Friday, August 30, 2024

Genetic Improvement is Bright

The future of poultry genetic improvement is bright

Poultry genetics companies are prioritizing new research and technology to meet the needs of hungry consumers worldwide.


Genetics have improved significantly across all sectors of agriculture over the last few decades, from feed ingredients, to cotton production and in animal agriculture, especially in the broiler industry, explained genetics company Cobb Research and Development Vice President William Herring at the 2024 Chicken Marketing Summit.

“We're becoming more efficient. We're doing more with less resources,” he stated. “I don't think there's any end in sight to genetic progress or genetic change. We don't see that across agriculture anywhere, and we certainly don't see it in broiler genetics.”

To help advance poultry genetics further, Cobb created a program called “Cobb’s Proving Grounds,” which is focused on generating a large amount of data to improve its currents products and create new ones.

According to Herring, the initiative is multiplying its product data capabilities by 50 to quickly improve the genetic progress it can make in poultry. To do so, Cobb is utilizing multiple genetic research facilities with large-scale capabilities and environments comparable to commercial production conditions around the world.

Herring said that Cobb placed the first parent stock in April 2024, the second in August 2024 and it plans to place a third in February 2025 to ensure its genetic improvements are scalable on a commercial level and product consistent results.

“It's meant to generate data that one can have confidence in,” he added.

Genetics has always been a subject that is slightly over my head; it can be confusing when you start talking about how it all really works. However, it is encouraging to see how far poultry genetics have come, witness the impact they have had on the supply chain and wonder about where they are going to go next.

Attend the 2025 Chicken Marketing Summit

The 2025 Chicken Marketing Summit will be held at the DeSoto Savannah in Savannah, Georgia, on July 28-30, 2025.

Serving a unique cross-section of the chicken supply chain, the Chicken Marketing Summit explores issues and trends in food marketing and consumer chicken consumption patterns and purchasing behavior.

dual gene therapy, FKRP, FST, LGMDR9, Limb-Girdle Muscular Dystrophy, ambulation, muscle strength, pathology, gene expression, mouse model, muscular dystrophy, Fukutin-Related Protein, Follistatin, therapeutic approach, muscle function, neuromuscular disease, genetic treatment, dystroglycanopathy, muscle regeneration, preclinical study,

#DualGeneTherapy, #FKRP, #FST, #LGMDR9, #MuscularDystrophy, #Ambulation, #MuscleStrength, #Pathology, #GeneExpression, #MouseModel, #FukutinRelatedProtein, #Follistatin, #TherapeuticApproach, #MuscleFunction, #NeuromuscularDisease, #GeneticTreatment, #Dystroglycanopathy, #MuscleRegeneration, #PreclinicalStudy, #GeneTherapy

International Conference on Genetics and Genomics of Diseases 




For Enquiries: genetics@healthcarek.com 

Get Connected Here 
--------------------------------- 
--------------------------------- 
facebook.com/profile.php?id=61555903296992 
tumblr.com/blog/dorita0211 

Thursday, August 29, 2024

Homozygosity primary ovarian insufficiency

Homozygosity for a stop-gain variant in CCDC201 causes primary ovarian insufficiency


Age at menopause (AOM) has a substantial impact on fertility and disease risk. While many loci with variants that associate with AOM have been identified through genome-wide association studies (GWAS) under an additive model, other genetic models are rarely considered. Here through GWAS meta-analysis under the recessive model of 174,329 postmenopausal women from Iceland, Denmark, the United Kingdom (UK; UK Biobank) and Norway, we study low-frequency variants with a large effect on AOM. We discovered that women homozygous for the stop-gain variant rs117316434(A) in CCDC201 (p.(Arg162Ter), minor allele frequency ~1%) reached menopause 9 years earlier than other women (P = 1.3 × 10−15). The genotype is present in one in 10,000 northern European women and leads to primary ovarian insufficiency in close to half of them. Consequently, homozygotes have fewer children, and the age at last childbirth is 5 years earlier (P = 3.8 × 10−5). The CCDC201 gene was only found in humans in 2022 and is highly expressed in oocytes. Homozygosity for CCDC201 loss-of-function has a substantial impact on female reproductive health, and homozygotes would benefit from reproductive counseling and treatment for symptoms of early menopause.

Main

Menopause is caused by the depletion of the primordial follicle pool. There is a broad variation in the age of menopause (AOM), and early menopause (EM) impacts health, quality of life (https://www.menopausemandate.com/) and fertility potential. It is estimated that natural fertility ends on average 10 years before menopause. At the extreme end of the AOM distribution is primary ovarian insufficiency (POI) with cessation of menses before the age of 40 years, which occurs in 1–4% of women. EM and POI are a well-known cause of infertility, which is increasingly relevant as women in many populations are choosing to have children later in life.

Through genome-wide association studies (GWAS), we and others have reported associations of rare and low-frequency variants with variation in AOM, mostly under an additive model. Rare variants in several genes have also been reported to cause Mendelian forms of POIalthough many are only reported in a small number of cases or in single families. Despite advances in understanding the genetic causes of EM and POI, genetic screening has mainly been focused on Turner syndrome, which has a prevalence of 1 in 2,000, and the FMR1 premutation, found in 1 in 8,000 women.

We performed a GWAS meta-analysis for AOM under the recessive model as well as the additive one (not affected by surgical procedures, such as hysterectomy and/or oophorectomy) on 174,329 postmenopausal women from Iceland, the United Kingdom (UK), Denmark and Norway (nIceland = 27,281, nUK = 137,906, nDenmark = 5,978 and nNorway = 3,161; Supplementary Tables). We tested 39.3 million sequence variants for associations with AOM.

