Phytoplankton, tiny plant-like organisms in the ocean, are incredibly important for life on Earth. They’re a major food source for many sea creatures and produce almost half the oxygen we breathe. They also help control the climate by soaking up a lot of carbon dioxide, a gas that contributes to global warming.

Scientists want to learn more about how these phytoplankton use sunlight to make energy and oxygen, which can be useful in the context of environmental monitoring during global climate change. However, it’s tricky to study this because the usual methods only give an overall average for a large group of phytoplankton, hiding the differences between individual cells, or they only give limited measurements of individual phytoplankton.

Now, researchers at The Hebrew University of Jerusalem in Israel have come up with a new way to study these organisms. They’ve built a system that can measure the light given off by individual phytoplankton cells, which tells them how efficiently each individual is using light.

This new technique will help scientists better understand how different types of phytoplankton react to changes in their environment. The work was presented at the 69th Biophysical Society Annual Meeting, held February 15–19, 2025 in Los Angeles.

“I look at how individual plankton react to changing conditions by looking at the light that they dispose of—or in scientific terms, I look at fluorescence lifetimes. Basically, it’s how the phytoplankton convert light to energy they can use later,” said Paul Harris, who led the study.

This new system uses a special microscope to get a close look at individual phytoplankton cells which are sent down tiny channels. It measures the different colors of light the cells give off, which tells scientists a lot about how they’re using light to make energy.

So far, Harris and colleagues have used the system to study three different types of phytoplankton, looking at how they change throughout the day and how they react to brighter light. What they found is that each type of phytoplankton has its own unique way of adjusting to changes in light, kind of like how some people put on sunglasses when it’s sunny, while others might opt for a hat. Each species has its own way of dealing with light, and uses different strategies for surviving sudden changes in their conditions.

“We need to understand how these phytoplankton respond in order to predict and observe what’s happening in the oceans, especially with regard to climate change as oceans warm,” said Harris. “We hope to give some insight into how species are going to change,” he said.

The system could also help in predicting harmful algal blooms, which can spell disaster for fish and other species in the ecosystem and even poison humans if consumed. “We could use this tool to give advanced warning of algal blooms,” he pointed out.

The ability to differentiate species and determine how they are using light and energy offers a powerful tool for assessing the health and productivity of phytoplankton populations, which are essential for marine food webs and global carbon cycling.

Chinese scientists identify food ingredient they say could reverse some autism symptoms

Scientists have identified a probiotic in dairy fermentation that may help alleviate and reverse some autism symptoms.

Currently patients can only use antipsychotics, antidepressants, stimulants and anti-anxiety medications for treatments, but the new study suggests a natural method could just as effective.

The discovery was made using genetically modified mice that were prone to autism-like symptoms.

When modified, the mice exhibited symptoms of the disorder like a reduced interest in social interactions and an imbalance in the key neurotransmitters crucial for functions like learning, memory and cognitive processes.

Researchers gave the animals a daily dose of the probiotic Lactobacillus murinus (a type of bacteria commonly found in dairy products like cheese and yogurt) for one month.

Following the experiment, the mice’s brains became more flexible and adaptable, which helped them learn and remember things better.

The treatment also led to recovery of the intestines and other genes that are impacted by the disorder.

Around one in 36 children has been diagnosed with autism spectrum disorder (ASD) in the US, according to the CDC’s most recent estimate.

What causes the disorder is unknown. Research has pointed to growing pollution and chemical contamination in food and water may allow toxins to infiltrate the bloodstream of pregnant mothers and travel to the brain of the developing fetus, causing inflammation that impairs nerve signals that lead to autism.

Most children cannot be diagnosed until at least four years old and the current treatments include behavioral and speech therapies.

But the Chinese scientists may have uncovered a treatment in common foods that could complement behavioral interventions with minimal side effects, they said.

The study used 34 mice that had 13 pairs from their CHD8 gene removed, which disrupted protein production, South China Morning Post reported.

The mice then began to show autism-like behaviors, including anxiety, problems with socializing and memory.

Other research has found that gut bacteria can impact brain functions through the gut-brain axis, the complex network of communication between the gastrointestinal tract and the central nervous system.

That led the team to test if Lactobacillus murinus could be a treatment for autism.

The probiotic is known for its anti‐inflammatory and antibacterial actions. It has also been found to ease depression.

The mice were given Lactobacillus murinus over the course of 30 days, allowing researchers to monitor their behavior and internal progress.

Not only did their social behaviors reverse, but the intestine started to return to normal.

A 2022 study conducted at China’s Zhujiang Hospital found underdeveloped intestines could be a significant factor in driving autism-related gut microbiota and behavioral issues.

The new research also found the excitatory/inhibitory (E/I) balance in the brain’s cortex showed a trend toward recovery.

This balance of neural activity in the brain involves excitation and inhibition and is a key factor in how the brain processes information and generates oscillatory activity.

Individuals with autism often have lower levels of dopamine D2 receptors, a protein that helps regulate movement, learning, memory, reward and attention.

But following the probiotic treatment, researchers observed a burst in levels that suggested the cells were recovering.

Several brain pathways related to behavior, synapse organization and inflammation also improved.

The team looked a genes that were rescued by the treatment, finding a large amount occurred in astrocytes that regulate things like neurotransmitters and metabolism.

While excitatory neurons held the largest proportion of recovery. These are involved in the transmission of signals in the brain.

