Item 92/100: Hydrogels Emerging in 3-D Cultures
Karlie Wysong, MS, Senior Biologist
ATCC® HB-8065™ – Hep G2
Three-dimensional (3-D) cell cultures are increasingly studied and utilized as they better replicate the complexities of the tumor microenvironment and overall physiological relevance as compared to traditional two-dimensional (2-D) methods.1 Hydrogels have emerged as a prominent 3-D cell culture scaffolding techniques, serving as an alternative to the extracellular matrix (ECM). Hydrogels are 3-D networks of hydrophilic polymers—natural, synthetic, or hybrid—that can absorb large amounts of biological fluids while still maintaining their structural integrity.1 This property enables cells to aggregate and grow into 3-D structures that mimic tissue and organ architecture.
In one study, synthetic supramolecular hydrogels were used with Hep G2 cells (ATCC® HB-8065™) to illustrate the potential for hydrogel matrices for liver tissue engineering.2 The hydrogel was formed from bis-urea amphiphiles containing lactobionic acid and maltobionic acid bioactive ligands; both carbohydrate ligands bind to a specific receptor in hepatic cells allowing them to form spheroids within the scaffolding. After 5 days, the cells were in spheroid formation and showed positive viability and functionality.2
In another study, a bioartificial hydrogel composed of poly(vinyl alcohol) (PVA) and gelatin was used with Hep G2 to model a hepatocellular carcinoma (HCC) in 3-D.3 This hydrogel composition proved effective in developing long-term HCC models, enabling investigations into important aspects of tumor biology and migration patterns.3 Beyond Hep G2 applications, the biocompatibility of hydrogels makes them suitable in diverse research applications like cancer biology, tissue engineering, and regenerative medicine.
Item 93/100: Trusted and Reliable: ATCC Powers Breast Cancer Research with Digital Biology
Rula Khairi, BS Pharm, MS, Senior Biologist
ATCC® HTB-26™ – MDA-MB-231
MDA-MB-231 (ATCC® HTB-26™) is a widely studied breast cancer cell line known for its triple-negative characteristics—the lack of estrogen receptors (ER), progesterone receptors (PR), and HER2 protein. This invasive form of cancer accounts for 10-15% of all breast cancers, especially in women under 40. As a result, MDA-MB-231 has become a critical model for researching triple-negative breast cancer (TNBC).4
The rise of digital biology is revolutionizing cancer research. By leveraging computational tools, artificial intelligence (AI), and data science, digital biology allows researchers to simulate and model biological systems, accelerating research while reducing reliance on traditional lab experiments. Digital platforms can replicate cell behaviors, predict responses to treatments, and even simulate disease progression—all in a computer environment.5 For these models to be effective, data quality is essential. MDA-MB-231 from ATCC provides a reliable, authenticated cell line, ensuring that researchers are working with accurate, contamination-free cells. ATCC guarantees the authenticity of these cells through rigorous testing like STR profiling and mycoplasma screening, which prevents misidentification and contamination. This level of assurance is crucial when using MDA-MB-231 cells to build AI-driven models or simulate biological processes. By integrating genomic data and clinical information, digital biology not only helps in understanding the mechanics of TNBC but also advances personalized treatment strategies. With the power of AI and high-quality cell lines like MDA-MB-231, scientists are creating smarter tools to predict, prevent, and treat aggressive breast cancers more effectively than ever before. Thus, as digital biology grows, the demand for dependable cell lines like MDA-MB-231 continues to shape the future of breast cancer research.
Item 94/100: Exploring S. equi and Advancing Veterinary Medicine
Jeanette Rimbey, MSc, Lead Biologist
ATCC® 9528™ – Streptococcus equi subsp. equi Sand and Jensen
Streptococcus equi subsp. equi (commonly known as S. equi) is responsible for strangles, a highly contagious and often serious upper respiratory disease in horses. The impact of strangles extends beyond animal health, causing substantial economic losses in the equine industry due to the need for quarantine, medical treatment, and prolonged recovery times. Developing vaccines for S. equi is difficult due to its ability to avoid immune detection and persist in carriers without symptoms. Its protective capsule and virulence factors limit long-term immunity, while strain differences and the need to protect against Streptococcus equi subsp. zooepidemicus add complexity.
