January 10, 2025

Autism and Brain Cancer

Youthful Brain Stem Cells Linked to Autism and Brain Cancer



Researchers have identified a unique stem cell in the young brain capable of maturing into multiple cell types, potentially explaining the origins of autism and glioblastoma. These stem cells show gene expression patterns that regulate early brain development and, when disrupted, could lead to neurological conditions.

The study provides a detailed gene expression map, linking autism-related genes to immature neurons active during brain growth. The findings open avenues for targeting glioblastoma’s origins and better understanding autism’s developmental roots.Stem Cell Discovery: A stem cell capable of maturing into three brain cell types may drive glioblastoma growth.

Autism Insight: 

Autism-associated genes are active during key stages of brain development, potentially affecting neuronal growth.

Innovative Mapping: 

A comprehensive map of brain cell gene expression reveals links between development and disease.

Source: UCSF

UCSF scientists have discovered a stem cell in the young brain that’s capable of forming the cells found in tumors. The breakthrough could explain how adult brain cells take advantage of developmental processes to instigate the explosive growth seen in deadly brain cancers like glioblastoma.

They made the discovery while taking a broad genomic survey of human brain cells from the first two decades of life. The findings appear Jan. 8 in Nature.

“Many brain diseases begin during different stages of development, but until now we haven’t had a comprehensive roadmap for simply understanding healthy brain development,” said Arnold Kriegstein, MD, PhD, professor of neurology at UCSF and co-corresponding author of the paper.

“Our map highlights the genetic programs behind the growth of the human brain that go awry during specific forms of brain dysfunction.”

The study measured gene expression in cells taken from donated brain samples. The researchers kept track of the original location of each cell to help explain how the brain creates connections.

In addition to the discovery of an early stem cell that could explain the genetics of glioblastoma in adulthood, the data contained hints about the origins of autism. The researchers have published the data as a resource for the field to use for understanding a wide range of other brain disorders.

“Our study paints one of the most detailed pictures of human brain development,” said Li Wang, PhD, postdoctoral researcher in Kriegstein’s laboratory and co-first and co-corresponding author of the paper.

“Theories based on observations in the clinic and laboratory can now be tested against this hard data, and we’re excited to see what else the field can do with it.”
Samples reveal a treasure trove
Most studies of the developing brain are carried out in animal models, which are at best loose proxies for the human brain.

The team, also led by co-first author Cheng Wang, PhD, and co-corresponding author Jingjing Li, PhD, wagered that valuable new insights could be made by studying the human brain itself. They worked with the National Institutes of Health’s NeuroBioBank and local hospitals associated with UCSF to obtain brain samples.

These samples, donated from 27 individuals from early life through adolescence, were sent to UCSF and analyzed for gene expression in thousands of individual cells.

Gene expression refers to how DNA, stored in chromosomes, is copied into RNA – short-lived genetic messages – which are then used as a template for building proteins. By measuring RNA, the researchers could peer into the behavior of those cells.

“RNA degrades quickly, and you need to have very pristine tissue in order to get usable data,” Kriegstein said.

“It was a huge advance for Li and his colleagues to perform such high-resolution genomic tests on this tissue, and we thank the community for supporting such critical research by donating this precious tissue.”

The researchers analyzed which parts of each chromosome were available for expressing genes in each cell. They also labeled where each cell had been taken from in the brain.

The scientists focused on cells taken from the front and the back of the cerebral cortex, regions that in humans are responsible for learning, memory and language.

“RNA alone doesn’t tell the entire story of a cell’s behavior,” Wang said.

“By measuring RNA and chromatin state at the same time in the same cell and then mapping each cell back into the brain’s structure, we could begin to understand the full story of brain development.”

A mélange of autism risk genes emerge in the developing brain

Autism isn’t caused by a single gene mutation, but rather by the combination of many gene mutations.

The researchers found that many of the genes that correlate with autism were turned on by immature neurons well before any symptoms would have manifested. Mutations in these genes, they said, could interfere with the growth of the young brain, leading to autism.

“These programs of gene expression became active when young neurons were still migrating throughout the growing brain and figuring out how to build connections with other neurons,” Wang said.

“If something goes wrong at this stage, those maturing neurons might become confused about where to go or what to do.”

Since the study didn’t look at tissue from individuals with autism, it’s still unclear exactly how autism unfolds in the brain. But the data link many genetic variations associated with autism to the cells that serve as the building blocks for the growing brain.

“People talk about connecting the dots to come up with a picture of how autism emerges, and in a sense, we’ve identified many of the dots driving autism during a critical point in development,” Kriegstein said.

