Tag Archives: mutations

Most “Silent” Mutations Are Actually Harmful

Synonymous mutations have long been thought to have relatively no impact.

A new study finds that most “silent” mutations are harmful rather than neutral.

Marshall Nirenberg, a University of Michigan alumni, and a small group of researchers cracked the genetic code of life in the early 1960s, figuring out the rule by which information stored in

Occasionally, single-letter misspellings in the genetic code, known as point mutations, occur. Nonsynonymous mutations are point modifications that alter the protein sequences that result from them, while silent or synonymous mutations do not change the protein sequences.

One-quarter to one-third of protein-coding DNA sequence point mutations are synonymous. They have often been thought to be neutral or almost neutral mutations ever since the genetic code was deciphered.

But in a study recently published Nature that involved the genetic manipulation of yeast cells in the laboratory, University of Michigan biologists show that most synonymous mutations are strongly harmful.

The strong non-neutrality of most synonymous mutations—if found to be true for other genes and in other organisms—would have major implications for the study of human disease mechanisms, population and conservation biology, and evolutionary biology, according to the study authors.

“Since the genetic code was solved in the 1960s, synonymous mutations have been generally thought to be benign. We now show that this belief is false,” said study senior author Jianzhi “George” Zhang, the Marshall W. Nirenberg Collegiate Professor in the U-M Department of Ecology and Evolutionary Biology.

“Because many biological conclusions rely on the presumption that synonymous mutations are neutral, its invalidation has broad implications. For example, synonymous mutations are generally ignored in the study of disease-causing mutations, but they might be an underappreciated and common mechanism.”

In the past decade, anecdotal evidence has suggested that some synonymous mutations are nonneutral. Zhang and his colleagues wanted to know if such cases are the exception or the rule.

They chose to address this question in budding yeast (Saccharomyces cerevisiae) because the organism’s short generation time (about 80 minutes) and small size allowed them to measure the effects of a large number of synonymous mutations relatively quickly, precisely, and conveniently.

They used CRISPR/Cas9 genome editing to construct more than 8,000 mutant yeast strains, each carrying a synonymous, nonsynonymous or nonsense mutation in one of 21 genes the researchers targeted.

Then they quantified the “fitness” of each mutant strain by measuring how quickly it reproduced relative to the nonmutant strain. Darwinian fitness, simply put, refers to the number of offspring an individual has. In this case, measuring the reproductive rates of the yeast strains showed whether the mutations were beneficial, harmful or neutral.

To their surprise, the researchers found that 75.9% of synonymous mutations were significantly deleterious, while 1.3% were significantly beneficial.

“The previous anecdotes of nonneutral synonymous mutations turned out to be the tip of the iceberg,” said study lead author Xukang Shen, a graduate student research assistant in Zhang’s lab.

“We also studied the mechanisms through which synonymous mutations affect fitness and found that at least one reason is that both synonymous and nonsynonymous mutations alter the gene-expression level, and the extent of this expression effect predicts the fitness effect.”

Zhang said the researchers knew beforehand, based on the anecdotal reports, that some synonymous mutations would likely turn out to be nonneutral.

“But we were shocked by the large number of such mutations,” he said. “Our results imply that synonymous mutations are nearly as important as nonsynonymous mutations in causing disease and call for strengthened effort in predicting and identifying pathogenic synonymous mutations.”

The U-M-led team said that while there is no particular reason why their results would be restricted to yeast, confirmations in diverse organisms are required to verify the generality of their findings.

Reference: “Synonymous mutations in representative yeast genes are mostly strongly non-neutral” by Xukang Shen, Siliang Song, Chuan Li, and Jianzhi Zhang, 8 June 2022, Nature.
DOI: 10.1038/s41586-022-04823-w

The study was funded by the U.S. National Institutes of Health. 



Read original article here

Autism-Related Mutations Inhibit Export of Key Protein From Endoplasmic Reticulum

Summary: Study finds an autism-related genetic mutation increases splicing errors and induces endoplasmic reticulum stress by activating the unfolded protein response.

Source: University of Tsukuba

Anyone who has ever gotten stuck in a traffic jam can attest to the disruption that it causes to your day. Now, researchers from Japan have found that an autism-associated mutation can cause a traffic jam of unfolded proteins that disrupts normal brain function.

In a study that was recently published in Scientific Reports, researchers from the University of Tsukuba reveal that a mutation in an autism-associated protein called Hevin impairs its normal processing and secretion.

Many gene mutations associated with autism spectrum disorder have been identified to date, including some mutations that are inherited. However, in most cases the functional effects of these mutations have not been determined.

“We previously found that mutation of the Usp15 gene, which is closely associated with autism, increases the probability of splicing errors and induces endoplasmic reticulum stress by activating the unfolded protein response,” explains Professor Fuminori Tsuruta. “However, it remained unclear how it causes these effects.”

To address this, the researchers looked for autism-associated variants that exhibited abnormal splicing in the absence of Usp15 in mouse brains and found that the tail end of the transcript encoding a protein called Hevin tends to be lacking. Intriguingly, a mutation in the same part of Hevin, known as the EF-hand motif, has been associated with a familial case of autism.

