header logo image


Page 34«..1020..33343536..40..»

Archive for the ‘Genetics’ Category

Why is your dog so aggressive? The answer is largely in its DNA – CTV News

Thursday, October 10th, 2019

TORONTO -- Dog owners, take note: If you're fed up with your pet's personality, there may not be much you can do about it.

A new study confirms something many people have long suspected, namely that canine behaviours are largely based on the animals' genetic makeup.

Researchers at four American universities analyzed genetic information and behaviour logs for 14,000 dogs from 101 breeds and determined that 60 to 70 per cent of the differences in personality traits between breeds can be explained by the genes they inherited from their parents.

Genetics were found to contribute most strongly to traits such as trainability, aggression toward strangers and attention-seeking. This fits with the idea that these were some of the most or least sought-after attributes during the early stages of breeding, making them essentially hard-wired into the breeds' DNA.

Because most of these breeds have only been in existence for 300 years or less, they have not had enough time to develop the sort of genetic diversity seen in species with longer histories.

This helps explain why greyhounds and Siberian huskies are some of the least aggressive dogs toward strangers, for example, or why Yorkshire terriers and toy poodles have trouble coping with separation.

The researchers were also able to find 131 genetic variations that appear to be linked to breed behaviour. They say this suggests that no individual gene is solely responsible for dogs' personality traits, and instead the relationships between genes come into play just as they do with humans.

Even some of the genes located by the researchers as being associated with dogs' neurological development mirror similar genes that have been found in humans.

"Dogs exhibit striking parallels to traits in humans," the study reads.

"For example, common genetic mechanisms contribute to individual differences in social behaviour in dogs and humans."

This suggests that further study of dog genetics could help illustrate how personality traits develop in humans, and which traits are more or less likely to be inherited, the researchers said.

The study was published Oct. 1 in the Proceedings of the Royal Society B.

Read the original here:
Why is your dog so aggressive? The answer is largely in its DNA - CTV News

Read More...

Blue Devil of the Week: Searching for Answers in the Genetic Code – Duke Today

Thursday, October 10th, 2019

Name: Sue Jinks-Robertson

Position: Professor, Vice Chair and Director of the Cell and Molecular Biology Program

Years at Duke: 13

What she does at Duke: Jinks-Robertson has many duties in the Department of Molecular Genetics and Microbiology. She oversees the Cell and Molecular Biology Training Program which features around 100 graduate students and involves around 130 faculty members. She also co-directs the Cancer Genetics and Genomics program at Duke Cancer Institute.

But Jinks-Robertson is most at home in her lab, where she studies the genetic makeup of yeast.

Her team examines yeast DNA, looking for the factors behind mutations or changes in sequence. This research is important is because the DNA of yeast is essentially the same that found in many other organisms, including humans.

If we understand how this works in yeast, we can get information about what can go wrong in humans, Jinks-Robertson said.

The research is of great value in the fight against cancer, since it can occur when cells with genetic flaws multiply. Therapies that help identify and repair these flaws can be critical in battling the disease.

The basis for the therapies comes from the very basic work done in the trenches with an organism like yeast, Jinks-Robertson said.

What she loves about Duke: When she arrived at Duke after two decades on the faculty at Emory University, Jinks-Robertson was struck by the affection and loyalty her new colleagues both staff and faculty had for the university.

Soon, she too came to have a similar relationship with the university. She said its hard to pin down one specific reason for her connection with Duke, but she knows its there.

I dont know if its some of the physical structures around, like the Gardens, the Chapel, theres a central focus, of course theres basketball, its hard to put your finger on what it is, Jinks-Robertson said. Its just a very nice place to work. You feel connected to something bigger than yourself.

A memorable day at work: This spring, Jinks-Robertson was preparing for a major grant application when she got a call from colleague Thomas Petes with exciting news.

Petes, the Minnie Gellar Professor of Molecular Genetics and Microbiology, told her that shed been elected to the National Academy of Sciences, a 156-year old organization comprised of the nations top scientific minds.

It was a big surprise, Jinks-Robertson said. If youre a scientist, at least in this country, its a great recognition.

Jinks-Robertson was one of two Duke scientists elected to the 2019 class. Susan Alberts, the Robert F. Durden Professor of Biology, also earned election to the academy.

The nicest part of it was that I was hit with a flood of emails and phone calls, it was really wonderful, Jinks-Robertson said.

Special object/memorabilia in her workspace: On a shelf in her office, Jinks-Robertson has a collection of gifts given to her by former students who came to Duke from other countries. Theres a statue of Saraswati, the Hindu goddess of learning, which was given to her by a student from India. Theres also a vase from Russia, a screen from China and small house from the Philippines.

I like to think it shows I was successful in training the next generation, Jinks-Robertson said.

First ever job: A native of Panama City, Florida, Jinks-Robertson grew up around the water. As a child, she swam and water skied often. After she graduated from high school, she spent the next two summers working as a mermaid at Gulf World Marine Park, a popular attraction in Panama City.

We didnt have tails, but we had on scuba tanks and dove in saltwater tanks and fed the fish as people watched, Jinks-Robertson said of the mermaid role, which also had her swimming with dolphins and sea lions. It was fun.

Best advice received: In 1986, when she was finishing up her time as a post-doctoral researcher at the University of Chicago working with Thomas Petes, who many years later helped bring her to Duke, Jinks-Robertson began looking for faculty positions.

I was pregnant with my first child and I was concerned about that, Jinks-Robinson said. His advice was, If its a problem, its not a place you want to be. He really put me at ease and told me I shouldnt worry about that. Hes always been very supportive of women in science.

Something most people dont know about her: Much of Jinks-Robertsons work is done with a sleeping labradoodle at her feet. With soft, curly light brown hair, Gracie is Jinks-Robertsons constant companion, often accompanying her to work.

Its calming, Jinks-Robertson said. I walk her every day, so it gets me moving and out of my chair. Shes good company.

Is there a colleague at Duke who has an intriguing job or goes above and beyond to make a difference? Nominate that person for Blue Devil of the Week.

Read the original here:
Blue Devil of the Week: Searching for Answers in the Genetic Code - Duke Today

Read More...

Advances in the Diagnosis of Type 1 von Willebrand Disease: Genetic Testing – Hematology Advisor

Thursday, October 10th, 2019

Von Willebrand disease (VWD) is the most common hereditary bleeding disorder but one of the most difficult to diagnose, especially type 1 VWD. Recurrent challenges include the need to complete several assays of von Willebrand factor (VWF) activity and lack of consensus surrounding the acceptable standard for diagnosis. Consequently, improving current diagnostic techniques, as well as implementing new methods, is essential to ensure patients are provided optimal care.

In a review article published in Current Opinion in Hematology, Veronica H Flood, MD, of the department of pediatrics at the Medical College of Wisconsin in Milwaukee, and colleagues summarized the current literature surrounding the diagnosis of type 1 VWD. They also reviewed new advances in genetic testing for VWF, which could serve as a potential alternative to conventional laboratory methods.

Overview of Genetic Dysfunction

In contrast to type 2 VWD, type 1 VWD may include genetic defects in the coding region of the VWF gene. These mutations vary from insertions and deletions to point mutations that produce missense or nonsense mutations. With conventional sequencing methods, insertions and deletions can be missed, which has historically precluded the clinical use of genetic-based diagnostic techniques. These limitations are not typically seen in type 2 VWD as genetic defects are usually present in the DNA region specific to the impacted functional region.

Because of the high degree of polymorphism seen in the VWF gene, entire genome or exome sequencing may be required for diagnosis; in other instances, the VWF gene may be analyzed directly if a particular coagulation defect is suspected. In type 1 VWD, certain high frequency variants have been linked to disease etiology, but recent data have highlighted the potential role of novel variants in type 1 VWD. The high degree of variability seen in the VWF gene is certainly a key contributor to the disease phenotype, but not all defects will ultimately lead to VWD.

Modifier Genes and Diagnosis

In addition to defects in VWF, several genes independent of the VWF locus have been shown to affect VWF levels. The most described modifier gene is ABO, though others such as CLEC4M, STAB2, and STXBP5 also exist. Blood group O levels of less than 50 IU/dL are routinely used to establish a diagnosis of VWD, but some individuals with blood type O also meet this criteria despite being healthy. Some experts have proposed that low VWF may be more suitably described as a risk factor for bleeding instead of as the basis for bleeding.

Originally posted here:
Advances in the Diagnosis of Type 1 von Willebrand Disease: Genetic Testing - Hematology Advisor

Read More...

Datar Cancer Genetics Announces Positive Results With 42.9% Objective Response Rate, and 90.5% Disease Control Rate in Heavily Pre-treated Patients…

Thursday, October 10th, 2019

LONDON, MUMBAI, India and BAYREUTH, Germany, Oct. 9, 2019 /PRNewswire/ --

- RESILIENT Protocol captures Encyclopedic information from tumors and uses Artificial Intelligence (AI) to optimize treatments;

- Objective Response and Disease Control Comparable to or better than most Immunotherapy options;

- Unique Ultra-personalised combination of drugs already approved for cancer;

- RESILIENT Protocol commercially launched

Datar Cancer Genetics, a cancer research company, today announced positive data from the phase II/III RESILIENT trial intended to validate clinical benefit for cancer patients who have run out of treatment options under the present standard of care guidelines. The study achieved its primary and secondary end points of Objective Response Rate, Progression Free Survival and Disease Control Rate respectively.

Drug resistant cancers present a serious clinical challenge since there are virtually no treatments available and the prognosis is invariably poor. As a large proportion of all patients with advanced cancers ultimately progress towards this phase, life extending treatment options for these patients are urgently required.

The RESILIENT Protocol is designed to analyse all functional layers of a cancer cell i.e., DNA, RNA, proteins and germline genetics as also the chemoresistance/sensitivity of live tumour cells. This data is integrated through an Artificial Intelligence algorithm to derive treatment regimens which are most efficacious and yet show the least risk of toxicity.

RESILIENT is the world's first and only prospective Precision Oncology trial where drug combinations were selected on multi-analyte integration. Most prior trials based on a single molecular alteration for drug selection had dismal outcomes. 143 patients started treatment and 126 patients were evaluable as per study criteria. All patients underwent PET-CT and Brain MRI scans prior to start of treatment to establish extent of disease. Treatment response was determined by follow-up PET-CT and MRI scans.

In the majority (90.5%) of patients, further spread of cancer was effectively halted. In 42.9% of patients, treatments also led to a significant decrease in the extent of cancer. Remarkably, among the 12 patients where disease progressed, no new metastases were reported in 9 patients. There were no serious treatment related adverse events or deaths. Most patients reported stable to improved quality of life.

The data of the RESILIENT Trial is published in the peer revived oncology journal 'Oncotarget' (https://doi.org/10.18632/oncotarget.27188)

"The RESILIENT trial marks a watershed moment for molecular oncology as it unequivocally proves that patients who have failed even 2 to 3 lines of treatment can benefit from already approved drugs if comprehensive tumour analysis is used to guide treatments. Patients in the United Kingdom and all over the world have much to gain from the outcome of this trial," said Dr. Tim Crook, Medical Oncologist at the St. Luke's Cancer Centre, Royal Surrey County Hospital, Guildford, UK, who is one of the authors of the publication.

Datar is a leading cancer research corporation specialising in tumour analysis for better diagnosis, treatment decisions, and management of cancer. Datar's research initiatives are poised to bring about meaningful, patient-friendly and practice changing advancements in cancer treatment. Datar is also pursuing Adoptive Cell Immunotherapy for Multiple Solid Organ Cancers.

For more information please contact:

Dr. Vineet Datta - drvineetdatta@datarpgx.com

Dr Stefan Schuster - drstefanschuster@datarpgx.com

Datar Cancer Genetics info@datarpgx.org

Related Links

https://doi.org/10.18632/oncotarget.27188

https://doi.org/10.1093/annonc/mdz268.061

Encyclopedic Tumor Analysis – Exacta

SOURCE Datar Cancer Genetics Ltd

Read the rest here:
Datar Cancer Genetics Announces Positive Results With 42.9% Objective Response Rate, and 90.5% Disease Control Rate in Heavily Pre-treated Patients...

Read More...

Genetics of Diabetes | ADA

Thursday, October 3rd, 2019

You've probably wondered how you developed diabetes. You may worry that your children will develop it too.

Unlike some traits, diabetes does not seem to be inherited in a simple pattern. Yet clearly, some people are born more likely to develop diabetes than others.

Type 1 andtype 2 diabeteshave different causes. Yet two factors are important in both. You inherit a predisposition to the disease, then something in your environment triggers it.

Genes alone are not enough. One proof of this is identical twins. Identical twins have identical genes. Yet when one twin hastype 1 diabetes, the other gets the disease at most only half the time. When one twin has type 2 diabetes, the other's risk is at most 3 in 4.

In most cases of type 1 diabetes, people need to inherit risk factors from both parents. We think these factors must be more common in whites because whites have the highest rate of type 1 diabetes.

Because most people who are at risk do not get diabetes, researchers want to find out what the environmental triggers are. One trigger might be related to cold weather. Type 1 diabetes develops more often in winter than summer and is more common in places with cold climates. Another trigger might be viruses. Perhaps a virus that has only mild effects on most people triggers type 1 diabetes in others.

Early diet may also play a role. Type 1 diabetes is less common in people who were breastfed and in those who first ate solid foods at laterages.

In many people, the development of type 1 diabetes seems to take many years. In experiments that followed relatives of people with type 1 diabetes, researchers found that most of those who later got diabetes had certain autoantibodies in their blood for years before. (Antibodies are proteins that destroy bacteria or viruses. Autoantibodies areantibodies'gone bad' that attack the body's own tissues.)

If you are a man with type 1 diabetes, the odds of your child developing diabetes are 1 in 17. If you are a woman with type 1 diabetes and your child was born before you were 25, your child's risk is 1 in 25; if your child was born after you turned 25, your child's risk is 1 in 100.

Your child's risk is doubled if you developed diabetes before age 11. If both you and your partner have type 1 diabetes, the risk is between 1 in 10 and 1 in 4.