Homozygosity (n = 27 women) for the low-frequency stop-gain variant p.(Arg162Ter) (A), chr7: 45863165; minor allele frequency (MAF) ~1%) in CCDC201 is associated with earlier AOM by 9 years than in heterozygotes and noncarriers (recessive effect = −1.59 s.d.; 95% confidence interval (CI): −1.98, −1.20), recessive P = 1.3 × 10−15. The effect of the variant did not differ between the four groups (Phet = 0.28. The association was genome-wide significant in the UK, the largest of the four groups (P = 3.6 × 10−13), and was also significant in the remaining three sample sets combined (P = 2.4 × 10−4. The effect of p.(Arg162Ter) in CCDC201 on AOM deviates from the additive model and is limited to homozygotes. We did not detect an association with AOM under the additive model (additive effect = 0.029 s.d. (95% CI: −0.0094, 0.066), P = 0.16). We did not find a significant association of the p.(Arg162Ter) variant with any case–control or quantitative traits under the additive model.

Methods

Study population

AOM was derived for individuals who were considered to have undergone natural menopause not affected by surgical procedures, such as hysterectomy and/or oophorectomy.

In Iceland, we used data on AOM obtained from the Icelandic Cancer Society’s Cancer Registry (n = 9,794) and from questionnaires from various genetic programs at deCODE genetics (n = 21,390), of which the majority was gathered through deCODE’s osteoporosis project and the deCODE Health study, which had also been genotyped. The Cancer Society’s data were collected from a questionnaire in the years 1964–1994, and deCODE genetics data from 1999 to 2022. All Icelandic data were collected through studies approved by the National Bioethics Committee (approvals VSN-15-198 and VSN-15-214) following review by the Icelandic Data Protection Authority. Participants donated blood or buccal samples after signing a broad informed consent allowing the use of their samples and data in all projects at deCODE genetics approved by the NBC. All personal identifiers of the participants’ data were encrypted by a third-party system, approved and monitored by the Icelandic Data Protection Authority.

The UK Biobank study is a large prospective cohort study of ~500,000 individuals in the age range of 40–69 years from across the UK. AOM (Data Field 3581) was collected from a touchscreen questionnaire at the UK Biobank assessment centers from 140,688 genotyped females who indicated that their periods had stopped (Data Field 2724). Only British individuals of European ancestry were included in the study. The UK Biobank data were obtained under application 56270. All phenotype and genotype data were collected following informed consent obtained from all participants. The North West Research Ethics Committee reviewed and approved the UK Biobank’s scientific protocol and operational procedures (REC reference: 06/MRE08/65).

Data on menopause status from Denmark were provided by the Danish Blood Donor Study (DBDS). Around 51% of participants were females with an age span at inclusion 18–70 years. The data were obtained from a paper questionnaire (v1) on self-reported health status and lifestyle sent to all participants in the DBDS (n = 110,000) from 2010 to mid-year 2015. Around 85,000 participants responded to it. In the end, AOM from 8,037 chip-typed females was used in the analysis. All participants signed an informed consent statement, and the DBDS genetic study was approved by the Danish National Committee on Health Research Ethics (NVK-1700407) and by the Danish Capital Region Data Protection Office (P-2019-99).

Data on female infertility from Denmark were provided by the Copenhagen Hospital Biobank (CHB) Reproduction Study, which involves a targeted selection of patients with reproductive phenotypes from the CHB, a biobank based on patient blood samples drawn in Danish hospitals.

The AOM data from Norway were provided by the Hordaland Health Studies (HUSK). The HUSK surveys are a collaborative project between the University of Bergen, the Norwegian Health Screening Service (SHUS) and the Municipal Health Service in Hordaland aimed at gathering information so that disease ultimately can be prevented. In the first phase of the studies (HUSK1), in 1992–1993, around 18,000 residents of Hordaland County born in 1925–1952 participated in the study. In 1997–1999 (HUSK2), previous participants born in 1950–1951 and 1925–1927 were re-invited, in addition to all residents in Hordaland County born in 1953–1957. In total, approximately 36,000 individuals participated in the study (18,000 in 1992–1993 and 26,000 in 1997–1999), with some participating at both times. Age at last menstruation (proxy for menopause) was collected from questionnaires sent to participants both in HUSK1 and HUSK2. All participants signed an informed consent statement, and the HUSKment study was approved by the Regional Committee for Medical Research Ethics Western Norway (REK Vest 10279 (2018/915)). In the end, AOM from 3,161 genotyped females was used in the analysis.

For all strata, in the case of repeat measurements, the mean age of menopause or the mean age at the last period was used to represent each individual’s AOM.
Rounding tendency in reported age of menopause

It has been observed that when women are asked to recall their AOM, they tend to report values ending in 0 or 5. Thus, we need to take into account the possibility that some women who reported menopause at the age of 40 years may not have been included as POI cases due to this tendency and could lead to an underestimation of the risk of POI in our study. Of the 27 homozygotes for p.(Arg162Ter) with AOM information, nine reported AOM before the age of 40, while seven reported experiencing menopause exactly at the age of 40. Assuming an equal probability of rounding reported AOM up or down to 40, we estimated the penetrance of POI among homozygotes as 46% ((9 + 3.5)/27). Likewise, for noncarriers and heterozygotes, the estimated penetrance of POI is 3.7% ((4,678 + 1,728.5)/174,302).
Estimating the proportion of POI explained by p.(Arg162Ter) homozygosity

Using AOM data to define POI as AOM before the age of 40 years, we can observe nine homozygotes among the 4,687 females with AOM before the age of 40 years. Thus, we estimate that the proportion of all POI cases caused by p.(Arg162Ter) homozygosity is around 0.19% (that is, 1 of 521). Similarly, taking into account rounding bias, the proportion of all POI cases estimated to be caused by homozygosity is also 0.19% (that is, 1 of 513, or (9 + 3.5)/(4,687 + 1728.5)).

In the UK Biobank 500k WGS set, one homozygote was observed among the 571 females with the ICD-10 diagnostic code E283, indicative of POI. Thus, the incidence of p.(Arg162Ter) homozygosity is 1 of 571 among POI cases.