“This part of development could be worthy of further investigation for untangling all the mysteries of autism.”

Could a program for early brain growth be coopted for tumor growth later in life?

As the researchers sifted through their data, Wang noticed a group of stem cells that seemed poised to do something unusual. These immature cells had begun to express genes normally found across three mature cell types.

Many stem cells in the developing brain mature into just one cell type, like a neuron or a support cell. Some can mature into two types. But these stem cells could mature into three lineages: two types of support cells known as glia, and one type of neuron.

The researchers thought this ability might enable it to give rise later in life to glioblastoma tumors, which contain three similar cell types.

“Glioblastoma has been challenging because it’s so heterogeneous,” Kriegstein said. “Li found a precursor capable of making all three glioblastoma cell types.”

The discovery validates a widely held theory that tumors hijack genetic growth programs for out-of-control growth in adulthood.

And it may provide a new entree for treating glioblastoma at its source: the “cancer stem cell.”

“By understanding the context in which one stem cell produces three cell types in the developing brain, we could be able to interrupt that growth when it reappears during cancer,” Wang said.

DNA repair, oxidative stress, genomic instability, cell cycle checkpoints, DNA double-strand breaks, single-strand breaks, DNA methylation, nucleotide excision repair, base excision repair, homologous recombination, non-homologous end joining, telomere shortening, replication stress, chromosomal aberrations, mutagenesis, apoptosis, tumorigenesis, reactive oxygen species, genetic mutations, cancer progression

#DNArepair, #GenomicInstability, #CellCycle, #DNADamage, #OxidativeStress, #ChromosomalAberrations, #Mutagenesis, #Telomeres, #HomologousRecombination, #NonHomologousEndJoining, #BaseExcisionRepair, #NucleotideExcisionRepair, #ReactiveOxygenSpecies, #ReplicationStress, #Tumorigenesis, #DNARepairMechanisms, #CancerResearch, #GeneticMutations, #Apoptosis, #GenomeIntegrity

January 09, 2025

DNA Damage

Novel Mechanism in DNA Damage Unraveled


A research team from the University of California (UC), Irvine, has revealed a previously unknown mechanism that triggers an inflammatory immune response in cells when their DNA is damaged. The finding provides a new understanding of a new type of cell signaling that may lead to more effective treatments for cancer.

The study is published in Nature Structural & Molecular Biology in an article titled, “ATM and IRAK1 orchestrate two distinct mechanisms of NF-κB activation in response to DNA damage.”

The researchers discovered that UV irradiation or certain chemotherapeutic drugs activate a specific response when cells are too damaged to be repaired correctly, preventing them from becoming cancerous.

“DNA damage in cells induces the expression of inflammatory genes,” the researchers wrote. “However, the mechanism by which cells initiate an innate immune response in the presence of DNA lesions blocking transcription remains unknown. Here we find that genotoxic stresses lead to an acute activation of the transcription factor NF-κB through two distinct pathways, each triggered by different types of DNA lesions and coordinated by either ataxia-telangiectasia mutated (ATM) or IRAK1 kinases.”

“This discovery could have significant implications for cancer treatment,” explained corresponding author Rémi Buisson, PhD, UC Irvine associate professor of biological chemistry. “Understanding how different cancer cells react to DNA damage could lead to more tailored and effective therapies, potentially reducing negative side effects and improving the quality of life for patients.”

Scientists have long understood that when both DNA strands are broken, the ATM enzyme triggers the activation of the protein NF-κB within the cell, leading to the production of inflammatory signals. In this study, spearheaded by postdoctoral fellow Elodie Bournique, PhD, and assisted by graduate student Ambrocio Sanchez, it was shown that when DNA damage occurs due to UV exposure or treatment with chemotherapeutic drugs such as actinomycin D or camptothecin, the IRAK1 enzyme induces NF-κB to send out signals to recruit immune cells.

The researchers developed an imaging technique to analyze how NF-κB is regulated at the cellular level. They were able to measure a cell’s response to damaged DNA at the single-cell level and observed a new pathway to the activation of NF-κB. They found that after specific types of injury, cells release the IL-1α protein. It doesn’t act on the cell itself but travels to neighboring cells, where it triggers the IRAK1 protein, which then initiates the NF-κB inflammatory response.

“Our findings will help us better understand the consequences of certain types of chemotherapeutic drugs that are used to treat patients and cause DNA damage. We’ve discovered that the IL-1α and IRAK1 proteins, which play a role in the immune process, vary significantly across different cancer cell types. This suggests that not all patients will react to treatment in the same way, Buisson said. “By assessing these protein levels ahead of time, doctors might be able to personalize therapies tailored to individual patients’ needs for improved success rates.”