Many gene mutations associated with autism spectrum disorder have been identified to date, including some mutations that are inherited. Image is in the public domain

“Analysis of the Hevin deletion mutant and the Hevin variant with a single point mutation showed that both mutants accumulated in the endoplasmic reticulum, leading to activation of the unfolded protein response,” says Professor Tsuruta.

Importantly, structural modeling of the Hevin mutation associated with familial autism showed that this single amino acid substitution triggers exposure of a hydrophobic amino acid to the surface. This change is likely to cause structural instability and interfere with export from the endoplasmic reticulum.

“Taken together, our findings suggest that the integrity of the EF-hand motif in Hevin is crucial for proper folding, and that autism-related mutations impair the export of Hevin from the endoplasmic reticulum,” says Professor Tsuruta.

Given that several other autism-related mutations have also been shown to promote the accumulation of synaptic proteins in the endoplasmic reticulum, it is possible that the resulting impairment in neuronal function contributes to autism pathogenesis.

Future studies may help reveal how the endoplasmic reticulum stress response affects neural circuits and brain homeostasis and clarify the link to autism development.

About this genetics and autism research news

Author: Press Office
Source: University of Tsukuba
Contact: Press Office – University of Tsukuba
Image: The image is in the public domain

Original Research: Open access.
“Autism-associated mutation in Hevin/Sparcl1 induces endoplasmic reticulum stress through structural instability” by Takumi Taketomi et al. Scientific Reports


Abstract

See also

Autism-associated mutation in Hevin/Sparcl1 induces endoplasmic reticulum stress through structural instability

Hevin is a secreted extracellular matrix protein that is encoded by the SPARCL1 gene. Recent studies have shown that Hevin plays an important role in regulating synaptogenesis and synaptic plasticity. Mutations in the SPARCL1 gene increase the risk of autism spectrum disorder (ASD).

However, the molecular basis of how mutations in SPARCL1 increase the risk of ASD is not been fully understood. In this study, we show that one of the SPARCL1 mutations associated with ASD impairs normal Hevin secretion.

We identified Hevin mutants lacking the EF-hand motif through analyzing ASD-related mice with vulnerable spliceosome functions. Hevin deletion mutants accumulate in the endoplasmic reticulum (ER), leading to the activation of unfolded protein responses.

We also found that a single amino acid substitution of Trp647 with Arg in the EF-hand motif associated with a familial case of ASD causes a similar phenotype in the EF-hand deletion mutant. Importantly, molecular dynamics (MD) simulation revealed that this single amino acid substitution triggers exposure of a hydrophobic amino acid to the surface, increasing the binding of Hevin with molecular chaperons, BIP.

Taken together, these data suggest that the integrity of the EF-hand motif in Hevin is crucial for proper folding and that ASD-related mutations impair the export of Hevin from the ER.

Our data provide a novel mechanism linking a point mutation in the SPARCL1 gene to the molecular and cellular characteristics involved in ASD.

Read original article here

Mutations thought to be harmless turn out to cause problems

Enlarge / The genetic code. Note that a lot of the amino acids (the outer layer, in grey) are encoded by several sets of three-base codes that share the first two letters.

Mutations are the raw ingredient of evolution, providing variation that sometimes makes an organism more successful in its environment. But most mutations are expected to be neutral and have no impact on an organism’s fitness. These can be incredibly useful since these incidental changes help us track evolutionary relationships without worrying about selection for or against the mutation affecting its frequency. All of the genetic ancestry tests, for example, rely heavily on tracking the presence of these neutral mutations.

But this week, a paper provided evidence that a significant category of mutations isn’t as neutral as we thought they were. The big caveat is that the study was done in yeast, which is a weird organism in a couple of ways, so we’ll have to see if the results hold in others.

True neutral?

One of the reasons that most mutations are neutral is that most of our DNA doesn’t seem to be doing anything useful. Only a few percent of the human genome is composed of the portion of genes that encode proteins, and only some of the nearby DNA is involved in controlling the activity of those genes. Outside of those regions, mutations don’t do much, either because the DNA there has no function or because the function isn’t very sensitive to having a precise sequence of bases in the DNA.

But even within the parts of genes that encode proteins, the precise sequence shouldn’t matter all that much. Each protein’s amino acid is encoded by a combination of three bases in DNA. That means there are 64 possible codes for amino acids—but we only use 20 different amino acids. As a result, there’s plenty of redundancy in the genetic code. For example, the base series ACG encode the amino acid threonine. So does the series ACA. And ACC. All told, four different codes will get you threonine.

The key thing to note is that all four codes start with AC. If you have a mutation in either of those two bases, you no longer get threonine. But if you get a mutation in the third position, it doesn’t matter—whatever you change the base to, you still get threonine. That should be a completely neutral mutation. And researchers have used the assumption that it is neutral to help them track protein evolution.

That’s the assumption that the new paper put to the test.