There is an exception to these numbers. About 1 in every 7 people with type 1 diabetes has a condition called type 2 polyglandular autoimmune syndrome. In addition to having diabetes, these people also have thyroid disease and a poorly working adrenalgland. Some also have otherimmune systemdisorders. If you have this syndrome, your child's risk of getting the syndromeincluding type 1 diabetesis 1 in 2.

Researchers are learning how to predict a person's odds of getting diabetes. For example, most whites with type 1 diabetes have genes called HLA-DR3 or HLA-DR4. If you and your child are white and share these genes, your child's risk is higher. (Suspect genes in other ethnic groups are less well studied. The HLA-DR7 gene may put African Americans at risk, and the HLA-DR9 gene may put Japanese at risk.)

Other tests can also make your child's risk clearer. A special test that tells how the body responds toglucosecan tell which school-aged children are most at risk.

Another more expensive test can be done for children who have siblings with type 1 diabetes. This test measures antibodies toinsulin, to islet cells in thepancreas, or to anenzymecalled glutamic acid decarboxylase. High levels can indicate that a child has a higher risk of developing type 1 diabetes.

Type 2 diabetes has a stronger link to family history and lineage than type 1, and studies of twins have shown that genetics play a very strong role in the development of type 2 diabetes.

Yet it also depends on environmental factors.Lifestyle also influences the development of type 2 diabetes.Obesitytends to run in families, and families tend to have similar eating and exercise habits.

If you have a family history of type 2 diabetes, it may be difficult to figure out whether your diabetes is due to lifestyle factors or genetic susceptibility. Most likely it is due to both. However, dont lose heart. Studies show that it is possible to delay or prevent type 2 diabetes by exercising and losing weight.

Have you recently been diagnosed with type 2 diabetes?Join our free Living With Type 2 Diabetes program and get the information and support you need to live well with diabetes.

Type 2 diabetes runs in families. In part, this tendency is due to children learning bad habitseating a poor diet, not exercisingfrom their parents. But there is also a genetic basis.

If you would like to learn more about the genetics of all forms of diabetes, the National Institutes of Health has publishedThe Genetic Landscape of Diabetes. This free online book provides an overview of the current knowledge about the genetics of type 1 and type 2 diabetes, as well other less common forms of diabetes. The book is written for healthcare professionals and for people with diabetes interested in learning more about the disease.

Continue reading here:
Genetics of Diabetes | ADA

Read More...

Ph.D. in Genetics at Texas A&M University

Thursday, October 3rd, 2019

Please save the date for oursecond Career Club of the fall semester!We are very excited to listen to Dr. Robyn Baldens talk about: Medical Science Liaison and other opportunities at Merck nextFriday,September 20th at12:00 PMinNMR/Rm. N127

Dr. Balden is a physician scientist and Regional Medical Scientific Director for Anesthesia/Surgery, South/Central US Medical Affairs division of Merck Research Labs. This role integrates internal and external scientific exchange and collaboration in order to facilitate and support clinical and drug development programs and maximize patient safety and outcomes related to existing pharmaceuticals including clinical trials, investigator-initiated studies, medical education, and scientific content creation.Her role at Merck began in 2018 as Associate Director, Medical Science Liaison for Anesthesia/Surgery, South/Central US, subsequent to gaining experience conducting medical research and directing business development for clinical trials at the Texas Center for Drug Development in Houston, TX. At the Texas Center for Drug Development she engaged in medical affairs focusing on coordination of clinical research for various therapeutic areas, serving as a supporting investigator for clinical trials, scientific discussion and account management with key physician leaders, and development of medical educational materials. Prior to this role she was a surgical intern, resident anesthesiologist, and clinical scholar at Cedars-Sinai Medical Center in Los Angeles, CA, where she initiated clinical studies for novel anesthetic regimens.

Dr. Balden received her MD and PhD in Neuroscience from Texas A&M Health Science Center College of Medicine. Her passions involve the intersection of medicine and science with neuroimmunology and neuroendocrinology. She also collaborates with advocacy and student organizations, has written several academic papers on Vitamin D, and served as a member of the Vitamin D Councils Board of Directors contributing as a volunteer writer, podcast contributor, and graphic designer for the Vitamin D Council. Shelives with her family in Houston, TX and enjoys painting, design, traveling, scuba diving, outdoors, live music, reading, cooking, and gardening.

Continue reading here:
Ph.D. in Genetics at Texas A&M University

Read More...

Efficient Implementation of Penalized … – genetics.org

Monday, May 6th, 2019

Abstract

Polygenic Risk Scores (PRS) combine genotype information across many single-nucleotide polymorphisms (SNPs) to give a score reflecting the genetic risk of developing a disease. PRS might have a major impact on public health, possibly allowing for screening campaigns to identify high-genetic risk individuals for a given disease. The Clumping+Thresholding (C+T) approach is the most common method to derive PRS. C+T uses only univariate genome-wide association studies (GWAS) summary statistics, which makes it fast and easy to use. However, previous work showed that jointly estimating SNP effects for computing PRS has the potential to significantly improve the predictive performance of PRS as compared to C+T. In this paper, we present an efficient method for the joint estimation of SNP effects using individual-level data, allowing for practical application of penalized logistic regression (PLR) on modern datasets including hundreds of thousands of individuals. Moreover, our implementation of PLR directly includes automatic choices for hyper-parameters. We also provide an implementation of penalized linear regression for quantitative traits. We compare the performance of PLR, C+T and a derivation of random forests using both real and simulated data. Overall, we find that PLR achieves equal or higher predictive performance than C+T in most scenarios considered, while being scalable to biobank data. In particular, we find that improvement in predictive performance is more pronounced when there are few effects located in nearby genomic regions with correlated SNPs; for instance, in simulations, AUC values increase from 83% with the best prediction of C+T to 92.5% with PLR. We confirm these results in a data analysis of a case-control study for celiac disease where PLR and the standard C+T method achieve AUC values of 89% and of 82.5%. Applying penalized linear regression to 350,000 individuals of the UK Biobank, we predict height with a larger correlation than with the best prediction of C+T (65% instead of 55%), further demonstrating its scalability and strong predictive power, even for highly polygenic traits. Moreover, using 150,000 individuals of the UK Biobank, we are able to predict breast cancer better than C+T, fitting PLR in a few minutes only. In conclusion, this paper demonstrates the feasibility and relevance of using penalized regression for PRS computation when large individual-level datasets are available, thanks to the efficient implementation available in our R package bigstatsr.

POLYGENIC risk scores (PRS) combine genotype information across many single-nucleotide polymorphisms (SNPs) to give a score reflecting the genetic risk of developing a disease. PRS are useful for genetic epidemiology when testing polygenicity of diseases and finding a common genetic contribution between two diseases (Purcell et al. 2009). Personalized medicine is another major application of PRS. Personalized medicine envisions to use PRS in screening campaigns in order to identify high-risk individuals for a given disease (Chatterjee et al. 2016). As an example of practical application, targeting screening of men at higher polygenic risk could reduce the problem of overdiagnosis and lead to a better benefit-to-harm balance in screening for prostate cancer (Pashayan et al. 2015). However, in order to be used in clinical settings, PRS should discriminate well enough between cases and controls. For screening high-risk individuals and for presymptomatic diagnosis of the general population, it is suggested that, for a 10% disease prevalence, the AUC must be >75% and 99%, respectively (Janssens et al. 2007).

Several methods have been developed to predict disease status, or any phenotype, based on SNP information. A commonly used method often called P+T or C+T (which stands for Clumping and Thresholding) is used to derive PRS from results of Genome-Wide Association Studies (GWAS) (Wray et al. 2007; Evans et al. 2009; Purcell et al. 2009; Chatterjee et al. 2013; Dudbridge 2013). This technique uses GWAS summary statistics, allowing for a fast implementation of C+T. However, C+T also has several limitations; for instance, previous studies have shown that predictive performance of C+T is very sensitive to the threshold of inclusion of SNPs, depending on the disease architecture (Ware et al. 2017). In parallel, statistical learning methods have also been used to derive PRS for complex human diseases by jointly estimating SNP effects. Such methods include joint logistic regression, Support Vector Machine (SVM) and random forests (Wei et al. 2009; Abraham et al. 2012, 2014; Botta et al. 2014; Okser et al. 2014; Lello et al. 2018; Mavaddat et al. 2019). Finally, Linear Mixed-Models (LMMs) are another widely used method in fields such as plant and animal breeding, or for predicting highly polygenic quantitative human phenotypes such as height (Yang et al. 2010). Yet, predictions resulting from LMM, known e.g., as gBLUP, have not proven as efficient as other methods for predicting several complex diseases based on genotypes [see table 2 of Abraham et al. (2013)].

We recently developed two R packages, bigstatsr and bigsnpr, for efficiently analyzing large-scale genome-wide data (Priv et al. 2018). Package bigstatsr now includes an efficient algorithm with a new implementation for computing sparse linear and logistic regressions on huge datasets as large as the UK Biobank (Bycroft et al. 2018). In this paper, we present a comprehensive comparative study of our implementation of penalized logistic regression (PLR), which we compare to the C+T method and the T-Trees algorithm, a derivation of random forests that has shown high predictive performance (Botta et al. 2014). In this comparison, we do not include any LMM method, yet, L2-PLR should be very similar to LMM methods. Moreover, we do not include any SVM method because it is expected to give similar results to logistic regression (Abraham et al. 2012). For C+T, we report results for a large grid of hyper-parameters. For PLR, the choice of hyper-parameters is included in the algorithm so that we report only one model for each simulation. We also use a modified version of PLR in order to capture not only linear effects, but also recessive and dominant effects.

To perform simulations, we use real genotype data and simulate new phenotypes. In order to make our comparison as comprehensive as possible, we compare different disease architectures by varying the number, size and location of causal effects as well as disease heritability. We also compare two different models for simulating phenotypes, one with additive effects only, and one that combines additive, dominant and interaction-type effects. Overall, we find that PLR achieves higher predictive performance than C+T except in highly underpowered cases (AUC values lower than 0.6), while being scalable to biobank data.

We use real genotypes of European individuals from a case-control study for celiac disease (Dubois et al. 2010). This dataset is presented in Supplemental Material, Table S1. Details of quality control and imputation for this dataset are available in Priv et al. (2018). For simulations presented later, we first restrict this dataset to controls from UK in order to remove the genetic structure induced by the celiac disease status and population structure. This filtering process results in a sample of 7100 individuals (see supplemental notebook preprocessing). We also use this dataset for real data application, in this case keeping all 15,155 individuals (4496 cases and 10,659 controls). Both datasets contain 281,122 SNPs.

We simulate binary phenotypes using a Liability Threshold Model (LTM) with a prevalence of 30% (Falconer 1965). We vary simulation parameters in order to match a range of genetic architectures from low to high polygenicity. This is achieved by varying the number of causal variants and their location (30, 300, or 3000 anywhere in all 22 autosomal chromosomes or 30 in the HLA region of chromosome 6), and the disease heritability (50 or 80%). Liability scores are computed either from a model with additive effects only (ADD) or a more complex model that combines additive, dominant and interaction-type effects (COMP). For model ADD, we compute the liability score of the i-th individual aswhere is the set of causal SNPs, are weights generated from a Gaussian distribution or a Laplace distribution , is the allele count of individual i for SNP j, corresponds to its standardized version (zero mean and unit variance for all SNPs), and follows a Gaussian distribution . For model COMP, we simulate liability scores using additive, dominant and interaction-type effects (see Supplemental Materials).

We implement three different simulation scenarios, summarized in Table 1. Scenario N1 uses the whole dataset (all 22 autosomal chromosomes 281,122 SNPs) and a training set of size 6000. For each combination of the remaining parameters, results are based on 100 simulations except when comparing PLR with T-Trees, which relies on five simulations only because of a much higher computational burden of T-Trees as compared to other methods. Scenario N2 consists of 100 simulations per combination of parameters on a dataset composed of chromosome six only (18,941 SNPs). Reducing the number of SNPs increases the polygenicity (the proportion of causal SNPs) of the simulated models. Reducing the number of SNPs (p) is also equivalent to increasing the sample size (n) as predictive power increases as a function of (Dudbridge 2013; Vilhjlmsson et al. 2015). For this scenario, we use the additive model only, but continue to vary all other simulation parameters. Finally, scenario N3 uses the whole dataset as in scenario N1 while varying the size of the training set in order to assess how the sample size affects predictive performance of methods. A total of 100 simulations per combination of parameters are run using 300 causal SNPs randomly chosen on the genome.

In this study, we use two different measures of predictive accuracy. First, we use the Area Under the Receiver Operating Characteristic (ROC) Curve (AUC) (Lusted 1971; Fawcett 2006). In the case of our study, the AUC is the probability that the PRS of a case is greater than the PRS of a control. This measure indicates the extent to which we can distinguish between cases and controls using PRS. As a second measure, we also report the partial AUC for specificities between 90 and 100% (McClish 1989; Dodd and Pepe 2003). This measure is similar to the AUC, but focuses on high specificities, which is the most useful part of the ROC curve in clinical settings. When reporting AUC results of simulations, we also report maximum achievable AUC values of 84% and 94% for heritabilities of 50% and 80%, respectively. These estimates are based on three different yet consistent estimations (see Supplemental Materials).

In this paper, we compare three different types of methods: the C+T method, T-Trees and PLR.