Genotyping

In Iceland, 34,453,001 sequence variants identified in WGS data from 63,460 Icelanders participating in various disease projects at deCODE genetics were tested. The samples were sequenced using standard TruSeq (Illumina) methodology to an average genome-wide coverage of 40×. SNPs and insertions and deletions (InDels) were identified, and their genotypes were called using joint calling with Graphtyper. Variant Effect Predictor from RefSeq was used to annotate the effects of sequence variants on protein-coding genes. We chip-typed 173,025 Icelanders (around 50% of the population) using Illumina SNP arrays, and the chip-typed individuals were long-range phased. The variants identified in the whole-genome sequencing of Icelanders were imputed into the chip-typed individuals. In addition, based on Icelandic genealogy, the genotype probabilities for 292,636 untyped close relatives of chip-typed individuals were calculated.

From the UK Biobank, we used data from around 428k WGS individuals who were of British/Irish ancestry. The WGS was performed using Illumina standard TruSeq methodology (mean depth of 32×) in a collaborative work between deCODE genetics in Iceland and The Wellcome Sanger Institute in the UK. Sequence variants from the WGS were identified and called jointly using Graphtyper. Phasing from previous chip-typing of the same sample was used as the basis to assign haplotypes.

From Denmark and Norway, we chip-typed 464,016 and 254,304 individuals, respectively. The samples were chip-typed by deCODE genetics using both Omni microarrays (Illumina) and Global Screening Array (Illumina). Graphtyper was used to identify SNPs and InDels and jointly call their genotypes. Using the identified variants, the samples were then phased (using SHAPEIT4) along with an international set of 1,041,174 genotyped individuals from 49 countries (including Denmark and Norway), chip-typed at deCODE genetics. For variant imputation, we compiled an international reference panel from 50,839 WGS individuals from 14 countries, including 10,985 from Denmark and 3,467 from Norway. The identified variants from WGS were subsequently imputed into the chip-typed individuals.

Association analysis

We performed a meta-analysis on GWAS on 180,564 females from Iceland, the UK, Denmark and Norway with self-reported AOM or age at last menstruation. We tested a total of 39,281,741 sequence variants (imputation info >0.80 and MAFIce > 0.02%, MAFUK > 0.01%, MAFDen > 0.1%, MAFNor > 0.2%), identified in the WGS, for association with AOM. The quantitative traits were transformed to a standard normal distribution. For the quantitative traits, the year of birth was included as a covariate in the analysis, with additional adjusting for the first 20 principal components in the UK, for population stratification. For each population, the quantitative traits were tested using a linear mixed model implemented in BOLT-LMM. For the meta-analysis, we used a fixed-effects inverse variance method based on effect estimates and s.e. from each population. For each study, we used linkage disequilibrium (LD) score regression to account for distribution inflation in the dataset due to cryptic relatedness and population stratification. Using a set of about 1.1 million sequence variants with available LD scores, we regressed the χ2 statistics from our GWAS scan against the LD score and used the intercept as a correction factor. The estimated correction factor for AOM, based on LD score regression, was 0.97 for the recessive model in the Icelandic sample, 1.01 in the UK, 1.01 in Denmark and 1.02 in Norway.

We report the effect estimates for POI and EM phenotypes against population controls and as a categorical trait among women who reported AOM (AOM < 40 versus AOM ≥ 40; AOM < 45 versus AOM ≥ 45. The effect estimates from the two methods do not differ significantly, and we have reached the same conclusion (Phet > 0.25).

Significance thresholds

We applied genome-wide significance thresholds corrected for multiple testing using an adjusted Bonferroni procedure weighted for variant classes and predicted functional impact. With 39,281,741 sequence variants being tested in the meta-analysis, the weights given in  were rescaled to control the family-wise error rate. The adjusted significance thresholds are 2.0 × 10−7 for variants with high impact (n = 9,910), 4.0 × 10−8 for variants with moderate impact (n = 202,465), 3.7 × 10−9 for low-impact variants (n = 3,244,032), 1.8 × 10−9 for other variants in DNase I hypersensitivity sites (n = 5,001,568) and 6.1 × 10−10 for all other variants (n =30,823,766).

Variant frequency map

UK Biobank participants were first grouped by birth country. We then defined regional ancestry groupings with the aim that the groups be representative of the region’s current population, be homogeneous by genetic ancestry and have at least 200 individuals (for accurate estimation of variant frequencies).

In some cases, we split off ancestry-based groupings representing distinct populations or unrepresentative migrant communities (for example, South Asian ancestry born in Africa and West Asia) to achieve homogeneous birthplace-based groupings. Groups depicted on the maps in Supplementary are those best representing the current demographic majority. If countries had fewer than 200 participant birthplaces, we merged them with neighboring countries with similar assessed ancestry profiles. Map geometries were obtained via R package maps and manipulated. The maps in Supplementary are sourced from Natural Earth (https://www.naturalearthdata.com/about/terms-of-use/).

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.

homozygosity, alleles, genetic disorders, recessive traits, consanguinity, inbreeding, isolated populations, homozygosity mapping, disease-causing genes, inherited disorders, genetic risk, population genetics, genetic variation, autosomal recessive, gene expression, genetic linkage, mutation, genome-wide studies, genetic counseling, targeted therapies,

#Homozygosity, #GeneticDisorders, #RecessiveTraits, #Consanguinity, #Inbreeding, #IsolatedPopulations, #HomozygosityMapping, #DiseaseCausingGenes, #InheritedDisorders, #GeneticRisk, #PopulationGenetics, #GeneticVariation, #AutosomalRecessive, #GeneExpression, #GeneticLinkage, #Mutation, #GenomeWideStudies, #GeneticCounseling, #TargetedTherapies, #GeneticResearch

International Conference on Genetics and Genomics of Diseases 




For Enquiries: genetics@healthcarek.com 

Get Connected Here 
--------------------------------- 
--------------------------------- 
facebook.com/profile.php?id=61555903296992 
tumblr.com/blog/dorita0211 

Wednesday, August 28, 2024

Researchers Discover New Genetic Disease

Hyderabad’s NIMS-CDFD researchers discover new genetic disease


NIMS and CDFD discover new genetic disease, ‘lethal serpinopathy’, expanding knowledge of genetic disorders

Hyderabad’s Nizam’s Institute of Medical Sciences (NIMS) and the Centre for DNA Fingerprinting & Diagnostics (CDFD) have identified a previously unknown genetic disease, expanding the understanding of the thousands of genetic disorders known to medical science.