Make all the mutations

To test neutral mutations, the researchers started with a panel of 21 yeast genes, chosen partly because they are involved in a wide variety of cellular activities. The other part behind their choice is that eliminating these genes doesn’t kill the yeast but makes it less healthy. That should make it easier to detect partial effects, where the mutation makes the yeast less healthy.

Within that stretch, the researchers picked a 150-base stretch in the DNA and made every single possible mutation, using DNA editing to make a yeast strain carrying the mutation. That is a total of over 9,000 individual yeast strains, with some carrying mutations that will change the amino acid sequence and others carrying mutations we’d expect to be neutral. But of course, this involved lab work, where things don’t work for random, unknown reasons, so the researchers had to settle for testing about 8,300 mutant yeast strains.

The test was pretty simple. Throw equal numbers of normal and mutant yeast in a flask, and let them grow for a bit. Then, sample the population, and check the relative levels of normal and mutant yeast. If the mutation lowered the fitness, you’d see more normal yeast when you sampled the flask.

That was true for mutations that changed an amino acid. These saw their relative fitness drop a bit, though not by much (their fitness was 0.988 that of the normal yeast). But the neutral mutations weren’t notably different—they also dropped the yeast’s fitness by a tiny amount relative to a normal strain. In effect, the mutations that didn’t change any amino acids were, on average, indistinguishable from the ones that did. Beyond this average, you could tell a slight difference. There were more amino acid-altering mutations that had a stronger deleterious effect on fitness, and more neutral ones that had a minimal effect. But it’s clear that, as a whole, the class expected to be neutral wasn’t.

Read original article here

Coronavirus mutations aren’t slowing down

Placeholder while article actions load

During those terrifying early days of the pandemic, scientists offered one piece of reassuring news about the novel coronavirus: It mutated slowly. The earliest mutations did not appear to be consequential. A vaccine, if and when it was invented, might not need regular updating over time.

This proved overly optimistic.

The coronavirus, SARS-CoV-2, has had billions of chances to reconfigure itself as it has spread across the planet, and it continues to evolve, generating new variants and subvariants at a clip that has kept scientists on their toes. Two-and-a-half years after it first spilled into humans, the virus has repeatedly changed its structure and chemistry in ways that confound efforts to bring it fully under control.

And it’s not showing signs of settling down into a drowsy old age. Even with all the changes so far, it still has abundant evolutionary space to explore, according to virologists who are tracking it closely. What that means in practical terms is that a virus that’s already extremely contagious could become even more so.

“This virus has probably got tricks we haven’t seen yet,” virologist Robert F. Garry of Tulane University said. “We know it’s probably not quite as infectious as measles yet, but it’s creeping up there, for sure.”

The latest member of the rogue’s gallery of variants and subvariants is the ungainly named BA.2.12.1, part of the omicron gang. Preliminary research suggests it is about 25 percent more transmissible than the BA.2 subvariant that is currently dominant nationally, according to the Centers for Disease Control and Prevention. The CDC said the subvariant has rapidly spread in the Northeast in particular, where it accounts for the majority of new infections.

“We have a very, very contagious variant out there. It is going to be hard to ensure that no one gets covid in America. That’s not even a policy goal,” President Biden’s new covid-19 coordinator, Ashish Jha, said in his inaugural news briefing Tuesday.

He was answering a question about Vice President Harris, who recently tested positive for the virus and went into isolation. Harris had recently been boosted for the second time — her fourth shot of vaccine.

Her case highlights what has become painfully obvious in recent months: No amount of vaccination or boosting can create a perfect shield against infection from SARS-CoV-2. What the vaccines do very well, however, is greatly reduce the risk of severe illness. That is hugely consequential as a matter of public health, as is the wider use of therapeutics, such as the antiviral Paxlovid.

The vaccines currently deployed were all based on the genomic sequence of the original strain of the virus that spread in late 2019 in Wuhan, China. They essentially mimic the spike protein of that version of the virus and trigger an immune response that is protective when the real virus shows up.

But the variants that have emerged can evade many of the neutralizing antibodies that are the immune system’s front line of defense.

“It’s evolving at a fairly rapid rate,” said Jesse Bloom, a computational biologist at the Fred Hutchinson Cancer Research Center in Seattle. “I do think we need to aggressively consider whether we should update vaccines, and do it soon.”

BA.2.12.1 brings the novel coronavirus up another step on the contagiousness scale. Its close relative, BA.2, was already more transmissible than the first omicron strain that hit the country in late 2021.

And omicron was more transmissible than delta, and delta was more transmissible than alpha, and alpha was more transmissible than earlier variants that did not have the glory of a Greek alphabet name.

Most mutations are not advantageous to the virus. But when a mutation offers some advantage, the process of natural selection will favor it.

There are two fundamental ways that the virus can improve its fitness through mutation. The first could be described as mechanical: It can become innately better at infecting a host. Perhaps it improves its ability to bind to a receptor cell. Or perhaps the mutation allows the virus to replicate in greater numbers once an infection has begun — increasing the viral load in the person and, commensurately, the amount of virus that is shed, potentially infecting other people.