Looking toward the future, the researchers will continue their work by testing their findings on mouse models that lack specific factors involved in the new pathway.

DNA repair, oxidative stress, genomic instability, cell cycle checkpoints, DNA double-strand breaks, single-strand breaks, DNA methylation, nucleotide excision repair, base excision repair, homologous recombination, non-homologous end joining, telomere shortening, replication stress, chromosomal aberrations, mutagenesis, apoptosis, tumorigenesis, reactive oxygen species, genetic mutations, cancer progression

#DNArepair, #GenomicInstability, #CellCycle, #DNADamage, #OxidativeStress, #ChromosomalAberrations, #Mutagenesis, #Telomeres, #HomologousRecombination, #NonHomologousEndJoining, #BaseExcisionRepair, #NucleotideExcisionRepair, #ReactiveOxygenSpecies, #ReplicationStress, #Tumorigenesis, #DNARepairMechanisms, #CancerResearch, #GeneticMutations, #Apoptosis, #GenomeIntegrity

January 08, 2025

Correcting Genetic Spelling Errors

Correcting Genetic Spelling Errors With Next-Generation Crispr


Sam Berns was my friend. With the wisdom of a sage, he inspired me and many others about how to make the most of life. Afflicted with the rare disease called progeria, his body aged at a rapid rate, and he died of heart failure at just 17, a brave life cut much too short.

My lab discovered the genetic cause of Sam’s illness two decades ago: Just one DNA letter gone awry, a T that should have been a C in a critical gene called lamin A. The same misspelling is found in almost all of the 200 individuals around the world with progeria.

The opportunity to address this illness by directly fixing the misspelling in the relevant body tissues was just science fiction a few years ago. Then Crispr came along—the elegant enzymatic apparatus that allows delivery of DNA scissors to a specific target in the genome. In December 2023, the FDA approved the first Crispr-based therapy for sickle cell disease. That approach required taking bone marrow cells out of the body, making a disabling cut in a particular gene that regulates fetal hemoglobin, treating the patient with chemotherapy to make room in the marrow, and then reinfusing the edited cells. A relief from lifelong anemia and excruciating attacks of pain is now being delivered to sickle cell patients, albeit at very high cost.

For progeria and thousands of other genetic diseases, there are two reasons why this same approach won’t work. First, the desired edit for most misspellings will not usually be achieved by a disabling cut in the gene. Instead, a correction is needed. In the case of progeria, the disease-causing T needs to be edited back to a C. By analogy with a word processor, what’s needed is not “find and delete” (first-generation Crispr), it’s “find and replace” (next-generation Crispr). Second, the misspelling needs to be repaired in the parts of the body that are most harmed by the disease. While bone marrow cells, immune cells, and skin cells can be taken out of the body to administer gene therapy, that won’t work when the main problem is in the cardiovascular system (as in progeria) or the brain (as in many rare genetic diseases). In the lingo of the gene therapist, we need in vivo options.

The exciting news in 2025 is that both of these barriers are starting to come down. The next generation of Crispr-based gene editors, pioneered particularly elegantly by David Liu of the Broad Institute, allows precise corrective editing of virtually any gene misspelling, without inducing a scissors cut. As for delivery systems, the family of adeno-associated virus (AAV) vectors already provides the ability to achieve in vivo editing in eye, liver, and muscle, though there is still much work to be done to optimize delivery to other tissues and ensure safety. Nonviral delivery systems such as lipid nanoparticles are under intense development and may displace viral vectors in a few years.

Working with David Liu, Sam Berns’ mom, and Leslie Gordon of the Progeria Research Foundation, my research group has already shown that a single intravenous infusion of an in vivo gene editor can dramatically extend the life of mice that have been engineered to carry the human progeria mutation. Our team is now working to bring this forward to a human clinical trial. We are truly excited about the potential for kids with progeria, but that excitement could have even greater impact. This strategy, if successful, could be a model for the approximately 7,000 genetic diseases where the specific misspelling that causes the disease is known, but no therapy exists.

There are many hurdles, cost being a major one as private investment is absent for diseases that affect only a few hundred individuals. However, success for a few rare diseases, supported by government and philanthropic funds, will likely lead to efficiencies and economies that will help with other future applications. This is the best hope for the tens of millions of children and adults who are waiting for a cure. The rare-disease community must press on. That’s what Sam Berns would have wanted.