The researchers said the findings show that Lactobacillus murinus ‘[improves ASD-related social behavior deficits through bacteria-gut-brain communication.’

‘Our findings align with and extend previous research, which has shown that Lactobacillus murinu rescues behavioral deficits.’

AI-Enabled Gene Editing Produces Fewer Off-Target Outcomes

Artificial intelligence (AI) is known for enabling deeper insights into drug development, identifying patterns and molecules that may otherwise go unnoticed. Now it is poised to make similar contributions to gene editing. A few companies are using AI to develop gene editing tools that are more specific and more efficacious.

CRISPR systems such as CRISPR-Cas9 revolutionized gene editing, but genomic rearrangements are becoming a real concern for in vivo therapies, and nonspecific editing has been a longstanding issue that affects subsequent generations of cells. Zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) also have challenges, thus underscoring the need for improvements.

Using rational design to find new gene editors, however, hasn’t yielded anything notably different from CRISPR-Cas9, says Chelsea Trengrove, PhD, CEO of Neoclease, and a platform approach to their development tends to limit efficacy and specificity.

AI technology is emerging as a possible solution to enhance the precision of multiple types of gene editors. And the addition of generative AI lets scientists look beyond what exists in nature.

GenAI-created editors

Neoclease’s custom AI model develops gene-specific editors in silico. Eventually, top candidates may direct gene editing for humans in vivo, using the CRISPR nucleases, ZFNs, TALENs, and other gene editing nucleases.

“We’re using a generative AI model,” Trengrove says. “This is a large language model that’s trained on millions of known proteins that cut DNA.” The idea, she adds, isn’t to create a workhorse enzyme that can do everything, but to optimize every editor for a specific gene of interest.

Trengrove explains that generative AI enables Neoclease to create a knowledge network of variables to understand how editors can be optimized, and to make a virtue of hallucination such that truly novel sequences can be generated. The goal, she stresses, is to generate additional editors that are “optimized and weighted in the direction we want to push them toward.”

“It’s almost like ChatGPT for proteins,” Trengrove remarks. “While some associate hallucinations with errors, we leverage them… as an innovation tool to generate novel and effective protein designs.”

Generating potential gene editors is just the first step. After tens of thousands of novel sequences have been generated that can be optimized toward specific features—certain binding energies, degrees of polarity, or domains, for example—the features are fed through a series of computational checkpoints. Those checkpoints identify which editors are best suited to advance into in vitro validation based upon their features and functionality. Of the tens of thousands of nucleases the company has created in silico, it has, to date, advanced about 7,000.

Some of these editors are about half the size of the CRISPR- Cas9 system, Trengrove notes. They include the miniaturized nucleases developed by Jin Liu, PhD, chief technology officer and co-founder of Neoclease and tenured professor of pharmaceutical sciences in Texas. According to Trengrove, Liu “has shown that some of her miniaturized editors have comparable cleavage, energy, and efficacy in vitro, and have reduced off-target effects by sixfold.”

These small editors can be packaged into adeno-associated vectors or similar vehicles to deliver them to tissues throughout the body. “We’re actually looking at targeting the brain for Parkinson’s disease,” Trengrove says.

Currently, most of the testing has been done in silico, with only limited in vitro validation. Neoclease plans to take these editors into mouse and zebrafish models in mid-2025, and that the company anticipates Investigational New Drug–enabling studies will begin in 2026. Trengove adds that the company is also “working on a small deal with a pharma company to evaluate thousands of nucleases in vitro.”

AI CRISPR-like technology

In 2017, Wayne Danter, MD, CEO of 123Genetix, pioneered the development of artificial human stem cells and organoids for medical research. This work led to the development of aiHumanoid simulations for virtual drug trials. “To produce a specific type of cell, I had to… alter the cell’s genetic makeup,” Danter says. “I did that by creating a symbolic representation of a gene and then adding to it or deleting it.” The AI system he developed to do that, DeepNEU, simulates the CRISPR-Cas9 enzyme.

DeepNEU is built around an intelligent database. It functions like a text editor for genes to enable rapid prototyping and quality checks. It is fully developed and is already in use as a complement to CRISPR-Cas9 gene editing.

The advantage of AI-enabled gene editing is specificity. Off-target effects are avoided so that when the virtual results are compared to those of CRISPR-Cas9 experiments, any differences can be identified and, perhaps, minimized or eliminated.

Rather than train algorithms on data and outcomes, DeepNEU makes use of a healthcare-oriented Wise Learning process. As Danter indicated in a recent bioRxiv preprint (DOI: 10.1101/2022.06.18.496679), Wise Learning “combines fuzzy cognitive map simulations, with data from multiple experts and a generic decision-making system.” He added that the Wise Learning process “should also explore available learning algorithms including deep learning methods when available.” Essentially, Wise Learning uses an unsupervised (untrained) approach based on experiences. According to Danter, AI technology that incorporates Wise Learning can emulate human thought more closely.

DeepNEU applications yield “a very large matrix of relationships and weights,” Danter says. “The basic information includes gene–gene and gene–protein relationships.” 123Genetix’s gene relationship network has approximately 65 million neurons.

Danter’s passion is to find effective treatments for rare diseases, and he is proud that DeepNEU has been used for multiple studies of rare diseases. He indicates that access to DeepNEU has been free for rare disease organizations, and that he is currently “bringing on board a number of pharma partners interested in using the technology.”