The other strategy involves the workaround of immunity. The human immune system, when primed by vaccines or previous infection to be alert for a specific virus, will deploy antibodies that recognize and neutralize it. But mutations make the virus less familiar to the immune system’s front-line defense.

The subvariants keep coming: Scientists in South Africa have identified BA.4 and BA.5, which have mutations that were seen in earlier variants and could lead to immune evasion.

The BA.2 “stealth” omicron variant is expected to soon become the dominant strain. Here is what you need to know about a possible new wave of infections. (Video: Brian Monroe, John Farrell/The Washington Post)

“The evolution is much more rapid and expansive than we initially estimated,” said Michael T. Osterholm, a University of Minnesota infectious-disease expert. “Every day I wake up, I fear there will be a new subvariant that we will have to consider. … We’re seeing subvariants of subvariants.”

Garry, the Tulane scientist, points out that mutations in the virus do not change its appearance dramatically. In fact, he said, even the heavily mutated variants don’t look much different from the original Wuhan strain, or different from other coronaviruses that cause common colds. These are subtle changes.

Garry has a software program that allows him to create a graphic image of the virus, and even rotate it, to observe the locations of mutations and draw inferences for why they matter. On Friday, asked about BA.2.12.1, and why it is spreading, he noted it has a mutation, named S704L, that probably destabilizes a portion of the spike protein on the virus’s surface. That essentially loosens up part of the spike in a way that facilitates infection.

This S704L mutation distinguishes this subvariant from BA.2.

The “704” refers to the 704th position for an amino acid on a chain of roughly 1,100 amino acids that form the protein. The S is one type of amino acid (“serine”) seen in the original strain of the virus, and the L (“leucine”) is what is there after the mutation. (The mutation is caused by a change in one nucleotide, or “letter,” in the genetic code of the virus; three nucleotides encode for an amino acid.)

The virus is spreading today in the United States on an immunological landscape much different from the one it first encountered in early 2020. Between vaccinations and infections, there aren’t many people entirely naive to the virus. The latest CDC data suggest the virus has managed to infect nearly 200 million people in the nation, which has a population of about 330 million. Among children and teenagers, about three out of four have been infected, the CDC estimates.

For the new CDC study, researchers looked at blood samples from thousands of people and searched for an antibody that is found after a natural infection, but not found after vaccination. The CDC concluded that the omicron variant managed to plow through the United States population during the winter almost as if it were an entirely new virus. The country by then was largely vaccinated. And yet 80 million people, approximately, became infected for the first time in that omicron wave.

On the family tree of this coronavirus, omicron is a distant cousin of delta, alpha and the other variants that had spread earlier — it came out of virologic left field. No one is sure of the origin of omicron, but many disease experts assume it came from an immunocompromised patient with a very lengthy illness, and the virus continued to use mutations to evade the immune system’s efforts to clear it.

Omicron was mercifully less likely to kill a person than previous variants. But infectious-disease experts are clear on this point: Future variants could be more pathogenic.

As if mutation wasn’t enough of a problem, the virus has another trick up its sleeve: recombination. It happens when two distinct strains infect a single host simultaneously and their genes becoming entangled. The recombination process is the origin of what’s known as omicron XE. That recombinant probably emerged from a person co-infected with the original omicron variant and the BA.2 subvariant.

It was always possible in theory, but the identification of actual recombinants provides “proof of concept,” as Jeremy Luban, a virologist at the University of Massachusetts Medical School, puts it.

The worst-case scenario would be the emergence of a variant or recombinant that renders current vaccines largely ineffective at blocking severe disease. But so far, that hasn’t happened. And no “recombinant” has spread like omicron or other recent variants and subvariants.

This is the first catastrophic pandemic to occur in the age of modern genomic sequencing. A century ago, no one knew what a coronavirus was, and even a “virus” was a relatively new concept. But today, with millions of samples of the virus analyzed at the genetic level, scientists can track mutations virtually in real time and watch the virus evolve. Scientists across the planet have uploaded millions of sequences to the database known as GISAID.

Genomic sequencing has a major limitation in that, although scientists can track changes in the genome, they don’t automatically know what each of those changes is doing to the virus. Which mutations matter most is a question that can be discerned through laboratory experiments, modeling or epidemiological surveillance, but it’s not always simple or obvious.

Erica Saphire, president of the La Jolla Institute for Immunology, speculates that omicron has mutations that have changed the virus in ways not yet understood but which make it more resistant to antibody-mediated neutralization.

“It may have acquired some new trick that we haven’t uncovered yet,” Saphire said. “It’s harder to neutralize than I would have expected, based on the number of mutations alone.”

A reality check comes from Jeremy Kamil, associate professor of microbiology and immunology at Louisiana State University Health Shreveport: “These are all SARS-CoV-2.”

What he means is that these are all variations of the same virus, despite what seems like a tremendous amount of mutation. Correspondingly, someone who gets infected with one of these new variants has the same disease as people who got infected previously.

“They got covid,” he said.

Read original article here

Mutations in Noncoding DNA Are Found to Protect the Brain From ALS

Summary: Mutations in the IL18RAP gene reduce inflammation and appear to protect the brain against ALS.