Globally, around 6,000 to 7,000 genetic diseases have been documented and the advancements in next-generation sequencing technology are enabling scientists to uncover new genetic conditions. In this study, the teams from NIMS and CDFD collaborated to investigate a couple’s tragic case, where their infants suffered from severe fluid accumulation in body cavities and organs, leading to premature death.

The discovery came after Head of the Department of Medical Genetics at NIMS Dr. Shagun Aggarwal along with Additional Professor Dr. Prajnya Ranganath and their team conducted comprehensive tests on the second foetus which was medically terminated. By utilising advanced DNA sequencing techniques, they identified a mutation in the SERPINA11 gene. This critical finding led to the recognition of a new disease.

Further analysis by the CDFD team, led by Dr. Rashna Bhandari and Dr. Ashwin Dalal, revealed that the gene’s malfunction affects multiple tissues, causing a deadly condition they have named ‘lethal serpinopathy’.

The results of this study, which were published in the September 2024 issue of Clinical Genetics, highlight the emergence of this new genetic disease in a family with recurrent pregnancy loss.

Telangana’s Minister for Health C. Damodar Raja Narasimha lauded the researchers for this exceptional achievement. “This research places NIMS Hospital and the state of Telangana on the global map for medical advancements,” he stated.

genetic disease, DNA, inherited disorders, gene mutations, single-gene disorders, chromosomal disorders, complex disorders, cystic fibrosis, sickle cell anemia, Huntington's disease, genetic testing, genetic counseling, gene therapy, personalized medicine, hereditary conditions, genetic screening, prenatal diagnosis, genomics, genetic research, ethical considerations,

#GeneticDisease, #DNA, #InheritedDisorders, #GeneMutations, #SingleGeneDisorders, #ChromosomalDisorders, #ComplexDisorders, #CysticFibrosis, #SickleCellAnemia, #HuntingtonsDisease, #GeneticTesting, #GeneticCounseling, #GeneTherapy, #PersonalizedMedicine, #HereditaryConditions, #GeneticScreening, #PrenatalDiagnosis, #Genomics, #GeneticResearch, #EthicalConsiderations

International Conference on Genetics and Genomics of Diseases 




For Enquiries: genetics@healthcarek.com 

Get Connected Here 
--------------------------------- 
--------------------------------- 
facebook.com/profile.php?id=61555903296992 
tumblr.com/blog/dorita0211 

Tuesday, August 27, 2024

Reduce Genetic Risk of High Cholesterol

Fish Oil Supplementation May Reduce Genetic Risk of High Cholesterol



Fish oil and other omega-3 supplements may reduce the genetic risk of high total cholesterol and low-density lipoprotein (LDL) cholesterol, according to data published in The American Journal of Clinical Nutrition. The authors’ findings indicate that individuals who reported regular supplementation of fish oil had lower blood lipid levels, especially for total cholesterol, LDL cholesterol, and triglycerides.

Many patients turn to fish oil supplements, and they are frequently suggested by pharmacists as a method for increasing omega-3 fatty acid intake. Studies indicate that omega-3s have anti-inflammatory properties that may provide relief from mild inflammation or joint pain, and fish oil supplementation, specifically eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), may protect the heart by reducing triglyceride concentrations. It is important to know which patients may benefit from omega-3 supplements the most, the supplements’ proper dosage, and the benefits that can be expected.Pharmacists can be essential sources of education for patients, offering them guidance on safe use, proper dosage, and the reasonings behind these recommendations to ensure positive outcomes.

The authors examined whether fish oil supplementation modifies the association between genetically predicted and observed concentrations of total cholesterol, LDL cholesterol, high-density lipoprotein (HDL) cholesterol, and triglycerides. Using UK Biobank, they collected and assessed complete genetic and phenotypic data from 441,985 individuals aged 37 to 73. Participants provided blood samples, as well as sociodemographic, lifestyle, and medical records information via questionnaires at baseline.

Fish oil supplementation was determined through a touchscreen questionnaire that asked about consumption of specific supplements; however, it did not collect the dose, frequency, or duration of supplementation. The study included the primary outcomes of 4 serum lipid parameters including total cholesterol, LDL cholesterol, HDL cholesterol, and triglycerides. Using multivariable linear regression models, the authors assessed the modifying impacts of fish oil supplementation on the associations between the polygenic scores of the 4 lipids and their observed concentrations. These analyses were performed separately for each lipid across 4 ancestry groups: European, African, Central/South Asian, and East Asian.

According to the data, 31.8% of the participants reported habitual use of fish oil, of which 56% were female with a mean age of 59 years. Participants who consumed fish oil were more likely to be older, female, current alcohol drinkers, statin users, engaged in high physical activity, and have higher concentrations of total cholesterol, LDL cholesterol, and HDL cholesterol. Among other ancestry groups, 2,289 (34.8%) African, 1,942 (22.5%) Central/South Asian, and 845 (31.6%) East Asian participants used fish oil.

Overall, the data indicated that fish oil supplementation weakened the association between genetically predicted and observed circulating concentrations of total cholesterol, LDL cholesterol, and triglycerides while strengthening the corresponding association for HDL cholesterol. This suggests the ability of fish oil to lower specific lipid levels, thereby reducing their influence on genetic risk, as well as enhance the body’s ability to maintain higher HDL levels.