Gene expression, Genetic variation, DNA sequencing, Gene therapy, Genomic editing, CRISPR-Cas9, Genetic disorders, Population genetics, Hereditary traits, Epigenetics, Genome-wide association study, Genetic linkage, Molecular genetics, Genetic engineering, Genomics, Chromosomal abnormalities, Mendelian inheritance, Genetic markers, Bioinformatics, Functional genomics

#Genetics, #DNA, #GeneTherapy, #CRISPR, #Epigenetics, #GenomeResearch, #GenomicScience, #PopulationGenetics, #GeneEditing, #MolecularBiology, #Bioinformatics, #HereditaryResearch, #GenomicMedicine, #GeneticMarkers, #FunctionalGenomics, #MendelianGenetics, #GeneticDisorders, #GeneticResearch, #ChromosomalStudies, #GeneticVariation

January 07, 2025

Genetic Disease Variability

New Insight into Genetic Disease Variability



New research reveals that certain cells inactivate one parent’s copy of a gene, leading to a bias in gene activity that may explain why some individuals with disease-causing mutations remain symptom-free. This selective gene inactivation, known as monoallelic expression, affects about 1 in 20 genes and varies between cell types.

The study shows that in families with genetic disorders, the active copy of a gene often determines disease severity. These findings challenge traditional genetic paradigms and suggest new approaches to diagnosing and treating inherited diseases.

Key Facts: 

Gene Inactivation: Cells can selectively inactivate one parent’s gene copy, influencing disease outcomes.
Active copies of genes determine the severity or absence of symptoms in genetic disorders.
Understanding this phenomenon could lead to therapies that adjust gene expression patterns.

Every biology student learns that each cell in our body (except sperm and eggs) contains two copies of each gene, one from each parent, and each copy plays an equal part in the cell.

The new study shows that some cells are often biased when it comes to some genes and inactivate one parent’s copy. The phenomenon was discovered about a decade ago, but the new study shows how it can influence disease outcomes.

The Columbia researchers looked at certain immune cells of ordinary people to get an estimate of the phenomenon and found that these cells had inactivated the maternal or paternal copy of a gene for one out of every 20 genes utilized by the cell.

“This is suggesting that there is more plasticity in our DNA than we thought before,” says study leader Dusan Bogunovic, professor of pediatric immunology at Columbia University Vagelos College of Physicians and Surgeons.

“So in some cells in your body every 20th gene can be a little bit more Mom, a little bit less Dad, or vice versa. And to make thing even more complicated, this can be different in white blood cells than in the kidney cells, and it can perhaps change with time.”

The results were published Jan. 1 in the journal Nature.

Why it matters

The new study explains a longstanding puzzle in medicine: why do some people who’ve inherited a disease-causing mutation experience fewer symptoms than others with the same mutation?

“In many diseases, we’ll see that 90% of people who carry a mutation are sick, but 10% who carry the mutation don’t get sick at all,” says Bogunovic, a scientist who studies children with rare immunological disorders at Columbia University Irving Medical Center.

Enlisting an international team of collaborators, the researchers looked at several families with different genetic disorders affecting their immune systems. In each case, the disease-causing copy was more likely to be active in sick patients and suppressed in healthy relatives who had inherited the same genes.

“There was some speculation that this bias toward one copy or the other could explain wide differences in the severity of a genetic disease, but no experimental evidence existed until now,” Bogunovic says.

Though the current work looked only at immune cells, Bogunovic says the selective bias for the maternal or paternal copy of a gene affected more than just immune-related genes.

“We don’t see a preference for immune genes or any other class of genes, so we think this phenomenon can explain the wide variability in disease severity we see with many other genetic conditions,” he says, adding “this could be just the tip of the iceberg.”

The phenomenon could help explain diseases with flares, like lupus, or those that emerge following environmental triggers. It could also play a role in cancer.

Changing the future of treatments for genetic diseases?

The study’s findings point to an entirely new paradigm for diagnosing and perhaps even treating inherited diseases.

The investigators propose expanding the standard characterization of genetic diseases to include patients’ “transcriptotypes,” their gene activity patterns, in addition to their genotypes.

“This changes the paradigm of testing beyond your DNA to your RNA, which as we’ve shown in our study, is not equal in all cell types and can change over time,” says Bogunovic.

If researchers can identify the mechanisms behind selective gene inactivation, they may also be able to treat genetic diseases in a new way, by switching a patient’s gene expression pattern to suppress the undesirable copy.