Source: Weizmann Institute of Science

Genetic mutations linked to a disease often spell bad news. Mutations in over 25 genes, for example, are associated with amyotrophic lateral sclerosis, or ALS, and they all increase the risk of developing this incurable disorder.

Now, a research team headed by Prof. Eran Hornstein of the Weizmann Institute of Science has linked a new gene to ALS, but this one contains mutations of a different sort: They seem to play a defensive rather than an offensive role in the disease.

The gene newly linked to ALS is located in the part of our genome once called “junk DNA.” This DNA makes up over 97 percent of the genome, but because it does not encode proteins, it used to be considered “junk.”

Today, though this noncoding DNA is still regarded as biological dark matter, it’s already known to serve as a crucial instruction manual. Among other things, it determines when genes within the coding DNA—the ones that do encode proteins—are turned on and off.

Hornstein’s lab in Weizmann’s Molecular Neuroscience and Molecular Genetics Departments studies neurodegenerative diseases—that is, diseases in which neurons degenerate and die. The team is focusing on our noncoding DNA.

“This massive, noncoding part of the genome has been overlooked in the search for the genetic origins of neurodegenerative diseases like ALS,” Hornstein explains.

“This is despite the fact that for most ALS cases, proteins cannot explain the emergence of the disease.”

Many people know about ALS thanks to the Ice Bucket Challenge that went viral a few years ago. This rare neurological disease attacks motor neurons, the nerve cells responsible for controlling voluntary muscle movement involved in everything from walking to talking and breathing.

The neurons gradually die off, ultimately causing respiratory failure and death. One of the symptoms of ALS is inflammation in the brain regions connected to the dying neurons, caused by immune mechanisms in the brain.

“Our brain has an immune system,” explains Dr. Chen Eitan, who led the study in Hornstein’s lab together with Aviad Siany. “If you have a degenerative disease, your brain’s immune cells, called microglia, will try to protect you, attacking the cause of the neurodegeneration.”

Credit: Weizmann Institute of Science

The problem is that in ALS, the neurodegeneration becomes so severe that the chronic microglial activation in the brain rises to extremely high levels, turning toxic. The immune system thus ends up causing damage to the brain it set out to protect, leading to the death of more motor neurons.

That’s where the new findings, published today in Nature Neuroscience, come in. The Weizmann scientists focused on a gene called IL18RAP, long known to affect microglia, and found that it can contain mutations that mitigate the microglia’s toxic effects. “We have identified mutations in this gene that reduce inflammation,” Eitan says.

After analyzing the genomes of more than 6,000 ALS patients and of more than 70,000 people who do not have ALS, the researchers concluded that the newly identified mutations reduce the risk of developing ALS nearly fivefold.

The gene newly linked to ALS is located in the part of our genome once called “junk DNA.” Image is in the public domain

It is therefore extremely rare for ALS patients to have these protective mutations, and those rare patients who do harbor them tend to develop the disease roughly six years later, on average, than those without the mutations. In other words, the mutations seem to be linked to a core ALS process, slowing the disease down.

To confirm the findings, the researchers used gene-editing technology to introduce the protective mutations into stem cells from patients with ALS, causing these cells to mature into microglia in a laboratory dish.

They then cultured microglia, with or without the protective mutations, in the same dishes with motor neurons. Microglia harboring the protective mutations were found to be less aggressive toward motor neurons than microglia that did not have the mutations.

“Motor neurons survived significantly longer when cultured with protective microglia, rather than with regular ones,” Siany says.

Eitan notes that the findings have potential implications for ALS research and beyond. “We’ve found a new neuroprotective pathway,” she says.

See also

“Future studies can check whether modulating this pathway may have a positive effect on patients. On a more general level, our findings indicate that scientists should not ignore noncoding regions of DNA—not just in ALS research, but in studying other diseases with a genetic component as well.”

About this genetics and ALS research news

Author: Press Office
Source: Weizmann Institute of Science
Contact: Press Office – Weizmann Institute of Science
Image: The image is in the public domain

Original Research: Closed access.
“Whole-genome sequencing reveals that variants in the Interleukin 18 Receptor Accessory Protein 3′UTR protect against ALS” by Chen Eitan et al. Nature Neuroscience


Abstract

Whole-genome sequencing reveals that variants in the Interleukin 18 Receptor Accessory Protein 3′UTR protect against ALS

The noncoding genome is substantially larger than the protein-coding genome but has been largely unexplored by genetic association studies.

Here, we performed region-based rare variant association analysis of >25,000 variants in untranslated regions of 6,139 amyotrophic lateral sclerosis (ALS) whole genomes and the whole genomes of 70,403 non-ALS controls.

We identified interleukin-18 receptor accessory protein (IL18RAP) 3′ untranslated region (3′UTR) variants as significantly enriched in non-ALS genomes and associated with a fivefold reduced risk of developing ALS, and this was replicated in an independent cohort. These variants in the IL18RAP 3′UTR reduce mRNA stability and the binding of double-stranded RNA (dsRNA)-binding proteins.

