Imagine a world where a devastating genetic disease like sickle-cell anemia is not just managed, but cured with a one-time treatment. Or where cancer cells are not bombarded with toxins, but are reprogrammed to self-destruct. This is not science fiction; it is the dawn of the gene editing era! Read more >>
CRISPR-Cas9 is now a familiar term, and this field is advancing at a remarkable pace. Originally an immune system in bacteria used to fight viruses, CRISPR-Cas9 has evolved into a sophisticated toolkit for rewriting human health [1,2]. Systems like CRISPR-Cas9 function as molecular scissors, guided to specific DNA sequences to cut, repair, or edit genes. However, recent high-profile studies and clinical approvals reveal that the real progress lies in refinement, making these "scissors" safer, smarter, and ready for the doctor's office.
To understand the field's trajectory, we can look to the work of pioneers like Dr. Wayne Ngo and Dr. Jennifer Doudna, who designed enveloped delivery vehicles (EDVs) to transport Cas9 ribonucleoproteins for therapeutic genome editing [3]. Further research highlights the pivotal shift from foundational discovery to applied therapeutic engineering. A compelling clinical example is a recent report by a team at Children's Hospital of Philadelphia, led by Drs. Rebecca Ahrens-Nicklas and Kiran Musunuru, which demonstrated the efficacy of a custom base editor, known as kayjayguran abengcemeran (k-abe), to correct a genetic "misspelling" in an infant's genome that caused CPS1 deficiency [4].
It is important to distinguish gene editing from newer techniques like base editing. As illustrated in this figure [5], CRISPR-Cas9 gene editing introduces double-stranded breaks in DNA break (a relatively "blunt", approach, which can lead to errors), whereas base editing and prime editing are far more precise. Think of base editing as a high-tech eraser and pen: it can directly convert one DNA base pair into another (e.g., A•T to C•G) without severing both DNA strands, thereby dramatically reducing unintended off-target effects. This precision is crucial for treating point mutations that cause disorders like sickle cell anemia and certain forms of thalassemia.
Despite this progress, significant challenges remain. For example, off-target effects occur when Cas9 acts on unintended genomic sites, causing modifications that may lead to adverse outcomes. Safely packaging and delivering CRISPR-Cas9 components to specific tissues in the body also presents a major engineering hurdle. Finally, the long-term impacts of these therapies are still largely unknown—could there be unforeseen effects decades later?
Researchers are actively tackling these challenges. One such scientist is Dr. Monica Sentmanat, the Research and Development Lead at the Genome Engineering & Stem Cell Center (GESC@MGI). In her talk at the 2025 Sequence to Function: Applications and Analysis for the Future (SFA2F) meeting, Monica highlighted her work investigating and resolving the on- and off-target effects of CRISPR-Cas9 therapies. She introduced two methods, Cas9-Seq and LOCK-Seq, to survey these effects during in vivo gene editing or the insertion ("knock-in") of large gene cassettes.
In her research, recombinant adeno-associated virus (rAAV) is used to deliver the CRISPR-Cas9 system into mouse embryos to knock in large gene cassettes. LOCK-seq, in particular, demonstrates significant advantages: it offers 10x higher capture efficiency, 20x greater read depth per sample, and superior multiplexing capability. In one example, LOCK-seq successfully identified imprecise knock-ins of an mCherry gene cassette. Furthermore, this rAAV delivery system showed efficient multiplexing, enabling the knock-in of at least three separate gene cassettes per vector.
Dr. Sentmanat's full SFA²F 2025 presentation may be found here, and you can read her recent preprint here.
Gene editing is rapidly transitioning from a revolutionary lab technique to a pillar of modern medicine. As diagnostic capabilities improve, we are moving toward a future where treatments address not just symptoms, but the root genetic cause. The era of precision genetic medicine is not just on the horizon, it is here, and its next chapter is being written in clinics and for patients today!
By Kayla Cross-Wiley and Kimberley S Ndlovu
We dedicate this day to our beloved canine family members. Whether you are a dog-parent, dog-sitter, or dog-enthusiast, we all gather to celebrate National Dog Day. As a pet-owner to two fur-babies, and their favorite sitter, Kim, we wanted to not only celebrate our furry companions, but also appreciate their contribution to scientific research.