“Our study shows that considering lifestyles will improve genetic prediction,” Kaixiong Ye, BS, PhD, corresponding author of the study and an assistant professor of genetics in Franklin College of Arts and Sciences at the University of Georgia in Athens, said in an interview with UGA Today. “Our findings also support that fish oil supplements may counteract the genetic predisposition to high cholesterol.”

The study had some potential limitations, such as unreliable self-reported fish oil intake, lack of dosage information, and the inability to evaluate the effects of fish oil for an extended duration of use. Studies with larger sample sizes and accurate dose information are needed to further expand the findings. However, the initial data supports the hypothesis that genetic effects on blood lipid concentrations could be modified by habitual fish oil supplementation.

genetic risk, high cholesterol, cardiovascular disease, cholesterol levels, lifestyle modifications, healthy diet, saturated fats, physical activity, smoking cessation, weight management, omega-3 fatty acids, cholesterol control, regular screenings, family history, statins, cholesterol medication, heart health, prevention, dietary changes, lipid levels,

#GeneticRisk, #HighCholesterol, #CardiovascularHealth, #CholesterolLevels, #HealthyLifestyle, #DietAndExercise, #SaturatedFats, #PhysicalActivity, #SmokingCessation, #WeightManagement, #Omega3FattyAcids, #CholesterolControl, #RegularScreenings, #FamilyHistory, #Statins, #HeartHealth, #CholesterolPrevention, #DietaryChanges, #LipidLevels, #CholesterolManagement

International Conference on Genetics and Genomics of Diseases 




For Enquiries: genetics@healthcarek.com 

Get Connected Here 
--------------------------------- 
--------------------------------- 
facebook.com/profile.php?id=61555903296992 
tumblr.com/blog/dorita0211 

Monday, August 26, 2024

Genes for Improved Cancer Diagnosis

Paige Launches AI Tool that Screens 505 Genes for Improved Cancer Diagnosis



AI startup Paige launched a new AI product this week. The New York City-based company unveiled an AI-powered biomarker module that the company says can evaluate more than 505 genes and identify 1,228 molecular biomarkers from standard pathology slides.

The new product, called OmniScreen, analyzes digital images of cancer tissue slides that are stained using hematoxylin and eosin (H&E).

“These slides contain important visual patterns linked to genetic changes in cancer cells. By training our AI on three million of these images, it learns to recognize patterns and in turn detects genetic mutations or biomarkers that are often used to guide cancer treatment decisions,” explained Razik Yousfi, Paige’s CEO and chief technology officer.

In oncology, gene mutations are important in determining what type of cancer a patient has and what treatments will be most effective, he noted.

When people hear “breast cancer,” most think it is just one disease, he said. But the reality is that it can be one of many different types, all of which require unique approaches to treatment, Yousfi pointed out.

“Omniscreen could be valuable in detecting these subtypes across many different types of cancer. Whereas other methods, such as gene sequencing, are expensive and time consuming, Omniscreen can be done quickly and more cost effectively directly from the tissue sample using AI,” he declared.

While Omniscreen is currently only available for research, it allows researchers to better characterize disease and develop improved therapies for patients, Yousfi added.

In his view, Omniscreen is cheaper and faster than other methods for detecting gene mutations and alterations. Rather than completely replacing these other methods, Yousfi believes there is an opportunity to decrease the costs associated with gene analysis for precision medicine.

“By screening patients quickly using Omniscreen, we could identify quickly which patients are negative for certain mutations and use these to select patients for more complex, expensive testing. In research, Omniscreen gives cancer researchers a convenient, fast and cheaper approach to studying cancer and building better and more effective markers to improve cancer care,” he remarked.

This applies to cancer research conducted at hospitals, universities and pharmaceutical facilities for the development of new cancer drugs, Yousfi noted.

X-chromosome inactivation, telomere length, recurrent pregnancy loss, RPL, miscarriage, skewed XCI, cellular aging, genetic factors, pregnancy outcomes, female fertility, chromosomal abnormalities, maternal age, reproductive health, embryonic development, immune response, telomere shortening, fertility treatment, genetic screening, epigenetics, pregnancy complications,

#XChromosomeInactivation, #TelomereLength, #RecurrentPregnancyLoss, #RPL, #Miscarriage, #SkewedXCI, #CellularAging, #GeneticFactors, #PregnancyOutcomes, #FemaleFertility, #ChromosomalAbnormalities, #MaternalAge, #ReproductiveHealth, #EmbryonicDevelopment, #ImmuneResponse, #TelomereShortening, #FertilityTreatment, #GeneticScreening, #Epigenetics, #PregnancyComplications


International Conference on Genetics and Genomics of Diseases 




For Enquiries: genetics@healthcarek.com 

Get Connected Here 
--------------------------------- 
--------------------------------- 
facebook.com/profile.php?id=61555903296992 
tumblr.com/blog/dorita0211 

Friday, August 23, 2024

Genetic Study Identifies Myeloma

12-year genetic study identifies unique types of multiple myeloma


An unprecedented effort to sequence the genome, exome and RNA in tumors from patients with multiple myeloma defines distinct subtypes of the disease, according to an international team of scientists led by researchers from the Translational Genomics Research Institute (TGen), part of the City of Hope.

The findings of the 12-year observational study provide a clearer picture of the genetic changes that may be important in each subtype of this cancer of our antibody producing plasma cells, which is treatable but incurable. This information could help guide more personalized treatments in the future, the researchers write in Nature Genetics.

The scientists identified the genetic basis for a high-risk patient population, named "PR," that is associated with a median survival of under two years compared to the average survival in the study that exceeds eight years. About 25% of patients transitioned to the PR subtype during the study, and those who transitioned to PR had worse outcomes, with a median survival of 88 days after the transition.

"We've known about this subtype for over a decade and now replicated that this is a high-risk group of patients with the therapies used today. Clearly, these are patients who we need to get on clinical trials because the current drugs just don't work for them," said Jonathan Keats, Ph.D., assistant professor, director of Bioinformatics and Collaborative Sequencing Center at TGen, and a senior author on the paper.