To support these efforts, ATCC plays a critical role by providing authenticated microbial strains and genomic resources essential for vaccine research. For example, S. equi strain 2/1/2023, isolated from a foal with strangles, has been used in proteomics and reverse vaccinology studies to identify extracellular vesicles as promising vaccine candidates. Beyond microbial cultures, ATCC offers genome sequenced reference materials and cell lines that enable researchers to study virulence factors, host-pathogen interactions, and immune responses. With the complete genome of S. equi available through the ATCC Genome Portal, scientists can explore its genetic evolution and develop targeted vaccines, improved diagnostics, and novel therapies—advancing the long-term goal of eliminating strangles as a global equine health threat.
Item 95/100: Microbes Mining Metals and Capturing Carbon
Leka Papazisi, PhD, Principal Scientist, Product Lifecycle Management, ATCC Research & Industrial Solutions
ATCC® 9844™ – Gluconobacter oxydans (Henneberg) De Ley
Imagine a world where microbes help us extract valuable metals from rocks while also fighting climate change. That’s the promise of biomining, a cutting-edge process where bacteria like Gluconobacter oxydans break down minerals to pull out elements such as copper, gold, and rare earths. This technique skips harsh chemicals, works at room temperature, and produces less pollution than traditional mining.6 What makes G. oxydans truly remarkable is its ability to produce acid that dissolves rocks and frees up metals for use in electronics, batteries, and solar panels. Scientists have genetically engineered the bacterium to enhance its capabilities, increasing its efficiency in leaching rare earth elements by up to 73% and doubling the extraction speed. Even more impressive, when these microbes interact with certain rocks, they accelerate the process of locking away carbon dioxide into stable minerals, doing it 58 times faster than nature alone.7
Biomining isn’t just for mining raw ore. The same microbes can help recycle metals from electronic waste and clean up contaminated sites. In fact, the world already uses biomining to harvest around 10–15% of the world’s copper, and the method accounts for 5% of global gold production.7 As the demand for metals grows and the need for sustainable technologies rises, these tiny miners may play a big role in both powering our future and cleaning our planet.
Item 96/100: The Matrix Behind the Model: Cell Basement Membrane in Organoid Development
Fernanda Ventura, BS, Biologist
ATCC® ACS-3035™ – Cell Basement Membrane
Human Cancer Models Initiative (HCMI) models are transforming how we study cancer, enabling researchers to identify novel therapies and accelerate translational research. ATCC is proud to be a provider of these models, offering a comprehensive and diverse portfolio from both common and rare cancers across various tissue types. Central to the cultivation of these models is the Cellular Basement Membrane (ATCC® ACS-3035™)—a Matrigel purified from murine Engelbreth-Holm-Swarm (EHS) tumors. This specialized matrix is essential for supporting the 3-D culture of primary tissue-derived human and mouse organoids, including patient-derived organoids. Cell Basement Membrane plays a pivotal role in advancing cancer research. Once warmed, it polymerizes into an active matrix that mimics the mammalian basement membrane, providing the structural and biochemical signals necessary for cell morphology, differentiation, and tumor growth. This environment enables organoids to develop their characteristic architecture, making them invaluable tools for modeling cancer in vitro.
Item 97/100: Desmoid Tumor Model Supports Rare Cancer Research
Matthew Graziano, BS, Biologist
ATCC® PDM-625™ – HCM-BROD-0762-C49
Desmoid tumors, also called desmoid fibromatosis, are locally aggressive growths that occur in connective tissue throughout the body.8 Despite being unable to metastasize, desmoid tumors may cause local damage to surrounding tissue, create functional problems in vital organs, and pose challenges through high rates of recurrence.9 Distinguishing between neoplastic and stromal cells in connective tissue tumors is challenging, and isolations of these neoplastic cells for disease modeling have historically limited access to verified desmoid tumor models.10
There is currently a lack of verified and standardized desmoid tumor cell models commercially available for the scientific community. Through ATCC’s collaboration with the Human Cancer Models Initiative (HCMI), ATCC is helping to enable research on rare cancer types by making them available for pharmacological screening applications while pairing these resources with directly linked clinical and molecular characterization.
HCM-BROD-0762-C49 is a patient-derived next-generation cancer model developed from primary abdominal fibromatosis. The cell model is positive for the CTNNB1 T41A mutation—a β-catenin mutation prevalent in desmoid-type fibromatosis.10 Additional molecular characterization on this model, including whole-genome sequencing, whole-exome sequencing, RNA-seq, and DNA methylation, is available through the Genomic Data Commons (GDC) as well as the HCMI Searchable Catalog. On the ATCC website, users will find all relevant product information, as well as free-to-access, optimized growth protocols and resources for model culture.