The C+T method directly derives PRS from the results of Genome-Wide Associations Studies (GWAS). In GWAS, a coefficient of regression (i.e., the estimated effect size ) is learned independently for each SNP j along with a corresponding P-value . The SNPs are first clumped (C) so that there remain only loci that are weakly correlated with one another (this set of SNPs is denoted ). Then, thresholding (T) consists in removing SNPs with P-values larger than a user-defined threshold . Finally, the PRS for individual i is defined as the sum of allele counts of the remaining SNPs weighted by the corresponding effect coefficientswhere are the effect sizes (P-values) learned from the GWAS. In this study, we mostly report scores for a clumping threshold at within regions of 500kb, but we also investigate thresholds of 0.05 and 0.8. We report three different scores of prediction: one including all the SNPs remaining after clumping (denoted C+T-all), one including only the SNPs remaining after clumping and that have a P-value under the GWAS threshold of significance (, C+T-stringent), and one that maximizes the AUC (C+T-max) for 102 P-value thresholds between 1 and (Table S2). As we report the optimal threshold based on the test set, the AUC for C+T-max is an upper bound of the AUC for the C+T method. Here, the GWAS part uses the training set while clumping uses the test set (all individuals not included in the training set).

T-Trees (Trees inside Trees) is an algorithm derived from random forests (Breiman 2001) that takes into account the correlation structure among the genetic markers implied by linkage disequilibrium (Botta et al. 2014). We use the same parameters as reported in table 4 of Botta et al. (2014), except that we use 100 trees instead of 1000. Using 1000 trees provides a minimal increase of AUC while requiring a disproportionately long processing time (e.g., AUC of 81.5% instead of 81%, data not shown).

Finally, for PLR, we find regression coefficients and that minimize the following regularized loss function(1)where , x denotes the genotypes and covariables (e.g., principal components), y is the disease status to predict, and are two regularization hyper-parameters that need to be chosen. Different regularizations can be used to prevent overfitting, among other benefits: the L2-regularization (ridge, Hoerl and Kennard (1970)) shrinks coefficients and is ideal if there are many predictors drawn from a Gaussian distribution (corresponds to in the previous equation); the L1-regularization (lasso, Tibshirani 1996) forces some of the coefficients to be equal to zero and can be used as a means of variable selection, leading to sparse models (corresponds to ); the L1- and L2-regularization (elastic-net, Zou and Hastie 2005) is a compromise between the two previous penalties and is particularly useful in the situation (p is the number of SNPs), or any situation involving many correlated predictors (corresponds to ) (Friedman et al. 2010). In this study, we use a grid search over . This grid-search is directly embedded in our PLR implementation for simplicity. Using should result in a model very similar to gBLUP.

To fit PLR, we use an efficient algorithm (Friedman et al. 2010; Tibshirani et al. 2012; Zeng and Breheny 2017) from which we derived our own implementation in R package bigstatsr. This algorithm builds predictions for many values of , which is called a regularization path. To obtain an algorithm that does not require to choose this hyper-parameter , we developed a procedure that we call Cross-Model Selection and Averaging (CMSA, Figure S1). Because of L1-regularization, the resulting vector of estimated effect sizes is sparse. We refer to this method as PLR in the results section.

To capture recessive and dominant effects on top of additive effects in PLR, we use simple feature engineering: we construct a separate dataset with three times as many variables as the initial one. For each SNP variable, we add two more variables coding for recessive and dominant effects: one variable is coded 1 if homozygous variant and 0 otherwise, and the other is coded 0 for homozygous referent and 1 otherwise. We then apply our PLR implementation to this dataset with three times as many variables as the initial one; we refer to this method as PLR3 in the rest of the paper.

We use Monte Carlo cross-validation to compute AUC, partial AUC, the number of predictors, and execution time for the original Celiac dataset with the observed case-control status: we randomly split 100 times the dataset in a training set of 12,000 individuals and a test set composed of the remaining 3155 individuals.

We compared PLR with the C+T method using simulations of scenario N1 (Table 1). When simulating a model with 30 causal SNPs and a heritability of 80%, PLR provides AUC of 93%, nearly reaching the maximum achievable AUC of 94% for this setting (Figure 1). Moreover, PLR consistently provides higher predictive performance than C+T across all scenarios considered, except in some cases of high polygenicity and small sample size, where all methods perform poorly (AUC values below 60% Figure 1 and Figure 3). PLR provides particularly higher predictive performance than C+T when there are correlations between predictors, i.e., when we choose causal SNPs to be in the HLA region. In this situation, the mean AUC reaches 92.5% for PLR and 84% for C+T-max (Figure 1). For the simulations, we do not report results in terms of partial AUC because partial AUC values have a Spearman correlation of 98% with the AUC results for all methods (Figure S3).

In practice, a particular value of the threshold of inclusion of SNPs should be chosen for the C+T method, and this choice can dramatically impact the predictive performance of C+T. For example, in a model with 30 causal SNPs, AUC ranges from <60% when using all SNPs passing clumping to 90% if choosing the optimal P-value threshold (Figure S4).

Concerning the threshold of the clumping step in C+T, we mostly used the common value of 0.2. Yet, using a more stringent value of 0.05 provides equal or higher predictive performance than using 0.2 in most of the cases we considered (Figure 2 and Figure 3).

Our implementation of PLR that automatically chooses hyper-parameter provides similar predictive performance than the best predictive performance of 100 models corresponding to different values of (Figure S8).

We tested the T-Trees method in scenario N1. As compared to PLR, T-Trees perform worse in terms of predictive ability, while taking much longer to run (Figure S5). Even for simulations with model COMP in which there are dominant and interaction-type effects that T-Trees should be able to handle, AUC is still lower when using T-Trees than when using PLR (Figure S5).

We also compared the two PLRs in scenario N1: PLR vs. PLR3 that uses additional features (variables) coding for recessive and dominant effects. Predictive performance of PLR3 are nearly as good as PLR when there are additive effects only (differences of AUC are always <2%) and can lead to significantly greater results when there are also dominant and interactions effects (Figures S6 and S7). For model COMP, PLR3 provides AUC values at least 3.5% higher than PLR, except when there are 3000 causal SNPs. Yet, PLR3 takes two to three times as much time to run and requires three times as much disk storage as PLR.

First, when reproducing simulations of scenario N1 using chromosome six only (scenario N2), the predictive performance of PLR always increase (Figure 2). There is a particularly large increase when simulating 3000 causal SNPs: AUC from PLR increases from 60% to nearly 80% for Gaussian effects and a disease heritability of 80%. On the contrary, when simulating only 30 or 300 causal SNPs with the corresponding dataset, AUC of C+T-max does not increase, and even decreases for a heritability of 80% (Figure 2). Second, when varying the training size (scenario N3), we report an increase of AUC with a larger training size, with a faster increase of AUC for PLR as compared to C+T-max (Figure 3).

Joint PLRs also provide higher AUC values for the Celiac data: 88.7% with PLR and 89.1% with PLR3 as compared to 82.5% with C+T-max (Figure S2 and Table 2). The relative increase in partial AUC, for specificities larger than 90%, is even larger (42 and 47%) with partial AUC values of 0.0411, 0.0426, and 0.0289 obtained with PLR, PLR3, and C+T-max, respectively. Moreover, logistic regressions use less predictors, respectively, at 1570, 2260, and 8360. In terms of computation time, we show that PLR, while learning jointly on all SNPs at once and testing four different values for hyper-parameter , is almost as fast as the C+T method (190 vs. 130 sec), and PLR3 takes less than twice as long as PLR (296 vs. 190 sec).

We tested our implementation on 656K genotyped SNPs of the UK Biobank, keeping only Caucasian individuals and removing related individuals (excluding the second individual in each pair with a kinship coefficient >0.08). Results are presented in Table 3.

Our implementation of L1-penalized linear regression runs in <1 day for 350K individuals (training set), achieving a correlation of >65.5% with true height for each sex in the remaining 24K individuals (test set). By comparison, the best C+T model achieves a correlation of 55% for women and 56% for men (in the test set), and the GWAS part takes 1 hr (for the training set). If using only the top 100,000 SNPs from a GWAS on the training set to fit our L1-PLR, correlation between predicted and true heights drops at 63.4% for women and 64.3% for men. Our L1-PLR on breast cancer runs in 13 min for 150K women, achieving an AUC of 0.598 in the remaining 39K women, while the best C+T model achieves an AUC of 0.589, and the GWAS part takes 15hr.

In this comparative study, we present a computationally efficient implementation of PLR. This model can be used to build PRS based on very large individual-level SNP datasets such as the UK biobank (Bycroft et al. 2018). In agreement with previous work (Abraham et al. 2013), we show that jointly estimating SNP effects has the potential to substantially improve predictive performance as compared to the standard C+T approach in which SNP effects are learned independently. PLR always outperforms the C+T method, except in some highly underpowered cases (AUC values always <0.6), and the benefits of using PLR are more pronounced with an increasing sample size or when causal SNPs are correlated with one another.

When there are many small effects and a small sample size, PLR performs worse than (the best result for) C+T. For example, this situation occurs when there are many causal variants (3K) to distinguish among many typed variants (280K) while using a small sample size (6K). In such underpowered scenarios, it is difficult to detect true causal variants, which makes PLR too conservative, whereas the best strategy is to include nearly all SNPs (Purcell et al. 2009).

When increasing sample size (scenario N3), PLR achieves higher predictive performance than C+T and the benefits of using PLR over C+T increase with an increasing sample size (Figure 3). Moreover, when decreasing the search space (total number of candidate SNPs) in scenario N2, we increase the proportion of causal variants and we virtually increase the sample size (Dudbridge 2013). In this scenario N2, even when there are small effects and a high polygenicity (3000 causal variants out of 18,941), PLR gets a large increase in predictive performance, now consistently higher than C+T (Figure 2).

The choice of hyper-parameter values is very important since it can greatly impact the performance of methods. In the C+T method, there are two main hyper-parameters: the and the thresholds that control how stringent are the C+T steps. For the clumping step, appropriately choosing the threshold is important. Indeed, on the one hand, choosing a low value for this threshold may discard informative SNPs that are correlated. On the other hand, when choosing a high value for this threshold, too much redundant information is included in the model, which adds noise to the PRS. Based on the simulations, we find that using a stringent threshold leads to higher predictive performance, even when causal SNPs are correlated. It means that, in most cases tested in this paper, avoiding redundant information in C+T is more important than including all causal SNPs. The choice of the threshold is also very important as it can greatly impact the predictive performance of the C+T method, which we confirm in this study (Ware et al. 2017). In this paper, we reported the maximum AUC of 102 different P-value thresholds, a threshold that should normally be learned on the training set only. To our knowledge, there is no clear standard on how to choose these two critical hyper-parameters for C+T. So, for C+T, we report the best AUC value on the test set, even if it leads to overoptimistic results for C+T as compared to PLR.

In contrast, for PLR, we developed an automatic procedure called CMSA that releases investigators from the burden of choosing hyper-parameter . Not only this procedure provides near-optimal results, but it also accelerates the model training thanks to the development of an early stopping criterion. Usually, cross-validation is used to choose hyper-parameter values and then the model is trained again with these particular hyper-parameter values (Hastie et al. 2008; Wei et al. 2013). Yet, performing cross-validation and retraining the model is computationally demanding; CMSA offers a less burdensome alternative. Concerning hyper-parameter that accounts for the relative importance of the L1 and L2 regularizations, we use a grid search directly embedded in the CMSA procedure.

We also explored how to capture nonlinear effects. For this, we introduced a simple feature engineering technique that enables PLR to detect and learn not only additive effects, but also dominant and recessive effects. This technique improves the predictive performance of PLR when there are nonlinear effects in the simulations, while providing nearly the same predictive performance when there are additive effects only. Moreover, it also improves predictive performance for the celiac disease.

Yet, this approach is not able to detect interaction-type effects. In order to capture interaction-type effects, we tested T-Trees, a method that is able to exploit SNP correlations and interactions thanks to special decision trees (Botta et al. 2014). However, predictive performance of T-Trees are consistently lower than with PLR, even when simulating a model with dominant and interaction-type effects that T-Trees should be able to handle.

The computation time of our PLR implementation mainly depends on the sample size and the number of candidate variables (variables that are included in the gradient descent). Indeed, the algorithm is composed of two steps: first, for each variable, the algorithm computes an univariate statistic that is used to decide if the variable is included in the model (for each value of ). This first step is very fast. Then, the algorithm iterates over a regularization path of decreasing values of , which progressively enables variables to enter the model (Figure S1). In the second step, the number of variables increases and computations stop when an early stopping criterion is reached (when prediction is getting worse on the corresponding validation set, see Figure S1).

For highly polygenic traits such as height and when using huge datasets such as the UK Biobank, the algorithm might iterate over >100,000 variables, which is computationally demanding. On the contrary, for traits like celiac disease or breast cancer that are less polygenic, the number of variables included in the model is much smaller so that fitting is very fast (only 13min for 150K women of the UK Biobank for breast cancer).

Memory requirements are tightly linked to computation time. Indeed, variables are accessed in memory thanks to memory-mapping when they are used (Priv et al. 2018). When there is not enough memory left, the operating system (OS) frees some memory for new incoming variables. Yet, if too many variables are used in the gradient descent, the OS would regularly swap memory between disk and RAM, severely slowing down computations. A possible approach to reduce computational burden is to apply penalized regression on a subset of SNPs by prioritizing SNPs using univariate tests (GWAS computed from the same dataset). Yet, this strategy was shown to reduce predictive power (Abraham et al. 2013; Lello et al. 2018), which we also confirm in this paper. Indeed, when using only the 100K most significantly associated SNPs, correlation between predicted and true heights is reduced from 0.656/0.657 to 0.634/0.643 within women/men. A key advantage of our implementation of PLR is that prior filtering of variables is no more required for computational feasibility, thanks to the use of sequential strong rules and early stopping criteria.

Our approach has one major limitation: the main advantage of the C+T method is its direct applicability to summary statistics, allowing to leverage the largest GWAS results to date, even when individual cohort data cannot be merged because of practical or legal reasons. Our implementation of PLR does not allow yet for the analysis of summary data, but this represents an important future direction. The current version is of particular interest for the analysis of modern individual-level datasets including hundreds of thousands of individuals.

Finally, in this comparative study, we did not consider the problem of population structure (Vilhjlmsson et al. 2015; Mrquez-Luna et al. 2017; Martin et al. 2017), and also did not consider nongenetic data such as environmental and clinical data (Van Vliet et al. 2012; Dey et al. 2013).

