures. Regular assessments, including cell viability checks and validation of spheroid morphology, are indispensable to uphold consistency. Likewise, validation studies are essential to ascertain the reproducibility and reliability of 3D HTS assays, often entailing comparisons with established 2D assays and in vivo data.

Additionally, it is vital to consider regulatory requirements when employing 3D cultures in drug development, as these may diverge from those applicable to 2D assays. By embracing these challenges and rigorously addressing them, the integration of 3D cultures into high throughput screening platforms promises to enhance the precision and relevance of pharmaceutical drug development processes.

Table 2. Critical parameters for high-throughput screening (HTS) in 3D cell culutres.
ParameterDescription
3D Culture Model SelectionChoose an appropriate 3D culture model (spheroids, organoids, hydrogels, etc.) based on your research objectives.
Cell Type and SourceSelect relevant cell types and sources that mimic the target tissue or disease.
Culture ConditionsOptimize culture conditions including media composition, pH, oxygen levels, and temperature for 3D growth.
Assay DevelopmentDevelop assays compatible with 3D cultures, ensuring they provide meaningful readouts for your screening goals.
AutomationImplement automated liquid handling and robotics to streamline handling of 3D cultures and assays.
Imaging and AnalysisUse high-content imaging systems and software for data acquisition and analysis of 3D structures.
Plate FormatChoose appropriate microplate formats (e.g., 96-well, 384-well) for your HTS to match the scale of your screening.
Assay ReadoutsSelect relevant endpoints, such as viability, proliferation, apoptosis, or specific biomarkers, based on your research objectives.
Replicates and ControlsEnsure sufficient replicates and positive/negative controls to assess assay robustness and reproducibility.
Compound LibrariesPrepare or acquire compound libraries for screening, considering the size and diversity of the library.
Data ManagementEstablish data storage, management, and analysis workflows to handle the large volume of data generated in HTS.
Hit Confirmation and ValidationConfirm hits from primary screens using secondary assays and validate potential drug candidates.
Data StandardizationImplement standardized data formats and quality control measures to enhance data comparability.
Hit PrioritizationDevelop criteria for prioritizing hits based on potency, specificity, and other relevant factors.
Hit CharacterizationCharacterize identified compounds for pharmacological properties and mechanism of action.
Data Visualization and ReportingCreate user-friendly data visualization tools and reports to facilitate decision-making.
Screening Assay MiniaturizationOptimize assays for miniaturization to reduce reagent and sample consumption, as well as screening costs.
Throughput and Screening CapacityDetermine the desired throughput (e.g., screens per day) and screening capacity of the HTS platform.
Data Integration with External SourcesIntegrate data from external sources, such as literature, to augment screening results and prioritize compounds.
Quality Control and QC MetricsEstablish quality control metrics and monitoring procedures to ensure assay performance and data reliability.
Ethical Considerations and ComplianceAdhere to ethical guidelines and regulatory compliance, especially when using patient-derived samples or clinical data.
Hit Validation in Relevant ModelsValidate hits in more complex, physiologically relevant models, including in vivo studies if applicable.

 

Peering into the Future of 3D Cell Cultures


Advancements in cellular biology suggest that the future of 3D cultures lies in even more sophisticated models. Concepts like organ-on-a-chip and multi-tissue systems are on the horizon. These intricate models, designed to simulate multiple organ systems simultaneously, promise to deliver unparalleled physiological relevance. Yet, their increased complexity will bring about new challenges in assay optimization, necessitating an interdisciplinary melding of cell biology, materials science, engineering, and computational analytics.

 

Conclusion


3D cell cultures represent a monumental evolution in cellular research, bridging the gap between the simplicity of 2D cultures and the elaborate dynamics of in vivo systems. They promise a closer approximation to physiological reality, making them indispensable tools in modern research. While the journey to full optimization might be laden with challenges, the potential benefits in terms of research depth, clarity, and relevance make it a journey worth undertaking. As we march into a future brimming with technological advancements, 3D cultures, with their blend of complexity and physiological relevance, are poised to be at the forefront of cellular research and drug discovery.

 

References

  1. Astashkina, A., Grainger, D. W., & Critical Path Institute. (2012). Critical challenges in high content screening for tissue engineering applications. Tissue Engineering Part C: Methods, 18(11), 837-842.
  2. Breslin, S., & O'Driscoll, L. (2013). Three-dimensional cell culture: the missing link in drug discovery. Drug Discovery Today, 18(5-6), 240-249.
  3. Chen, Y., Li, A., Tan, X., Chen, Z., Ding, K., & Li, R. (2015). 3D culture models of tissues under tension for regenerative medicine and cancer research. BioMed Research International, 2015.
  4. Edmondson, R., Broglie, J. J., Adcock, A. F., & Yang, L. (2014). Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay and Drug Development Technologies, 12(4), 207-218
  5. Fennema, E., Rivron, N., Rouwkema, J., van Blitterswijk, C., & de Boer, J. (2013). Spheroid culture as a tool for creating 3D complex tissues. Trends in Biotechnology, 31(2), 108-115.
  6. Huh, D., Leslie, D. C., Matthews, B. D., Fraser, J. P., Jurek, S., Hamilton, G. A., ... & Ingber, D. E. (2012). A human disease model of drug toxicity–induced pulmonary edema in a lung-on-a-chip microdevice. Science Translational Medicine, 4(159), 159ra147-159ra147.
  7. Kapałczyńska, M., Kolenda, T., Przybyła, W., Zajączkowska, M., Teresiak, A., Filas, V., ... & Lamperska, K. (2018). 2D and 3D cell cultures—a comparison of different types of cancer cell cultures. Archives of Medical Science, 14(4), 910-919.
  8. Riss, T. L., Moravec, R. A., Niles, A. L., Duellman, S., Benink, H. A., Worzella, T. J., ... & Minor, L. (2004). Cell viability assays. In Assay Guidance Manual (pp. 1-25). Eli Lilly & Company and the National Center for Advancing Translational Sciences.
  9. Yin, Z., & Wang, C. (2014). Microfluidic cell culture systems for drug research. Lab on a Chip, 14(6), 1039-1042.

 

Products


Cat No.Product NameUnit Size
22768Cell Meter™ Colorimetric MTT Cell Proliferation Kit1000 Tests
22769Cell Meter™ Colorimetric MTT Cell Proliferation Kit5000 Tests
23015Cell Meter™ Live Cell ATP Assay Kit100 Tests
21617PhosphoWorks™ Colorimetric ATP Assay Kit100 Tests
21620PhosphoWorks™ Fluorimetric ATP Assay Kit100 Tests
21609PhosphoWorks™ Luminometric ATP Assay Kit *Extended Luminescence*1 Plates
21608PhosphoWorks™ Luminometric ATP Assay Kit *Extended Luminescence*10 Plates
21610PhosphoWorks™ Luminometric ATP Assay Kit *Maximized Luminescence*1 Plates
21621PhosphoWorks™ Luminometric ATP Assay Kit *Maximized Luminescence*10 Plates
21612PhosphoWorks™ Luminometric ATP Assay Kit *DTT-Free*10 Plates
21613PhosphoWorks™ Luminometric ATP Assay Kit *DTT-Free*10 Plates
21601ReadiUse™ Rapid Luminometric ATP Assay Kit100 Tests
21602ReadiUse™ Rapid Luminometric ATP Assay Kit1000 Tests
21603ReadiUse™ Rapid Luminometric ATP Assay Kit10000 Tests