Item 98/100: State-Of-The-Art In Vitro Liver Models
Changsuk Moon, PhD, Senior Scientist
ATCC® PCS-450-010™ – HepatoXcell™ Plus: Normal Human Hepatocytes
The liver is the largest internal organ in the human body and is responsible for over 500 metabolic functions. Accurate liver models are essential for studying liver physiology, disease mechanism, drug metabolism, and toxicity, which are critical areas in drug discovery and development. Traditionally, animal models and conventional 2-D cell cultures have been used for this purpose. However, both approaches have significant limitations in replicating human-specific physiological responses, reducing their predictive value in clinical research. While 2-D cultures of primary human hepatocytes are considered the gold standard for in vitro liver models, it has notable limitations, including a short culture lifespan, lack of structural and functional complexity, and a gradual loss of liver-specific functions over time.
Microphysiological systems (MPS) are cutting-edge in vitro platforms designed to mimic the biochemical, electrical, and mechanical properties of human organs. These advanced technologies employ 3-D cell cultures integrated within microfluidic devices to recreate a physiologically relevant microenvironment, enabling more accurate modeling of organ-level functions and responses. By replicating physiologically relevant 3-D tissue architecture, MPS supports long-term culture durations while enabling controlled mechanical stimulation and complex multicellular interactions, effectively replicating the dynamic and physiologically relevant environment of human organs.
Freshly isolated primary human hepatocytes are cryopreserved to preserve their viability and functionality. High-quality HepatoXcell™ primary human hepatocytes and validated liver-specific media kits provide a comprehensive culture system for establishing robust in vitro liver models. These systems are well-suited for integration into MPS systems, offering a promising alternative to animal models by enabling physiologically relevant and ethically responsible research. Furthermore, the ability to interconnect multiple organ models allows for integrated, systemic studies, facilitating more accurate pharmacokinetic and pharmacodynamic assessments across organ systems.
Item 99/100: Aeromonas: From Symbiont to Pathogen and Beyond
James Budnick, PhD, Senior Scientist, Microbiology R&D, ATCC
ATCC® 35624™ – Aeromonas veronii Hickman-Brenner et al.
In the natural world, leeches have their own microbial partner: Aeromonas, a genus of gram-negative, facultatively anaerobic bacteria commonly found in aquatic environments. While many species are harmless, some can cause infections in both humans and animals, ranging from gastroenteritis and wound infections to septicemia and respiratory illnesses.11
Aeromonas veronii is a dominant species within the microbiota of the leech gut. This bacterium aids the leech in digesting blood meals by producing enzymes that break down complex molecules and by secreting antimicrobial compounds that prevent spoilage of stored blood. Due to its simple and stable microbiome, the medicinal leech is considered an excellent model for studying host-microbe interactions. Researchers use it to explore microbial transmission, immune modulation, and evolutionary stability of symbiosis.12 While A. veronii is beneficial to leeches, it can be opportunistic in humans. Medicinal leech therapy, used to relieve venous congestion post-surgery, has been associated with infections in up to 20% of cases if prophylactic antibiotics are not used.13 This dual nature—a symbiont in leeches and a pathogen in humans—makes Aeromonas a subject of intense study.
In aquaculture, Aeromonas is a major concern due to its role in diseases like hemorrhagic septicemia. These bacteria possess a wide array of virulence factors, including hemolysins, proteases, enterotoxins, and the ability to form biofilms.11 Their genetic plasticity also allows for horizontal gene transfer, contributing to the spread of antibiotic resistance. Recent studies have highlighted the growing issue of multidrug resistance in Aeromonas, including resistance to beta-lactams, carbapenems, and colistin.14,15 Aeromonas spp. also play a role in environmental monitoring. Its presence in drinking water systems and wastewater is used as an indicator of microbial contamination and water quality. Molecular techniques such as qPCR and gene profiling have improved detection and risk assessment of toxigenic strains in water sources.16
Aeromonas spp. show promise in biotechnology. Certain strains have been explored for their ability to produce industrial enzymes and biopolymers or for their use in bioremediation strategies. Recent advances have even positioned Aeromonas as a fast-growing alternative for recombinant protein production, rivaling traditional systems like E. coli.17
Item 100/100: Next-Generation Organoid Models for Colorectal Cancer Research
Matthew Graziano, BS, Biologist
ATCC® PDM-43™ – HCM-SANG-0266-C20
Colorectal cancer (CRC) is the second leading cause of cancer-related mortality worldwide. CRC cases may present asymptomatic, leading to advanced clinical stages with limited treatment options. ATCC currently offers a variety of tools available to support colorectal cancer drug discovery and disease modeling, including advanced models in addition to traditional colorectal cancer cell line alternatives. Advanced cancer models provided from The Human Cancer Models Initiative (HCMI) better represent in vivo physiology, genetic diversity, and clinical functionality. In collaboration between the National Cancer Institute, Cancer Research UK, Wellcome Sanger Institute, Hubrecht Organoid Technology, ATCC has generated hundreds of novel patient-derived cancer models that are supported with clinical and molecular annotation. These models were manufactured, characterized, and validated at ATCC and are available for academic and commercial use to support cancer research and drug discovery.