This fall, 123Genetix plans to release a version of the aiHumanoid that includes Serious Second Look. This addition enables the AI system to pause to consider whether it accurately answered the question before presenting results. If the results fall short, the AI reoptimizes on subsequent attempts.

Danter is also validating an AI system that is designed to use simulated sentience to make ethical decisions, specifically, decisions in line with the “first do no harm” principle of the Hippocratic Oath. Danter notes that the system is not self-aware.

Zinc finger improvements

Marcus Noyes, PhD, co-founder of newly formed TBG Tx and assistant professor of biochemistry and molecular pharmacology at New York University Langone Health, is developing an AI-enabled gene editor for ZFNs with his collaborator and co-founder, Philip Kim, PhD, professor of molecular genetics and professor of computer science at the University of Toronto. This gene editor, ZFDesign, is ready for commercial use.

Since publishing ZFDesign in 2023, Noyes and his team have increased the editor’s precision. “The first version of the model was trained to understand how to design an array of ZFs, but it didn’t really know which of the thousands of designs returned for each target would be the most specific,” Noyes says. “We needed to teach the model which target sequences and which proteins will provide the most precise activity genome wide.”

The latest iteration of ZFDesign incorporates several improvements. “We added more interface data to increase our understanding of compatibility,” Noyes details. “We also screened the specificity of hundreds of ZFNs to train the model.” As a result of this work, ZFDesign can identify the most precise options and thus reduce off-targeting. “We’ve also modified the model to express all the ZFNs in the array continuously, rather than skipping bases between pairs,” Noyes adds. “This reduces the modularity in the design.” He says he expects to publish the updated model in 2025.

The most notable aspect of ZFDesign, Noyes says, is the gene editor’s ability to understand whether trends regarding modifications to a ZFN could be generalized to subsequent designs: “In the past, you could ask questions about how modifications of a designed ZFN array might change its on- or off-target activity, but it was never really clear if the trends were generalizable or were specific to just that protein, because you would need to design, validate, and test several arrays. By contrast, ZFDesign allows the simple design of any number of proteins for any number of target sequences, making the confirmation of generalizability a trivial process.”

How well this model works depends on function and precision requirements. Regarding activation and repression—the areas for which he has the most data.” Noyes says, “In general, about 80% of the designs will produce a change in target gene expression.”

About 30% of the designs have more than fivefold activation and more than 70% repression when assayed by transient transfection. Precision for highly functional designs appears high. However, Noyes cautions, “We have only tested off-target activity for around 20 constructs designed with the new model.” About half have shown minimal to no off-target activity without optimization. And according to Noyes, even better results are obtained with optimization: “Typically, we can develop a candidate for any target gene with single-target resolution. … If we design 10, we expect about 8 will do something, 3 will be really good, and those 3 should have limited off-target activity.”

ZFDesign is being used in the research community now. “One scientist tried three activators in cardiomyocytes, and two worked very well,” Noyes reports. “Another group created a nearly complete set of precise probes that bind each of the human centromeres, allowing them to be labeled in live cells. Yet another group found four potent repressors in neurons from a screen of 12 candidates.

“We are finding that the amount of off-target activity is often tied to the mechanism. For example, activation, repression, labeling, and cutting all seem to have different optimal affinity regimes. Moving forward, we hope our model will be precise enough that users will only need to test a few designs, and that any optimization will be a straightforward affinity adjustment to match the mechanism.”

Additional tools

Several other companies are creating tools that support the use of AI for gene editing. In October, Shape Therapeutics published two preprints. One detailed how it engineered guide RNA to fit into adeno-associated viruses. The other discussed how the Sharpes’s system, which is based on the company’s DeepREAD technology, allows therapeutic guide RNA to be expressed within cells.

Last spring, Profluent announced that its open source, AI-based gene editor, OpenCRISPR-1, successfully edited the human genome. The company reported the gene editor generates “millions of diverse CRISPR-like proteins that do not occur in nature.”

AI tools for gene editing are helping scientists enact more precise edits, which lowers off-target effects for multiple gene editing technologies. Ultimately, this may help make gene editing more accessible.

History May Not Be Repeating but It Is Certainly Rhyming

Trofim Lysenko, the mid-20th century anti-Mendel scientist and Stalin favorite, is poised for a comeback. Millions of Soviet citizens died as a result of his government-backed pseudoscientific biological ideas. Even though he was eventually dismissed in shame, Russian science tanked for several generations, struggling to catch up with the rest of the world to the present day.

How did so many people, including so many scientists, become so willing to abandon the scientific method and support Lysenko, helping pave the way for catastrophe after catastrophe? Clearly many were scared into accepting his ideas, aligned as they were with Soviet philosophy and policies (and the government’s ruthless enforcement of them). But that still doesn’t fully account for his rapid rise and—coming way too late for many—his dramatic fall.

My recent interest is more than just historical curiosity. When one looks at the incoming administration’s nominees for Health and Human Services, NIH, FDA, NASA, CDC, etc., one could be forgiven for fearing that ideology is again ascendant over scientific reality: The very dynamic that led to Lysenkoism less than a century ago. These possible “science” leaders are indeed a threat, particularly if they get a high level of popular support for “theories” they have publicly announced that defy years of scientific discovery. What is going on in the American public?

What is wrong with us?

It is too easy to place the blame on a poor education system for the deep suspicion of science and scientists in many parts of our country. But asking “what is wrong with them?” should be secondary to the far more important question of “what is wrong with us?” Scientists and their supporters may be about to reap a whirlwind that we have at least partially sown.