"What we clearly showed is that there is a genetic basis for people being in the PR subgroup."

The study also "helps to highlight that late-stage disease is not like early-stage disease, and in an era where we're moving into early detection, drugs that work in the early stage may not be the drugs that work at the very late stage," Keats added.

The Multiple Myeloma Research Foundation (MMRF) Relating Clinical Outcomes in Multiple Myeloma to Personal Assessment of Genetic Profile (CoMMpass) study included 1,143 patients from 84 clinical sites located in the United States, Canada, Spain and Italy, enrolled between 2011 and 2015 and followed patients for at least eight years after diagnosis.

It is the largest single sequencing study of multiple myeloma patients to date, based on the number of patients and the number of sequencing assays performed.

"There's also clinical data on these patients collected every three months throughout their entire disease course, not just when they're genetically profiled, which is an absolute treasure trove," said Keats.

Two main groups of multiple myeloma patients have been known for some time: a hyperdiploid group with extra copies of chromosomes in tumor cells, and a non-hyperdiploid group with chromosomal rearrangements. The approach used by the current team of researchers helped them identify different subgroups within these broad categories.

"A lot of studies have looked at one method—they look at copy number state, or another one looks at if there are mutations—very few actually integrate the two to really understand how an individual gene is functioning in that patient," Keats noted. "For the field, we've provided a very robust list of candidate genes that are important in the disease."

Keats said the findings could change how doctors speak with their patients about crafting a treatment plan. Although the common belief is that hyperdiploid patients have more favorable overall survival, for instance, the researchers found that "hyperdiploid" encompasses five different groups of patients—one of which has significantly worse outcomes and could benefit from aggressive therapy.

One of the next challenges is to match medicines with specific genetic features, Keats said. "We've definitely answered the question of what different types there are, but not which drugs they best respond to."

TGen is building on this pioneering work to bring personalized medicine to multiple myeloma patients by launching clinical-grade whole genome sequencing in the TGen Clinical Lab. This test will provide patients and their care team with a complete picture of the genetics of each tumor along with prognostic information and therapeutic recommendations within 48 hours.

genetic study, myeloma, multiple myeloma, plasma cells, genetic mutations, blood cancer, therapeutic targets, genetic predisposition, chromosomal abnormalities, gene expression, personalized treatment, patient outcomes, genetic markers, myeloma progression, cancer research, targeted therapy, oncogenes, tumor suppressor genes, genomics, hematologic malignancies,

#GeneticStudy, #Myeloma, #MultipleMyeloma, #PlasmaCells, #GeneticMutations, #BloodCancer, #TherapeuticTargets, #GeneticPredisposition, #ChromosomalAbnormalities, #GeneExpression, #PersonalizedTreatment, #PatientOutcomes, #GeneticMarkers, #CancerResearch, #TargetedTherapy, #Oncogenes, #TumorSuppressorGenes, #Genomics, #HematologicMalignancies, #CancerGenetics

International Conference on Genetics and Genomics of Diseases 




For Enquiries: genetics@healthcarek.com 

Get Connected Here 
--------------------------------- 
--------------------------------- 
facebook.com/profile.php?id=61555903296992 
tumblr.com/blog/dorita0211 


Wednesday, August 21, 2024

Genes to cope with Infections

The bacteria that write new genes to cope with infections



Amid the unprecedented challenges presented by the COVID-19 pandemic, a once obscure enzyme found itself in the spotlight: reverse transcriptase. As laboratories worldwide rushed to develop reliable diagnostic tests, techniques using the enzyme became the gold standard to detect the SARS-2 virus, and a cornerstone of molecular diagnostics. This remarkable enzyme didn’t only facilitate rapid and accurate testing; along with another powerful approach — genome-sequencing — it also helped track the virus’s spread, paving the way for surveillance, better public healthcare, and vaccine development.

The discovery of reverse transcriptase is a story unto itself. Researchers in the labs of Howard Temin and David Baltimore independently discovered it and published their findings in back-to-back articles in the journal Nature in 1970. In his paper, Dr. Baltimore suggested that in the vesicular stomatitis virus, a protein called RNA polymerase was involved in reverse-translating RNA to DNA.

A molecular biology revolution

The discovery was transformative. The prevailing belief at the time was that in all living beings, hereditary information flowed only from DNA to RNA and from RNA to protein (a.k.a. the ‘Central Dogma’). The discoveries of Drs. Temin and Baltimore et al. showed information could flow the other way, too, with RNA giving ‘rise’ to DNA. The name “reverse transcriptase” was however coined by the editor of Nature, in an article discussing the significant advance in an accompanying column.

The discovery’s impact was also immediate. The ability of cells to create DNA copies from RNA revolutionised research methods in molecular biology, where researchers could reverse-transcribe messenger RNAs to pieces of DNA, clone that DNA into bacterial vectors, and study the function of the corresponding genes. In diagnostics, clinicians used reverse transcriptase to convert RNA to DNA and thus estimate the amount of viral material in a given sample. This technique quickly found wide application and use in the study of RNA viruses, including hepatitis B and the human immunodeficiency virus (HIV).

Indeed, the discovery of reverse transcriptase had a significant effect on the management and treatment of HIV infections, including Acquired Immunodeficiency Syndrome (AIDS), in the 1980s. A generation of antiviral agents that specifically targeted the reverse transcriptase enzyme helped convert an otherwise deadly disease to one that could be managed, translating to improving the long-term outcomes and survival of people living with AIDS.

Subsequent studies of the reverse transcriptase enzyme since the 1970s led to mechanistic insights into how viruses use this enzyme to replicate, as well.

Retroelements in the human genome

Reverse transcriptases also had a significant role in shaping the human genome.