Finally, the variants of the IL18RAP 3′UTR confer a survival advantage for motor neurons because they dampen neurotoxicity of human induced pluripotent stem cell (iPSC)-derived microglia bearing an ALS-associated expansion in C9orf72, and this depends on NF-κB signaling.

This study reveals genetic variants that protect against ALS by reducing neuroinflammation and emphasizes the importance of noncoding genetic association studies.

Read original article here

Strange Evolutionary Pace of SARS-CoV-2 Mutations Is Finally Revealed in New Study

The timeline of the COVID-19 pandemic has been marked by a series of catastrophic waves: surging crests of infection spreading around the world, often spearheaded by newly evolved variants of the pathogen, such as Delta and Omicron.

 

This is simply how viral evolution can play out, of course. But SARS-CoV-2 is an unusually successful and dangerous virus; part of what makes the ‘novel coronavirus’ so formidable is its ceaseless novelty – the unusually rapid pace at which new variants appear to be spawning.

“What we were seeing with the variants of SARS-CoV-2, particularly the variants of concern, is that they have undergone many more mutations than we would expect under the normal evolutionary pace of similar coronaviruses,” explains infectious disease researcher Sebastian Duchene from the Peter Doherty Institute for Infection and Immunity in Australia.

Ordinarily, Duchene notes, viruses tend to mutate at a relatively constant pace, taking perhaps a year or longer for a new viral variant to emerge. But the coronavirus doesn’t seem to stick to that calendar.

“The Delta variant, for example, emerged within just six weeks from its ancestral form,” Duchene says.

In a new study, Duchene and fellow researchers sought to investigate where this dramatically accelerated timeframe comes from.

They analyzed SARS-CoV-2 genome sequence data to examine how the emergence of variants of concern (VOCs, the most virulent and harmful lineages) might be linked to changes in the substitution rate of the virus: the rate at which new mutations arise in the pathogen’s genetic code.

 

According to the researchers, the background substitution rate of SARS-CoV-2 suggests the virus accrues approximately two mutations each month.

But VOCs are a different beast, with variants such as Alpha, Beta, Gamma, and Delta acquiring numerous mutations in relatively short timeframes, each of which can alter things like the variants’ infectiousness, ability to replicate, level of fitness, and so on.

“The sheer number of mutations observed in these four VOCs is much higher than what would be expected under phylogenetic estimates of the nucleotide evolutionary rate of SARS-CoV-2,” the researchers explain in their paper, led by first author John Tay, a bioinformatics researcher at the Doherty Institute.

According to the team, the secret of the VOCs’ accelerated mutation is not a constant, ongoing phenomenon, but rather something that appears to happen temporarily in the virus’s evolution, taking place shortly before variants emerge.

“We find compelling evidence that episodic, instead of long term, increases in the substitution rate underpin the emergence of VOCs,” the team writes.

The increased rate of substitutions is about four times higher than the background phylogenetic rate estimate for SARS-CoV-2, but the analysis suggests the accrual of mutations happens in a compressed burst: perhaps as short as four weeks for the Beta variant, and six weeks for the Delta variant.

 

Other variants took longer, with the Gamma variant thought to have evolved over the course of 17 weeks, while Alpha required 14 weeks.

That’s the how of it, but as for why these mutation bursts occur at all, we’re not entirely sure.

The researchers say the emergence of VOCs is probably driven by natural selection. Other relevant factors could include infections in unvaccinated populations – which may enable the virus to spread and evolve more easily – and persistent infections in particular individuals, such as immunocompromised patients, which may also lead to altered viral dynamics.

While there’s still much we don’t fully understand about what triggers so many rapid mutations in SARS-CoV-2, the fact that we can see and track this happening means ongoing genomic monitoring of the virus is crucial.

Doing so might just give us a chance to stop the next wave – instead of catching it.

“This makes the case for very good genomic surveillance, because we didn’t catch the intermediate forms of Omicron, and surely there were a few,” Duchene told The Sydney Morning Herald.

“Imagine if you could have detected Omicron in the first few patients – if you could prevent it spreading from there, then we wouldn’t be in the situation we are now.”

The findings are reported in Molecular Biology and Evolution.

 

Read original article here

Are genetic mutations random? Israeli study says no

Is the view by neo-Darwinists, that genetic mutations in human genes are inherently randomized, true? A study by a team of researchers from Israel and Ghana seemingly refutes this argument.

For the past century, an assumption central to Charles Darwin’s evolutionary theory is that mutations are random and accidental and that natural selection favors such accidents. In an article published in the scientific peer-reviewed journal Genome Research, researchers have found the first evidence of non-random mutations in human genes.

Using a new and innovative method, the researchers – led by Haifa University’s Prof. Adi Livnat – have managed to prove that the rate of generation of the human hemoglobin S (HbS) mutation which protects one from malaria is higher in people from Africa in contrast to people from Europe. In other words, the mutation is not random but rather exists preferentially within the population of Africa where it is more needed.