In this comparative study, we have presented a computationally efficient implementation of PLR that can be used to predict disease status based on genotypes. A similar penalized linear regression for quantitative traits is also available in R package bigstatsr. Our approach solves the dramatic memory and computational burdens faced by standard implementations, thus allowing for the analysis of large-scale datasets such as the UK biobank (Bycroft et al. 2018).

We also demonstrated in simulations and real datasets that our implementation of penalized regressions is highly effective over a broad range of disease architectures. It can be appropriate for predicting autoimmune diseases with a few strong effects (e.g., celiac disease), as well as highly polygenic traits (e.g., standing height) provided that sample size is not too small. Finally, PLR as implemented in bigstatsr can also be used to predict phenotypes based on other omics data, since our implementation is not specific to genotype data.

We are grateful to Flix Balazard for useful discussions about T-Trees, and to Yaohui Zeng for useful discussions about R package biglasso. We are grateful to the two anonymous reviewers who contributed to improving this paper. The authors acknowledge LabEx Pervasive Systems and Algorithms (PERSYVAL)-Lab [Agence Nationale de Recherche (ANR)-11-LABX-0025-01] and ANR project French Regional Origins in Genetics for Health (FROGH) (ANR-16-CE12-0033). The authors also acknowledge the Grenoble Alpes Data Institute, which is supported by the French National Research Agency under the Investissements davenir program (ANR-15-IDEX-02). This research was conducted using the UK Biobank Resource under Application Number 25589.

Available freely online through the author-supported open access option.

This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Read more from the original source:
Efficient Implementation of Penalized ... - genetics.org

Read More...

Biology for Kids: Genetics – ducksters.com

Saturday, March 23rd, 2019

");}

Genetics is the study of genes and heredity. It studies how living organisms, including people, inherit traits from their parents. Genetics is generally considered part of the science of biology. Scientists who study genetics are called geneticists.

What are genes?

Genes are the basic units of heredity. They consist of DNA and are part of a larger structure called the chromosome. Genes carry information that determine what characteristics are inherited from an organism's parents. They determine traits such as the color of your hair, how tall you are, and the color of your eyes.

What are chromosomes?

Chromosomes are tiny structures inside cells made from DNA and protein. The information inside chromosomes acts like a recipe that tells cells how to function. Humans have 23 pairs of chromosomes for a total of 46 chromosomes in each cell. Other plants and animals have different numbers of chromosomes. For example, a garden pea has 14 chromosomes and an elephant has 56.

What is DNA?

The actual instructions inside the chromosome is stored in a long molecule called DNA. DNA stands for deoxyribonucleic acid.

Gregor Mendel is considered the father of the science of genetics. Mendel was a scientist during the 1800s who studied inheritance by experimenting with pea plants in his garden. Through his experiments he was able to show patterns of inheritance and prove that traits were inherited from the parents.

See original here:
Biology for Kids: Genetics - ducksters.com

Read More...

Genetics | The Institute for Creation Research

Friday, January 25th, 2019

For over 150 years, Darwins hypothesis that all species share a common ancestor has dominated the creation-evolution debate. Surprisingly, when Darwin wrote his seminal work, he had no direct evidence for these genealogical relationships. Now, with online databases full of DNA-sequence information from thousands of species, the direct testing of Darwins hypothesis has finally commenced. More...

Authentic speciation is a process whereby organisms diversify within the boundaries of their gene pools, and this can result in variants with specific ecological adaptability. While it was once thought that this process was strictly facilitated by DNA sequence variability, Darwin's classic example of speciation in finches now includes a surprisingly strong epigenetic component as well. More...

One of the rapidly expanding and exciting research fields in molecular biology is the area of epigenetics. In the study of epigenetic modifications, scientists analyze DNA that has been modified in such a way that its chemistry is changed, but not the actual base pairs that make up the genetic code of the sequence. Its like a separate control code and system imposed upon and within the standard code of DNA sequence.

Because epigenetic modifications in the genome are related to gene expression, researchers have been using highly advanced technologies for comparing these differences in humans and chimps for regions of the genome that they both have in common. More... More...

Living things develop partly according to genetic instructions encoded on their DNA. The study of inheritance has widened the paradigms from genes to genomes, and now recent research indicates that critical biological information is carried from one generation to the next in systems additional to DNA, called epigenetic factors.

So, where did this information come from? More...

Genes could be thought of as brick molds, used to construct materials for building the physical structures of living organisms. They carry the codes to help make proteins, which then make up different cells that are combined together to form mega-structures called tissues.

New research has shed more light on how genes are used by cells to build the different tissues needed by complex living creatures. More...

Indiana University researchers discovered that certain genes used in developing horned beetle larvae are re-used later to make horns in their adult stage. The studys authors called the genes co-opted, indicating their belief that evolution decided to give them a secondary use. The authors suggestion that gene co-opting offers a possible explanation for the development of novel traits comes up short, however. More...

One of the past arguments for evidence of biological evolution in the genome has been the concept of pseudogenes. These DNA sequences were once thought to be the defunct remnants of genes, representing nothing but genomic fossils in the genomes of plants and animals. More...

Amazingly, scientists documented the activity of 2,082 distinct pseudogenes in the human genome whose aberrant levels of activity were directly associated with cancer-specific pathologies. More...

Proteins do most of the required metabolic tasks within each of the trillions of cells in the human body. However, only about four percent of human DNA contains coded instructions that specify proteins.

So what is the purpose of the remaining 96 or so percent? More...

A research team recently characterized a group of genes in humans and other mammals that not only defies evolutionary models but vindicates the Bibles prediction of the uniqueness of created kinds with distinct genetic features. More...

Read more from the original source:
Genetics | The Institute for Creation Research

Read More...

Human Genetics – medschool.ucla.edu

Friday, January 11th, 2019

A hub of deep expertise, the Department of Human Genetics helps partners across UCLA interpret data and leverage genomic technology to improve study design and solve medical problems.

We demystify genetic complexities to provide vital insights for a range of clinical and research applications. We strive to improve the care of as many patients as possible by pushing our capabilities, developing novel ways to address unanswered questions.

Your next collaboration is right down the street.

Our enviable proximity to the worlds brightest scientific minds enables both thriving scheduled events and impromptu sidewalk powwows. A casual conversation during your coffee run could lead to your next big publication.

Come find out why innovation lives here.

LEARN MORE

Julian Martinez-Agosto, MD, PhDGenetic sequencing unravels rare disease mysteries; among the first medical centers to use exome sequencing.Learn More

Jingyi "Jessica" Li, PhDStatistics professor honored as a leading woman in STEM at the intersection of statistics and biology.Learn More

Aldons J. Lusis, PhDScientists identify 2 hormones that burn fat faster, prevent and reverse diabetes in mice.Learn More

Daniel Geschwind, MD, PhDAutism, schizophrenia, bipolar disorder share molecular traits, study finds.Learn More

Here is the original post:
Human Genetics - medschool.ucla.edu

Read More...

My genetics – How I Recovered

Wednesday, September 19th, 2018

CYP1A1*2C A4889Grs1048943CTT-/-CYP1A1*4 C2453Ars1799814TGG-/-CYP1A2 C164Ars762551CAC+/-CYP1B1 L432Vrs1056836CCG+/-CYP1B1 N453Srs1800440CTT-/-CYP1B1 R48Grs10012CGG-/-CYP2A6*2 A1799Trs1801272TAA-/-CYP2C19*17rs12248560TCC-/-CYP2C9*2 C430Trs1799853TCC-/-CYP2C9*3 A1075Crs1057910CAA-/-CYP2D6 S486Trs1135840GGG+/+CYP2D6 T100Crs1065852AGG-/-CYP2D6 T2850Crs16947AAA+/+CYP2E1*1B G9896Crs2070676GCC-/-CYP2E1*4 A4768Grs6413419AGG-/-CYP3A4*1Brs2740574CTT-/-CYP3A4*3 M445Trs4986910GAA-/-CYPs are primarily membrane-associatedproteins located either in the inner membrane ofmitochondriaor in theendoplasmic reticulumof cells. CYPs metabolize thousands ofendogenousandexogenouschemicals. Some CYPs metabolize only one (or a very few) substrates, such asCYP19(aromatase), while others may metabolize multiple substrates. Both of these characteristics account for their central importance inmedicine. Cytochrome P450 enzymes are present in most tissues of the body, and play important roles inhormonesynthesis and breakdown includingestrogenandtestosteronesynthesis and metabolism,cholesterolsynthesis, andvitamin Dmetabolism. Cytochrome P450 enzymes also function to metabolize potentially toxic compounds, includingdrugsand products of endogenous metabolism such asbilirubin, principally in theliver.rs762551 (C) allele is a slow metabolizer or of certain substrates including caffeine which means Im more stimulated by it than most people.rs1056836 increases susceptibility to lung and breast cancer, blocks testosterone and inhibits mitochondrial function.rs1135840 is involved in the metabolism of approximately 25% of all medications and most psych meds including antipsychotics and antidepressants.GPX3rs8177412CTT-/-GSTM1rs12068997TCC-/-GSTM1rs4147565AGG-/-GSTM1rs4147567GAA-/-GSTM1rs4147568ATT-/-GSTM1rs1056806TCC-/-GSTM1rs12562055ATT-/-GSTM1rs2239892GAA-/-GSTP I105Vrs1695GAG+/-GSTP1 A114Vrs1138272TCC-/-GSTP genes encode the Glutathione S-transferase P enzyme. Glutathione S-transferases (GSTs) are a family of enzymes that play an important role in detoxification by catalyzing the conjugation of manyhydrophobic and electrophilic compounds with reducedglutathione. Mutations here will increase your need for glutathione and importance of chelating out mercury.rs1695 influences asthma risk.NAT1 A560G(?) (R187Q)rs4986782AGG-/-NAT2 A803G (K268R)rs1208GGG+/+NAT2 C190T (R64W)rs1805158TCC-/-NAT2 G590A (R197Q)rs1799930AGG-/-NAT2 G857A (G286E)rs1799931AGG-/-NAT2 T341C (I114T)rs1801280CCC+/+NAT2 encodes N-acetyltransferases which are enzymes acting primarily in the liver to detoxify a large number of chemicals, includingcaffeineand several prescribed drugs. The NAT2 acetylation polymorphism is important because of its primary role in the activation and/or deactivation of many chemicals in the bodys environment, including those produced by cigarettes as well as aromatic amine and hydrazine drugs used medicinally. In turn, this can affect an individualscancerrisk.I have a particular combination of NAT2 polymorphisms rs1801280 (C) +rs1208 (G) which makes me a slow metabolizer. In general, slow metabolizers have higher rates of certain types ofcancerand are more susceptible to side effects from chemicals (known as MCS) metabolized by NAT2.SOD2rs2758331AAA+/+SOD2rs2855262TCT+/-SOD2 A16Vrs4880GGG+/+SOD2 gene is a member of the iron/manganesesuperoxide dismutasefamily and may be one of the key sources of my troubles. This protein transforms toxic superoxide, a byproduct of the mitochondrial electron transport chain, intohydrogen peroxideand diatomicoxygen. In simpler terms, the more energy your mitochondria produce, the more byproducts (also called free radicals) get produced. These toxic byproducts tear up cell membranes and walls through a process called oxidative stress.Mutations in the SOD2 gene diminish your ability to transform these toxic byproducts into harmless components. People with SOD2 polymorphisms may not tolerate nitrates or fish oil well. Mutations in this gene have been associated withidiopathic cardiomyopathy(IDC), sporadic motor neuron disease, and cancer.

Now what about SOD1 & 3? I dont know why it doesnt appear on this report but I was able to get some information on it from Livewello and it looks like I am much better off there. Heres my SOD1 and SOD3 status. Just for kicks, I decided to run SOD2 and I find it shows a much different picture than sterlings app: my SOD 2 on Livewello. Notice how it shows that I do have some working SOD2 genes!

Here is the original post:
My genetics - How I Recovered

Read More...

Genetics | Definition of Genetics by Merriam-Webster

Saturday, August 25th, 2018

Suddenly, Soo-Kyung, 42, and her husband Jae Lee, 57, another genetics specialist at O.H.S.U., had to transform from dispassionate scientists into parents of a patient, desperate for answers.

Among the brightest of those homegrown stars is Zhao Bowen, a Chinese science prodigy who dropped out of high school to start running a genetics lab.

Among the brightest of those homegrown stars is Zhao Bowen, a Chinese science prodigy who dropped out of high school to start running a genetics lab.

Krainer, a molecular genetics professor at Cold Spring Harbor Laboratory on Long Island, N.Y., had worked on the scientific underpinnings of the medicine for more than 15 years.

Since these discoveries, the field of genetics has expanded even furtherall the way to our own front doors, in fact, thanks to at-home genetic tests such as 23andMe.

Coral genetics is a field of increasing interest to scientists.

Krainer, a molecular genetics professor at Cold Spring Harbor Laboratory on Long Island, N.Y., had worked on the scientific underpinnings of the medicine for more than 15 years.

His father retired as a genetics professor at Northern Illinois University, also in DeKalb.

Read the rest here:
Genetics | Definition of Genetics by Merriam-Webster

Read More...

Hundreds of Thousands of Species in a Few Thousand Years?

Sunday, July 29th, 2018

A recent1 review paper proposed a controversial claimthat the vast majority of animal species arose contemporary with modern humans. Not surprisingly, this claim was met with backlash from the evolutionary community. On what basis did the authors make this wide-reaching claim? Is their assertion true? Furthermore, what ramifications do their data have for the creationist explanation of the origin of species from the originally created min or kinds?

The main focus of Stoeckle and Thalers paper is genetics. Specifically, they focus on a subset of DNA in human and animal cells, termed mitochondrial DNA (mtDNA). Their analysis of mtDNA is clear, straightforward, and carefully justifiedso much so that I will summarize their arguments by liberally quoting from their paper.