The human genome is interspersed in many places with sequences, called elements, that appear to have originated from retroviruses. Thus researchers call them retroelements. Evolutionary biologists believe these retroelements to have been transferred horizontally during the course of millions of years of evolution. (Horizontal gene transfer refers to genes ‘jumping’ between organisms rather than from parent to offspring.) And until recently, researchers also considered them to be “junk” elements: they were repeated through the genome and they seemingly did not confer any function to the human organism.

However, recent evidence has suggested that these retroelements could really have had a profound impact on human biology and evolution, and that they play important roles in a variety of physiological processes.

In a recent paper in the journal Nature Communications, researchers extensively studied the expression of genes in different parts of the human brain from post-mortem brain samples. They reported that the expression of more than a thousand human endogenous retroviruses — a major class of retroelements in the human genome — could be associated with a risk of neuropsychiatric diseases in humans.

Retroelements in the human genome and bacterial reverse transcriptases have a common evolutionary history as well as share functional mechanisms. Bacterial reverse transcriptases — believed to be the precursors of their eukaryotic counterparts — exhibit analogous mechanisms.

The discovery of reverse transcriptase activity across the different domains of life underscores the enzyme’s fundamental role in both prokaryotic and eukaryotic systems as well as a remarkable evolutionary continuity and functional versatility.

Writing genes using reverse transcriptase

Researchers widely believed that bacterial reverse transcriptases were the precursors of their eukaryotic counterparts. They discovered the first reverse transcriptase in bacteria in 1989, with papers published back to back in the journals Science and Cell. In bacteria, as in the case of humans, retroelements are categorised as belonging to three broad groups: the Group II introns, the retrons, and the diversity generating retroelements.

In a preprint paper uploaded to the bioRxiv preprint server on May 8, researchers at Columbia University in New York, led by Stephen Tang and Samuel Sternberg, suggested that when the bacteria Klebsiella pneumoniae is infected by bacteriophages — viruses that infect bacteria — they use a non-coding RNA with specific motifs (or structures) that could bind to reverse transcriptase and instruct cells to create DNA. This DNA copy has multiple copies of a gene that can create a specific protein.

The researchers dubbed this protein ‘Neo’ for “never-ending open-reading frame”. It could place the bacterial cell in a state of suspended animation, blocking its replication, and thus stalling the replication of the invading bacteriophage as well. Thus, the infection is stopped in its tracks.

Recent discoveries — including the role of reverse transcriptase in bacterial defence against bacteriophages — hint at the potential of innovative applications in biotechnology and medicine, especially in the context of emerging antimicrobial resistance, the ability of disease-causing microbes to resist the effects of substances designed to incapacitate or kill them. Further exploring reverse transcriptases could also reveal novel mechanisms of genetic evolution and viral resistance, potentially leading to new therapeutic strategies and biotechnological tools.

International Conference on Genetics and Genomics of Diseases 




For Enquiries: genetics@healthcarek.com 

Get Connected Here 
--------------------------------- 
--------------------------------- 
facebook.com/profile.php?id=61555903296992 
tumblr.com/blog/dorita0211 

Sunday, August 18, 2024

Genetic Causes of Colorectal Cancer

Study reveals previously unknown genetic causes of colorectal cancer


Cancers develop partly through genetic abnormalities within cells of the body. Colorectal cancer is a major cause of death worldwide, but we don’t yet have a full understanding of the genetic changes that cause it to grow. New research - published today in Nature - delivers an unprecedented view of the genetic landscape of CRC and its responses to treatment.

Utilising data from 2,023 bowel cancers from the 100,000 Genomes Project led by Genomics England and NHS England, the research team has identified new gene faults that lead to CRC. They’ve also uncovered new CRC cancer sub-groups (categories of cancer with specific genetic characteristics that affect how cancer behaves and responds to treatment). These findings offer profound insights into the disease's development and potential treatment strategies.

Key Findings of the Study:

• Identification of Over 250 Key Genes: The study has pinpointed more than 250 genes that play a crucial role in CRC, the great majority of which have not been previously linked to CRC or other cancers, expanding our understanding of how CRC develops.
• New Sub-Groups of CRC: Four novel, common sub-groups of CRC have been discovered based on genetic features. In addition, several rare CRC sub-groups have been identified and characterised. These groups have different patient outcomes and may respond differently to therapy.
• Genetic Mutation Causes: The research reveals a variety of genetic changes across different regions of the colorectum, highlighting differences in CRC causes between individuals. For example, a process has been found that is more active in younger CRC patients’ cancers; the cause is unknown, but might be linked to diet and smoking.
• New Treatment Pathways: Many identified mutations could potentially be targeted with existing treatments currently used across other cancers.

Commenting on the findings, co-lead researcher, Ian Tomlinson, Professor of Cancer Genetics at the University of Oxford, said: 'Our findings represent a significant advancement in understanding colorectal cancer. By better understanding the genetic changes in CRC, we can better predict patient outcomes and identify new treatment strategies, quite possibly including the use of anti-cancer drugs that are not currently used for CRC.'

The research provides a vital resource for the scientific community and a promising foundation for future studies. The results from the study are available to other researchers, who are invited to build on the data by undertaking more focussed projects based on the CRC genome.

Co-lead researcher, Professor Richard Houlston, Professor of Cancer Genomics at The Institute of Cancer Research, London, said: 'This research is a great insight into the biology of colorectal cancer, uncovering the clues as to how it develops, grows, and responds to treatments. I look forward to seeing future studies use these findings to develop tailored treatments for people with colorectal cancer, based on their genetics.'

Co-lead researcher, Professor David Wedge, Professor of Cancer Genomics and Data Science at the University of Manchester, said: 'This is the first really large study to come out of the 100,000 Genomes Project led by Genomics England and NHS England. In the coming months and years, I expect it to be followed by many more studies of different types of cancer as well as combined studies across all types of cancer, fuelled by the fantastic data resource provided by Genomics England.'