Malaria is endemic in Africa, highly common around the entire continent; the more common development of a malaria-resistant mutation specific to the region where it is most needed cannot be explained by the traditional neo-Darwinist theories.

“We hypothesize that evolution is influenced by two sources of information: external information that is natural selection, and internal information that is accumulated in the genome through the generations and impacts the origination of mutations,” explained Livnat.

While the theory of evolution is widely accepted in the scientific community, the small details have been put under a microscope for quite some time. For example, there are some impressively quick adaptations of wildlife to their changing surroundings and conditions that suggest that, if natural selection were fully true, the random accidents mentioned earlier were happening at an astonishingly fast pace.

Until now, the only response to this proposed problem was Lamarckism, which claims that the physical changes in organisms which occur during their lifetimes can be passed along genetically to offspring. Since this was not proven to work, the random mutation was maintained as the prominently held belief.

Livnat, alongside lab manager Dr. Daniel Melamed, managed to develop a new record-breakingly accurate method of detecting random mutations which they applied in their research to track the development of the HbS mutation. If random, the mutation should appear relatively equally throughout both Europe and Africa.

“Contrary to the widely accepted expectations, the results supported the nonrandom pattern,” Haifa University announced. “The HbS mutation originated de novo not only much faster than expected from random mutation but also much faster in the population (in sub-Saharan Africans as opposed to Europeans) and in the gene (in the beta-globin as opposed to the control delta-globin gene) where it is of adaptive significance.”

These results effectively contradicted the commonly-held random mutation belief held by Darwinists.

“The results suggest that complex information that is accumulated in the genome through the generations impacts mutation, and therefore mutation-specific origination rates can respond in the long-term to specific environmental pressures,” said Prof. Livnat, whose study was funded by a grant given by the  John Templeton Foundation. “Mutations may be generated nonrandomly in evolution after all, but not in the way previously conceived. We must study the internal information and how it affects mutation, as it opens the door to evolution being a far bigger process than previously conceived.”



Read original article here

How Omicron’s Mutations Allow It To Thrive

Because an immunocompromised host doesn’t produce a lot of antibodies, many viruses are left to propagate. And new mutant viruses that resist the antibodies can multiply.

A mutation that allows a virus to evade antibodies isn’t necessarily advantageous. It could make the virus’s spike protein unstable so that it can’t latch quickly onto a cell, for example. But inside someone with a weak immune system, viruses may be able to gain a new mutation that stabilizes the spike again.

Similar mutations could have built upon themselves again and again in the same person, Dr. Pond speculates, until Omicron evolved a spike protein with just the right combination of mutations to allow it to spread supremely well among healthy people.

“It certainly seems plausible,” said Sarah Otto, an evolutionary biologist at the University of British Columbia who was not involved in the study. But she said scientists still needed to run experiments to rule out alternative explanations.

It’s possible, for example, that the 13 spike mutations offer no benefit to Omicron at all. Instead, some of the other spike mutations could be making Omicron successful, and the 13 are just along for the ride.

“I would be cautious about interpreting the data to indicate that all of these previously deleterious mutations have been adaptively favored,” Dr. Otto said.

Dr. Pond also acknowledged that his hypothesis still has some big gaps. For example, it’s not clear why, during a chronic infection, Omicron would have gained an advantage from its new “bubble” method for getting into cells.

“We just lack imagination,” Dr. Pond said.

James Lloyd-Smith, a disease ecologist at U.C.L.A. who was not involved in the study, said that the research revealed just how hard it is to reconstruct the evolution of a virus, even one that arose recently. “Nature is certainly doing its part to keep us humble,” he said.

Read original article here

High number of Omicron mutations render antibodies ineffective – study

The highly contagious COVID-19 Omicron variant has a large number of uniquely specific mutations that allow it to evade pre-existing antibodies in the human body, accounting for its high rate of infection, new research carried out by the University of Minnesota has found.

The peer-reviewed study titled “Omicron SARS-CoV-2 variant: Unique features and their impact on pre-existing antibodies” was first published in the Journal of Autoimmunity and was produced by Kamlendra Singh, a professor in the University of Missouri College of Veterinary Medicine and assistant director of the college’s Molecular Interactions Core and Bond Life Sciences Center investigator.

The research team set out to gather data on the mutations found in the spike protein (S-protein) of the Omicron variant. An S-protein refers to a large structure projecting from the surface of the virus’s outermost layer, and they are most commonly associated with all forms of coronavirus cells.

The research team found an unprecedented number of mutations in the Omicron S-protein. They analyzed the available sequences of the virus along with the structural data on the spike protein in order to understand the possible impact that the high number of mutations could have on the binding of antibodies to the virus.

Antibodies allow the human body to fight off viruses that enter the system, preventing them from entering the immune system. While earlier in the COVID-19 pandemic it was thought that being infected with COVID-19, or being vaccinated against it, would provide enough antibodies to prevent reinfection, the Omicron variant has proved otherwise, as high amounts of people are being re-infected, or infected despite being fully vaccinated.