It is a tragedy that so many people I talk with have experienced science in their school education as only a set of facts to be memorized and spit back out at test time. I think we can all agree that is not good science, or even science at all. No, the best science is built on the twin pillars of scientific method and scientific language, both of which have tremendous power to enlighten reality and unite or—sadly—to obscure and fragment.

What we say and do matters

Scientific method at its best is an intellectual openness to considering, debating, and testing new findings that enhance or even contradict current understandings. That philosophy is obviously inimical to more dogmatic belief systems, whose proponents simply dismiss scientists and scientific discoveries (e.g., vaccines) as incompatible with deeply held “truths.” The growing out-of-hand dismissal of science at least partially reflects a failure by us scientists to impart in both our communications and our actions just how freeing and even existentially satisfying the scientific method is.

The other pillar, scientific language, evolved out of necessity for scientists to communicate new concepts and discoveries accurately with each other as succinctly as possible. But for the non-scientist (or even the scientist in a different field of expertise), scientific language might as well be Sumerian when used outside the relevant laboratory. Modern day scientists tend to be particularly guilty of relying on professional language even in settings in which they may be the only one fluent, basically rendering concepts and ideas inaccessible to others, and thus more easily dismissed as “just another belief system.”

As a result of our unintentional obscuring of science, all manner of crazy ideas and charlatans can gain political and social ascendancy thanks to the relative accessibility of their mendacious language that offers “insider” knowledge and comfort. Lysenko’s twisting of method and language gave him (initially) great power over the Soviet population and even over many scientists. Those few scientists and science-supporters who opposed him did so initially only from inside the scientific ivory tower, and they were quickly and easily silenced. Others simply fled or hid, hoping he wouldn’t last long, which ended up only prolonging his tenure.

All of us must step up

Are we on the verge of a new American version of Lysenkoism? Maybe not, but there is no question we are facing at the very least a rough few years ahead for science. The temptation is strong to lay low until this madness is over. But that is what most scientists did in the face of Lysenkoism, to their own detriment. If we truly believe science is a critical element of a healthy and just society, scientists and their supporters cannot hide now.

The December 9th letter to the U.S. Senate from 75+ Nobel Laureates requesting that the senators turn down the nomination Robert F. Kennedy, Jr., to head the Department of Health and Human Services was a first step in the right direction. But all of us must push back, individually and collectively, on the emerging anti-scientific forces. Each of us needs to engage a much wider audience—using more accessible language—than our usual comfortable setting. In short, we must model a fearless commitment to good science and undertake a clear unmasking of bad science, no matter the political winds.

An alternate version of this article first appeared on December 11, 2024, in the Boulder Daily Camera under the title “This is no time for scientists and science-supporters to hide.”

AcrVIB1: The unexpected anti-CRISPR protein that tightens RNA binding

The CRISPR-Cas gene scissors offer a wide range of potential applications, from the treatment of genetic diseases to antiviral therapies and diagnostics. However, to safely harness their powers, scientists are searching for mechanisms that can regulate or inhibit the systems’ activity. Enter the anti-CRISPR protein AcrVIB1, a promising inhibitor whose exact function has remained a mystery—until now.

A research team from the Helmholtz Institute for RNA-based Infection Research (HIRI) in Würzburg, in collaboration with the Helmholtz Centre for Infection Research (HZI) in Braunschweig, has uncovered the precise way AcrVIB1 works that expands the known means by which Acrs can shut down CRISPR. The results are published in the journal Molecular Cell.

Bacteria and their viruses, known as phages, are locked in an age-old arms race. To defend against phage attacks, bacteria have evolved sophisticated mechanisms to recognize and counteract invading viruses. In turn, phages have developed innovative strategies to evade these defenses. A prime example of this ongoing battle is the CRISPR-Cas defense system in bacteria, countered by anti-CRISPR proteins (Acrs) in phages, which specifically block these bacterial “gene scissors.”

Apart from their counter-defensive function, anti-CRISPR proteins hold great promise for enabling more precise control over CRISPR technologies. The research team has now further elucidated the function of an important yet so far uncharacterized anti-CRISPR protein.

“In a previous study, we used a deep learning algorithm to predict new Acrs. This led to the identification of AcrVIB1, the first anti-CRISPR protein targeting the Cas13b nuclease,” says HIRI department head Prof. Chase Beisel, who led the study together with the department of Prof. Wulf Blankenfeldt at HZI.

“The nuclease Cas13b can recognize and cut RNA. It is currently used to silence genes, whether to study their function, clear viruses, or counteract genetic diseases linked to the gene.” However, how the protein AcrVIB1 inhibits Cas13b remained unknown until now. In their study, the research team presents this entirely new blocking mechanism.

An RNA dead end

The Cas13b nuclease operates by interacting with a CRISPR ribonucleic acid (crRNA), which serves as a guide to identify and bind to complementary RNA sequences, for example those from phages. Once the target RNA is bound, Cas13b can cleave and degrade not only these complementary RNA molecules but also all other RNAs in the vicinity.

While most known anti-CRISPR proteins block steps along this path such as crRNA binding or target recognition, AcrVIB1 adopts a radically different strategy: Rather than blocking the binding of the crRNA to Cas13b, AcrVIB1 even improves it. The formed pair is dysfunctional though, meaning that the enzyme cannot begin degrading RNAs even when its target is present. Furthermore, the bound crRNA becomes vulnerable to attack by cellular ribonucleases, which break down RNA molecules.