Dr Henry Wood, Lecturer in Translational Bioinformatics from Pathology in the University of Leeds’ School of Medicine, said: 'This study is the first to provide in-depth, whole-genome sequencing and characterisation of the microbiome - the community of bacteria and viruses that live in the gut - in a large number of cases of bowel cancer. This means that we are now in a position to investigate the importance of the microbiome in the development of these cancers, and whether we can change it to influence the tumour and improve patient outcomes.'

The paper, 'The genomic landscape of 2,023 colorectal cancers', is published in Nature.

β-Sn texture, micro solder joints, heredity, multi-reflow process, cooling rate, reflow temperature, grain orientation, magnetic field, electric field, rare earth elements, nanoparticles, substrate type, joint geometry, solder composition, grain boundary engineering, thermal cycling, defects, mechanical properties, microelectronics, durability,

#βSnTexture, #MicroSolderJoints, #Heredity, #MultiReflow, #CoolingRate, #ReflowTemperature, #GrainOrientation, #MagneticField, #ElectricField, #RareEarthElements, #Nanoparticles, #SubstrateType, #JointGeometry, #SolderComposition, #GrainBoundaryEngineering, #ThermalCycling, #Defects, #MechanicalProperties, #Microelectronics, #Durability


International Conference on Genetics and Genomics of Diseases 




For Enquiries: genetics@healthcarek.com 

Get Connected Here 
--------------------------------- 
--------------------------------- 
facebook.com/profile.php?id=61555903296992 
tumblr.com/blog/dorita0211 
 


Friday, August 16, 2024

Genes Linked to Relapse

Scientists identify genes linked to relapse in the most common form of childhood leukemia

Researchers identified genetic predictors of relapse in standard risk B-cell acute lymphoblastic leukemia highlighting the role of genetic testing in tailoring therapy.





Scientists from St. Jude Children’s Research Hospital, Seattle Children’s and the Children’s Oncology Group (COG) have identified novel genetic variations that influence relapse risk in children with standard risk B-cell acute lymphoblastic leukemia (SR B-ALL), the most common childhood cancer. The identification of genomic predictors of relapse in SR B-ALL provides a basis for improved diagnosis, precise tailoring of treatment intensity and potentially the development of novel treatment approaches. The study was published today in the Journal of Clinical Oncology.

Standard risk ALL has an excellent prognosis, with remission rates over 90%. However, around 15% of patients who achieve remission later experience a relapse. Previous studies examining genomic alterations to predict relapse risk have primarily focused on high-risk ALL subgroups. SR B-ALL represents a larger group of patients and accounts for approximately half of children with ALL that relapse. This study is one of the first to systematically examine genetic factors on a large scale that influence relapse risk in SR B-ALL.

“ALL, as the most common childhood cancer, is a great success story with over 90% of children cured. But there remains a population of children whose disease is not fully cured, and we've not completely understood why that's the case,” said co-senior author, Charles Mullighan, MBBS (Hons), MSc, MD, St. Jude Comprehensive Cancer Center Deputy Director and Department of Pathology member. “This study focused on that group of poorly understood cases, where we know less about the features that influence the risk of treatment not working and the disease coming back.”
Genomic profiling identifies specific genetic alterations associated with cancer susceptibility, relapse risk and how tumors respond to therapeutics. These studies allow scientists and clinicians to predict how patients are likely to respond to therapy, providing insights that shape the treatment of childhood ALL. Results from this collaborative study demonstrate the importance of genomic profiling in accurately determining patient risk in B-ALL, in conjunction with traditional criteria.

“We are planning to reduce conventional therapies in the future for children with ALL because we know that many patients can be cured with less therapy,” explained co-senior author, Mignon Loh, MD, leader of Seattle Children’s Cancer and Blood Disorders Center, COG ALL Committee chair emeritus, Seattle Children’s Ben Towne Center for Childhood Cancer Research director, and head of Seattle Children’s Division of Pediatric Hematology, Oncology, Bone Marrow Transplant and Cellular Therapy. “We want to make sure we accurately identify those children, and because of the special design of the study, this project allowed us to do just that.”

The scientists conducted genome and transcriptome sequencing on both SR B-ALL samples that relapsed and samples that remained in complete remission in a one:two ratio. They found that ALL subtypes, genetic alterations and patterns of aneuploidy (extra or missing chromosomes) were associated with the risk of relapse and time to relapse. Some B-ALL subtypes, such as hyperdiploid and ETV6::RUNX1 ALL, had a low frequency of relapse, but others including PAX5-altered, TCF3/4::HLF, ETV6::RUNX1-like and BCR::ABL1-like were associated with an increased risk of relapse. Notably, the specific type of genetic changes within those B-ALL subtypes further influenced the risk of relapse. This work demonstrated that genetic variations and cancer subtypes influence relapse risk in SR B-ALL, and patients classified as standard-risk can have tumors with high-risk features.

“Whole genome sequencing was important to accurately and comprehensively identify these changes, and they could not all have been identified without it,” explained Mullighan. “Children with SR ALL should have their tumor cell genome sequenced upon their initial diagnosis to identify if their tumor cells have these high-risk features, so that their initial therapy intensity can be increased.”

“Beyond conventional therapies, this information could also be used to develop and explore novel, personalized treatment strategies,” added Loh.

childhood leukemia, cancer, blood and bone marrow, diagnosis, treatment, survival rates, genetic mutations, environmental factors, family history, chemotherapy, radiation therapy, stem cell transplants, child, family, support systems, targeted therapies, immunotherapy, long-term follow-up, recurrence, side effects.


International Conference on Genetics and Genomics of Diseases 




For Enquiries: genetics@healthcarek.com 

Get Connected Here 
--------------------------------- 
--------------------------------- 
facebook.com/profile.php?id=61555903296992 
tumblr.com/blog/dorita0211 

Genetic Insights Into Alopecia Areata

New Genetic Insights Into Alopecia Areata Alopecia areata has a genetic relationship with trace elements, serum metabolites, and inflammato...