About 15 years ago, DNA barcoding was first proposed as a tool for practical taxonomy.2 Taxonomy is the field of science concerned with the classification of life, and scientists thought that taking small subsets of DNA would aid in identifying and classifying species. The particular mitochondrial sequence that has become the most widely used is the 648 base pair (bp) [think of base pairs as DNA letters] segment of the gene [a subsection of DNA sequence] encoding mitochondrial cytochrome c oxidase subunit I (COI).3

With a subset of a subset of DNA, Skeptics of COI barcoding raised a number of objections about its power and/or generality as a single simple metric applicable to the entire animal kingdom, including: the small fraction of the genome (about 5% of the mitochondrial genome and less than one millionth of the total organisms genome [total DNA in an organism]) might not be sensitive or representative.4

A simple example from humans illustrates this concern. For instance, on average any two humans differ at 0.2%0.5% of their mtDNA base pairs. Theoretically, if all mtDNA differences are evenly distributed around the human mtDNA genome, you would expect 12 mtDNA differences in each individuals 648 bp COI barcode. With numbers this low, one generation of an extra mutation or two in the COI barcode sequence might throw a real classification pattern (i.e., one based on comparisons of hundreds of anatomical and physiological features) into confusion.

However, since the early days of DNA barcoding, such objections have been mostly mollified. I can attest to this from my own experience in handling thousands of mtDNA sequences. As a representative of the mtDNA diversity among species and individuals, a subset of mtDNA sequence is a good first approximation. Though subsets arent always perfect representations of the whole sequence, they are good initial data points.

Furthermore, over several decades of mtDNA barcoding, scientists have discovered a specific clustering pattern among mtDNA barcodes from individuals across diverse species: a general observation is that barcode clusters correspond best to species in well-studied animal groups, where taxonomists have mostly decided and agreed upon what species are. Thus there is good support in several major phyla, including Chordata [e.g., vertebrates and a handful of other species], Arthropoda [e.g., insects, arachnids, and crustaceans], Mollusca [e.g., shellfish, octopi], Echinodermata [e.g., starfish]. We note that these phyla are estimated to contain about 34 of named animal species.5

This fact has two major ramifications: First, the cluster structure of the animal world found in COI barcode analysis is independent of any definition(s) of species. Second, domain experts judgments of species tend to agree with barcode clusters and many apparent deviations turn out to be exceptions that prove the rule.6 In other words, the initial fears of those skeptical of DNA barcoding have not been met. Instead, barcoding has been very successful.

In light of these successes, the authors acknowledge the unexpected implications for explanations for the origin of species: At its origin DNA barcoding made no claim of contributing to evolutionary theory,7 yet the pattern of DNA barcode variance is the central fact of animal life that needs to be explained by evolutionary theory.8

Expanding our scope beyond the narrow evolutionary focus of the authors, we can generalize their statement: These mtDNA barcode patterns need to be explained by any model purporting to account for the origin of species.

The barcode patterns take a very specific form: the clustering structure of COI barcodessmall variance within species and often but not always sequence gaps among nearest neighbor species is the primary fact that a model of evolution and speciation must explain. Furthermore, the average pairwise difference among individuals (APD; equivalent to population genetics parameter ) within animal species is between 0.0% and 0.5%. The most data are available for modern humans, who have an APD of 0.1% calculated in the same way as for other animals.9

Stoeckle and Thaler recognize the sweeping potential in these patterns: The agreement of barcodes and domain experts implies that explaining the origin of the pattern of DNA barcodes would be in large part explaining the origin of species. Understanding the mechanism by which the near-universal pattern of DNA barcodes comes about would be tantamount to understanding the mechanism of speciation.10

In their evolutionary model, Stoeckle and Thaler invoke two hypotheses account for the barcode cluster patterns: Either 1) COI barcode clusters represent species-specific adaptations, OR 2) extant populations have recently passed through diversity-reducing regimes whose consequences for sequence diversity are indistinguishable from clonal bottlenecks.11

Their conclusion? Modern human mitochondria and Y chromosome [another subset of DNA, but inherited paternally] originated from conditions that imposed a single sequence on these genetic elements between 100,000 and 200,000 years ago.12 In other words, to account for human CO barcode patterns, they favor the second hypothesissome sort of population dynamic (contraction) that reduced the genetic diversity of the population.

Stoeckle and Thaler then extrapolate their conclusions to controversial heights. To justify their extrapolation, they caution that one should not as a first impulse seek a complex and multifaceted explanation for one of the clearest, most data rich and general facts in all of evolution. Then they draw a parallel: The simple hypothesis is that the same explanation offered for the sequence variation found among modern humans applies equally to the modern populations of essentially all other animal species. Namely that the extant population, no matter what its current size or similarity to fossils of any age, has expanded from mitochondrial uniformity within the past 200,000 years.13 In other words, based on mtDNA barcodes, Stoeckle and Thaler claim that the vast majority of species have originated contemporary with modern humans.

Though Stoeckle and Thaler dont perform this step, lets revisit their data and take their results to the next logical conclusion. We can do this because creationists have no problems with the observations that Stoeckle and Thaler describe. Ive already mentioned that my own experience with mtDNA matches theirsbarcodes are a useful first approximation and should be treated as such. Yet this first approximation has revealed a consistent patternlow numbers of mtDNA differences within species and higher numbers of mtDNA differences between species.

Furthermore, since Stoeckle and Thaler explore the origin of individual speciesrather than the origin of whole classification groups, like mammalstheir reasoning applies almost seamlessly to the creationist explanation for the origin of species. Their claim that species arose recently is one that focuses on species within kindsnot one that explores changes from one kind into another. In other words, for Stoeckle and Thalers particular question, evolutionists and creationists agree on the question of common ancestry.

Nevertheless, they differ sharply on the question of timewhen these individual species arose. Unlike Stoeckle and Thaler, creationists invoke not two, but three potential explanations for low numbers of mtDNA sequence differences within species: (1) species-specific adaptations; (2) changing population sizes or past bottlenecks (see especially the discussion of American bison (Bison bison) mtDNA and African buffalo (Syncerus caffer) mtDNA in this paper; (3) time recent origin (e.g., within the last 4,5006,000 years).

We now have two decades worth of direct measurements of the rate at which human mtDNA mutates, and it matches exactly the 6,000-year timescale and rejects the evolutionary timescale (see Genetics Confirms the Recent, Supernatural Creation of Adam and Eve and references therein). Thus, taking Stoeckle and Thalers results to their logical conclusion, we can revise their statement to Modern human [mitochondrial DNA] originated from conditions that imposed a single sequence on these genetic elements14 about 6,000 years ago.

Lets now re-extrapolate these results to other species. The simple hypothesis is that the same explanation offered for the sequence variation found among modern humans applies equally to the modern populations of essentially all other animal species. Namely that the extant population, no matter what its current size or similarity to fossils of any age, has expanded from mitochondrial uniformity within the past 6,000 years.

We can refine this conclusion even more, with more spectacular implications for the creationist model: In the last two decades, the mtDNA mutation rate in a handful of invertebrate species has also been directly measured, and these rates14 are around 10 times higher (or more!) than the human mtDNA mutation rate (again, see this article and references therein). This would imply that multiple species within a genus (or perhaps even a family) have originated within the last 6,000 years.

In other words, these broad mtDNA barcode results suggest that, in general, the predictions15 I made for mtDNA mutation rates in diverse species are likely to be fulfilled. This is good evidence that Darwins ideas are well on their way to being replaced.

As this article was going to press, the theistic evolutionary organization BioLogos posted a critique of Stoeckle and Thalers paper. More specifically, BioLogos posted a critique of creationist responses to Stoeckle and Thaler. BioLogos took strong exception to the type of thesis that I advanced above. For example, consider the following quote from BioLogos: "Did Stoeckel [sic] and Thaler conclude that 90% of animal species appeared at same time as humans? The answer is No [emphasis theirs].

Did I miss a key element of the Stoeckle and Thaler paper?

Lets take a look at the BioLogos article, which was written by PhD biologist and professor Joel Duff. Duff clearly desired to minimize the implications of Stoeckle and Thalers paper. For example, Duff characterized the journal in which it was published as a low-profile Italian journal. He also downplayed the impact, saying that the extended press release didnt generate much reaction inside or outside of the scientific community. More strongly, Duff denounced claims like the one I made above as mischaracterization of the original research. He said it was an incorrect claim that most species originated about the same time.

Why?

To support his assertion, Duff proposed an examination of the original intent of the authors of this paper. Since an authors intent is invisible unless the author clearly states it, Duffs suggested methodology to justify his strong critique is a creative way to tackle a scientific controversy.

After examining Stoeckle and Thalers intent to Duffs satisfaction, Duffs journalism gets more questionable. Weve already examined his emphatic assertion: Did Stoeckel [sic] and Thaler conclude that 90% of animal species appeared at same time as humans? The answer is No. Duff justifies his forceful condemnation with a quote from Stoeckle and Thalers paper: the extant population, no matter what its current size or similarity to fossils of any age, has expanded from mitochondrial uniformity within the past 200,000 years.16 In light of this quote, Duff concludes, In other words, the genetic diversity observed in mitochondrial genomes of most species alive today can be attributed to the accumulation of mutations from an ancestral genome within the past 200,000 years, and Duff asserts that the authors never claim that most species came into existence within the past 200,000 years.

For a critique that began with a proposal to examine intent, Duff seems to have missed the actual intent of the authors. The title of their paper is, Why should mitochondria define species? After discussing and justifying at length the observation that mtDNA differences do, in fact, delineate species, the authors then make a startling statement: The pattern of DNA barcode variance is the central fact of animal life that needs to be explained by evolutionary theory17 [emphasis theirs]. In case the intent of their statement wasnt transparent, the authors make it explicit: The agreement of barcodes and domain experts implies that explaining the origin of the pattern of DNA barcodes would be in large part explaining the origin of species. Understanding the mechanism by which the near-universal pattern of DNA barcodes comes about would be tantamount to understanding the mechanism of speciation.18 They then spend the next chunk of their paper discussing what mtDNA barcodes imply about the mechanism of speciation. Clearly, Stoeckle and Thaler are concerned with much more than just the accumulation of mutations from an ancestral genome within the past 200,000 years. Instead, they have a strong focus on the origin of species.

But did the authors never claim that most species came into existence within the past 200,000 years? In one sense, if we split hairs, Duff is technically correct: In their paper, Stoeckle and Thaler never say so explicitly. Yet as weve just observed, the conclusion about the timing of the origin of species is implied. Furthermore, Thaler makes the conclusion explicit in the press releasethe very one that Duff cited:

Our paper strengthens the argument that the low variation in the mitochondrial DNA of modern humans also explains the similar low variation found in over 90% of living animal specieswe all likely originated by similar processes and most animal species are likely young19. [emphasis added]

How did Biologos miss this?

Duff advances a second argument in his critique of the implications of Stoeckle and Thalers paper. He says that the mtDNA results at best, [tell] us the minimum age of the species. It tells us little to nothing about the maximum age of a species [emphasis his]. For the maximum age, Duff thinks the fossil record is essential. Furthermore, he states that an examination of the mitochondrial genome of any species will only tell us when the common ancestor of all modern members of this species existed, which will almost invariably occur after the evolutionary origin of the species.

But how does Duff know that this is true? Ive already documented that fossils do not directly record genealogical relationships; only DNA does. Why would Duff defer the genealogical question of ancestry (a.k.a. the question of the origin of species) to an indirect field of science (paleontology) when a direct field (geneticsmtDNA) gives a clear answer?

Ive also documented that the process of speciation involves several stepsat a minimum, (1) the formation of one or more distinct individuals, (2) the multiplication of these distinct individuals into a population, and (3) the isolation of this distinct population from the parent species. How does Duff know that the supposed ancestors (recorded by fossils) of modern species were isolated enough from the other populations alive at the time to be called a new species? Duff is trying to win a scientific argument, not by data and by experimentation, but by assertion. This is not a scientific way to resolve the controversy.

BioLogos response is sad, if not ironic. Weve already documented the fact that our evolutionary opponents dont read our literature (Duff included , despite BioLogos professed commitment to dialogue with those who hold other views); yet they call us liars. Sometimes I wonder if they carefully read even the evolutionary literature. Either way, BioLogos main critique (of the implications of Stoeckle and Thalers paper) amounts to misrepresentation and speculation even approaching outright denial. If this is the best that the evolutionary community can do, then perhaps my scientific conclusions (above) are even stronger than they first appear.

See more here:
Hundreds of Thousands of Species in a Few Thousand Years?

Read More...

New hybrid whale-dolphin discovered in Hawaii

Sunday, July 29th, 2018

Last year, a team of scientists spotted what they believed was a hybrid animal off the coast of Kauai, Hawaii.A new report from Cascadia Research Collectiveconfirms they did and the new sea creature is the result of a whale and a dolphin mating, the teams head researcher told CBS News.

What the researchers discovered was a hybrid of a melon-headed whale and a rough-toothed dolphin. In an interview with local newspaperThe Garden Island,the head of the project said the discovery is their most unusual finding. We had the photos and suspected it was a hybrid from morphological characteristics intermediate between species, Robin Baird said.

During their two-week project, scientists were able to get a biopsy sample from the creature and study its genetics. They were able to confirm that the animal was a hybrid. Based on the genetics, the father was a rough-toothed dolphin and the mother a melon-headed whale, Baird told CBS News via email.

One of the species that makes up this hybrid is very rare in Hawaii. Melon-headed whales usually dont swim in these waters, so when scientists spotted the whale, they put satellite tags on the animal. During this two-week study, scientists also spotted another rare species in the water, pantropical spotted dolphins, which they also tagged.

Bairds research team is going to be back in Kauais waters next month, when they hope to get more photos of the new hybrid whale-dolphin and water samples. They also hope to do testing on other species in the area.

Were hoping that just by talking to some tour operators and fishermen we might get tips and encounter something like pilot whales, Baird said.

See the article here:
New hybrid whale-dolphin discovered in Hawaii

Read More...

LR hospital hires cancer institute chief – arkansasonline.com

Sunday, July 29th, 2018

The outgoing director of the Winthrop P. Rockefeller Cancer Institute at the University of Arkansas for Medical Sciences has accepted a job at CHI St. Vincent.