Read more >>
I am the proud owner of MJ and Ella. MJ is quite skilled at swiping food off the counter, whether it’s freshly-baked cake, my Chipotle bowl, or a juicy hamburger. Ella is a professional shoe thief, and I’ve spent too much time searching for them. I named Ella after Legionella pneumophila (legionELLA), a microbe that I worked on for many days (and nights) during my undergrad studies
Kim and I had the pleasure of speaking with Dr. Vanessa Hale at The Ohio State University (OSU), who researches the canine urine microbiome, termed the "urobiome." The Hale Lab approaches the urobiome with a OneHealth perspective, where they recognize the interconnectedness of humans, animals, and environmental health.
For example, the dog is an excellent model to study bladder cancer. Did you know that the presentation of bladder cancer in humans and dogs is very similar? Bladder cancer is a disease linked to environmental exposures, and pets interact in the same environment that humans do, whether inside or outside the home. Dogs are exposed to many of the same exposures that we are, including, but not limited to, smoking, pollution, pesticides and drinking water quality. Given their naturally shorter lifespans, relative to humans, dogs provide a unique opportunity to study disease trajectories that would take decades to observe in humans.
Furthermore, dogs and their human companions share microbes, with some human-originating strains appearing in their dogs’ ‘biome.’ That’s right! The next time your dog gives you a surprise kiss on the face or gets all up into your personal space, remember all those microbes swapping back and forth in your microbiomes. This shared microbial landscape highlights how our pets are truly a part of our hearts and families, not just our homes.
Dr. Hale’s innovative research focuses on the microbes within the bladder, and how these microbes might mediate cancer development through their metabolic activity. The bladder was once thought to be sterile, however, advancements in NGS and improved sample collection revealed a healthy and stable urobiome.
We are still in the early stages of elucidating the role of the urobiome in health and disease, but with advancements in protocols and sequencing, and willing pet-parents, the potential for scientific advancements in the urobiome space is enormous. Even though urobiome research is still new, its future is exciting.
Dr. Hale is particularly excited about exploring the gut-bladder axis, and asks “can oral administration of microbe-based treatments influence the urobiome and therefore improve bladder cancer outcomes?” She is also excited about the potential of urobiome transplants (like fecal microbiota transplants, but for the bladder)--but emphasized the critical need for more research to better understand which strains are safe and effective.
One important takeaway that Dr. Hale wishes all pet parents knew was to be cautious about the overwhelming amount of information and packaging promoting pet probiotics. Although the marketing for pet probiotics is massive, more scientific validation is needed to know the effectiveness of these pet probiotics. She recommended seeking evidence-backed solutions and a trained vet professional on whether or not your pet requires these pet probiotics.
Working with and being a pet-parent to dogs has its unique rewards and challenges. Dr. Hale shared that her favorite part of her work is the deep connection between dogs and their owners. She loves hearing updates from pet parents on how their dog is doing post clinical trials. On the other hand, we know the trouble that our furry friends can get into.
I can’t count the number of times that MJ has been caught red-handed, stealing cake off the kitchen counter or my other dog, Ella, has been stealing my shoes to start her own collection on my bed. In one of Dr. Hale’s pet-parent stories, she shared that her dog, Kota, ate a corn cob out of the composting bin, which got stuck in her digestive tract. Dr. Hale immediately took her dog to the Ohio State Veterinary Medical Center where Kota had surgery to have the corn cob removed.
Thanks to Dr. Hale’s quick actions and the veterinary team at the Ohio State University, Kota is healthy and happy. Her dog’s legacy will live on in the Ohio State Vet School Curriculum, where her dog's abdominal radiograph presents a unique picture of “Corn Cob Gut Obstruction”. Look closely where the yellow circle is to see the corn cob. Note that Kota was adopted as an adult dog with an unknown history. The small bright white circles in this radiograph are bird shot. We didn’t know until we saw this radiograph that she’d previously been shot and she has no signs of being affected by the bird shot otherwise. Rest assured, she is a very happy, healthy, loved dog!
This National Dog Day, let’s celebrate not just our pets, but also the incredible scientific advances that they are contributing to. Happy Dog Day to all!
A special thank you to Dr. Vanessa Hale for giving us her time and her stories. You can connect with her here on LinkedIn or via email (hale.502@osu.edu). If you’re interested in learning more about her work, you can view her ongoing bladder cancer trial here, as well as her work with Dr. Jenessa Winston on two dog owners who love microbes!
Curious about how pet-parents are instructed to collect urine samples from their pets? Watch this video (skip to minute 3:20) and see how Dr. Vanessa Hale followed her dog, Kota, around to collect a urine sample. Ah, the things we do for our pets (and science).
By Kimberley S Ndlovu and Kayla Cross-Wiley
Kayla and I have been conference buddies for our past three conferences and this year we had the pleasure and privilege of attending the 19th Annual Sequence to Function: Applications and Analysis for the Future (SFA2F) meeting for the first time. It was an enriching experience and perfect opportunity for us as early-career researchers to present our work; mine on the vaginal microbiome of pregnant people living with HIV and Kayla on investigating host-microbe interactions using the cockroach gut. We encourage everyone in genomics, proteomics, metagenomics, and related fields to put SFA2F at the top of their conference schedule for 2026, so we wanted to share highlights from the 2025 meeting... Read more >>
At the apex of its spring season, the city of Santa Fe welcomed more than 200 attendees for SFA²F 2025. The conference, held annually at the unique La Fonda on the Plaza hotel in beautiful and vibrant Santa Fe, brings together scientists from various fields of study and different sectors for three days of intensive science presentations, panel discussions and networking events. This year, SFA²F celebrated its 19th year, showcasing a breadth of compelling and insightful presentations covering topics in genomics, oncology, biosurveillance, microbiome, new technologies, and more!
Day 1
Keynote speaker Dr. Richard Gibbs (Baylor College of Medicine) opened Day 1 by addressing the ongoing challenges of understanding common diseases, emphasizing how insights from rare Mendelian diseases and somatic mutations are ushering in a "golden era" for deciphering complex illnesses and somatic cell mosaicism.
Subsequent presentations explored a wide range of topics, including:
Using RNA-Seq to map network topology in Alzheimer's Disease (Dr. John J. Sninsky, Stanford University, Molecular Stethoscope)
Innovative AI frameworks for protein design (Dr. Arvind Ramanathan, Argonne National Laboratory)
High-risk mutation prediction (Dr. Adela Habib, LANL)
Advancements in nanopore sequencing with 2D materials for real-time protein analysis (Dr. Katie Young, GTRI)
High-throughput sequencing by expansion (SBX) technology (Dr. Yashashree S. Joshi, Roche )
Day 1 concluded with a lively poster session that provided a platform for trainees to share their research and interact with scientists of various career stages and domains. Posters that stood out to us were those of:
Dr. Juliette Lewis (CSU), who presented her work on generating the first reference-quality African #horseshoebat genomes, crucial for species identification and viral analyses
Austin Hendricks (UNM), who showcased her research on microbial symbioses in ptinid beetles, untangling beetle phylogeny and identifying fungal symbionts to open new avenues for pest control
Andre Watson (LANL), who has developed of a new version of the EDGE bioinformatics platform, significantly improving efficiency and maintainability for next-generation sequencing data analysis
Day 2
Day 2 began with Dr. Richard K. Wilson's compelling narrative about Nationwide Children's Hospital's pioneering use of genome sequencing to save children's lives, including an emotional case where rapid genomic analysis identified mutations enabling life-saving treatment of a 2-year old child with sudden cardiac arrest.
A diverse range of topics in genomic research were covered during the rest of the day. Presentations that stood out were:
Somatic mosaicism as a driver of neurological disease (Dr. Katherine Miller, NCH)
The evolution and regulation of complex genetic loci (Alasdair Taylor, Texas A&M University)
Optimized Nanopore sequencing workflows for rapid avian influenza surveillance (Dr. Iryna Goraichuk, USDA)
A global wastewater microbiome survey for tracking pathogens (Nelson Ruth, University of Oregon)
Real-time biosecurity threat detection software (Caitlin Jagla, Raytheon)
Bioinformatics pipelines for tick-borne virus analysis (Colin Price, MRIGlobal)
Efforts to characterize background bioaerosol content across the U.S. for pathogen identification (Dr. Kamil Khanipov, The University of Texas Medical Branch)
Studies on the genomic diversity and antimicrobial resistance of Klebsiella aerogenes (Leslie Huggins, UNM)
The transformative impact of targeted next-generation sequencing for detecting antimicrobial resistance genes, pathogen profiling, and identifying cancer-related methylation signatures (Dr. Alison Enk, Daicel Arbor Biosciences).
To celebrate the release of the initial draft of the first human genome a quarter century ago, this year’s meeting included a very special “fireside chat” with Dr. Richard Gibbs and Dr. Richard Wilson. During this session, these key contributors to the Human Genome Project (HGP) reflected on the profound influence of the HGP and the rapid advancements in sequencing technology. They also provided valuable insights on the importance of teamwork and how far technology has progressed since the project began.
Day 3
The third and final day of the conference included amazing talks on diverse microbiomes – from those in the human gut, cockroach, ocean, and vagina. The keynote address by Dr. Rob Knight (UC San Diego) explored how microbiome research has advanced from a data-limited to a data-enabled field and how we can continue to advance to a data-driven field.
Dr. Knight was followed by Viome Life Sciences' Dr. Momchilo (Momo) Vuyisich, sharing thoughts on data-driven, personalized diet recommendations using gut microbiome, with a special focus on the importance of the metatranscriptome in understanding how our microbiome affects our health. Conner Mertz from UNM rounded out the microbiome session by explaining how stable isotope tracing can be used to reveal synthesis of essential amino acids by gut microbes.
Many new tools were introduced in the subsequent bioinformatics session, including:
GenomeOcean, a genome foundation model trained on large scale metagenomic assemblies (Dr. Zhong Wang, DOE JGI & Lawrence Berkeley National Lab)
MetaAdapt, a metatranscriptomics analysis pipeline of microbes in extreme environments (Dr. Atul Chander, University of Mississippi).
The final session of the day featured talks on:
Characterization of the cis and trans interactions of the integrated Hepatitis B Virus with the human genome (Dr. Yih-Ping Su, Drexel University College of Medicine)
Phylogenomic analyses of non toxigenic Corynebacterium diphtheriae (Rachel Sidebottom, UNM)
Updates on metagenome assembly of long reads from complex microbiomes (Dr. Daniel Portik, PacBio).
The meeting concluded with a lovely networking lunch.
It is worth mentioning that SFA²F is not just about science! The highlight of the event--and I’m sure the attendees will agree--was the Donkey Happy Hour networking event where attendees had the opportunity to snuggle with Rosita the donkey, while enjoying pizza, drinks and the spring sunset!
This was truly a great experience for our professional development and we plan to be back in Santa Fe for the 20th Anniversary SFA²F meeting from May 19-21, 2026. So, join us for a 3-day long invigorating science discussion next year and perhaps you'll also get a chance to see Rosita!
As 2024 drew to a close, we celebrated another year of accomplishments in Genomics, NGS Applications, and Data Science with a summary of five must-read articles from this year. Read more >>
Machine-guided design of cell-type-targeting cis-regulatory elements
Gosai SJ, Castro RI, Fuentes N, et al. | Nature 634: 1211–1220
doi: 10.1038/s41586-024-08070-z
Researchers from Yale University, the Broad Institute of MIT and Harvard, and The Jackson Laboratory developed a method to effectively control gene expression by using AI-designed DNA switches, allowing them to "flip genes on and off" in specific cell types within an organism, potentially leading to new avenues for targeted gene therapies and medical research.
The technology utilizes an AI platform called "Computational Optimization of DNA Activity (CODA)" capable of designing synthetic DNA sequences that act as highly specific gene switches. These AI-designed switches can be used to activate genes in only certain targeted cell types. This could revolutionize gene therapy by allowing for more precise delivery of treatments, potentially targeting diseases like metabolic disorders without causing unwanted side effects in healthy cells.
Longitudinal multi-omics analysis of host microbiome architecture and immune responses during short-term spaceflight
Tierney BT, Kim J, Overbey EG, et al. | Nat Microbiol 9: 1661–1675
doi: 10.1038/s41564-024-01635-8
As the number of commercial space missions increase and space tourism grows, more people with diverse medical histories and microbiomes will be sent into space. It is critical to gain an understanding of the complex interactions between the space environment and the composition of the hosts' microbiota in order to be able to avoid putting lives at risk.
NASA is employing metagenomics to identify and quantify microbes on spacecraft surfaces and in cleanrooms. The goal is to assess potential risks associated with microbes that can survive in space, such as those that are resistant to radiation, desiccation, and ionization.
In this article, results are presented from the analysis of swabs collected from ten different areas on the body to study three types of microbial ecosystems (oral, nasal, skin) at eight different time points. Astronauts were found to experience changes in their microbiota and immune response during spaceflight. For example, the amount of skin bacteria associated with skin rashes or hypersensitivity increased. The oral microbiome displayed mission-dependent changes in metatranscriptomic expression of bacteria associated with tooth decay and biofilm formation. The number of gut bacteria associated with intestinal inflammation increased, while the number of bacteria with anti-inflammatory properties decreased.
These changes can be transient or more long-lasting and vary by astronaut, study, and body location. Astronaut microbiota, however, typically returns to its natural baseline when they return to Earth.
Additional experiments and missions are needed to further test the microbiome-derived theory of spaceflight-associated immune changes.
Brain aging patterns in a large and diverse cohort of 49,482 individuals
Yang Z, Wen J, Erus G, et al. | Nat Med 30: 3015–3026
doi: 10.1038/s41591-024-03144-x
It has long been known that aging causes changes in brain anatomy, some of which can be detected using magnetic resonance imaging (MRI). However, the patterns of systematic brain changes are invisible to the human eye and remain largely unexplored.
Studies using data from a restricted number of people have shown that machine learning methods can help to solve this problem by extracting subtle fingerprints of aging from MRI data. In this paper, the deep-learning algorithm, Surreal-GAN -- previously developed by the first author of the article -- was trained on MRI scans of the brains of 1,150 healthy people aged 20 to 49 and 8,992 older people, many of whom showed reduced cognitive abilities. The trained algorithm was subsequently applied to nearly 50,000 people participating in various studies of aging and neurological health.
This analysis identified five discrete patterns of brain atrophy, various combinations of which were associated with different types of age-related brain degeneration. For example, dementia and its precursor, mild cognitive impairment, were associated with three of the five patterns. Other patterns were associated with conditions such as Parkinson's disease and Alzheimer's disease, and one combination of three patterns was highly predictive of mortality.
The data analysis also showed that the identified models could potentially be used to estimate the likelihood of greater brain degeneration in the future. Five distinct patterns of brain atrophy associated with aging and neurodegenerative diseases were also linked to factors such as smoking and alcohol consumption, genetic factors, and blood markers that influence health and disease risk.
This new approach can significantly advance our understanding of the subtle mechanisms of age-related changes in brain anatomy and advance the possibility of their targeted control.
Nucleotide Transformer: building and evaluating robust foundation models for human genomics
Dalla-Torre H, Gonzalez L, Mendoza-Revilla J, et al. | Nat Methods 22: 287–297
doi: 10.1038/s41592-024-02523-z
The authors of the article developed the "Nucleotide Transformer", a series of the generalized AI models for genomics applications. Such models are pre-trained on DNA sequences using self-supervised learning, which then extracts context-sensitive representations of nucleotide sequences suitable for accurate predictions of molecular phenotypes from genotypes from publicly available genomic data.
The training was performed on 128 GPUs across 16 compute nodes. This process lasted 28 days, during which models with 2.5 billion parameters were generated. Following this, a robust benchmarking and statistical evaluation pipeline was created to evaluate the analysis design used for various tasks related to the identification of chromatin features, DNA regulatory elements, and splice sites in the human genome.
Benchmarking demonstrated Nucleotide Transformer models to perform better than self-supervised approaches such as Hyena-DNA and DNABERT-2. During the pre-training process, Nucleotide Transformer models acquired biological knowledge applying, for example, to regulatory elements and differentiation between coding and non-coding regions.
Results confirm the promise of the developed approach, despite a number of limitations related to the limited sizes of nucleotide sequences that they could process, how effectively they capture long-range interactions, and the lack of cell type-specific information.
It should be noted that Nucleotide Transformer models are currently used in agricultural applications for annotating genomes at single-nucleotide resolution and for solving specific problems on proteins from their corresponding coding sequences. The paper also provides other examples of the acquisition of knowledge about genomic elements by Nucleotide Transformer models.
Training and application of fundamental models in genomics provides a widely applicable approach for accurate prediction of molecular phenotype from DNA sequence.
Personalized pangenome references
Sirén J, Eskandar P, Ungaro MT, et al. | Nat Methods 21: 2017–2023
doi: 10.1038/s41592-024-02407-2
A pangenome is the complete set of common and unique genomes present in a given species. It combines the genetic information of all sampled genomes, resulting in a large and diverse range of genetic material. The pangenome concept--first proposed in 2005 for bacterial studies--is applicable to many other species, including humans.
Pangenomes reduce reference bias by representing genetic diversity better than a single reference sequence. However, when comparing a new sample to a pangenome, misleading variants may arise, causing, for example, false read mappings. Such irrelevant variants are usually rare and were previously addressed by filtering them out. The same time, this approach did not remove all irrelevant variants and lost some relevant ones.
The authors developed a k-mer-based method for sampling local haplotypes similar to the sequenced genome. Using a subgraph based on sampled haplotypes as a reference for read mapping yields more accurate results than the full graph or a frequency-filtered graph containing only common variants.
The approach is implemented in the vg toolkit (https://github.com/vgteam/vg) for the Giraffe short read aligner and its accuracy was assessed while compared with modern methods using human pangenome graphs from the Human Pangenome Reference Consortium.
The authors showed that the proposed approach reduces small variant genotyping errors by four times compared to a set of genome analysis tools, making genotyping of structural variants of short reads competitive with long read variant detection methods. This represents an improvement over previous filter-based methods, but there are some limitations to overcome.
The mysterious and marvelous microbiome is one of a wide range of topics covered at the Sequencing to Function: Applications & Analysis for the Future (SFA2F) meeting every year. We asked Dr Momo Vuyisich, Co-founder, Chief Science Officer, and Head of Clinical Research at Viome Life Sciences and long-standing member of the SFA2F community to share a few thoughts on the state of the art and his research. Read more >>
Where does your passion for microbiome science come from?
"The microbiome is an integral part of our body, and microbial biochemical functions are essential determinants of our health. These microbial functions are heavily controlled by the foods we eat. Therefore, we can start to control our health with personalized nutritional recommendations based on quantifying each person’s biology, which should include both human and microbial components."
"Metabolomics and metatranscriptomics are the most useful technologies for studying the microbiome, because they identify the microbial functions that determine our health and disease status. DNA-based microbiome analysis methods have a fundamental limitation in that they cannot quantify the active microbial functions (biochemical activities)."
"The fact that it’s been co-evolving with us for millions of years, contributing to our mental, physical, immune, and cognitive health, yet we only started understanding it about 15 years ago."
"Developing a practical technology (universal metatranscriptomic analysis of any clinical sample) for digitizing the human body by quantifying molecules (human and microbial transcripts) that (i) are very relevant for our health, and (ii) can be modulated with diet and lifestyle. This technology is, for the first time, enabling the systems biology approach to controlling our health."
"Every person now has an opportunity to increase their health span by taking care of their microbiome. Healthy microbiome functions (e.g. butyrate production) make us healthy, and unhealthy microbiome functions (e.g. LPS or TMA production) can make us chronically sick. Ensuring you have a diverse microbiome with many functions, then supporting the healthy ones with personalized diet is critical for healthy aging."
"It is already affordable for many people. My company, Viome, is working very hard to make the tests free to the consumer. Once we show that data-driven personalized nutrition is truly a preventive medicine, the payers will offer Viome tests for free to their users/members."
"The microbiome field has become too big to recommend just a few resources. There are >340,000 publications in NCBI. I would recommend that each person simply google “microbiome” and whatever aspect they are interested in. Some examples are “microbiome blood pressure”, “microbiome metabolites heart disease”, “microbiome depression”, “microbiome neurotransmitters”, etc. Google will find both consumerized and scientific sources. All consumerized articles should be cross-checked with peer-reviewed literature."