While emphasizing that such strategies are still far from clinical use, Bogunovic is optimistic: “At least in cell culture in the lab we can do it, so manipulation in that way is something that could turn somebody’s genetic disease into non-disease, assuming we are successful.”

genotype-phenotype relationships, mutational hotspots, epigenetic regulation, gene-environment interaction, allelic heterogeneity, modifier genes, pleiotropy, polygenic traits, copy number variations, genomic imprinting, mosaicism, single nucleotide polymorphisms (SNPs), penetrance, expressivity, genomic instability, rare genetic disorders, multifactorial inheritance, molecular diagnostics, precision medicine, genetic counseling

#GeneticVariability #GeneticDiseases #GenomicResearch #GeneEnvironmentInteraction #Epigenetics #MolecularGenetics #AllelicDiversity #GenotypePhenotype #HumanGenomics #PrecisionMedicine #RareDiseases #GenomicImprinting #SNPAnalysis #GenomicInstability #ModifierGenes #Pleiotropy #MultifactorialDiseases #GeneticCounseling #MolecularDiagnostics #EpigenomicStudies

January 06, 2025

Unborn Babies Use Genes

Unborn Babies Use Genes From Dad to “Remote-Control” Mothers for Extra Food


Scientists at Cambridge have unveiled a fascinating mechanism where fetuses use a paternal gene to control the mother’s nutrient release during pregnancy.

This “remote control” system involves hormonal signals from the placenta, which ensure the fetus grows optimally by altering the mother’s metabolic processes. Remarkably, this battle for nutrients is a delicate balance, crucial not just for fetal growth but also for the mother’s health and her future reproductive potential.

Nutritional Control in Pregnancy

Cambridge scientists have discovered that fetuses use a gene inherited from their father to influence their mother’s body into providing more nutrients during pregnancy.

This creates a kind of “nutritional tug of war,” where the unborn baby ‘remote controls’ its mother’s metabolism to maximize its growth, while the mother’s body balances her own need to maintain health. The mother must ensure enough glucose and fats remain available for her energy needs, to sustain the pregnancy, support breastfeeding, and allow for future pregnancies.

Hormonal Signaling by the Placenta

A University of Cambridge study explored how the placenta plays a key role in this process. By releasing specific hormones, the placenta communicates with the mother’s body to prioritize the baby’s growth. This vital organ, which develops alongside the fetus, supports fetal development in humans and other mammals. In experiments with pregnant mice, scientists modified the signaling cells in the placenta that regulate how nutrients are allocated to the fetus.

Professor Amanda Sferruzzi-Perri, Professor in Fetal and Placental Physiology, a Fellow of St John’s College and co-senior author of the paper, said: “It’s the first direct evidence that a gene inherited from the father is signaling to the mother to divert nutrients to the fetus.”

Gene Wars: Maternal vs Paternal Influences

Dr. Miguel Constancia, MRC Investigator based at the Wellcome-MRC Institute of Metabolic Science and co-senior author of the paper, said: “The baby’s remote control system is operated by genes that can be switched on or off depending on whether they are a ‘dad’s’ or ‘mum’s’ gene’, the so-called imprinted genes.

“Genes controlled by the father are ‘greedy’ and ‘selfish’ and will tend to manipulate maternal resources for the benefit of the fetuses, so to grow them big and fittest. Although pregnancy is largely cooperative, there is a big arena for potential conflict between the mother and the baby, with imprinted genes and the placenta thought to play key roles.”

The findings by researchers from the Centre for Trophoblast Research at Cambridge’s Department of Physiology, Development and Neuroscience and the Medical Research Council Metabolic Diseases Unit, part of the Wellcome-MRC Institute of Metabolic Science, have been published in Cell Metabolism.

The baby’s genes controlled by the father tend to promote fetal growth and those controlled by the mother tend to limit fetal growth.

Professor Sferruzzi-Perri explained: “Those genes from the mother that limit fetal growth are thought to be a mother’s way of ensuring her survival, so she doesn’t have a baby that takes all the nutrients and is too big and challenging to birth. The mother also has a chance of having subsequent pregnancies potentially with different males in the future to pass on her genes more widely.”

Genetic Manipulation and Nutrient Allocation

Researchers deleted the expression of an important imprinted gene called Igf2, which provides instructions for making a protein called ‘Insulin Like Growth Factor 2’. Similar to the hormone insulin, which is responsible for making and controlling glucose levels in our circulation, the gene promotes fetal growth and plays a key part in the development of fetal tissues including the placenta, liver and brain.