Dr. Peter Emanuel turned in his resignation letter on May 4. His last day at UAMS is Tuesday.

Emanuel, 59, will join CHI St. Vincent on Sept. 1, according to a statement from the hospital. His position was not specified.

At the time of his resignation, he declined to give the reason for his departure, only citing unspecified challenges. He could not be reached for comment Friday afternoon.

UAMS is conducting a national search for a new cancer institute director, said Leslie Taylor, vice chancellor of communications and marketing. Dr. Laura Hutchins was appointed interim director in June. Hutchins is a professor in the College of Medicine Division of Hematology/Oncology, where she was division director from 1998 until September 2013.

Emanuel is a widely recognized expert in leukemia and lymphoma, a UAMS website states. He joined UAMS in 2007 after leaving the University of Alabama at Birmingham, where he was a professor of medicine, genetics and biochemistry.

From 2004 to 2006 he was the acting director of the National Cancer Institute-designated Comprehensive Cancer Center at the Alabama university.

During his time at UAMS, he oversaw the addition of the cancer institute's 12-story research and treatment tower, which opened in 2010. His annual salary was $500,000.

His resignation in May followed UAMS' decision to temporarily suspend its cardiac surgery program due to lead surgeon Dr. Gareth Tobler's retirement. That program restarted at the beginning of July, with the hospital contracting with four new physicians.

UAMS also laid off almost 260 employees in January to curb an anticipated $72.3 million deficit. Those layoffs included one full-time physician -- a general ear, nose and throat doctor who did not work at the cancer institute.

News of Emanuel's new role comes one day after an invoice that his wife, Carla Emanuel, sent seeking reimbursement from the Winthrop P. Rockefeller Cancer Institute became public.

The $4,000 bill lists events that she attended, phone calls she made and work she did to resolve problems with donors. Taylor said UAMS was not going to pay the bill because state procedures regarding vendors and invoices were not followed.

Taylor added that the invoice was the first one she was aware of from a spouse, and the institution does not normally reimburse people for attending fundraising events.

The Arkansas Times first reported on the invoice on Thursday.

Metro on 07/28/2018

Originally posted here:
LR hospital hires cancer institute chief - arkansasonline.com

Read More...

TOday’s Movers: Seattle Genetics (NASDAQ:SGEN) Stock …

Sunday, July 29th, 2018

July 27, 2018 - By Vernon Prom

Investors sentiment increased to 1.61 in Q1 2018. Its up 0.38, from 1.23 in 2017Q4. It is positive, as 24 investors sold Seattle Genetics, Inc. shares while 53 reduced holdings. 31 funds opened positions while 93 raised stakes. 159.52 million shares or 12.47% more from 141.83 million shares in 2017Q4 were reported.

California State Teachers Retirement System reported 165,312 shares. 13,084 are held by Bluecrest Cap Ltd. Pictet Asset invested in 0.1% or 786,323 shares. Swiss Bankshares owns 0.02% invested in Seattle Genetics, Inc. (NASDAQ:SGEN) for 349,100 shares. Keybank National Association Oh invested in 0% or 8,414 shares. 4,998 were accumulated by Shell Asset Mngmt Company. Pnc Financial holds 6,727 shares. Utah Retirement Sys holds 0.02% of its portfolio in Seattle Genetics, Inc. (NASDAQ:SGEN) for 19,600 shares. Wells Fargo And Co Mn, a California-based fund reported 306,681 shares. The Connecticut-based Bridgewater Associate L P has invested 0.01% in Seattle Genetics, Inc. (NASDAQ:SGEN). Amundi Pioneer Asset Management has 21,523 shares. National Bank Of America Corp De accumulated 496,573 shares. Daiwa Securities accumulated 4,395 shares. Zurcher Kantonalbank (Zurich Cantonalbank), Switzerland-based fund reported 23,953 shares. Pub Employees Retirement Association Of Colorado invested in 20,183 shares or 0.01% of the stock.

Since February 1, 2018, it had 3 buys, and 12 sales for $266.62 million activity. Cline Darren S also sold $497,983 worth of Seattle Genetics, Inc. (NASDAQ:SGEN) shares. The insider SIEGALL CLAY B sold 18,832 shares worth $951,393. The insider HIMES VAUGHN B sold 5,000 shares worth $290,604. 10,457 shares were sold by DRACHMAN JONATHAN G, worth $552,452.

JP Morgan now has a $77 target on the $11.55 billion market cap company or 5.51 % upside potential. In analysts note issued to clients on Friday, 27 July, Seattle Genetics (NASDAQ:SGEN) shares have had their Overweight Rating kept by professional analysts at JP Morgan.

Among 8 analysts covering Seattle Genetics (NASDAQ:SGEN), 7 have Buy rating, 0 Sell and 1 Hold. Therefore 88% are positive. Seattle Genetics has $77.0 highest and $60.0 lowest target. $68.13s average target is -6.65% below currents $72.98 stock price. Seattle Genetics had 12 analyst reports since January 31, 2018 according to SRatingsIntel. SunTrust maintained it with Hold rating and $60.0 target in Wednesday, February 7 report. The stock of Seattle Genetics, Inc. (NASDAQ:SGEN) earned Buy rating by Needham on Wednesday, February 7. J.P. Morgan upgraded the shares of SGEN in report on Wednesday, February 14 to Buy rating. The rating was maintained by Morgan Stanley on Wednesday, March 21 with Overweight. The firm has Buy rating by RBC Capital Markets given on Tuesday, March 20. The firm has Buy rating given on Monday, June 11 by SunTrust. The company was maintained on Wednesday, February 7 by H.C. Wainwright. On Thursday, February 1 the stock rating was maintained by H.C. Wainwright with Buy. The stock of Seattle Genetics, Inc. (NASDAQ:SGEN) has Neutral rating given on Wednesday, February 7 by Bank of America. The firm has Overweight rating by JP Morgan given on Wednesday, February 14.

The stock increased 2.43% or $1.73 during the last trading session, reaching $72.98. About 1.60M shares traded or 72.55% up from the average. Seattle Genetics, Inc. (NASDAQ:SGEN) has declined 15.50% since July 28, 2017 and is downtrending. It has underperformed by 28.07% the S&P500.

Seattle Genetics, Inc., a biotechnology company, develops and commercializes targeted therapies to treat cancer worldwide. The company has market cap of $11.55 billion. It markets ADCETRIS, an antibody-drug conjugate for relapsed Hodgkin lymphoma and relapsed systemic anaplastic large cell lymphoma. It currently has negative earnings. The firm also develops SGN-CD33A that is in Phase III clinical trial to evaluate SGN-CD33A in combination with hypomethylating agents in previously untreated older patients, as well as in Phase 1/2 clinical trial for patients with relapsed or refractory acute myeloid leukemia ; ASG-22ME, which is in Phase I clinical trial for Nectin-4-positive solid tumors, including bladder cancer; SGN-LIV1A that is in Phase 1 clinical trial for patients with LIV-1-positive metastatic breast cancer; and SGN-CD19A, which is in Phase II clinical trial for patients with relapsed DLBCL, as well as in Phase II trial for patients with newly diagnosed DLBCL.

More notable recent Seattle Genetics, Inc. (NASDAQ:SGEN) news were published by: Streetinsider.com which released: Seattle Genetics (SGEN) Adcetris On-going Launch in 1L cHL is Positive Says SunTrust. on July 02, 2018, also Benzinga.com with their article: Benzingas Daily Biotech Pulse: Biogen, AC Immune Slip Despite Positive Trials, Sarepta Slapped With Clinical Hold published on July 26, 2018, Seekingalpha.com published: Mid-stage study underway for Seattle Genetics tisotumab vedotin in solid tumors; shares up 1% premarket on July 12, 2018. More interesting news about Seattle Genetics, Inc. (NASDAQ:SGEN) were released by: Seekingalpha.com and their article: Dont Sell Axon Enterprise Cramers Lightning Round (7/11/18) published on July 12, 2018 as well as Benzinga.coms news article titled: Benzingas Daily Biotech Pulse: Achaogen To Trim Workforce By 28%, Amgens Beat-And-Raise Quarter with publication date: July 27, 2018.

Receive News & Ratings Via Email - Enter your email address below to receive a concise daily summary of the latest news and analysts' ratings with our FREE daily email newsletter.

See the original post here:
TOday's Movers: Seattle Genetics (NASDAQ:SGEN) Stock ...

Read More...

Seattle Genetics (SGEN) "Buy" Rating Reaffirmed Today By H …

Sunday, July 29th, 2018

July 27, 2018 - By Mona Holcomb

Investors sentiment increased to 1.61 in Q1 2018. Its up 0.38, from 1.23 in 2017Q4. It is positive, as 24 investors sold Seattle Genetics, Inc. shares while 53 reduced holdings. 31 funds opened positions while 93 raised stakes. 159.52 million shares or 12.47% more from 141.83 million shares in 2017Q4 were reported.

Pub Employees Retirement Association Of Colorado holds 20,183 shares. Jgp Global Gestao De Recursos Ltda reported 22,334 shares or 0.47% of all its holdings. Jane Street Gru Limited Company invested in 3,903 shares or 0% of the stock. Prudential Fincl, New Jersey-based fund reported 6,451 shares. 7,900 were reported by Ellington Management Grp. Caisse De Depot Et Placement Du Quebec reported 5,300 shares or 0% of all its holdings. D E Shaw & stated it has 168,554 shares. Td Asset owns 0.01% invested in Seattle Genetics, Inc. (NASDAQ:SGEN) for 154,016 shares. Virtu Fincl Ltd Liability Corporation reported 10,922 shares stake. Stratos Wealth Limited holds 0% of its portfolio in Seattle Genetics, Inc. (NASDAQ:SGEN) for 1,213 shares. World Asset Inc stated it has 3,870 shares. State Of Alaska Department Of Revenue reported 9,710 shares stake. Franklin Res has 108,400 shares for 0% of their portfolio. Zurcher Kantonalbank (Zurich Cantonalbank) owns 23,953 shares. 205,300 are owned by California Pub Employees Retirement.

Since February 1, 2018, it had 3 insider buys, and 12 insider sales for $266.62 million activity. $936,818 worth of stock was sold by SIEGALL CLAY B on Friday, February 9. On Thursday, March 15 HIMES VAUGHN B sold $290,604 worth of Seattle Genetics, Inc. (NASDAQ:SGEN) or 5,000 shares. On Wednesday, May 9 the insider Cline Darren S sold $497,983. DRACHMAN JONATHAN G sold $552,452 worth of stock or 10,457 shares.

EU: In an analyst report issued to investors and clients on 27 July, H.C. Wainwright reiterated their Buy rating on Seattle Genetics (SGEN) shares. They now have a $98.0 target price on the firm. H.C. Wainwrights target indicates a potential upside of 37.54 % from the companys last price.

The stock increased 2.04% or $1.45 during the last trading session, reaching $72.7. About 995,861 shares traded or 7.56% up from the average. Seattle Genetics, Inc. (SGEN) has declined 15.50% since July 27, 2017 and is downtrending. It has underperformed by 28.07% the S&P500.

Seattle Genetics, Inc., a biotechnology company, develops and commercializes targeted therapies to treat cancer worldwide. The company has market cap of $11.50 billion. It markets ADCETRIS, an antibody-drug conjugate for relapsed Hodgkin lymphoma and relapsed systemic anaplastic large cell lymphoma. It currently has negative earnings. The firm also develops SGN-CD33A that is in Phase III clinical trial to evaluate SGN-CD33A in combination with hypomethylating agents in previously untreated older patients, as well as in Phase 1/2 clinical trial for patients with relapsed or refractory acute myeloid leukemia ; ASG-22ME, which is in Phase I clinical trial for Nectin-4-positive solid tumors, including bladder cancer; SGN-LIV1A that is in Phase 1 clinical trial for patients with LIV-1-positive metastatic breast cancer; and SGN-CD19A, which is in Phase II clinical trial for patients with relapsed DLBCL, as well as in Phase II trial for patients with newly diagnosed DLBCL.

More notable recent Seattle Genetics, Inc. (NASDAQ:SGEN) news were published by: Streetinsider.com which released: Seattle Genetics (SGEN) Adcetris On-going Launch in 1L cHL is Positive Says SunTrust. on July 02, 2018, also Seekingalpha.com with their article: Dont Sell Axon Enterprise Cramers Lightning Round (7/11/18) published on July 12, 2018, Seekingalpha.com published: Mid-stage study underway for Seattle Genetics tisotumab vedotin in solid tumors; shares up 1% premarket on July 12, 2018. More interesting news about Seattle Genetics, Inc. (NASDAQ:SGEN) were released by: Benzinga.com and their article: Benzingas Daily Biotech Pulse: Biogen, AC Immune Slip Despite Positive Trials, Sarepta Slapped With Clinical Hold published on July 26, 2018 as well as Benzinga.coms news article titled: Benzingas Daily Biotech Pulse: Achaogen To Trim Workforce By 28%, Amgens Beat-And-Raise Quarter with publication date: July 27, 2018.

Receive News & Ratings Via Email - Enter your email address below to receive a concise daily summary of the latest news and analysts' ratings with our FREE daily email newsletter.

More here:
Seattle Genetics (SGEN) "Buy" Rating Reaffirmed Today By H ...

Read More...