HCM-SANG-0266-C20 is a next-generation patient-derived cancer model developed from primary adenocarcinoma of rectum. Whole-exome and whole-transcriptome profiling reveal that HCM-SANG-0266-C20 harbors key driver mutations in relevant oncogenes including KRAS, APC, SMAD4, and PIK3CA. HCM-SANG-0266-C20, identified as a KRAS G12D mutant, exhibited heightened sensitivity to MRTX1133, a potent KRAS G12D inhibitor in a series of drug sensitivity screenings.18 Meanwhile, CRC organoids tested (n=28) exhibited more homogenous sensitivities to broad chemotherapeutic agents like cisplatin and 5-fluorouracil. In combination with available sequencing data, drug sensitivity screenings suggest potential utility of HCMI organoid models in the drug discovery pipeline.18
Additional clinical information and molecular characterization on this model, including whole-genome sequencing, whole-exome sequencing, RNA-seq, and DNA methylation, is available through the Genomic Data Commons (GDC) as well as the HCMI Searchable Catalog. On the ATCC website, users will find all relevant product information, as well as free-to-access, optimized growth protocols and resources for model culture.19 Through ATCC’s collaboration with the Human Cancer Models Initiative (HCMI), ATCC is helping to enable research on next-generation 3-D organoids by making them commercially available for pharmacological screening applications, while pairing these resources with directly linked clinical and molecular characterization.
Did you know?
The future of science is fueled by discovery and there is still a wealth of organisms out there to be identified. The National Science Foundation estimates that between 15,000 and 20,000 new species are named each year.20
Read our other posts from the 100 for 100 series
100 for 100: A Centennial Celebration of the ATCC Collection
In this first post in our 100 for 100 series, learn about the microorganisms that are special to our staff.
More100 for 100: Looking back at ATCC’s most influential deposits
In this second post in our 100 for 100 series, learn about our most influential cell lines and microorganisms.
More100 for 100: Toxicology
In this third post in our 100 for 100 series, learn about ATCC's reliable tools for toxicology.
More100 for 100: Cancer
In this fourth post in our 100 for 100 series, learn about ATCC's advanced cancer models.
More100 for 100: Bioinformatics at ATCC
In this fifth post in our 100 for 100 series, explore interesting whole-genome sequenced items in our collection.
More100 for 100: Vector-Borne Diseases
In this sixth post in our 100 for 100 series, learn about ATCC's vast portfolio of vector-borne disease pathogens.
More100 for 100: Harnessing the Power of Microorganisms for Environmental Sustainability
In this seventh post in our 100 for 100 series, learn about ATCC's strains that support environmental sustainability.
More100 for 100: Tiny Organisms, Big Impact
In this eighth post in our 100 for 100 series, we reflect on the incredible range of organisms housed at ATCC and their role in driving scientific progress.
More100 for 100: Bioproduction Powerhouses
In this ninth post in our 100 for 100 series, we showcase how ATCC’s diverse resources are powering the future of food, fuel, pharmaceuticals, and beyond.
More100 for 100: Microbes, Mutations, Moonshots and More
In this tenth post in our 100 for 100 series, we showcase the power of microorganisms and cell models to shape research and innovation.
More100 for 100: Shaping the Future of Antimicrobial Resistance Research
In this eleventh post in our 100 for 100 series, we explore the cutting-edge science and bold innovation shaping the fight against antimicrobial resistance.