Dr. Jorge Lopez-Tello, a lead author of the study based at the University’s Department of Physiology, Development and Neuroscience, said: “If the function of Igf2 from the father is switched off in signaling cells, the mother doesn’t make enough amounts of glucose and lipids – fats – available in her circulation. These nutrients therefore reach the fetus in insufficient amounts and the fetus doesn’t grow properly.”

The scientists found that deleting Igf2 from the placenta’s signaling cells affects the production of other hormones that modulate the way the mother’s pancreas produces insulin, and how her liver and other metabolic organs respond.

“We found Igf2 controls the hormones responsible for reducing insulin sensitivity in the mother during pregnancy. It means the mother’s tissues don’t absorb glucose so nutrients are more available in the circulation to be transferred to the fetus,” said Professor Sferruzzi-Perri.

Babies with Igf2 gene defects can be overgrown or growth-stunted. “Until now, we didn’t know that part of the Igf2 gene’s role is to regulate signaling to the mother to allocate nutrients to the fetus,” added Professor Sferruzzi-Perri.

The mice studied were smaller at birth and their offspring showed early signs of diabetes and obesity in later life.

Professor Sferruzzi-Perri said: “Our research highlights how important the controlled allocation of nutrients to the fetus is for the lifelong health of the offspring, and the direct role the placenta plays.

“The placenta is an amazing organ. At the end of pregnancy, the placenta is delivered by the mother, but the memories of how the placenta was functioning leaves a lasting legacy on the way those fetal organs have developed and then how they’re going to function through life.”

The next step is to understand how placental hormones are controlled by Igf2 and what those hormones are doing. Future research could help scientists discover new strategies to target the placenta to improve health outcomes for mums and babies.

embryonic development, fetal genetics, gene expression, DNA signaling, prenatal growth, genetic markers, cellular differentiation, genomic regulation, epigenetics, maternal inheritance, developmental biology, CRISPR technology, hereditary traits, organogenesis, genomic imprinting, fetal adaptation, molecular pathways, neurogenesis, gene editing, placental biology.

#EmbryonicDevelopment, #FetalGenetics, #GeneExpression, #PrenatalResearch, #DNA, #GeneticScience, #Epigenetics, #DevelopmentalBiology, #MaternalHealth, #CRISPR, #GeneEditing, #Neurogenesis, #Organogenesis, #GenomicImprinting, #HereditaryTraits, #FetalHealth, #MolecularBiology, #GeneticsForAll, #ScientificDiscovery, #HealthyBabies

January 04, 2025

DNA Methylation

Histone markers predict human age with accuracy comparable to DNA methylation clocks


In a recent study published in the journal Science Advances, a group of researchers investigated the role of histone modifications in human aging by developing and evaluating histone-specific age prediction models across tissues and cell types.

Background

Aging involves complex cellular and molecular changes, including epigenetic modifications like Deoxyribonucleic acid (DNA) methylation and histone marks. Age predictors, or "clocks," have been developed using DNA methylation, transcriptomics, and blood chemistry data, with methylation-based models achieving a median absolute error of ~4 years.

While histone marks offer an interpretable framework based on the histone code, their potential for constructing accurate age predictors remains underexplored. Research has shown age-related shifts in histone modifications, suggesting their utility in modeling aging. However, the study emphasizes that sample size plays a critical role in determining the accuracy and reliability of such predictors.

Further research is needed to fully understand their role and to establish histone-based clocks comparable to existing methylation-based predictors.

About the Study

Researchers collected 1,814 human tissue chromatin immunoprecipitation sequencing (ChIP-seq) samples from the Encyclopedia of DNA Elements (ENCODE) project in bigWig format to generate and interpret histone-based age predictors.

The samples included seven histone modifications: histone H3 lysine 4 trimethylation (H3K4me3), histone H3 lysine 27 acetylation (H3K27ac), histone H3 lysine 27 trimethylation (H3K27me3), histone H3 lysine 4 monomethylation (H3K4me1), histone H3 lysine 36 trimethylation (H3K36me3), histone H3 lysine 9 trimethylation (H3K9me3), and histone H3 lysine 9 acetylation (H3K9ac).

Data processing involved averaging the negative base-10 logarithm of P-values’ signals across gene bodies using Homo sapiens annotations from Ensembl release 105. Samples with substantial missing features were discarded, and missing values were encoded as zero.

Various genomic regions were analyzed, including intergenic regions and Cytosine-phosphate-Guanine (CpG) dinucleotides. Embryonic samples were encoded with gestational week adjustments, while anonymized samples over 90 were assigned an age of 90.