“The tighter binding between nuclease and guide RNA was entirely unexpected. The simpler and therefore initially expected mechanism would have been to just prevent the guide RNA from binding in the first place,” says first author Dr. Katharina Wandera, who completed her doctorate in Chase Beisel’s laboratory.

“Nevertheless, the path taken by AcrVIB1 appears to be more effective: AcrVIB1 binds tightly to and thereby renders Cas13b inactive. At the same time, it increases the turnover of guide RNAs, making Cas13b a dead end for crRNAs.”

Chase Beisel’s team at HIRI and the laboratory of Wulf Blankenfeldt at HZI have joined forces to decipher the structure of the inhibition mechanism more precisely. Using cryo-electron microscopy, Blankenfeldt’s group showed that AcrVIB1 binds to Cas13b, leaving the crRNA-binding domain untethered.

“Our finding provides a blueprint for the development of molecules that could mimic or modify the function of the anti-CRISPR protein,” says Blankenfeldt. These are the first data from the HZI’s new cryo-electron microscopy facility to be published.

Diet drug boom weighs heavily on state budgets

Colorado’s spending on highly effective but costly weight-loss drugs for state workers more than quadrupled from 2023 to 2024 — and costs have been doubling every six months.

Now, the state wants to scrap the benefit, arguing that it’s financially unsustainable.

But the potential removal of the popular benefit has sparked blowback among state workers. The state employees’ union argues ending coverage will cost the health plan in other ways — increasing spending on obesity-related diseases and leading to a less healthy workforce.

“[State employees] are very upset about this,” said Hilary Glasgow, executive director of the union, Colorado WINS. “Long-term obesity drives a lot of the major fatal diseases in America, and the employees I’ve talked to feel like they’re losing a lifeline that got thrown to them.”

The Colorado case underscores the broader struggle of states — many of which are facing budget shortfalls this year — that cover weight-loss drugs like Ozempic and Wegovy for state workers. Over the past three years, a growing number of states have taken leaps of faith in covering the expensive class of drugs, hoping that the benefit would lead to healthier workforces that would be less expensive to insure.

But with the number of prescriptions skyrocketing past projections — more than 1 in 10 Americans reported taking a GLP-1 drug, according to a May 2024 KFF Health Tracking poll — some states are limiting or ending the benefit.

At least two others — North Carolina and West Virginia — have already eliminated coverage due to cost concerns. That means state employees seeking the drugs for weight loss in those states will have to pay up to $1,500 a month for the treatments.

The issue is forcing states into difficult deliberations: Keep covering the drugs and risk depleting their budgets — and potentially increase premiums for everyone on their plans — or eliminate a benefit that many employees now rely on.

The dilemma also comes as states face their toughest budgetary pressures in years, in large part because federal cash they received during the pandemic has disappeared. And the Trump administration’s aggressive efforts to slash federal funding threatens to create further fiscal duress for states.

In a Fox News interview that aired Sunday, President Donald Trump lambasted the high price of the drugs.

“In London you get it for $88. In New York you get it for $1,200,” he said. “It’s very unfair.”

And if Trump follows through on his threat to impose tariffs on Denmark, the move could further drive up the costs of Ozempic and Wegovy, which are manufactured by Danish drugmaker Novo Nordisk — and create even greater fiscal challenges for state policymakers.

“The problem is the near-term cost is so high, and the benefits that you would gain are over a longer-term period,” said Charles Sallee, director of the New Mexico Legislative Finance Committee, which is exploring options to reduce the costs of covering the drugs for state employees. “But is that person still going to be in your health plan five years from now?”

Balancing costs and benefits

Because states can choose whether to cover GLP-1 drugs for weight loss, there is a patchwork of coverage for state employees across the country. At least 17 state employee health plans — an almost even mix of red and blue states — cover the drugs for weight loss, according to the Leverage Obesity Coverage Nexus.

The potential market for the drugs is massive — more than 40 percent of adults in the U.S. are obese — and has seen explosive growth. The number of Americans taking GLP-1s for weight loss rose more than 700 percent between 2019 and 2023, according to a recent study.

North Carolina opted to end coverage of the drugs for weight loss last year after estimating that the costs over the next six years could reach more than $1 billion. And West Virginia last year abruptly ended a subsidy program that helped state employees pay for the weight-loss drugs, saying the treatments were too expensive. Both states still cover the drugs for other conditions, like diabetes and cardiovascular disease.

Delaware started covering GLP-1s for weight loss for state employees in 2023, expecting to spend $1.8 million that year. The state ended up spending $14.2 million. For 2025, the plan expects to spend $52.8 million on weight-loss drugs.

The plan raised premiums 27 percent for fiscal year 2025 to address rising costs of prescription drugs and increased health care expenses overall. The state committee that manages the health plan has had discussions about limiting, or potentially ending, coverage. But adding restrictions raises another risk: Drugmakers might threaten to pull back tens of millions of dollars in rebates that plans receive for offering coverage.

“Once you’ve done it, it is difficult to take it away,” said Shaun O’Brien, a member of the Delaware State Employee Benefits Committee and the policy director of the American Federation of State, County, and Municipal Employees.