Gregor Mendel – Wikipedia

Wednesday, June 27th, 2018

Gregor Johann Mendel (Czech: eho Jan Mendel;[1] 20 July 1822[2] 6 January 1884) (English: ) was a scientist, Augustinian friar and abbot of St. Thomas' Abbey in Brno, Margraviate of Moravia. Mendel was born in a German-speaking family[3] in the Silesian part of the Austrian Empire (today's Czech Republic) and gained posthumous recognition as the founder of the modern science of genetics. Though farmers had known for millennia that crossbreeding of animals and plants could favor certain desirable traits, Mendel's pea plant experiments conducted between 1856 and 1863 established many of the rules of heredity, now referred to as the laws of Mendelian inheritance.[4]

Mendel worked with seven characteristics of pea plants: plant height, pod shape and color, seed shape and color, and flower position and color. Taking seed color as an example, Mendel showed that when a true-breeding yellow pea and a true-breeding green pea were cross-bred their offspring always produced yellow seeds. However, in the next generation, the green peas reappeared at a ratio of 1 green to 3 yellow. To explain this phenomenon, Mendel coined the terms recessive and dominant in reference to certain traits. (In the preceding example, the green trait, which seems to have vanished in the first filial generation, is recessive and the yellow is dominant.) He published his work in 1866, demonstrating the actions of invisible factorsnow called genesin predictably determining the traits of an organism.

The profound significance of Mendel's work was not recognized until the turn of the 20th century (more than three decades later) with the rediscovery of his laws.[5] Erich von Tschermak, Hugo de Vries, Carl Correns and William Jasper Spillman independently verified several of Mendel's experimental findings, ushering in the modern age of genetics.[4]

Mendel was born into a German-speaking family in Hynice (Heinzendorf bei Odrau in German), at the Moravian-Silesian border, Austrian Empire (now a part of the Czech Republic).[3] He was the son of Anton and Rosine (Schwirtlich) Mendel and had one older sister, Veronika, and one younger, Theresia. They lived and worked on a farm which had been owned by the Mendel family for at least 130 years.[6] During his childhood, Mendel worked as a gardener and studied beekeeping. As a young man, he attended gymnasium in Opava (called Troppau in German). He had to take four months off during his gymnasium studies due to illness. From 1840 to 1843, he studied practical and theoretical philosophy and physics at the Philosophical Institute of the University of Olomouc, taking another year off because of illness. He also struggled financially to pay for his studies, and Theresia gave him her dowry. Later he helped support her three sons, two of whom became doctors.

He became a friar in part because it enabled him to obtain an education without having to pay for it himself. As the son of a struggling farmer, the monastic life, in his words, spared him the "perpetual anxiety about a means of livelihood."[8] He was given the name Gregor (eho in Czech)[1] when he joined the Augustinian friars.

When Mendel entered the Faculty of Philosophy, the Department of Natural History and Agriculture was headed by Johann Karl Nestler who conducted extensive research of hereditary traits of plants and animals, especially sheep. Upon recommendation of his physics teacher Friedrich Franz,[10] Mendel entered the Augustinian St Thomas's Abbey in Brno (called Brnn in German) and began his training as a priest. Born Johann Mendel, he took the name Gregor upon entering religious life. Mendel worked as a substitute high school teacher. In 1850, he failed the oral part, the last of three parts, of his exams to become a certified high school teacher. In 1851, he was sent to the University of Vienna to study under the sponsorship of Abbot C. F. Napp so that he could get more formal education. At Vienna, his professor of physics was Christian Doppler.[12] Mendel returned to his abbey in 1853 as a teacher, principally of physics. In 1856, he took the exam to become a certified teacher and again failed the oral part. In 1867, he replaced Napp as abbot of the monastery.[13]

After he was elevated as abbot in 1868, his scientific work largely ended, as Mendel became overburdened with administrative responsibilities, especially a dispute with the civil government over its attempt to impose special taxes on religious institutions.[14] Mendel died on 6 January 1884, at the age of 61, in Brno, Moravia, Austria-Hungary (now Czech Republic), from chronic nephritis. Czech composer Leo Janek played the organ at his funeral. After his death, the succeeding abbot burned all papers in Mendel's collection, to mark an end to the disputes over taxation.[15]

Gregor Mendel, who is known as the "father of modern genetics", was inspired by both his professors at the Palack University, Olomouc (Friedrich Franz and Johann Karl Nestler), and his colleagues at the monastery (such as Franz Diebl) to study variation in plants. In 1854, Napp authorized Mendel to carry out a study in the monastery's 2 hectares (4.9 acres) experimental garden,[16] which was originally planted by Napp in 1830.[13] Unlike Nestler, who studied hereditary traits in sheep, Mendel used the common edible pea and started his experiments in 1856.

After initial experiments with pea plants, Mendel settled on studying seven traits that seemed to be inherited independently of other traits: seed shape, flower color, seed coat tint, pod shape, unripe pod color, flower location, and plant height. He first focused on seed shape, which was either angular or round. Between 1856 and 1863 Mendel cultivated and tested some 28,000 plants, the majority of which were pea plants (Pisum sativum).[18][19][20] This study showed that, when true-breeding different varieties were crossed to each other (e.g., tall plants fertilized by short plants), in the second generation, one in four pea plants had purebred recessive traits, two out of four were hybrids, and one out of four were purebred dominant. His experiments led him to make two generalizations, the Law of Segregation and the Law of Independent Assortment, which later came to be known as Mendel's Laws of Inheritance.[21]

Mendel presented his paper, "Versuche ber Pflanzenhybriden" ("Experiments on Plant Hybridization"), at two meetings of the Natural History Society of Brno in Moravia on 8 February and 8 March 1865. It generated a few favorable reports in local newspapers,[23] but was ignored by the scientific community. When Mendel's paper was published in 1866 in Verhandlungen des naturforschenden Vereines in Brnn,[24] it was seen as essentially about hybridization rather than inheritance, had little impact, and was only cited about three times over the next thirty-five years. His paper was criticized at the time, but is now considered a seminal work.[25] Notably, Charles Darwin was unaware of Mendel's paper, and it is envisaged that if he had, genetics as we know it now might have taken hold much earlier.[26][27] Mendel's scientific biography thus provides an example of the failure of obscure, highly original, innovators to receive the attention they deserve.[28]

Mendel began his studies on heredity using mice. He was at St. Thomas's Abbey but his bishop did not like one of his friars studying animal sex, so Mendel switched to plants. Mendel also bred bees in a bee house that was built for him, using bee hives that he designed.[30] He also studied astronomy and meteorology,[13] founding the 'Austrian Meteorological Society' in 1865.[12] The majority of his published works was related to meteorology.[12]

Mendel also experimented with hawkweed (Hieracium)[31] and honeybees. He published a report on his work with hawkweed,[32] a group of plants of great interest to scientists at the time because of their diversity. However, the results of Mendel's inheritance study in hawkweeds was unlike his results for peas; the first generation was very variable and many of their offspring were identical to the maternal parent. In his correspondence with Carl Ngeli he discussed his results but was unable to explain them.[31] It was not appreciated until the end of the nineteen century that many hawkweed species were apomictic, producing most of their seeds through an asexual process.

None of his results on bees survived, except for a passing mention in the reports of Moravian Apiculture Society.[33] All that is known definitely is that he used Cyprian and Carniolan bees,[34] which were particularly aggressive to the annoyance of other monks and visitors of the monastery such that he was asked to get rid of them.[35] Mendel, on the other hand, was fond of his bees, and referred to them as "my dearest little animals".[36]

He also described novel plant species, and these are denoted with the botanical author abbreviation "Mendel".[37]

It would appear that the forty odd scientists who listened to Mendel's two path-breaking lectures failed to understand his work. Later, he also carried a correspondence with Carl Naegeli, one of the leading biologists of the time, but Naegli too failed to appreciate Mendel's discoveries. At times, Mendel must have entertained doubts about his work, but not always: "My time will come," he reportedly told a friend.[8]

During Mendel's lifetime, most biologists held the idea that all characteristics were passed to the next generation through blending inheritance, in which the traits from each parent are averaged. Instances of this phenomenon are now explained by the action of multiple genes with quantitative effects. Charles Darwin tried unsuccessfully to explain inheritance through a theory of pangenesis. It was not until the early twentieth century that the importance of Mendel's ideas was realized.

By 1900, research aimed at finding a successful theory of discontinuous inheritance rather than blending inheritance led to independent duplication of his work by Hugo de Vries and Carl Correns, and the rediscovery of Mendel's writings and laws. Both acknowledged Mendel's priority, and it is thought probable that de Vries did not understand the results he had found until after reading Mendel.[5] Though Erich von Tschermak was originally also credited with rediscovery, this is no longer accepted because he did not understand Mendel's laws.[38] Though de Vries later lost interest in Mendelism, other biologists started to establish modern genetics as a science.[5] All three of these researchers, each from a different country, published their rediscovery of Mendel's work within a two-month span in the Spring of 1900.

Mendel's results were quickly replicated, and genetic linkage quickly worked out. Biologists flocked to the theory; even though it was not yet applicable to many phenomena, it sought to give a genotypic understanding of heredity which they felt was lacking in previous studies of heredity which focused on phenotypic approaches.[40] Most prominent of these previous approaches was the biometric school of Karl Pearson and W. F. R. Weldon, which was based heavily on statistical studies of phenotype variation. The strongest opposition to this school came from William Bateson, who perhaps did the most in the early days of publicising the benefits of Mendel's theory (the word "genetics", and much of the discipline's other terminology, originated with Bateson). This debate between the biometricians and the Mendelians was extremely vigorous in the first two decades of the twentieth century, with the biometricians claiming statistical and mathematical rigor,[41] whereas the Mendelians claimed a better understanding of biology.[42][43] (Modern genetics shows that Mendelian heredity is in fact an inherently biological process, though not all genes of Mendel's experiments are yet understood.)[44][45]

In the end, the two approaches were combined, especially by work conducted by R. A. Fisher as early as 1918. The combination, in the 1930s and 1940s, of Mendelian genetics with Darwin's theory of natural selection resulted in the modern synthesis of evolutionary biology.[46][47]

In 1936, R.A. Fisher, a prominent statistician and population geneticist, reconstructed Mendel's experiments, analyzed results from the F2 (second filial) generation and found the ratio of dominant to recessive phenotypes (e.g. green versus yellow peas; round versus wrinkled peas) to be implausibly and consistently too close to the expected ratio of 3 to 1.[48][49][50] Fisher asserted that "the data of most, if not all, of the experiments have been falsified so as to agree closely with Mendel's expectations,"[48] Mendel's alleged observations, according to Fisher, were "abominable", "shocking",[51] and "cooked".[52]

Other scholars agree with Fisher that Mendel's various observations come uncomfortably close to Mendel's expectations. Dr. Edwards,[53] for instance, remarks: "One can applaud the lucky gambler; but when he is lucky again tomorrow, and the next day, and the following day, one is entitled to become a little suspicious". Three other lines of evidence likewise lend support to the assertion that Mendels results are indeed too good to be true.[54]

Fisher's analysis gave rise to the Mendelian Paradox, a paradox that remains unsolved to this very day. Thus, on the one hand, Mendel's reported data are, statistically speaking, too good to be true; on the other, "everything we know about Mendel suggests that he was unlikely to engage in either deliberate fraud or in unconscious adjustment of his observations."[54] A number of writers have attempted to resolve this paradox.

One attempted explanation invokes confirmation bias.[55] Fisher accused Mendel's experiments as "biased strongly in the direction of agreement with expectation... to give the theory the benefit of doubt".[48] This might arise if he detected an approximate 3 to 1 ratio early in his experiments with a small sample size, and, in cases where the ratio appeared to deviate slightly from this, continued collecting more data until the results conformed more nearly to an exact ratio.

In his 2004, J.W. Porteous concluded that Mendel's observations were indeed implausible.[56] However, reproduction of the experiments has demonstrated that there is no real bias towards Mendel's data.[57]

Another attempt[54] to resolve the Mendelian Paradox notes that a conflict may sometimes arise between the moral imperative of a bias-free recounting of one's factual observations and the even more important imperative of advancing scientific knowledge. Mendel might have felt compelled to simplify his data in order to meet real, or feared, editorial objections.[53] Such an action could be justified on moral grounds (and hence provide a resolution to the Mendelian Paradox), since the alternativerefusing to complymight have retarded the growth of scientific knowledge. Similarly, like so many other obscure innovators of science,[53][28] Mendel, a little known innovator of working-class background, had to break through the cognitive paradigms and social prejudices of his audience.[53] If such a breakthrough could be best achieved by deliberately omitting some observations from his report and adjusting others to make them more palatable to his audience, such actions could be justified on moral grounds.[54]

Daniel L. Hartl and Daniel J. Fairbanks reject outright Fisher's statistical argument, suggesting that Fisher incorrectly interpreted Mendel's experiments. They find it likely that Mendel scored more than 10 progeny, and that the results matched the expectation. They conclude: "Fisher's allegation of deliberate falsification can finally be put to rest, because on closer analysis it has proved to be unsupported by convincing evidence."[51][58] In 2008 Hartl and Fairbanks (with Allan Franklin and AWF Edwards) wrote a comprehensive book in which they concluded that there were no reasons to assert Mendel fabricated his results, nor that Fisher deliberately tried to diminish Mendel's legacy.[59] Reassessment of Fisher's statistical analysis, according to these authors, also disprove the notion of confirmation bias in Mendel's results.[60][61]

More here:
Gregor Mendel - Wikipedia

Read More...

Plant genetics – Wikipedia

Tuesday, June 26th, 2018

Plant genetics is the study of genes, genetic variation, and heredity specifically in Plants.[1][2] It is generally considered a field of biology and botany, but intersects frequently with many other life sciences and is strongly linked with the study of information systems. Plant genetics is similar in many ways to animal genetics but differs in a few key areas.

The discoverer of genetics is Gregor Mendel, a late 19th-century scientist and Augustinian friar. Mendel studied "trait inheritance", patterns in the way traits are handed down from parents to offspring. He observed that organisms (pea plants) inherit traits by way of discrete "units of inheritance". This term, still used today, is a somewhat ambiguous definition of what is referred to as a gene. Much of Mendel's work with plants still forms the basis for modern plant genetics.

Plants, like all known organisms, use DNA to pass on their traits. Animal genetics often focuses on parentage and lineage, but this can sometimes be difficult in plant genetics due to the fact that plants can, unlike most animals, can self-fertilize. Speciation can be easier in many plants due to unique genetic abilities, such as being well adapted to polyploidy. Plants are unique in that they are able to make their own food via photosynthesis, a process which is achieved by use of a structure mostly exclusive to plants: chloroplasts. Chloroplasts, like the superficially similar mitochondria, possess their own DNA. Chloroplasts thus provide an additional reservoir for genes and genetic diversity, and an extra layer of genetic complexity not found in animals.