MoreReferences
- Abuwatfa WH, Pitt WG, Husseini GA. Scaffold-based 3D cell culture models in cancer research. J Biomed Sci 31(1): 7, 2024. PubMed: 38221607
- Liu J, et al. Hepatic Spheroid Formation on Carbohydrate-Functionalized Supramolecular Hydrogels. Biomacromolecules 24(6): 2447-2458, 2023. PubMed: 37246400
- Moscato S, Ronca F, Campani D, Danti S. Poly(vinyl alcohol)/gelatin Hydrogels Cultured with HepG2 Cells as a 3D Model of Hepatocellular Carcinoma: A Morphological Study. J Funct Biomater 6(1):16-32, 2015. PubMed: 25590431
- American Cancer Society. (n.d.). Triple-negative breast cancer: Details, diagnosis, and signs. Details, Diagnosis, and Signs . https://www.cancer.org/cancer/types/breast-cancer/about/types-of-breast-cancer/triple-negative.html
- Digital Biology: Evolution, components, applications, and more. The Lifesciences Magazine. (June 7, 2024). https://thelifesciencesmagazine.com/digital-biology-data-and-life-sciences/
- Lee JJ, et al. Bio-accelerated weathering of ultramafic minerals with Gluconobacter oxydans. Sci Rep 15(1): 15134, 2025. PubMed: 40307501
- Schmitz AM, et al. High efficiency rare earth element bioleaching with systems biology guided engineering of Gluconobacter oxydans. Commun Biol 8(1): 815, 2025. PubMed: 40425722
- Mayo Clinic, Mayo Foundation for Medical Education and Research. Desmoid Tumors. (December 6, 2023) https://www.mayoclinic.org/diseases-conditions/desmoid-tumors/symptoms-causes/syc-20355083.
- The Desmoid Tumor Research Foundation. What Is a Desmoid Tumor? (February 25, 2025). https://www.desmoidtumors.com/about-desmoid-tumors/.
- Timbergen MJM, et al. Differentially Methylated Regions in Desmoid-Type Fibromatosis: A Comparison Between CTNNB1 S45F and T41A Tumors. Front Oncol 10: 565031, 2020. PubMed: 33194643
- Majeed S, et al. Occurrence of potential virulence determinants in Aeromonas spp. isolated from different aquatic environments.J Appl Microbiol 134(3): lxad031, 2023. PubMed: 36809788
- Tasiemski A, et al. Reciprocal immune benefit based on complementary production of antibiotics by the leech Hirudo verbana and its gut symbiont Aeromonas veronii. Sci Rep 5: 17498, 2015. PubMed: 26635240
- Graf J. Symbiosis of Aeromonas veronii Biovar sobria and Hirudo medicinalis, the Medicinal Leech: a Novel Model for Digestive Tract Associations. Infect Immun 67(1): 1–7, 1999. PubMed: 9864188
- Pineda-Reyes R, et al. Clinical Presentation, Antimicrobial Resistance, and Treatment Outcomes of Aeromonas Human Infections: A 14-Year Retrospective Study and Comparative Genomics of 2 Isolates From Fatal Cases. Clin Infect Dis 79(5): 1144–1152, 2024. PubMed: 38759099
- Gray HK, et al. Nosocomial infections by diverse carbapenemase-producing Aeromonas hydrophila associated with combination of plumbing issues and heat waves. Am J Infect Control 52(3): 337–343, 2024. PubMed: 37778710
- Robertson BK. Molecular Detection, Quantification, and Toxigenicity Profiling of Aeromonas spp. in Source- and Drinking-Water. Open Microbiol J 8: 32-39, 2014. PubMed: 24949108
- Tang M-X, et al. Aeromonas spp. as a fast-growing high-performance chassis for protein production. Appl Environ Microbiol 91(7): e0078025, 2025. PubMed: 40459293
- Clinical, Molecular, and Functional Characterization of a Diverse Collection of Patient-Derived Colorectal Cancer Organoids from the Human Cancer Models Initiative. www.atcc.org/resources/posters/2025-posters/patient-derived-colorectal-cancer-organoids.
- National Institutes of Health, U.S. Department of Health and Human Services. HCMI Catalog. https://hcmi-searchable-catalog.nci.nih.gov/
- US National Science Foundation. Life in the fast lane: Racing to identify species as biodiversity shrinks. ( January 4, 2013). https://www.nsf.gov/news/life-fast-lane-racing-identify-species