The health plan for New Mexico’s nearly 60,000 state and local government workers is projecting a $85 million budget shortfall in fiscal year 2025, according to a presentation from the state Health Care Authority obtained by POLITICO. The agency, which runs the plan, estimates GLP-1 costs for weight loss alone may exceed $20 million in 2025.

The Health Care Authority is projecting a $87 million budget shortfall and a more than 10 percent increase in drug spending in fiscal year 2026, largely driven by GLP-1 costs, according to the presentation. That means the plan would need a 29 percent premium increase in 2026 to cover the rising costs of medical and prescription drug claims.

New Mexico is struggling to contain the costs without ending coverage, said Sallee, the director of the Legislative Finance Committee. The panel is working with the Health Care Authority on options — including more restrictions on who can access the drugs, like a higher BMI threshold.

“Taking away what is widely seen as a highly effective drug at helping people lead healthier lives purely based on cost of a drug could be challenging,” said Sallee. “We should be working on getting better prices for the drugs.”

The committee has also floated purchasing compounded versions of the drugs, which can be cheaper than brand-name versions sold by drugmakers like Novo Nordisk and Eli Lilly. Compounded drugs contain the same basic ingredients as the brand-name versions, but the FDA doesn’t review them for safety and efficacy, posing risks for states looking to purchase them directly.

Colorado’s state employee union is trying to get Democratic Gov. Jared Polis to keep coverage of the drugs, which his administration proposed scrapping in its latest budget request. The state is facing a $1 billion budget shortfall, and the threat of suspension of some federal funding is compounding the financial challenges. Colorado WINS and the governor’s office are weighing how to continue covering the drugs against how much premiums might increase for everyone on the plan due to rising costs.

“To eliminate GLP-1s is short-sighted because of the cost of life-saving drugs later on for things like heart disease, cancer — the myriad of diseases that are driven by obesity or have obesity as a factor,” said Glasgow, the union executive director.

“Going in the wrong direction”

Connecticut’s state employee health plan last year took an experimental approach to coverage — requiring employees to enroll in a lifestyle management program called Flyte before receiving the medications. The program offers tools for weight management and personalized care plans. The state hoped adding the requirement would reverse the drugs’ cost trend, which was projected to reach $30 million in 2023.

The program showed early success in reducing the number of GLP-1 prescriptions, cutting the cost trend in half. The experiment also offered a potential road map for other organizations working through how to afford the drugs.

And clinically, the program has been a success: Enrollees have had a 16 percent drop in weight and BMI and a 14 percent drop in blood pressure. The outcomes have led to a happier and more productive workforce, said Connecticut State Comptroller Sean Scanlon.

But 19 months in, more and more state employees are enrolling in the program. Between October and January, more than 3,500 state employees joined Flyte, bringing the total to nearly 12,000 members. And while the program initially slashed the state’s $30 million cost for the drugs in half, those costs are creeping back up. The state now projects it will spend $23 million on the treatments in fiscal year 2025.

“Our cost trend is kind of now going back in the wrong direction,” said Scanlon. “But at the end of the day, I still believe that this is the right policy.”

Could yogurt help protect against colorectal cancer?

Colorectal cancer is the third most common cancer worldwide, with the World Health Organization (WHO) reportingTrusted Source that it makes up more than 10% of all cancer diagnoses.

Lifestyle factors, such as being sedentary, smoking, obesity and excessive alcohol consumption, as well as high intake of processed meats and low intake of fruits and vegetables, can all increase a person’s risk of developing colorectal cancer.

Eating a healthy, high-fiber diet, with plenty of fresh fruits and vegetables, whole grains, calcium and dairy products is associated with a lower colorectal cancer risk.

A study led by researchers from Mass General Brigham — and published in the journal Gut Microbes — has now found that yogurt could also decrease the risk of some types of colorectal cancer.

The researchers found that people who ate 2 or more servings of yogurt containing live bacteria each week had a 20% lower risk of developing colorectal tumors that were positive for Bifidobacterium, a strain of bacteria that is common in the gut microbiome.

Gemma Balmer-Kemp, PhD, Head of Research at Cancer Research UK’s Cancer Grand Challenges, one of the funders of the study, told Medical News Today that:

“Endogenous bacterial species are of significant interest for their application in human health. This study provides new evidence about the potential benefit of yogurt (which contains live bacteria) in reducing risk of a certain subtype of colorectal cancer.”

“While this study has shown a correlation between long-term consumption of yogurt and lower rates of proximal colorectal cancer positive for Bifidobacterium, more work is required to understand any causative role of Bifidobacterium and the mechanisms involved if so,” she added.

Weight-loss, diabetes drugs linked to vision problems in small study

Popular drugs for diabetes and weight loss could have an unexpected side effect.

Glucagon-like peptide 1 (GLP-1) receptor agonists, which are used to treat type 2 diabetes and/or obesity, were linked to vision problems in a small study at the University of British Columbia.

Some common GLP-1 drugs include Ozempic and Wegovy, which contain semaglutide as the active ingredient, and Mounjaro and Zepbound, which contain tirzepatide.

In the study, nine patients who were using a GLP-1 developed “ophthalmic complications,” according to the researchers. The average age of the patients was 57.4 years, according to the study findings.

Seven of the patients had nonarteritic anterior ischemic optic neuropathy (NAION), which causes vision loss in one eye.

One patient developed bilateral papillitis, which involves swollen optic nerves that can cause impaired vision, and another had paracentral acute middle maculopathy, which leads to a blind spot in the retina.