To test in vitro performance, 568 additional samples spanning 12 histone marks were collected. Imputed data from ENCODE’s Avocado dataset added 1,379 samples, enhancing the training dataset. Age predictors employed Elastic Net regularization-based feature selection, principal component analysis (PCA) with truncated support vector decomposition, and automatic relevance determination regression, all implemented in Python. Performance evaluation used 10-fold nested cross-validation to prevent artificially inflated accuracy metrics, excluding cancer samples.

Histone-based predictors were compared to DNA methylation-based predictors, with the study noting the impact of differences in sample size and dataset distributions on the comparison. Predictor interpretation involved gene set enrichment analysis (GSEA), selecting genes significantly contributing to age prediction accuracy. Statistical analyses employed Python packages, ensuring validation.

Study Results

Focusing on seven histone marks (H3K4me3, H3K27ac, H3K9ac, H3K9me3, H3K27me3, H3K36me3, and H3K4me1), researchers used data from 1,814 human tissue samples spanning 82 tissues and age groups ranging from embryonic stages to 90-plus years. The samples represented diverse biological contexts and were processed using standardized methods to ensure consistency and reliability.

To create age predictors, researchers reduced the dimensionality of the data by averaging negative log-transformed P-values for each histone modification across gene bodies. These values were then transformed to stabilize the variance. Uniform manifold approximation and projection (UMAP) and PCA revealed distinct clustering based on histone type, with some age-related trends emerging, particularly for samples over 70 years old.

Histone marks showed significant correlations with age, particularly the repressive marks H3K9me3 and H3K27me3, which decreased with age, and the activating mark H3K4me3, which increased. Notably, the study observed that signal variance for all histone marks increased with age, highlighting epigenetic drift as a key factor in declining regulation. These observations informed the development of multivariate age predictors using ElasticNet for feature selection, principal component analysis to reduce noise, and automatic relevance determination regression for age estimation.

The histone-specific age predictors demonstrated robust performance, with H3K4me3 achieving the highest accuracy (Pearson’s r = 0.94, median absolute error = 4.31 years). Comparisons with DNA methylation-based predictors indicated comparable accuracy, particularly for activating histone marks, though the paper notes that DNA methylation predictors often have a younger skew in sample age distributions, which can affect performance comparisons. Additional experiments with imputed and primary cell data confirmed the reliability and accuracy of the histone mark predictors.

GSEA and pathway analyses highlighted developmental processes, transcriptional regulation, and ribonucleic acid (RNA)-related pathways as key contributors to age prediction. Histone-coding genes and age-related genes such as Homeobox D8 (HOXD8), Thioredoxin Interacting Protein (TXNIP), and Period Circadian Regulator 1 (PER1) were strongly associated with histone mark changes.

The study also introduced a pan-histone, pan-tissue age predictor, which leverages shared age-related trends across histone marks. This model not only performed comparably to histone-specific predictors but also emphasized the shared epigenetic patterns across the genome that underpin aging.
Conclusions

Since the development of DNA methylation-based age predictors, biohorology has rapidly expanded, offering biomarkers like telomere length, transcriptomics, and proteomics. While DNA methylation clocks are accurate, interpreting them is often challenging due to ambiguous gene associations. In contrast, histone mark-based predictors reveal genes linked to development, circadian regulation, and aging. Using ChIP-seq data, researchers created age predictors from seven histone marks.

Crucially, the study demonstrated that models trained on one histone mark could predict age using another, underscoring the shared epigenetic information across histone modifications. This research highlights the interpretability and potential of histone mark-based predictors as a robust tool for understanding epigenetic aging and developing age estimation models.

DNA methylation, epigenetics, gene expression, cytosine modification, methyltransferases, CpG islands, histone modification, gene silencing, transcription regulation, genome stability, 5-methylcytosine, DNA demethylation, epigenomic profiling, chromatin remodeling, DNA methylation biomarkers, environmental epigenetics, DNA methylation and aging, epigenetic inheritance, epigenetic therapy, cancer epigenetics

#DNAmethylation, #Epigenetics, #GeneExpression, #CytosineModification, #Methyltransferases, #CpGIslands, #HistoneModification, #GeneSilencing, #TranscriptionRegulation, #GenomeStability, #5mC, #DNADeMethylation, #Epigenomics, #ChromatinRemodeling, #DNABiomarkers, #EnvironmentalEpigenetics, #EpigeneticsAndAging, #EpigeneticInheritance, #EpigeneticTherapy, #CancerEpigenetics

January 03, 2025

Mitochondrial Mutations

Sweet Spot of Mitochondrial Mutations Fuels Cancer Growth

Moderate mitochondrial DNA mutations enhance leukemia growth, while high mutation levels halt tumor development.

Mitochondria are vital to energy production in cells and so play a key role in fueling cancer growth. However, how mitochondrial DNA (mtDNA) contributes to cancer has been unclear. Scientists at St. Jude Children’s Research Hospital studied varying levels of mutated mtDNA to see their effect on leukemia cells. They found that while cancer growth was blocked in cells in which all mitochondria contained mutated mtDNA, it was notably increased in cells with moderate amounts of mutated mtDNA. By amplifying an enzyme vital to energy production, the researchers we also able to restart cancer growth in leukemia cells with fully mutated mtDNA. Collectively, these findings highlight an unexplored connection between mitochondrial DNA and cancer cells’ metabolic function. The findings were published today in Science Advances.

mtDNA is found exclusively within mitochondria and contains just 37 genes, which are largely responsible for energy production. Mutations occur to this DNA in the same way as DNA found in the nucleus, but studying the effect these mutations have on cancer is much more challenging. Recent advances have allowed Mondira Kundu, MD, PhD, St. Jude Department of Cell & Molecular Biology, to begin to address this knowledge gap.

“The role of mitochondrial DNA mutations in cancer is controversial,” said Kundu. “Some papers suggest they are pro-tumorigenic, and others say they have no impact. It’s essentially been unknown.”

Leukemia thrives in mtDNA mutation ‘sweet spot’

Introducing individual mutations to mtDNA is challenging due to the large number of mitochondria within each cell. Instead, the researchers used a leukemia mouse model with a defective genetic proofreading system called Polg, which gradually accrues mtDNA mutations. By disrupting Polg’s proofreading function in either one (heterozygous) or both (homozygous) parental lines, the researchers could look at the burden that mtDNA mutations place on tumor growth based on the number of mitochondria with mutated mtDNA.

The researchers found that heterozygous mice (those with a moderate number of mutated mitochondria) seemed to amplify leukemia growth. Homozygous mice with a high number of mutations had the opposite effect, blocking tumor growth.

“Until now, researchers have been focusing on an all-or-nothing approach, thinking that a lot of mutation impairs tumor function,” Kundu explained, “but in terms of leukemia, our findings suggest that an intermediate level of mitochondrial mutations might promote leukemogenesis.”

This effect may be related to the ability of leukemia cells to reprogram their metabolism to thrive in a harsh tumor microenvironment (their plasticity). “The amount of metabolic stress [from mtDNA mutation] increases the plasticity of the cells,” she explained. “So, exposure to a little bit of metabolic stress in the heterozygous mice may increase the susceptibility to transformation by different oncogenes, whereas in the homozygous mice, they are basically shutting down. The impact on metabolism was so severe that it could not be overcome.”

Metabolic plasticity connects mtDNA and tumor growth

To explore the mechanisms behind this, the researchers looked at an enzyme called pyruvate dehydrogenase. This enzyme links the two stages of cellular respiration: glycolysis and the citric acid cycle. In doing so, pyruvate dehydrogenase helps regulate the metabolic plasticity of cells. The researchers found that by blocking the kinase “off switch” of pyruvate dehydrogenase, they could restore leukemia cells’ plasticity in the homozygous (high mutation) mice. These results suggest that the citric acid cycle shuts down in the homozygous models, so promoting it restores the growth of those cells.

Collectively, the findings provide clear evidence that low to medium levels of mtDNA mutations can contribute to leukemogenesis and that complete disruption of mitochondrial function can have the opposite effect, essentially halting tumor growth.

Mitochondrial DNA mutations, oxidative phosphorylation, mitochondrial genome, heteroplasmy, mtDNA copy number, mitochondrial dysfunction, mitochondrial diseases, somatic mutations, inherited mutations, oxidative stress, ATP production, bioenergetics, mitochondrial biogenesis, electron transport chain, mitochondrial ROS, mitochondrial repair mechanisms, mitochondrial dynamics, mitochondrial fusion, mitochondrial fission, mitochondrial quality control.

#Mitochondria #GeneticMutations #MitochondrialDNA #MitoDiseases #OxidativeStress #EnergyProduction #MitochondrialHealth #Bioenergetics #MitochondrialResearch #mtDNA #CellMetabolism #Genomics #MitochondrialFunction #MitoBiology #DNARepair #MitochondrialFusion #MitochondrialFission #OxPhos #ReactiveOxygenSpecies #HealthandGenetics

Autism and Brain Cancer

Youthful Brain Stem Cells Linked to Autism and Brain Cancer Researchers have identified a unique stem cell in the young brain capable of mat...