Technicians carry out a diagnostic test for Covid-19 in a lab at Leumit Health Care Services branch in Or Yehuda, on January 21, 2022. (credit: YOSSI ZELIGER/FLASH90)

Using complete sequences of the Omicron variant, the research team identified a total of 46 signature mutations within the variant, 23 of which were completely unique and had not been identified in any of the earlier variants of the virus. Two of the mutations had first been recorded in the Delta or Delta Plus variant which preceded Omicron by several months.

Of the 46 mutations found, 30 were identified in the S-protein while the remainder were located elsewhere in the virus cell.

Having identified the unique mutations found in the Omicron variant, the team turned to researching whether or not they were responsible for the lack of antibody response against the variant.

Using a preexisting S-protein structure taken from the Protein Data Bank, one which would theoretically prevent the binding of antibodies to a virus, they worked to assess whether or not the Omicron mutations would similarly affect the COVID-19 S-protein, thus rendering antibodies ineffective.

Through this method, the team discovered that specific mutations create interference in the surface of the virus, preventing antibodies from binding to it, while others result in a complete loss of interaction between the antibodies and the virus, thereby rendering the antibodies ineffective against the highly-mutated variant.

This, the study assessed, suggests that preexisting immunization (whether from vaccination or previous infection) may no longer be able to provide optimal protection against the Omicron variant, allowing it to bypass antibodies and enter into the immune system.

“The purpose of antibodies is to recognize the virus and stop the binding, which prevents infection,” said Singh of the research. “However, we found many of the mutations in the Omicron variant are located right where the antibodies are supposed to bind, so we are showing how the virus continues to evolve in a way that it can potentially escape or evade the existing antibodies, and therefore continue to infect so many people.”



Read original article here

DNA Mutations Do Not Occur Randomly – Discovery Transforms Our View of Evolution

Beating the Odds in Mutation’s Game of Chance

Discovery that plants protect their most essential genes transforms our view of evolution.

Mutations of

The thale cress (Arabidopsis thaliana). Credit: Max Planck Institute for Biology Tübingen

Protecting plants with harmful mutations

Researchers grew specimens of the widely distributed weed Arabidopsis thaliana in a sheltered lab environment, where all plants, including ones with harmful mutations, could reproduce. Such harmful mutations would normally be quickly removed by the selection pressures that prevail in nature and therefore disappear before they could be observed. By analyzing the genomes of hundreds of lab grown plants, the scientist could identify thousands of mutations as they arose.

Sophisticated statistical analyses revealed that these mutations were by no means randomly distributed in the genome, as the researchers had expected. Instead, they found stretches of the genome where mutations were rare, and others where mutations were much more common. In those regions with few mutations, genes needed in every cell and thus essential for the survival of every plant were greatly overrepresented. “These are regions of the genome most sensitive to harmful effects of new mutations,” Weigel says, “and DNA damage repair seems therefore to be particularly effective in these regions.”  It is as if evolution were playing with loaded dice – it minimizes the risk of damaging the most vital genes.

Breeding of the thale cress under laboratory conditions in the greenhouse. Credit: Max Planck Institute for Biology Tübingen

A new perspective on classical evolutionary theory

The scientists found that the different types of proteins around which DNA is wrapped in the cell nucleus are highly correlated with the appearance of mutations. “It gives us a good idea of what’s going on, so that we can predict which genes are more likely to mutate than others,” Monroe says.

Weigel stressed how entirely unexpected the results were in the light of classical evolutionary theory: “It has long been known that during the course of evolution certain regions of the genome accumulate more mutations than other regions do. At first glance, what we found seemed to contradict accepted wisdom that this just reflects natural selection removing most mutations before they can actually be observed,” he explains. However, despite the uneven distribution of mutations in a typical genome, the important regions are not entirely devoid of them, and these regions can therefore also evolve, although at a slower pace than other parts of the genome. 

Future uses in breeding and medical research

“The plant has evolved a way to protect its most important genes from mutation,” Monroe says. “This is exciting because we could even use these discoveries to think about how to protect human genes from mutation.” In the future, one might use them to predict which genes are best targets for breeding because they evolve fast, or which are most likely to cause disease in humans.

Reference: “Mutation bias reflects natural selection in Arabidopsis thaliana” by J. Grey Monroe, Thanvi Srikant, Pablo Carbonell-Bejerano, Claude Becker, Mariele Lensink, Moises Exposito-Alonso, Marie Klein, Julia Hildebrandt, Manuela Neumann, Daniel Kliebenstein, Mao-Lun Weng, Eric Imbert, Jon Ågren, Matthew T. Rutter, Charles B. Fenster and Detlef Weigel, 12 January 2022, Nature.
DOI: 10.1038/s41586-021-04269-6

Most of the work was carried out at the Max Planck Institute for Biology (formerly the Max Planck Institute for Developmental Biology), and it is now being continued both there and at UC Davis. Researchers from the Carnegie Institution for Science, Stanford University, Westfield State University, University of Montpellier, Uppsala University, College of Charleston, and South Dakota State University also contributed to the work. Funding came from the Max Planck Society, with additional funding from the National Science Foundation and the German Research Foundation.



Read original article here

The Ultimate News Site