All the patients had a history of type 2 diabetes, hyperlipidemia (high lipids or fats in the blood), hypertension and/or sleep apnea.

The findings were published in JAMA Ophthalmology.

“In one of the cases presented, the patient was taking the drugs for weight loss and did not have a prior history of diabetes (which can also be linked to the condition),” Mahyar Etminan, associate professor of medicine at the University of British Columbia, told Fox News Digital. (Etminan was author of the commentary on the study.)

“In another case, when the drug was stopped and reintroduced, the condition reappeared, strengthening a causal link.”

Ziyad Al-Aly, a clinical epidemiologist at Washington University in St. Louis, was not involved in the study but shared his comments on the findings.

“This is a very small study and it was uncontrolled — meaning it did not include people who were not using GLP-1 drugs,” he told Fox News Digital.

“The story of GLP-1 is still being written — and we are learning something new about these drugs every day.”

“This makes it impossible to know whether the reported eye problems are caused by these drugs.”

Nevertheless, the doctor noted, “the story of GLP-1 is still being written — and we are learning something new about these drugs every day. The findings in this study should be pursued further.”

Etminan also acknowledged the study’s limitations.

“This data was derived from a series of individual cases and was not an epidemiologic study,” he noted. “However, another recent epidemiologic study also confirmed an increase in risk.”

Al-Aly called for large, controlled studies — including people who take the drug and a control group of people who are not using the drug — to evaluate the long-term health effects of these medications, including eye problems.

“In the meantime, for people who may be at risk of vision problems, or who already have vision problems, caution is advised,” he added. “People should discuss with their doctors to determine if GLP-1 is the right medication for them.”

Etminan echoed that cautionary guidance.

“Those taking these drugs for diabetes should probably continue taking them for their cardiovascular benefits, but be aware of the signs of NAION,” he advised.

“Healthy individuals taking them to lose a few pounds for an event might want to more carefully weigh the risks versus the benefits of taking these drugs.”

“Most of the vision side effects appear to resolve when the medication is stopped.”

Dr. Seth Kipnis, medical director of bariatric and robotic surgery at Hackensack Meridian Jersey Shore University Medical Center, noted that there have been “rare and unusual side effects” from this class of medications, but he believes the vision changes seem to be more related to the rapid blood sugar changes caused by the medications than to the medications themselves.

“We have encouraged any patients who are on these types of medications to report any unusual symptoms to their prescribing doctors,” Kipnis, who also was not involved in the research, told Fox News Digital.

“Most of the vision side effects appear to resolve when the medication is stopped.”

Kipnis emphasized that these drugs should only be taken under the care of a healthcare professional and that “good and consistent follow-up for dose adjustments with monitoring for side effects” is critical.

When contacted by Fox News Digital, Novo Nordisk (maker of Ozempic and Wegovy) provided the following statement.

“NAION is a very rare eye disease, and it is not an adverse drug reaction for the marketed formulations of semaglutide (Ozempic®, Rybelsus®and Wegovy®) as per the approved labels. After a thorough evaluation of studies from the University of Southern Denmark and Novo Nordisk’s internal safety assessment, Novo Nordisk is of the opinion that the benefit-risk profile of semaglutide remains unchanged.”

The company also noted that eye conditions are “well-known comorbidities” for people living with diabetes.

“Any decision to start treatment with prescription-only medicines should be made in consultation with a healthcare professional who should do a benefit-risk evaluation for the patient in question, weighing up the benefits of treatment with the potential risks,” Novo Nordisk added.

Enzymes are the engines of life—machine learning could help scientists design new ones

Enzymes are molecular machines that carry out the chemical reactions that sustain all life, an ability that has captured the attention of scientists like me.

Consider muscle movement. Your body releases a molecule called acetylcholine to trigger your muscle cells to contract. If acetylcholine sticks around for too long, it can paralyze your muscles—including your heart muscle cells—and, well, that’s that. This is where the enzyme acetylcholinesterase comes in. This enzyme can break down thousands of acetylcholine molecules per second to ensure muscle contraction is stopped, paralysis avoided and life continued. Without this enzyme, it would take a month for a molecule of acetylcholine to break down on its own—about 10 billion times slower.

You can imagine why enzymes are of particular interest to scientists looking to solve modern problems. What if there were a way to break down plastic, capture carbon dioxide or destroy cancer cells as fast as acetylcholinesterase breaks down acetylcholine? If the world needs to take action quickly, enzymes are a compelling candidate for the job—if only researchers could design them to handle those challenges on demand.

Designing enzymes, unfortunately, is very hard. It’s like working with an atom-sized Lego set, but the instructions were lost and the thing won’t hold together unless it’s assembled perfectly. Newly published research from our team suggests that machine learning can act as the architect on this Lego set, helping scientists build these complex molecular structures accurately.

What’s an enzyme?

Let’s take a closer look at what makes up an enzyme.

Enzymes are proteins—large molecules that do the behind-the-scenes work that keep all living things alive. These proteins are made up of amino acids, a set of building blocks that can be stitched together to form long strings that get knotted up into specific shapes.

The specific structure of a protein is key to its function in the same way that the shapes of everyday objects are. For example, much like a spoon is designed to hold liquid in a way that a knife simply can’t, the enzymes involved in moving your muscles aren’t well suited for photosynthesis in plants.

For an enzyme to function, it adopts a shape that perfectly matches the molecule it processes, much like a lock matches a key. The unique grooves in the enzyme—the lock—that interact with the target molecule—the key—are found in a region of the enzyme known as the active site.

The active site of the enzyme precisely orients amino acids to interact with the target molecule when it enters. This makes it easier for the molecule to undergo a chemical reaction to turn into a different one, making the process go faster. After the chemical reaction is done, the new molecule is released and the enzyme is ready to process another.

How do you design an enzyme?

Scientists have spent decades trying to design their own enzymes to make new molecules, materials or therapeutics. But making enzymes that look like and go as fast as those found in nature is incredibly difficult.

Enzymes have complex, irregular shapes that are made up of hundreds of amino acids. Each of these building blocks needs to be placed perfectly or else the enzyme will slow down or completely shut off. The difference between a speed racer and slowpoke enzyme can be a distance of less than the width of a single atom.

Initially, scientists focused on modifying the amino acid sequences of existing enzymes to improve their speed or stability. Early successes with this approach primarily improved the stability of enzymes, enabling them to catalyze chemical reactions at a higher range of temperatures. But this approach was less useful for improving the speed of enzymes. To this day, designing new enzymes by modifying individual amino acids is generally not an effective way to improve natural enzymes.

Researchers found that using a process called directed evolution, in which the amino acid sequence of an enzyme is randomly changed until it can perform a desired function, proved much more fruitful. For example, studies have shown that directed evolution can improve chemical reaction speed, thermostability, and even generate enzymes with properties that aren’t seen in nature. However, this approach is typically labor-intensive: You have to screen many mutants to find one that does what you want. In some cases, if there’s no good enzyme to start from, this method can fail to work at all.

Both of these approaches are limited by their reliance on natural enzymes. That is, restricting your design to the shapes of natural proteins likely limits the kinds of chemistry that enzymes can facilitate. Remember, you can’t eat soup with a knife.

Is it possible to make enzymes from scratch, rather than modify nature’s recipe? Yes, with computers.

Designing enzymes with computers

The first attempts to computationally design enzymes still largely relied on natural enzymes as a starting point, focusing on placing enzyme active sites into natural proteins.

This approach is akin to trying to find a suit at a thrift store: It is unlikely you will find a perfect fit because the geometry of an enzyme’s active site (your body in this analogy) is highly specific, so a random protein with a rigidly fixed structure (a suit with random measurements) is unlikely to perfectly accommodate it. The resulting enzymes from these efforts performed much more slowly than those found in nature, requiring further optimization with directed evolution to reach speeds common among natural enzymes.

Recent advances in deep learning have dramatically changed the landscape of designing enzymes with computers. Enzymes can now be generated in much the same way that AI models such as ChatGPT and DALL-E generate text or images, and you don’t need to use native protein structures to support your active site.

Our team showed that when we prompt an AI model, called RFdiffusion, with the structure and amino acid sequence of an active site, it can generate the rest of the enzyme structure that would perfectly support it. This is equivalent to prompting ChatGPT to write an entire short story based on a prompt that only says to include the line “And sadly, the eggs never showed up.”

We used this AI model specifically to generate enzymes called serine hydrolases, a group of proteins that have potential applications in medicine and plastic recycling. After designing the enzymes, we mixed them with their intended molecular target to see whether they could catalyze its breakdown. Encouragingly, many of the designs we tested were able to break down the molecule, and better than previously designed enzymes for the same reaction.

To see how accurate our computational designs were, we used a method called X-ray crystallography to determine the shapes of these enzymes. We found that many of them were a nearly perfect match to what we digitally designed.

Our findings mark a key advance in enzyme design, highlighting how AI can help scientists start to tackle complex problems. Machine learning tools could help more researchers access enzyme design and tap into the full potential of enzymes to solve modern-day problems.

 

AI model generates antimicrobial peptide structures for screening against treatment-resistant microbes

A team of microbiologists, chemists and pharmaceutical specialists at Shandong University, Guangzhou Medical University, Second Military Medical University and Qingdao University, all in China, has developed an AI model that generates antimicrobial peptide structures for screening against treatment-resistant microbes.

In their study published in the journal Science Advances, the group developed a compression method to reduce the number of elements needed in training data for an AI system, which helped to reduce diversification issues with current AI models.

Prior research has suggested that drug-resistant microbes are one of the most pressing problems in medical science. Researchers around the world have been looking for new ways to treat people infected with such microbes—one approach involves developing antimicrobial peptides, which work by targeting bacterial membranes.

Unfortunately, developing or finding peptides has proven to be too slow to address the crisis. So researchers have turned to AI-based approaches to aid in finding such peptides. But that approach has encountered problems, as well, the biggest being the lack of a large training base, which leads to peptide discovery that lacks diversity.

In this new study, the researchers in China found a way around this problem by developing a compression technique that reduces the number of elements needed to train their AI system.

The researchers call their system a two-stage AI pipeline leverage diffusion model. The first stage works by compressing data describing 2.8 million known peptides into a numerical form by amplifying signal noise randomly. The second stage then pulls new peptides from the simplified data, removes the noise, and decompresses the data used to describe its peptide sequence.

In testing their new system, the research team found that it was able to filter peptides listed in a training database down to a reasonable number of those most likely to have antimicrobial properties. In looking at 600,000 of them, the team experimentally tested 40 peptides and found 25 that showed promise in combating bacterial and fungal pathogens.

error: Content is protected !!