The study of plant genetics has major economic impacts: many staple crops are genetically modified to increase yields, confer pest and disease resistance, provide resistance to herbicides, or to increase their nutritional value.

The field of plant genetics began with the work of Gregor Mendel, who is often called the "father of genetics". He was an Augustinian priest and scientist born on 20 July 1822 in Austria-Hungary. He worked at the Abbey of St. Thomas in Brno , where his organism of choice for studying inheritance and traits was the pea plant. Mendel's work tracked many phenotypic traits of pea plants, such as their height, flower color, and seed characteristics. Mendel showed that the inheritance of these traits follows particular laws, which were later named after him. His seminal work on genetics was published in 1866, but went almost entirely unnoticed until 1900. Mendel died in 1884. The significance of Mendel's work was not recognized until the turn of the 20th century. Its rediscovery prompted the foundation of modern genetics.

Deoxyribonucleic acid (DNA) is a nucleic acid that contains the genetic instructions used in the development and functioning of all known living organisms and some viruses. The main role of DNA molecules is the long-term storage of information. DNA is often compared to a set of blueprints or a recipe, or a code, since it contains the instructions needed to construct other components of cells, such as proteins and RNA molecules. The DNA segments that carry this genetic information are called genes, but other DNA sequences have structural purposes, or are involved in regulating the use of this genetic information. Geneticists, including plant geneticists, use this sequencing of DNA to their advantage as they splice and delete certain genes and regions of the DNA molecule to produce a different or desired genotype and thus, also producing a different phenotype.

Plants, like all other known living organisms, pass on their traits using DNA. Plants however are unique from other living organisms in the fact that they have Chloroplasts. Like mitochondria, chloroplasts have their own DNA. Like animals, plants experience somatic mutations regularly, but these mutations can contribute to the germ line with ease, since flowers develop at the ends of branches composed of somatic cells. People have known of this for centuries, and mutant branches are called "sports". If the fruit on the sport is economically desirable, a new cultivar may be obtained.

Some plant species are capable of self-fertilization, and some are nearly exclusively self-fertilizers. This means that a plant can be both mother and father to its offspring, a rare occurrence in animals. Scientists and hobbyists attempting to make crosses between different plants must take special measures to prevent the plants from self-fertilizing. In plant breeding, people create hybrids between plant species for economic and aesthetic reasons. For example, the yield of Corn has increased nearly five-fold in the past century due in part to the discovery and proliferation of hybrid corn varieties.[3] Plant genetics can be used to predict which combination of plants may produce a plant with Hybrid vigor, or conversely many discoveries in Plant genetics have come from studying the effects of hybridization.

Plants are generally more capable of surviving, and indeed flourishing, as polyploids. Polyploid organisms have more than two sets of homologous chromosomes. For example, humans have two sets of homologous chromosomes, meaning that a typical human will have 2 copies each of 23 different chromosomes, for a total of 46. Wheat on the other hand, while having only 7 distinct chromosomes, is considered a hexaploid and has 6 copies of each chromosome, for a total of 42.[4] In animals, inheritable germline polyploidy is less common, and spontaneous chromosome increases may not even survive past fertilization. In plants however this is no such problem, polyploid individuals are created frequently by a variety of processes, however once created usually cannot cross back to the parental type. Polyploid individuals, if capable of self-fertilizing, can give rise to a new genetically distinct lineage, which can be the start of a new species. This is often called "instant speciation". Polyploids generally have larger fruit, an economically desirable trait, and many human food crops, including wheat, maize, potatoes, peanuts,[5] strawberries and tobacco, are either accidentally or deliberately created polyploids.

Arabidopsis thaliana, also known as thale cress, has been the model organism for the study of plant genetics. As Drosphila, a species of fruit fly, was to the understanding of early genetics, so has been arabidopsis to the understanding of plant genetics.

Genetically modified (GM) foods are produced from organisms that have had changes introduced into their DNA using the methods of genetic engineering. Genetic engineering techniques allow for the introduction of new traits as well as greater control over traits than previous methods such as selective breeding and mutation breeding.[6]

Genetically modifying plants is an important economic activity: in 2017, 89% of corn, 94% of soybeans, and 91% of cotton produced in the US were from genetically modified strains[7]. Since the introduction of GM crops, yields have increased by 22%, and profits have increased to farmers, especially in the developing world, by 68%. An important side effect of GM crops has been decreased land requirements, [8]

Commercial sale of genetically modified foods began in 1994, when Calgene first marketed its unsuccessful Flavr Savr delayed-ripening tomato.[9][10] Most food modifications have primarily focused on cash crops in high demand by farmers such as soybean, corn, canola, and cotton. Genetically modified crops have been engineered for resistance to pathogens and herbicides and for better nutrient profiles.[11] Other such crops include the economically important GM papaya which are resistant to the highly destructive Papaya ringspot virus, and the nutritionally improved golden rice (it is however still in development).[12]

There is a scientific consensus[13][14][15][16] that currently available food derived from GM crops poses no greater risk to human health than conventional food,[17][18][19][20][21] but that each GM food needs to be tested on a case-by-case basis before introduction.[22][23] Nonetheless, members of the public are much less likely than scientists to perceive GM foods as safe.[24][25][26][27] The legal and regulatory status of GM foods varies by country, with some nations banning or restricting them, and others permitting them with widely differing degrees of regulation.[28][29][30][31] There are still ongoing public concerns related to food safety, regulation, labeling, environmental impact, research methods, and the fact that some GM seeds are subject to intellectual property rights owned by corporations.[32]

Genetic modification has been the cause for much research into modern plant genetics, and has also lead to the sequencing of many plant genomes. Today there are two predominant procedures of transforming genes in organisms: the "Gene gun" method and the Agrobacterium method.

The gene gun method is also referred to as "biolistics" (ballistics using biological components). This technique is used for in vivo (within a living organism) transformation and has been especially useful in monocot species like corn and rice.This approach literally shoots genes into plant cells and plant cell chloroplasts. DNA is coated onto small particles of gold or tungsten approximately two micrometres in diameter. The particles are placed in a vacuum chamber and the plant tissue to be engineered is placed below the chamber. The particles are propelled at high velocity using a short pulse of high pressure helium gas, and hit a fine mesh baffle placed above the tissue while the DNA coating continues into any target cell or tissue.

Transformation via Agrobacterium has been successfully practiced in dicots, i.e. broadleaf plants, such as soybeans and tomatoes, for many years. Recently it has been adapted and is now effective in monocots like grasses, including corn and rice. In general, the Agrobacterium method is considered preferable to the gene gun, because of a greater frequency of single-site insertions of the foreign DNA, which allows for easier monitoring. In this method, the tumor inducing (Ti) region is removed from the T-DNA (transfer DNA) and replaced with the desired gene and a marker, which is then inserted into the organism. This may involve direct inoculation of the tissue with a culture of transformed Agrobacterium, or inoculation following treatment with micro-projectile bombardment, which wounds the tissue.[33] Wounding of the target tissue causes the release of phenolic compounds by the plant, which induces invasion of the tissue by Agrobacterium. Because of this, microprojectile bombardment often increases the efficiency of infection with Agrobacterium. The marker is used to find the organism which has successfully taken up the desired gene. Tissues of the organism are then transferred to a medium containing an antibiotic or herbicide, depending on which marker was used. The Agrobacterium present is also killed by the antibiotic. Only tissues expressing the marker will survive and possess the gene of interest. Thus, subsequent steps in the process will only use these surviving plants. In order to obtain whole plants from these tissues, they are grown under controlled environmental conditions in tissue culture. This is a process of a series of media, each containing nutrients and hormones. Once the plants are grown and produce seed, the process of evaluating the progeny begins. This process entails selection of the seeds with the desired traits and then retesting and growing to make sure that the entire process has been completed successfully with the desired results.

Domingo, Jos L.; Bordonaba, Jordi Gin (2011). "A literature review on the safety assessment of genetically modified plants" (PDF). Environment International. 37: 734742. doi:10.1016/j.envint.2011.01.003. PMID21296423.

Krimsky, Sheldon (2015). "An Illusory Consensus behind GMO Health Assessment" (PDF). Science, Technology, & Human Values. 40: 132. doi:10.1177/0162243915598381.

And contrast:

Panchin, Alexander Y.; Tuzhikov, Alexander I. (January 14, 2016). "Published GMO studies find no evidence of harm when corrected for multiple comparisons". Critical Reviews in Biotechnology: 15. doi:10.3109/07388551.2015.1130684. PMID26767435.

and

Yang, Y.T.; Chen, B. (2016). "Governing GMOs in the USA: science, law and public health". Journal of the Science of Food and Agriculture. 96: 18511855. doi:10.1002/jsfa.7523. PMID26536836.

Pinholster, Ginger (October 25, 2012). "AAAS Board of Directors: Legally Mandating GM Food Labels Could "Mislead and Falsely Alarm Consumers"". American Association for the Advancement of Science. Retrieved February 8, 2016.

Go here to read the rest:
Plant genetics - Wikipedia

Read More...

Myriad Genetics (MYGN) versus Quotient (QTNT) Head-To-Head …

Tuesday, June 26th, 2018

Myriad Genetics (NASDAQ: MYGN) and Quotient (NASDAQ:QTNT) are both medical companies, but which is the superior stock? We will contrast the two businesses based on the strength of their profitability, dividends, analyst recommendations, earnings, institutional ownership, risk and valuation.

Risk & Volatility

Myriad Genetics has a beta of 0.55, meaning that its stock price is 45% less volatile than the S&P 500. Comparatively, Quotient has a beta of 0.25, meaning that its stock price is 75% less volatile than the S&P 500.

This table compares Myriad Genetics and Quotients net margins, return on equity and return on assets.

Insider & Institutional Ownership

61.5% of Quotient shares are owned by institutional investors. 6.7% of Myriad Genetics shares are owned by company insiders. Comparatively, 29.0% of Quotient shares are owned by company insiders. Strong institutional ownership is an indication that endowments, large money managers and hedge funds believe a stock is poised for long-term growth.

Analyst Recommendations

This is a summary of current ratings and price targets for Myriad Genetics and Quotient, as reported by MarketBeat.

Myriad Genetics currently has a consensus price target of $30.91, suggesting a potential downside of 20.48%. Quotient has a consensus price target of $11.50, suggesting a potential upside of 30.68%. Given Quotients stronger consensus rating and higher probable upside, analysts plainly believe Quotient is more favorable than Myriad Genetics.

Earnings and Valuation

This table compares Myriad Genetics and Quotients gross revenue, earnings per share and valuation.

Myriad Genetics has higher revenue and earnings than Quotient. Quotient is trading at a lower price-to-earnings ratio than Myriad Genetics, indicating that it is currently the more affordable of the two stocks.

Summary

Myriad Genetics beats Quotient on 8 of the 13 factors compared between the two stocks.

About Myriad Genetics

Myriad Genetics, Inc., a molecular diagnostic company, focuses on developing and marketing novel predictive medicine, personalized medicine, and prognostic medicine tests worldwide. The company offers molecular diagnostic tests, including myRisk Hereditary Cancer, a DNA sequencing test for hereditary cancers; BRACAnalysis, a DNA sequencing test to assess the risk of developing breast and ovarian cancer; BART, a DNA sequencing test for hereditary breast and ovarian cancer; BRACAnalysis CDx, a DNA sequencing test for use as a companion diagnostic with the platinum based chemotherapy agents and poly ADP ribose inhibitor Lynparza; and Tumor BRACAnalysis CDx, a DNA sequencing test that is designed to be utilized to predict response to DNA damaging agents. It also provides COLARIS, a DNA sequencing test for colorectal and uterine cancer; COLARIS AP, a DNA sequencing test for colorectal cancer; Vectra DA, a protein quantification test for assessing the disease activity of rheumatoid arthritis; Prolaris, a RNA expression test for assessing the aggressiveness of prostate cancer; and EndoPredict, a RNA expression test for assessing the aggressiveness of breast cancer. In addition, the company offers myPath Melanoma, a RNA expression test for diagnosing melanoma; myChoice HRD, a companion diagnostic to measure three modes of homologous recombination deficiency; and GeneSight, a DNA genotyping test to optimize psychotropic drug selection for neuroscience patients. Further, it provides biomarker discovery, and pharmaceutical and clinical services to the pharmaceutical, biotechnology, and medical research industries; and operates an internal medicine emergency hospital primarily for internal medicine and hemodialysis. The company has collaboration with AstraZeneca for the development of an indication for BRACAnalysis CDx. Myriad Genetics, Inc. was founded in 1991 and is headquartered in Salt Lake City, Utah.

About Quotient

Quotient Limited, a commercial-stage diagnostics company, develops, manufactures, and commercializes conventional reagent products used for blood grouping in the transfusion diagnostics market worldwide. The company is developing MosaiQ, a proprietary technology platform, which provides tests for blood grouping and serological disease screening. It also develops, manufactures, and commercializes conventional reagent products for blood grouping, including antisera products that are used to identify blood-group antigens; reagent red blood cells, which enable the identification of blood-group antibodies; whole blood control products for use as daily quality assurance tests; and ancillary products that are used to support blood grouping. The company sells its products to donor collection agencies and testing laboratories, hospitals, independent patient testing laboratories, reference laboratories, blood banking operations, and other diagnostic companies, as well as to original equipment manufacturers. Quotient Limited was founded in 2007 and is based in Penicuik, the United Kingdom.

Receive News & Ratings for Myriad Genetics Daily - Enter your email address below to receive a concise daily summary of the latest news and analysts' ratings for Myriad Genetics and related companies with MarketBeat.com's FREE daily email newsletter.

See the original post:
Myriad Genetics (MYGN) versus Quotient (QTNT) Head-To-Head ...

Read More...

Page 34«..1020..33343536..40..»


2024 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick