header logo image


Page 37«..1020..36373839..»

Archive for the ‘Genetics’ Category

Genetics may have a role in Uddanam kidney disease, claim Harvard experts – The News Minute

Tuesday, August 1st, 2017

The News Minute
Genetics may have a role in Uddanam kidney disease, claim Harvard experts
The News Minute
A two-member committee from Harvard University has found that genetic predisposition and a strong genetic founder effect could be the reason for nephropathy (kidney disease) in Uddanam, according to a Times of India report. The hypothesis is based on ...

and more »

Follow this link:
Genetics may have a role in Uddanam kidney disease, claim Harvard experts - The News Minute

Read More...

The DNA of ancient Canaanites lives on in modern-day Lebanese, genetic analysis shows – Los Angeles Times

Tuesday, August 1st, 2017

The Canaanites lived at the crossroads of the ancient world. They experienced wars, conquests and occupations for millennia, and as a result evolutionary geneticists expected that their DNA would become substantially mixed with incoming populations.

Astonishingly, new genetic analysis shows that scientists were wrong. According to a new study in the American Journal of Human Genetics, todays Lebanese share a whopping 93% of their DNA with the ancient Canaanites.

The study also found that the Bronze Age inhabitants of Sidon, a major Canaanite city-state in modern-day Lebanon, have the same genetic profile as people living 300 to 800 years earlier in present-day Jordan.

Later known as Phoenicians, the Canaanites have a murky past. Nearly all of their own records have been destroyed over the centuries, so their history has been mostly pieced together from archaeological records and the writings of other ancient peoples.

Archaeologists at the Sidon excavation site have been unearthing ancient Canaanite secrets for the last 19 years in the still-inhabited Lebanese port city. The team has uncovered 160 burials from the Canaanite period alone, said Claude Doumet-Serhal, director of the excavation. They have found people of all ages in these Canaanite burials, she said children were buried in jars and adults were placed in sand.

Claude Doumet-Serhal / The Sidon Excavation

An aerial view of the Sidon excavation site.

An aerial view of the Sidon excavation site. (Claude Doumet-Serhal / The Sidon Excavation)

Aided by new DNA sampling techniques, a team of evolutionary geneticists including Marc Haber and Chris Tyler-Smith from the Wellcome Trust Sanger Institute stepped in.

They sequenced the whole genomes of five individuals found in Sidon who lived about 3,700 years ago. The team then compared the genomes of these ancient Canaanites with those of 99 Lebanese people currently living in the country, along with the previously published genetic information from modern and ancient populations across Europe and Asia.

First, they investigated the genetic ancestry of the Canaanites themselves. They found that these Bronze Age inhabitants of Sidon shared about half their DNA with local Neolithic peoples and the other half with Chalcolithic Iranians. Interestingly, this genetic profile is nearly identical to the one evolutionary geneticist Iosif Lazaridis and his team found last year in Bronze Age villagers near Ain Ghazal in modern-day Jordan.

This suggests that Canaanite-related ancestry was spread across a wide region during the Bronze Age and was shared between urban societies on the coast and farming societies further inland. This evidence supports the idea that different Levantine cultural groups such as the Moabites, Israelites, and Phoenicians may have had a common genetic background, the authors said.

The researchers were also able to determine that the genetic mixing of the Levantine and Iranian peoples happened between 6,600 and 3,550 years ago, a range they would be able to narrow down with more ancient DNA samples from the region.

Claude Doumet-Serhal / The Sidon Excavation

The buried remains of a Canaanite adult whose DNA was sequenced in the study.

The buried remains of a Canaanite adult whose DNA was sequenced in the study. (Claude Doumet-Serhal / The Sidon Excavation)

Next, the team wanted to compare the Canaanite genome with the genetic makeup of the people who currently inhabit the ancient Canaanite cities. To do this, they collected DNA from 99 Lebanese people Druze, Muslim, and Christian alike.

As expected, they found some new additions to the modern Lebanese genome since the Bronze Age. About 7% of modern Lebanese DNA originates from eastern Steppe peoples found in what is now Russia, but wasnt represented in the Bronze Age Canaanites or their ancestors. What surprised the team was what was missing from their genetic data.

If you look at the history of Lebanon after the Bronze Age, especially it had a lot of conquests, Haber said. He and Tyler-Smith expected to see greater genetic contributions from multiple conquering peoples, and were surprised that as much as 93% of the Lebanese genome is shared with their Canaanite predecessors.

Though a 7% genetic influx from the Steppe seems very small, that number might be covering some hidden complexities, said Lazaridis, who worked on the Bronze Age Jordanian samples but was not involved in the new study.

Not much is known about the migrations of these eastern Steppe populations, he said. If the genomes of the incoming people were only half Steppe, for example, 14% of the Lebanese genome could have come from the new migrants.

Haber and Tyler-Smith said they want to explore this complexity further. Who were those eastern migrants? Where did they come from? And why did they migrate toward the Levant region? Haber asked. Analyzing more samples from different locations and periods could lead to an answer.

The team also wanted to know if the individuals from Sidon are more similar to modern-day Lebanese than to other modern Eurasian populations.

Despite small genetic variations between the three religious groups caused by preferential mating over time, the Lebanese genome is not widely varied. As a whole, the Lebanese people have more genetic overlap with the Canaanites from Sidon than do other modern Middle Eastern populations such as Jordanians, Syrians or Palestinians.

The difference is small, but its possible that the Lebanese population has remained more isolated over time from an influx of African DNA than other Levantine peoples, Lazaridis suggested.

Claude Doumet-Serhal - The Sidon Excavation

An archaeologist sorts pottery at the Sidon excavation site.

An archaeologist sorts pottery at the Sidon excavation site. (Claude Doumet-Serhal - The Sidon Excavation)

The findings have powerful cultural implications, Doumet-Serhal said. In a country struggling with the ramifications of war and a society fiercely divided along political and sectarian lines, religious groups have often looked to an uncertain history for their identities.

When Lebanon started in 1929, Doumet-Serhal said, the Christians said, We are Phoenician. The Muslims didnt accept that and they said, No, we are Arab.

But from this work comes a message of unity. We all belong to the same people, Doumet-Serhal said. We have always had a difficult past but we have a shared heritage we have to preserve.

mira.abed@latimes.com

Twitter: @mirakatherine

ALSO:

Fake news about statins is discouraging the use of these life-saving drugs, expert warns

See original here:
The DNA of ancient Canaanites lives on in modern-day Lebanese, genetic analysis shows - Los Angeles Times

Read More...

Genetics and Genomics in Racing: Speed Isn’t Everything – TheHorse.com

Wednesday, July 12th, 2017

Testing mares and stallions can help ensure foals are born free or at low risk of some genetic diseases.

Photo: iStock

Genetics refers to the study of genes and the way traits of conditions are passed down from one generation to another. Genomics, on the other hand, describes the study of all genes (the genome) including interactions of genes with each other and the environment. Although much of the genetic and genomic research done in Thoroughbreds is applied to racing performance, the full breadth of application of genetic and genomic research goes beyond that of faster horses.

Genetics and genomics allow for a more complete understanding of both simple and complex diseases. From a genetic perspective, simple is a term used to describe a disease that follows a single gene pattern of inheritance. These diseases are controlled by one gene, with other genes and outside factors having very little influence (i.e., the presence of the gene = disease). Diseases inherited this way are typically qualitative, where an animal either has the disease or it doesnt (i.e., lethal white foal syndrome).

Complex diseases, on the other hand, are usually controlled by not one, but many different genes and are often affected by environmental factors, such as nutrition and living conditions (e.g., cervical vertebral stenotic myelopathy, or wobbler syndrome). This combination of both genetic and environmental factors results in complex or multifactorial diseases. Basically, three different scenarios determine the manifestation of a complex disease:

As a result, complex genetic diseases can be extremely difficult to diagnose early and/or prevent using tradition methods such as pedigree analyses and veterinary evaluations. In some instances, a simple disease might even be classified as complex based on the inability of epidemiological studies and pedigree analyses to find common factors among cases.

Hydrocephalus, for example, is a developmental disorder that often results in stillbirth of foals and dystocia (difficult birth) in dams. Possible causes of the defect in horses could not be prove based on field data and pedigree analyses suggested the disorder to be complex. With this in mind, a genomic scan of 82 horses (13 cases and 69 controls) was performed and a small section of the genome was identified. Genomic sequencing was then performed on 10 horses (four cases and six controls) and the genetic cause of the disorder was pinpointed.

Figure 1

Two copies of a mutation that changes a "C" nucleotide to a "T" nucleotide results in hydrocephalus.

Photo: Equine Disease Quarterly

Ultimately, two copies of a mutation that changed a C nucleotide to a T nucleotide (Figure 1) resulted in the disorder. Although previously believed to be a complex disease, genetic and genomic methods were able to prove that the disorder was, in fact, simple, leading to the development of a genetic test that can help breeders avoid the disorder.

It is important to note the difference between a genetic test of a simple disease, such as hydrocephalus, and a genetic test for a complex disease (e.g., osteochondrosis). Genetic tests for simple diseases can confirm or rule out a genetic condition; however, genetic tests for complex diseases only help to determine an individuals chance of developing a genetic disorderan important distinction when genetic tests are used to help make breeding decisions. In either scenario, genetics and genomics in Thoroughbreds have far-reaching potential beyond that of breeding and selecting faster horses.

Understanding diseases caused by a single gene as well as complex diseases caused by multiple genes and the environment can lead to early diagnosis and targeted treatments. While the list of reasons a racehorse never reaches its potential might seem endless, genetics and genomics provide an opportunity to cross certain disorders off that list, thereby helping to eliminate or reduce the occurrence of those diseases.

CONTACTBrandon D. Velie, MS, PhDbrandon.velie@slu.seSwedish University of Agricultural Sciences Department of Animal Breeding and Genetics, Uppsala, Sweden

This is an excerpt from Equine Disease Quarterly, funded by underwriters at Lloyds, London.

Original post:
Genetics and Genomics in Racing: Speed Isn't Everything - TheHorse.com

Read More...

Hendrix Genetics announce new US turkey hatchery investment – Poultry World (subscription)

Wednesday, July 12th, 2017

Investment to the tune of US$25m has been announced by Hendrix Genetics in the form of a new commercial turkey hatchery in South Dakota, United States.

The hatchery will provide up to 30m day-old Hybrid poults and lead to the addition of 100 new temporary and permanent jobs at the site at Beresford.

The new hatchery will have the capacity for 35m hatching eggs and will be fitted with cutting edge equipment to ensure the highest biosecurity and poult quality.

Dave Libertini, Hendrix Genetics managing director, said: The new hatchery allows us to continue to deliver on our core focus of providing top quality genetics that match the needs of the market and our customers. Photo: ANP / Justin Sullivan

It is part of a substantial investment plan by Hendrix, which includes new hatcheries, egg production facilities and a modern transportation fleet and follows previous investments in grandparent facilities in Kansas and Nebraska.

Dave Libertini, Hendrix Genetics managing director, said: The new hatchery allows us to continue to deliver on our core focus of providing top quality genetics that match the needs of the market and our customers. This is a critical component of our plan to modernise the commercial turkey distribution infrastructure in the USA.

Matt McCready, Hendrix Genetics director of business development, added the hatchery would join the network of owned, aligned and contracted hatchery capacity set up to supply the strong demand for Hybrid genetics in the USA.

To comment, login here Or register to be able to comment.

Go here to see the original:
Hendrix Genetics announce new US turkey hatchery investment - Poultry World (subscription)

Read More...

Genetics of tongue twisting: Why some people do it while others can’t – Genetic Literacy Project

Wednesday, July 12th, 2017

Afsaneh Khetrapal | July 12, 2017 | News Medical

The term tongue twisting comprises rolling, folding, rotating, adjusting, and turning of the tongueAll aspects of a person are in some way influenced by genetics. Likewise, the tongue structure or its movement is controlled by ones respective gene patternTongue twisting is not a genetic disease or disorder, but a unique activity by a person using his tongue.

The early history of tongue genetics stated that the ability of tongue twisting is due to the influence of traitsThe tongue rolling ability occurs due to the influence of a dominant allele of the gene. A person who has either one or two copies of the dominant allele will be able to twist their tongue. In the case that a person is born with two recessive alleles, they cannot twist their tongue. In most cases, parents with a twisting-tongue ability can give birth to non-tongue twisters, and vice versa.

After a long-drawn out struggle, geneticists and researchers have finally proved that tongue twisting does not occur by genetic transformation. Genetic inheritance has only a minimal role to play in tongue twisting skills.

The GLP aggregated and excerpted this blog/article to reflect the diversity of news, opinion, and analysis. Read full, original post:Genetics of Tongue Twisting

Related Stories

See the article here:
Genetics of tongue twisting: Why some people do it while others can't - Genetic Literacy Project

Read More...

ABS Global launches NuEra Genetics beef program – Feedstuffs

Wednesday, July 12th, 2017

ABS Global announced July 12 the launch of NuEra Genetics, a new brand that encompasses all proprietary ABS beef breeding programs, evaluations, and indexes in order to deliver differentiated and superior terminal genetics for beef supply chain profitability.

ABS said NuEra Genetics symbolizes the next chapter in the history of ABS beef genetics and a new era of ABS beef genetic improvement.

This new brand provides customers access to continuous genetic improvement and a wider array of tailored evaluations and indexes that deliver genetic progress and profit faster, ABS said.

How will customers benefit? According to ABS, the answer is found in the NuEra Genetics tagline: Efficiency. Profitability. Sustainability. With NuEra, the goal is to optimize the customers efficiency, leading to greater profitability, and ultimately allowing for a sustainable system.

The new brand will strive to:

Customers should look for new products to be released under the NuEra Genetics brand in the coming months. Such products include proprietary indexes tailored to specific customer needs, making it easier for customers to select the most efficient and profitable genetics. In the long-term, NuEra will consistently deliver robust genetic improvement, year-on-year, to our customers, raising the bar of what is possible for them to achieve.

As we see dairymen focus the adoption of sexed genetics on their high-ranking females, NuEra Genetics will provide these producers with elite beef genetics for those lower-ranking cows, adding a significant revenue stream to their businesses, said Nate Zwald, chief operating officer of ABS Globals dairy division.

Jerry Thompson, chief operating officer of ABS Globals beef division, said NuEra Genetics has the potential to add significant value to the beef industry globally. Weve only really just scratched the surface and creating our own genetics to drive customer profitability will help us tap into many areas of growth opportunity.

Excerpt from:
ABS Global launches NuEra Genetics beef program - Feedstuffs

Read More...

Greenwood native doing high-level genetics work in hometown – Index-Journal

Wednesday, July 12th, 2017

Ah, summer vacation. The time of year when high school students laze on the beach, sleep in and stay up late binge watching their favorite TV shows.

But Greenwood native Emelee Guest is doing none of that. Her days have been occupied looking for mutations on Cullin-4B, a gene that has been linked to intellectual disabilities.

Ive been working with gene CUL4B and basically studying mutation E900X, which is basically a truncating mutation and Ive been studying the effects that a treatment called G-418 has on the mutation, is how the 17-year-old describes her work.

An incoming senior at the Governors School for Science and Mathematics in Hartsville, the 17-year-old is taking part in an intensive six-week research experience at the Greenwood Genetic Center under the guidance of Dr. Anand Srivastava, associate director for the facilitys Center for Molecular Studies.

Guest, who spent her freshman and sophomore years at Greenwood High School, said she was drawn to the field of genetics because she has relatives living with cystic fibrosis and epilepsy.

Part of my motivation is to help them. Ive watched them grow up with them, Guest said.

Guest is in elite company. Just one of 12 specialized, residential high schools in the country, the Governors School for Science and Mathematics which has an acceptance process offers more than 50 STEM classes with college and graduate-level opportunities and AP classes that enable students to earn multiple credits before starting their high education careers.

At my old school, we would sit there and learn, but there would be no application of it and there, we have labs once a week and they have more biology electives, Guest said. People say, get there, get anywhere, and its the hardest two years of your life.

More from this section (Story continues below)

Srivastava, whos taken summer interns from the Governors School for more than 10 years, said the arrangement is mutually beneficial for research institutions like the Genetic Center, which gets access to the next generation of scientists while providing real-world experience for students.

They are very committed, they desire to learn because they have some goals, Srivastava said. We try to design a project that is part of some ongoing project and they get to work with somebody in my lab, which allows them to learn and become independent.

Guest, who is undecided between pursuing a career in genetics and ecology, said the practical skills of working at summer internship in a high pressure laboratory setting are impactful.

I dont think I could have guessed what it was going to be like, just because I havent a lot of experience just some little things in class and its nothing like the real thing, she said. Its a little stressful sometimes because its a lot of small things that have to correctly but once you get used to it, its really exciting.

Contact staff writer Adam Benson at 864-943-5650 or on Twitter @ABensonIJ.

Original post:
Greenwood native doing high-level genetics work in hometown - Index-Journal

Read More...

Oxford Genetics gets 500000 from Mercia Technologies – Tech City News

Tuesday, June 6th, 2017

BioTech firm Oxford Genetics has raised 500,000 from Mercia Technologies.

The news comes after the company, which specialises in synthetic biology and DNA design, raised 1m from Mercia, which has a direct equity stake of 47.9% in the firm in October last year.

Oxford Genetics has so far raised 5.8m through a combination of grants and external investments and says it will use this latest round to expand its reach in the US market and further its growth.

Oxfords AI firm Oxbotica gets 8.6m to lead driverless car consortium

Dr Ryan Cawood, CEO of Oxford Genetics, commented on the raise: Mercias continued support has been instrumental in helping us to achieve the significant progress to date.

Our turnover has doubled in the last year and with this additional capital, we will be able to further expand the team, giving us the ability to build the most innovative technologies in the DNA and protein design market.

Dr Mark Payton, CEO of Mercia Technologies PLC, spoke about the companys trajectory over the past year.

Oxford Genetics has clearly demonstrated its ability to create market leading technologies and has been bolstered by an industry leading research and development team.

Payton went on to note that life sciences and bio-sciences continued to be a key sector for Mercia and one which they believe would deliver significant shareholder value over the medium term.

Follow Yessi Bello Perez on Twitter @yessibelloperez

Read this article:
Oxford Genetics gets 500000 from Mercia Technologies - Tech City News

Read More...

Why Herbalife, JinkoSolar, and NewLink Genetics Slumped Today … – Motley Fool

Tuesday, June 6th, 2017

The stock market closed Monday with modest losses, sending the Dow, S&P 500, and Nasdaq Composite lower from their record closes last Friday. Nevertheless, the declines were all less than 0.2%, and investors appeared to be in a wait-and-see mode as they look forward to more momentous news due out later this month. Among the top potential market movers for June will be the U.K. elections later this week and the Federal Reserve's meeting to determine the future course of interest rates. Yet company-specific items were in the spotlight today, and some stocks posted significant losses. Herbalife (NYSE:HLF), JinkoSolar (NYSE:JKS), and NewLink Genetics (NASDAQ:NLNK) were among the worst performers on the day. Below, we'll look more closely at these stocks to tell you why they did so poorly.

Shares of Herbalife dropped nearly 7% after the company said this morning that it would have to revise its financial expectations. Citing the need for its distributors to learn, teach, and implement new technology and processes, Herbalife said that it now expects net sales to fall 2% to 6% in the second quarter, with volume falling 4% to 8%. For the full year, Herbalife thinks it will be able to grow revenue 0.5% to 3.5% despite seeing volume come in a range between -1% and +2%. Upward adjustments to earnings guidance weren't enough to satisfy shareholders, and activist investor Bill Ackman spoke out against the company's news. Herbalife is a controversial company, but when negative things that get said about the business pan out in its financials, the seller of nutritional supplements and other consumer products needs to take steps to remedy the situation.

Image source: Herbalife.

JinkoSolar stock fell 8% in the wake of the company's first-quarter financial results. The Chinese solar company said that solar shipments jumped by nearly 30% from year-ago levels, topping the 2-gigawatt mark. Revenue was also up by double-digit percentages, but adjusted net income was down sharply, falling more than 80%. CEO Kangping Chen said that falling selling prices of solar modules led to gross margin contraction, which in turn resulted in bottom-line weakness. Chen remained optimistic about JinkoSolar's prospects for the remainder of the year, but investors didn't seem as confident that the company would be able to improve margins and capitalize on building demand in China. With JinkoSolar having been involved in big projects in the Persian Gulf region, it's possible that diplomatic tensions in the area also weighed on the stock.

Finally, shares of NewLink Genetics finished down 12%. The company said over the weekend that a phase 2 study of its breast cancer candidate treatment indoximod in combination with taxane chemotherapy failed to reach its intended endpoints. In particular, NewLink was trying to establish a statistically significant difference as to progression-free survival, overall survival, and objective response rate. Without achieving those goals, investors aren't certain what the next step forward is for NewLink. Still, with other studies having shown more encouraging results, NewLink might still end up being a potential takeover target from larger players in the biotech space.

Dan Caplinger has no position in any stocks mentioned. The Motley Fool has no position in any of the stocks mentioned. The Motley Fool has a disclosure policy.

Read the rest here:
Why Herbalife, JinkoSolar, and NewLink Genetics Slumped Today ... - Motley Fool

Read More...

New CEO for tilapia genetics firm – Fish Update

Tuesday, June 6th, 2017

GENOMAR Genetics, which specialises in the tilapia industry, has appointed Alejandro Tola Alvarez as its CEO.

Alvarez (pictured), who took up his new role on June 1, will be responsible for innovation, operations and business development within the company, which is part of the EW Group.

He hasbeen part of the Genomar group since 2006, based in South-East Asia as chief operational officer and in Norway as chief technical officer.

We were very pleased to find a highly qualified internal candidate for the CEO position, said chairman Odd Magne Rdseth.

Alejandro has played a major role in both R&D and commercial development of the most reputable and professional genetic brands in global tilapia aquaculture.

He comes with a deep understanding of the tilapia operating environments and the opportunities of modern breeding technologies, such as genomics, to improve economic and environmental performance of the industry.

Alvarez is a qualified vet and has masters degrees in aquaculture and business administration.

GenoMar Genetics, based in Oslo with its main operation in Luzon, Philippines, has developed the Genomar Supreme Tilapia strain (GST) through more than 25 years of selective breeding.

The company was part of the Norway Fresh Group until March 2017 when EW Group concluded an agreement to acquire 100 per cent of GenoMar Genetics shares.

EW Group, based in Visbek, Germany, is a family owned holding company with more than 120 subsidiaries in over 30 countries.

The core business of the group, which has 9,000 employees worldwide, is animal breeding, animal nutrition and animal health.

Over the past 10 years, the group has expanded into the aquaculture sector and includes companies such as AquaGen, Aquabel, GenoMar Genetics and Vaxxinova.

Read the rest here:
New CEO for tilapia genetics firm - Fish Update

Read More...

Plant genetics Branching out for crop improvement – Nature.com

Tuesday, June 6th, 2017

Nature.com
Plant genetics Branching out for crop improvement
Nature.com
Inflorescence architecture is the arrangement of flowers and their underlying stem branching patterns, and it has important effects on the yield of the fruits or grains from agricultural plants. A new study dissects key genetic underpinnings of tomato ...

See more here:
Plant genetics Branching out for crop improvement - Nature.com

Read More...

Genetics/Reproduction – beefmagazine.com

Monday, January 30th, 2017

A cowherds ability to maintain a high weaning rate with minimal supplementation of harvested feeds is a key contributor to a ranchs...

BEEFs 3rd annual Seedstock 100 listing, which ranks seedstock producers by number of bulls sold, offers you a...

Can you breed cattle to follow the road less traveled and graze hillsides? Research says its possible.

Sign Up for the BEEF Daily newsletter today!

This exclusive gallery features photos and information on the 100+ operations that make up the annual Seedstock 100 listing.

Bull buying season is nigh, and since your bull battery contributes 75% of your genetics, taking a little time to prepare ahead of the sale is time well spent. Those tips and more await you in this weeks Trending Headlines.

With most genetics available to everyone, increasingly, the primary point of differentiation among seedstock suppliers is their understanding of customer needs. That takes a relaetionship.

As the art and science of genomics becomes more accurate, cow-calf producers benefit. While cow-calf producers wont directly participate in genomic evaluation now that single-step evaluation is a reality, theyll be able to buy bulls with more...

Welcome to the 3rd annual edition of BEEF magazine's Seedstock 100, a listing of the biggest seedstock producers in the beef...

There will be plenty of bulls available this year, and while average prices will be lower than last year, the better bulls will...

Mary Lou Bradley-Henderson of Bradley 3 Ltd. at Memphis, Texas, offers her advice for getting the best bulls for your cowherd...

Engage your kids over the holiday break with an essay contest that asks the question, What does it mean to be a beef breeder in the 21st century?

Its not just chickens that have the dilemma of which came first. Every cow-calf producer faces a similar dilemma...

A North Dakota study shows smaller cows can produce more ranch profit, even when feedlot closeouts are applied to their steer calves...

Read the rest here:
Genetics/Reproduction - beefmagazine.com

Read More...

Cancer Genetics Overview (PDQ)Health Professional Version …

Thursday, January 26th, 2017

Introduction

[Note: Many of the medical and scientific terms used in this summary are found in the NCI Dictionary of Genetics Terms. When a linked term is clicked, the definition will appear in a separate window.]

[Note: A concerted effort is being made within the genetics community to shift terminology used to describe genetic variation. The shift is to use the term variant rather than the term mutation to describe a difference that exists between the person or group being studied and the reference sequence. Variants can then be further classified as benign (harmless), likely benign, of uncertain significance, likely pathogenic, or pathogenic (disease causing). Throughout this summary, we will use the term pathogenic variant to describe a disease-causing mutation. Refer to Table 1, Variant Classification for Pathogenicity for more information.]

The etiology of cancer is multifactorial, with genetic, environmental, medical, and lifestyle factors interacting to produce a given malignancy. Knowledge of cancer genetics is rapidly improving our understanding of cancer biology, helping to identify at-risk individuals, furthering the ability to characterize malignancies, establishing treatment tailored to the molecular fingerprint of the disease, and leading to the development of new therapeutic modalities. As a consequence, this expanding knowledge base has implications for all aspects of cancer management, including prevention, screening, and treatment.

Genetic information provides a means of identifying people who have an increased risk of cancer. Sources of genetic information include biologic samples of DNA, information derived from a persons family history of disease, findings from physical examinations, and medical records. DNA-based information can be gathered, stored, and analyzed at any time during an individuals life span, from before conception to after death. Family history may identify people with a modest to moderately increased risk of cancer or may serve as the first step in the identification of an inherited cancer predisposition that confers a very high lifetime risk of cancer. For an increasing number of diseases, DNA-based testing can be used to identify a specific pathogenic variant as the cause of inherited risk and to determine whether family members have inherited the disease-related variant.

The proportion of individuals carrying a pathogenic variant who will manifest the disease is referred to as penetrance. In general, common genetic variants that are associated with cancer susceptibility have a lower penetrance than rare genetic variants. This is depicted in Figure 1. For adult-onset diseases, penetrance is usually described by the individual carrier's age and sex. For example, the penetrance for breast cancer in female carriers of BRCA1/BRCA2 pathogenic variants is often quoted by age 50 years and by age 70 years. Of the numerous methods for estimating penetrance, none are without potential biases, and determining an individual carrier's risk of cancer involves some level of imprecision. Enlarge

Figure 1. Genetic architecture of cancer risk. This graph depicts the general finding of a low relative risk associated with common, low-penetrance genetic variants, such as single-nucleotide polymorphisms identified in genome-wide association studies, and a higher relative risk associated with rare, high-penetrance genetic variants, such as pathogenic variants in the BRCA1/BRCA2 genes associated with hereditary breast and ovarian cancer and the mismatch repair genes associated with Lynch syndrome.

Genetic variants, or changes in the usual DNA sequence of a particular gene, can have harmful, beneficial, neutral, or uncertain effects on health and may be inherited as autosomal dominant, autosomal recessive, or X-linked traits. Pathogenic variants that cause serious disability early in life are usually rare because of their adverse effect on life expectancy and reproduction. However, if the pathogenic variant is autosomal recessivethat is, if the health effect of the variant is caused only when two copies (one from each parent) of the altered gene are inherited carriers of the pathogenic variant (healthy people carrying one copy of the altered gene) may be relatively common in the general population. Common in this context refers, by convention, to a prevalence of 1% or more. Pathogenic variants that cause health effects in middle and older age, including several pathogenic variants known to cause a predisposition to cancer, may also be relatively common. Many cancer-predisposing traits are inherited in an autosomal dominant fashion, that is, the cancer susceptibility occurs when only one copy of the altered gene is inherited. For autosomal dominant conditions, the term carrier is often used in a less formal manner to denote people who have inherited the genetic predisposition conferred by the pathogenic variant. (Refer to individual PDQ summaries focused on the genetics of specific cancers for detailed information on known cancer-susceptibility syndromes.)

Increasingly, the public is turning to the Internet for information related both to familial and genetic susceptibility to cancer and to genetic risk assessment and testing. Direct-to-consumer marketing of genetic testing for hereditary breast and colon cancer is also taking place in some communities. This wider availability of information related to inherited cancer risk may raise concerns among persons previously unaware of the implications inherent in their family histories and may lead some of these individuals to consult their primary care physicians for management advice and recommendations. In many instances, the evaluation and advice will be relatively straightforward for physicians with a basic knowledge of familial cancer. In a subset of patients, the evaluation may be more complex, calling for referral to genetics professionals for further evaluation and counseling.

Correctly recognizing and identifying individuals and families at increased risk of developing cancer is one of countless important roles for primary care and other health care providers. Once identified, these individuals can then be appropriately referred for genetic counseling, risk assessment, consideration of genetic testing, and development of a management plan. When medical and family histories reveal cardinal clues to the presence of an underlying familial or genetic cancer susceptibility disorder (see list below),[1] further evaluation may be warranted. (Refer to the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information about the components of a genetics cancer risk assessment.)

Features of hereditary cancer include the following:

Concluding that an individual is at increased risk of developing cancer may have important, potentially life-saving management implications and may lead to specific interventions aimed at reducing risk (e.g., tamoxifen for breast cancer, colonoscopy for colon cancer, or risk-reducing salpingo-oophorectomy for ovarian cancer). Information about familial cancer risk may also inform a persons ability to plan for the future (lifestyle and health care decisions, family planning, or other decisions). Genetic information may also provide a direct health benefit by demonstrating the lack of an inherited cancer susceptibility. For example, if a family is known to carry a cancer-predisposing variant in a particular gene, a family member may experience reduced worry and lower health care costs if his or her genetic test indicates that he or she does not carry the familys disease-related variant. Conversely, information about familial cancer risk may have psychological effects or social costs (e.g., worry, guilt, or increased health care costs). Family dynamics also may be affected. For instance, the involvement of one or more family members may be required for genetic testing to be informative, and parents may feel guilt about passing inherited risk on to their children.

Knowledge about a cancer-predisposing variant can be informative not only for the individual tested but also for other family members. Family members who previously had not considered the implications of their family history for their own health may be led to do so, and some will undergo genetic testing, resulting in more definitive information on whether they are at increased genetic risk. Some relatives may learn their carrier status without being directly tested, for example, when a biological parent of a child who is a known carrier of a pathogenic variant is identified as an obligate carrier. Founder effects may result in the recognition that specific ethnic groups have a higher prevalence of certain pathogenic variants, knowledge that can be either clinically useful (permitting more rational genetic testing strategies) or potentially stigmatizing. Testing may reveal the presence of nonpaternity in a family. There is the theoretical possibility that genetic information may be misused, and concerns about the potential for insurance and/or employment discrimination may arise. Genetic information may also affect medical and lifestyle decisions.

Refer to individual PDQ summaries for available evidence addressing all ancillary issues.

Genetic counseling is a process of communication between genetics professionals and patients with the goal of providing individuals and families with information on the relevant aspects of their genetic health, available testing and management options, and support as they move toward understanding and incorporating this information into their daily lives. Genetic counseling generally involves the following six steps:

Genetic evaluation involves an interaction with a medical geneticist or other genetics professional and may include a physical examination and diagnostic testing, in addition to genetic counseling. The principles of voluntary and informed decision making, nondirective and noncoercive counseling, and protection of client confidentiality and privacy are central to the philosophy of genetic counseling.[1-5] (Refer to the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information on the nature and history of genetic counseling.)

From the mid-1990s to the mid-2000s, genetic counseling expanded to include discussion of genetic testing for cancer risk, as more genes associated with inherited cancer risk were discovered. Cancer genetic counseling often involves a multidisciplinary team of health professionals that may include a genetic counselor, an advanced practice genetics nurse, or a medical geneticist; a mental health professional; and various medical experts such as an oncologist, surgeon, or internist. The process of counseling may require a number of visits to address medical, genetic testing, and psychosocial issues. Even when cancer risk counseling is initiated by an individual, inherited cancer risk has implications for the entire family. Because genetic risk affects an unknown number of biological relatives, contact with these relatives is often essential to collect accurate family and medical histories. Cancer genetic counseling may involve several family members, some of whom will have had cancer and others who have not.

The impact of risk assessment and predisposition genetic testing is improved health outcomes. The information derived from risk assessment and/or genetic testing allows the health care provider to tailor an individual approach to health promotion and optimize long-term health outcomes through the identification of at-risk individuals before cancer develops. The health care provider can thus intervene earlier either to reduce the risk or diagnose a cancer at an earlier stage, when the chances for effective treatment are greatest. The information may be used to modify the management approach to an initial cancer, clarify the risks of other cancers, or predict the response of an existing cancer to specific forms of treatment, all of which may alter treatment recommendations and long-term follow-up.

Individual PDQ summaries focused on the genetics of specific cancers contain detailed information about many known cancer susceptibility syndromes. Although this is not a complete list, the following cancer susceptibility syndromes are discussed in the PDQ cancer genetics summaries (listed in parentheses after the syndromes):

The methods described in this section are intended to provide a brief background about the genetic analysis and discovery approaches that have been used during the past 10 to 15 years for identifying disease susceptibility genes. These methods led to important cancer gene discoveries such as BRCA1 and breast cancer risk. Since then, genetic analysis techniques have transitioned to next-generation sequencing methods as described in the Clinical Sequencing section of this summary.

The recognition that cancer clusters within families has led many investigators to collect data on multiple-case families with the goal of localizing cancer susceptibility genes through linkage studies.

Linkage studies are typically performed on high-risk kindreds, in whom multiple cases of a particular disease have occurred, in an effort to identify disease susceptibility genes. Linkage analysis statistically compares the genotypes between affected and unaffected individuals and looks for evidence that known genetic markers are inherited along with the disease trait. If such evidence is found (linkage), it provides statistical data that the chromosomal region near the marker also harbors a disease susceptibility gene. Once a genomic region of interest has been identified through linkage analysis, additional studies are required to prove that there truly is a susceptibility gene at that position. Linkage analysis is affected by the following:

An additional issue in linkage studies is the background rate of sporadic cancer in the context of family studies. For example, because a mans lifetime risk of prostate cancer is one in eight,[1] it is possible that families under study have both inherited and sporadic prostate cancer cases. Thus, men who do not inherit the prostate cancer susceptibility gene that is segregating in their family may still develop prostate cancer.

One way to address inconsistencies between linkage studies is to require inclusion criteria that defines clinically significant disease.[2-6] This approach attempts to define a homogeneous set of cases/families to increase the likelihood of identifying a linkage signal. It also prevents the inclusion of cases that may be considered clinically insignificant that were identified by screening in families.

GWAS are identifying common, low-penetrance susceptibility alleles for many complex diseases,[7] including cancer. This approach can be contrasted with linkage analysis, which searches for genetic-risk variants cosegregating within families that have a high prevalence of disease. While linkage analyses are designed to uncover rare, highly penetrant variants that segregate in predictable heritance patterns (e.g., autosomal dominant, autosomal recessive, X-linked, and mitochondrial), GWAS are best suited to identify multiple, common, low-penetrance genetic polymorphisms. GWAS are conducted under the assumption that the genetic underpinnings of complex phenotypes, such as prostate cancer, are governed by many alleles, each conferring modest risk. Most genetic polymorphisms genotyped in GWAS are common, with minor allele frequencies greater than 1% to 5% within a given population (e.g., men of European ancestry). GWAS capture a large portion of common variation across the genome.[8,9] The strong correlation between many alleles located close to one another on a given chromosome (called linkage disequilibrium) allows one to scan the genome without having to test all 10 million known single nucleotide polymorphisms (SNPs). With GWAS, researchers can test approximately 1 million to 5 million SNPs per study and ascertain almost all common inherited variants in the genome.

In a GWAS, allele frequency for each SNP is compared between cases and controls. Promising signalsin which allele frequencies deviate significantly in case compared to control populationsare validated in replication cohorts. To have adequate statistical power to identify variants associated with a phenotype, large numbers of cases and controls, typically thousands of each, are studied. Because up to 1 million SNPs are evaluated in a GWAS, false-positive findings are expected to occur frequently when using standard statistical thresholds. Therefore, stringent statistical rules are used to declare a positive finding, usually using a threshold of P < 1 10-7.[10-12]

To date, hundreds of cancer-risk variants have been identified by well-powered GWAS and validated in independent cohorts.[13] These studies have revealed consistent associations between specific inherited variants and cancer risk. However, the findings should be qualified with a few important considerations:

The implications of these points are discussed in greater detail in the PDQ summaries on Genetics of Breast and Gynecologic Cancers; Genetics of Colorectal Cancer; and Genetics of Prostate Cancer. Additional details can be found elsewhere.[18]

Broad-scale genome sequencing approaches, including multigene (panel) testing, whole-exome sequencing (WES), and whole-genome sequencing (WGS), are rapidly being developed and incorporated into a spectrum of clinical oncologic settings, including cancer therapeutics and cancer risk assessment. Several institutions and companies offer tumor sequencing, and some are developing precision medicine programs that sequence tumor genomes to identify driver genetic alterations that are targetable for therapeutic benefit to patients.[1-3] Many of these tumor-based approaches use germline DNA sequences as a reference to discriminate between DNA changes only within the tumor and those that are potentially inherited. In the genetic counseling and cancer risk assessment setting, the use of multigene testing to evaluate inherited cancer risk is becoming more common and may become routine in the near future, with institutions and companies offering multigene testing to detect alterations in a host of cancer riskassociated genes.

These advances in gene sequencing technologies also identify variants in genes related to the primary indication for ordering genetic sequence testing, along with findings not related to the disorder being tested. The latter genetic findings, termed incidental or secondary findings, are currently a source of clinical, ethical, legal, and counseling debate. The American College of Medical Genetics and Genomics (ACMG) and the Presidential Commission for the Study of Bioethical Issues have published literature that address some of these issues and provide guidance and recommendations for their use.[4-7] However, controversy continues about when and what results to provide to patients and their health care providers. This section was created to provide information about genomic sequencing technologies in the context of clinical sequencing and highlights additional areas of clinical uncertainty for which further research and approaches are needed.

DNA sequencing technologies have undergone rapid evolution, particularly since 2005 when massively parallel sequencing, or next-generation sequencing (NGS), was introduced.[8]

Automated Sanger sequencing is considered the first generation of sequencing technology.[9] Sanger cancer gene sequencing uses polymerase chain reaction (PCR) amplification of genetic regions of interest followed by sequencing of PCR products using fluorescently labeled terminators, capillary electrophoresis separation of products, and laser signal detection of nucleotide sequence.[10,11] While this is an accurate sequencing technology, the main limitations of Sanger sequencing include low throughput, a limited ability to sequence more than a few genes at a time, and the inability to detect structural rearrangements.[10]

NGS refers to high throughput DNA sequencing technologies that are capable of processing multiple DNA sequences in parallel.[11] Although platforms differ in template generation and sequence interrogation, the overall approach to NGS technologies involves shearing and immobilizing DNA template molecules onto a solid surface, which allows separation of molecules for simultaneous sequencing reactions (millions to billions) to be performed in a parallel fashion.[10,12] Thus, the major advantages of NGS technologies include the ability to sequence thousands of genes at one time, a lower cost, and the ability to detect multiple types of genomic alterations, such as insertions, deletions, copy number alterations, and rearrangements.[10] Limitations include the possibility that specific gene regions may be missed, turnaround time can be lengthy (although it is decreasing), and informatics support to handle massive amounts of genetic data has lagged behind the sequencing capability. A well-recognized bottleneck to utilizing NGS data is the lack of advanced computational infrastructure to preserve, process, and analyze the vast amount of genetic data. The magnitude of the variants obtained from NGS is exponential; bioinformatics approaches need to evaluate genetic variants for predicted functional consequence in disease biology. There is also a need for user-friendly bioinformatics pipelines to analyze and integrate genetic data to influence the scientific and medical community.[11,13]

The following terms are defined to better understand the clinical application of NGS testing and implications of results reported.

NGS has multiple potential clinical applications. In oncology, the two dominant applications are: 1) the assessment of somatic alterations in tumors to inform prognosis and/or targeted therapeutics; and 2) the assessment of the germline to identify cancer risk alleles.

There are multiple approaches to tumor testing for somatic alterations. With targeted multigene testing, a number of different genes can be assessed simultaneously. These targeted multigene tests can differ substantially in the genes that are included, and they can be tailored to individual tumor types. Targeted multigene testing limits the data to be analyzed and includes only known genes, which makes the interpretation more straightforward than in whole exome or whole genome techniques. In addition, greater depth of coverage is possible with targeted multigene testing than with WES or WGS. Depth of coverage refers to the number of times a nucleotide has been sequenced; a greater depth of coverage has fewer sequencing errors. Deep coverage also aids in differentiating sequencing errors from single nucleotide polymorphisms.

WES and WGS are far more extensive techniques and aim to uncover variants in known genes and in genes not suspected a priori. The discovery of a variant that is unexpected for a particular tumor type can lead to the use of a directed therapeutic, which could improve patient outcome. WES generates sequence data of the coding regions of the genome (representing approximately 1% of the human genome), rather than the entire genome (WGS). Consequently, WES is less expensive than WGS.

Noncoding variants can be identified using WGS but cannot be identified using WES. The use of WGS is limited by cost and the vast bioinformatics needed for interpretation. Although the costs of sequencing have dropped precipitously, the analysis remains formidable.[14]

Although the goal of WES and WGS is to improve patient care by detecting actionable genetic variants (mutations that can be targeted therapeutically), a number of issues warrant consideration. This testing may detect pathogenic variants, variants of uncertain significance (VUS), or no detectable abnormalities. In addition, pathogenic variants can be found in genes that are thought to be clearly related to tumorigenesis but can also be detected in genes with unclear relevance (particularly with WES and WGS approaches). VUS have unclear implications as they may, or may not, disrupt the function of the protein. The definition of actionable can vary, but often this term is used when an aberration, if found, would lead to recommendations against certain treatments (such as variants in ras) for which a clinical trial is available, or for which there is a known targeted drug. Although there are case reports of success with this approach, it is unlikely to be straightforward. Studies are ongoing.

Some commercial and single-institution assays test only the tumor. Clearly pathogenic variants found in important genes in the tumor can be somatic but could also be from the germline. In situations in which somatic analysis is paired with a germline analysis, it can be determined whether an identified alteration is inherited. A study that estimated the prevalence of germline variants from patients undergoing tumor sequencing with matched, normal DNA sequencing reported that cancer susceptibility genes were identified in 198 of 1,566 individuals (12.6%). Only 81 of these 198 individuals (40.9%) had pathogenic variants in cancer susceptibility genes concordant with their tumor type. When expanding to include known noncancer-related Mendelian disease genes, 246 of 1,566 individuals (15.7%) had pathogenic or presumed pathogenic germline variants identified.[15]

Sequencing tumors may lead to the identification of hereditary (germline) pathogenic variants.[16] Founder pathogenic variants in well-characterized cancer susceptibility genes are highly suggestive of a germline pathogenic variant. Hypermutated tumor phenotype may suggest an underlying constitutional defect in DNA repair. Clinical characteristics that fit with a particular genetic predisposition, such as family history, young age at diagnosis, or specific tumor type, may also raise the suspicion of a germline variant correlating with a tumor variant. A high variant allele fraction may also indicate a germline variant. All of these factors signify a potential need for patients to undergo genetic counseling and to consider confirmatory germline genetic testing.

The absence of a variant in a gene assessed as part of somatic testing does not rule out the presence of an inherited susceptibility. All patients whose personal and family histories are suggestive of hereditary cancer should consider germline testing regardless of their somatic results.

Ongoing clinical trials, such as the NCI Molecular Analysis for Therapy Choice (NCI-MATCH) Trial, are examining the value of somatic sequencing to find actionable targets. Germline sequencing is occurring as a component of this study.

The goal of germline testing is to identify pathogenic variants associated with an inherited risk of cancer and to guide cancer riskmanagement decisions. Also, germline testing can aid in some management decisions at the time of diagnosis (e.g., decisions about colectomy in Lynch syndromerelated colon cancer and contralateral mastectomy in carriers of BRCA1/2 pathogenic variants). In addition, there are emerging data that germline status may help determine systemic therapy (e.g., the use of cisplatin or PARP inhibitors in BRCA1/2-related cancer).

To date, most germline genetic testing has been performed in a targeted manner, looking for variants in the gene(s) associated with a clinical picture (e.g., BRCA1 and BRCA2 in hereditary breast and ovarian cancer; or the mismatch repair [MMR] genes in Lynch syndrome). However, targeted multigene tests now available commercially or within an institution contain different sets of genes. Some are targeted to all cancers, others to specific cancers (e.g., breast, colon, or prostate cancers). The genes on the multigene tests include high-penetrance genes related to the specific tumor (such as BRCA1/2 on a breast cancer panel); high penetrance genes related to a different type of cancer but with a more moderate risk for the tumor of reference (such as CDH1 or MSH6 on a breast cancer panel); and moderate penetrance genes for which clinical utility is uncertain (such as NBN on a breast cancer panel). Because multiple genes are included on these panels, it is anticipated that many, and perhaps most, individuals undergoing testing using these panels will be found to have at least one VUS. As it is not possible to do standard pretest counseling models for a panel of 20 genes, new counseling models are needed. Ethical issues of whether patients can opt out of specific results (such as TP53 or CDH1 in breast cancer) and how this would be done in clinical practice are unresolved.

Refer to the Multigene (panel) testing section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information about the use of targeted multigene tests.

WES for inherited cancer susceptibility is also commercially available. Secondary findings are likely and management of such findings is evolving.

The ACCE model uses four main components to evaluate new genetic tests: analytic validity; clinical validity; clinical utility; and ethical, legal, and social issues.[17]

The ACCE model's framework has been adopted worldwide for the evaluation of genetic tests.

Several layers of complexity exist in managing NGS in the clinical setting. At the purely technical level, improvements in the sequencing technique have allowed for sequencing across the entire genome, not merely the exome. As the costs decrease, exomic and genomic sequencing of tumor and normal tissue can be expected to become more routine.

With routine use of WGS, major challenges in interpretation emerge. Foremost is the matter of determining which sequence variations in known cancer predisposition genes are pathologic, which are harmless, and which variations require further evaluation as to their significance. This is not a new challenge. Various groups are developing processes for the interpretation and curation of a growing database of variants and their significance. For example, the International Society for Gastrointestinal Hereditary Tumors has developed such a process for the MMR genes in concert with the Human Variome Project and International Mismatch Repair Consortium.

These processes may serve as a framework for the emerging challenge of interpreting the significance of sequence variations in genes of uncertain or unknown function in regulation of neoplastic progression or other diseases. Larger cancer predisposition multigene tests have been developed by commercial laboratories, with their own process for interpretation. To the extent that increasingly larger multigene tests include genes of unknown significance, governance of the interpretation process requires that academic institutions offering their own multigene tests or using external proprietary panels develop a deliberative process for managing the quality assurance for test performance (including Clinical Laboratory Improvement Amendments [CLIA], where appropriate) and interpretation.

ACMG has issued the following updated guidelines for achieving accountability in interpreting and reporting secondary findings:[4,18]

Concerns remain that the routine reporting of germline variants in the context of tumor sequencing would require laboratories to conduct results review with germline and tumor genome expertise, which would be expected to increase costs, laboratory efforts, and turnaround time for results reporting. The nature of discussions between oncologists and patients would be altered to include the multiple facets involved with germline testing and potential results. Pre- and post-test discussions would also potentially require involvement of genetic counselors and geneticists, who are a limited resource in oncology practices. Recent expert comment stated that more data are needed about the benefits of return of secondary germline findings to cancer patients undergoing tumor sequencing, citing a need for recommendations by experts in the oncology and genetics communities.[19]

It is still very early in the development processes for oversight at the institutional level. As an example, at one high-volume cancer center, the following process has been used:

Informed consent for the sequencing of highly penetrant disease genes has been conducted since the mid-1990s in the contexts of known or suspected inherited diseases within selected families. However, the best methods and approaches for educating and counseling individuals about the potential benefits, limitations, and harms of genetic testing to facilitate informed decisions have not been fully elucidated or adequately tested. New informed consent challenges arise as NGS technologies are applied in clinical and research settings. Challenges to facilitating informed consent include the following:

The increased availability and decreased cost of NGS technology are expanding the use of genome-wide testing of tumors, with the goal of identifying somatic variants as potential targets for cancer treatment. While identifying germline pathogenic variants may be considered secondary to the main purpose of testing tumors, the possibility of identifying actionable secondary findings of pathogenic variants in cancer predisposition genes supports the need for genetic counseling in this context. Approaches for genetic counseling and informed consent in the context of tumor sequencing have been proposed.[20,21]

Advances in genetic sequencing technologies have dramatically reduced the cost of sequencing an individual's full genome or exome. WGS and WES are increasingly being employed in the clinical setting in testing for both somatic and germline variants. In addition, multigene tests are now available commercially or within an institution. Considerable debate surrounds the clinical, ethical, legal, and counseling aspects associated with NGS and gene panels. Future research is warranted to address these issues.

PDQ cancer genetics summaries focus on the genetics of specific cancers, inherited cancer syndromes, and the ethical, social, and psychological implications of cancer genetics knowledge. Sections on the genetics of specific cancers include syndrome-specific information on the risk implications of a family history of cancer, the prevalence and characteristics of cancer-predisposing variants, known modifiers of genetic risk, opportunities for genetic testing, outcomes of genetic counseling and testing, and interventions available for people with increased cancer risk resulting from an inherited predisposition.

The source of medical literature cited in PDQ cancer genetics summaries is peer-reviewed scientific publications, the quality and reliability of which is evaluated in terms of levels of evidence. Where relevant, the level of evidence is cited, or particular strengths of a study or limitations of the evidence are described.

Refer to the Levels of Evidence for Cancer Genetics Studies summary for more information on the levels of evidence utilized in the PDQ cancer genetics summaries.

Health care providers who deliver genetic services, including genetic counseling, can be located through local, regional, and national professional genetics organizations and through NCI's Cancer Genetics Services Directory website. Providers of cancer genetic services are not limited to one specialty and include medical geneticists, genetic counselors, advanced practice genetics nurses, oncologists (medical, radiation, or surgical), other surgeons, internists, pediatricians, family practitioners, and mental health professionals. A cancer genetics health care provider will assist in constructing and evaluating a pedigree, eliciting and evaluating personal and family medical histories, and calculating and providing information about cancer risk and/or probability of a pathogenic variant being associated with cancer in the family. In addition, if a genetic test is available, these providers can assist in pretest counseling, laboratory selection, informed consent, test interpretation, posttest counseling, and follow-up.

This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about cancer genetics. It is intended as a resource to inform and assist clinicians who care for cancer patients. It does not provide formal guidelines or recommendations for making health care decisions.

This summary is reviewed regularly and updated as necessary by the PDQ Cancer Genetics Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).

Board members review recently published articles each month to determine whether an article should:

Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.

Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.

Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Cancer Genetics Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.

PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as NCIs PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].

The preferred citation for this PDQ summary is:

PDQ Cancer Genetics Editorial Board. PDQ Cancer Genetics Overview. Bethesda, MD: National Cancer Institute. Updated . Available at: http://www.cancer.gov/about-cancer/causes-prevention/genetics/overview-pdq. Accessed . [PMID: 26389204]

Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.

The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.

More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the websites Email Us.

Excerpt from:
Cancer Genetics Overview (PDQ)Health Professional Version ...

Read More...

Genetics of Skin Cancer (PDQ)Health Professional Version …

Sunday, December 25th, 2016

Executive Summary

This executive summary reviews the topics covered in this PDQ summary on the genetics of skin cancer, with hyperlinks to detailed sections below that describe the evidence on each topic.

More than 100 types of tumors are clinically apparent on the skin; many are known to have familial and/or inherited components, either in isolation or as part of a syndrome with other features. Basal cell carcinoma (BCC) and squamous cell carcinoma (SCC), which are known collectively as nonmelanoma skin cancer, are two of the most common malignancies in the United States and are often caused by sun exposure, although several hereditary syndromes and genes are also associated with an increased risk of developing these cancers. Melanoma is less common than nonmelanoma skin cancer, but 5% to 10% of all melanomas arise in multiple-case families and may be inherited in an autosomal dominant fashion.

Several genes and hereditary syndromes are associated with the development of skin cancer. Basal cell nevus syndrome (BCNS, caused by pathogenic variants in PTCH1 and PTCH2) is associated with an increased risk of BCC, while syndromes such as xeroderma pigmentosum (XP), oculocutaneous albinism, epidermolysis bullosa, and Fanconi anemia are associated with an increased risk of SCC. The major tumor suppressor gene associated with melanoma is CDKN2A; pathogenic variants in CDKN2A have been estimated to account for 35% to 40% of all familial melanomas. Pathogenic variants in many other genes, including CDK4, CDK6, BAP1, and BRCA2, have also been found to be associated with melanoma.

Genome-wide searches are showing promise in identifying common, low-penetrance susceptibility alleles for many complex diseases, including melanoma, but the clinical utility of these findings remains uncertain.

Risk-reducing strategies for individuals with an increased hereditary predisposition to skin cancer are similar to recommendations for the general population, and include sun avoidance, use of sunscreen, use of sun-protective clothing, and avoidance of tanning beds. Chemopreventive agents such as isotretinoin and acitretin have been studied for the treatment of BCCs in patients with BCNS and XP and are associated with a significant decrease in the number of tumors per year. Vismodegib has also shown promise in reducing the per-patient annual rate of new BCCs requiring surgery among patients with BCNS. Isotretinoin has also been shown to reduce SCC incidence among patients with XP.

Treatment of hereditary skin cancers is similar to the treatment of sporadic skin cancers. One study in an XP population found therapeutic use of 5-fluorouracil to be efficacious, particularly in the treatment of extensive lesions. In addition to its role as a therapeutic and potential chemopreventive agent, vismodegib is also being studied for potential palliative effects for keratocystic odontogenic tumors in patients with BCNS.

Most of the psychosocial literature about hereditary skin cancers has focused on patients with familial melanoma. In individuals at risk of familial melanoma, psychosocial factors influence decisions about genetic testing for inherited cancer risk and risk-management strategies. Interest in genetic testing for pathogenic variants in CDKN2A is generally high. Perceived benefits among individuals with a strong family history of melanoma include information about the risk of melanoma for themselves and their children and increased motivation for sun-protective behavior. A number of studies have examined risk-reducing and early-detection behaviors in individuals with a family history of melanoma. Overall, these studies indicate inconsistent adoption and maintenance of these behaviors. Intervention studies have targeted knowledge about melanoma, sun protection, and screening behaviors in family members of melanoma patients, with mixed results. Research is ongoing to better understand and address psychosocial and behavioral issues in high-risk families.

[Note: Many of the medical and scientific terms used in this summary are found in the NCI Dictionary of Genetics Terms. When a linked term is clicked, the definition will appear in a separate window.]

[Note: A concerted effort is being made within the genetics community to shift terminology used to describe genetic variation. The shift is to use the term variant rather than the term mutation to describe a difference that exists between the person or group being studied and the reference sequence. Variants can then be further classified as benign (harmless), likely benign, of uncertain significance, likely pathogenic, or pathogenic (disease causing). Throughout this summary, we will use the term pathogenic variant to describe a disease-causing mutation. Refer to the Cancer Genetics Overview summary for more information about variant classification.]

[Note: Many of the genes described in this summary are found in the Online Mendelian Inheritance in Man (OMIM) database. When OMIM appears after a gene name or the name of a condition, click on OMIM for a link to more information.]

The genetics of skin cancer is an extremely broad topic. There are more than 100 types of tumors that are clinically apparent on the skin; many of these are known to have familial components, either in isolation or as part of a syndrome with other features. This is, in part, because the skin itself is a complex organ made up of multiple cell types. Furthermore, many of these cell types can undergo malignant transformation at various points in their differentiation, leading to tumors with distinct histology and dramatically different biological behaviors, such as squamous cell carcinoma (SCC) and basal cell cancer (BCC). These have been called nonmelanoma skin cancers or keratinocyte cancers.

Figure 1 is a simple diagram of normal skin structure. It also indicates the major cell types that are normally found in each compartment. Broadly speaking, there are two large compartmentsthe avascular epidermis and the vascular dermiswith many cell types distributed in a largely acellular matrix.[1]

Figure 1. Schematic representation of normal skin. The relatively avascular epidermis houses basal cell keratinocytes and squamous epithelial keratinocytes, the source cells for BCC and SCC, respectively. Melanocytes are also present in normal skin and serve as the source cell for melanoma. The separation between epidermis and dermis occurs at the basement membrane zone, located just inferior to the basal cell keratinocytes.

The outer layer or epidermis is made primarily of keratinocytes but has several other minor cell populations. The bottom layer is formed of basal keratinocytes abutting the basement membrane. The basement membrane is formed from products of keratinocytes and dermal fibroblasts, such as collagen and laminin, and is an important anatomical and functional structure. Basal keratinocytes lose contact with the basement membrane as they divide. As basal keratinocytes migrate toward the skin surface, they progressively differentiate to form the spinous cell layer; the granular cell layer; and the keratinized outer layer, or stratum corneum.

The true cytologic origin of BCC remains in question. BCC and basal cell keratinocytes share many histologic similarities, as is reflected in the name. Alternatively, the outer root sheath cells of the hair follicle have also been proposed as the cell of origin for BCC.[2] This is suggested by the fact that BCCs occur predominantly on hair-bearing skin. BCCs rarely metastasize but can invade tissue locally or regionally, sometimes following along nerves. A tendency for superficial necrosis has resulted in the name "rodent ulcer."[3]

Some debate remains about the origin of SCC; however, these cancers are likely derived from epidermal stem cells associated with the hair follicle.[4] A variety of tissues, such as lung and uterine cervix, can give rise to SCC, and this cancer has somewhat differing behavior depending on its source. Even in cancer derived from the skin, SCC from different anatomic locations can have moderately differing aggressiveness; for example, SCC from glabrous (smooth, hairless) skin has a lower metastatic rate than SCC arising from the vermillion border of the lip or from scars.[3]

Additionally, in the epidermal compartment, melanocytes distribute singly along the basement membrane and can undergo malignant transformation into melanoma. Melanocytes are derived from neural crest cells and migrate to the epidermal compartment near the eighth week of gestational age. Langerhans cells, or dendritic cells, are another cell type in the epidermis and have a primary function of antigen presentation. These cells reside in the skin for an extended time and respond to different stimuli, such as ultraviolet radiation or topical steroids, which cause them to migrate out of the skin.[5]

The dermis is largely composed of an extracellular matrix. Prominent cell types in this compartment are fibroblasts, endothelial cells, and transient immune system cells. When transformed, fibroblasts form fibrosarcomas and endothelial cells form angiosarcomas, Kaposi sarcoma, and other vascular tumors. There are a number of immune cell types that move in and out of the skin to blood vessels and lymphatics; these include mast cells, lymphocytes, mononuclear cells, histiocytes, and granulocytes. These cells can increase in number in inflammatory diseases and can form tumors within the skin. For example, urticaria pigmentosa is a condition that arises from mast cells and is occasionally associated with mast cell leukemia; cutaneous T-cell lymphoma is often confined to the skin throughout its course. Overall, 10% of leukemias and lymphomas have prominent expression in the skin.[6]

Epidermal appendages are also found in the dermal compartment. These are derivatives of the epidermal keratinocytes, such as hair follicles, sweat glands, and the sebaceous glands associated with the hair follicles. These structures are generally formed in the first and second trimesters of fetal development. These can form a large variety of benign or malignant tumors with diverse biological behaviors. Several of these tumors are associated with familial syndromes. Overall, there are dozens of different histological subtypes of these tumors associated with individual components of the adnexal structures.[7]

Finally, the subcutis is a layer that extends below the dermis with varying depth, depending on the anatomic location. This deeper boundary can include muscle, fascia, bone, or cartilage. The subcutis can be affected by inflammatory conditions such as panniculitis and malignancies such as liposarcoma.[8]

These compartments give rise to their own malignancies but are also the region of immediate adjacent spread of localized skin cancers from other compartments. The boundaries of each skin compartment are used to define the staging of skin cancers. For example, an in situ melanoma is confined to the epidermis. Once the cancer crosses the basement membrane into the dermis, it is invasive. Internal malignancies also commonly metastasize to the skin. The dermis and subcutis are the most common locations, but the epidermis can also be involved in conditions such as Pagetoid breast cancer.

The skin has a wide variety of functions. First, the skin is an important barrier preventing extensive water and temperature loss and providing protection against minor abrasions. These functions can be aberrantly regulated in cancer. For example, in the erythroderma (reddening of the skin) associated with advanced cutaneous T-cell lymphoma, alterations in the regulations of body temperature can result in profound heat loss. Second, the skin has important adaptive and innate immunity functions. In adaptive immunity, antigen-presenting cells engender T-cell responses consisting of increased levels of TH1, TH2, or TH17 cells.[9] In innate immunity, the immune system produces numerous peptides with antibacterial and antifungal capacity. Consequently, even small breaks in the skin can lead to infection. The skin-associated lymphoid tissue is one of the largest arms of the immune system. It may also be important in immune surveillance against cancer. Immunosuppression, which occurs during organ transplant, is a significant risk factor for skin cancer. The skin is significant for communication through facial expression and hand movements. Unfortunately, areas of specialized function, such as the area around the eyes and ears, are common places for cancer to occur. Even small cancers in these areas can lead to reconstructive challenges and have significant cosmetic and social ramifications.[1]

While the appearance of any one skin cancer can vary, there are general physical presentations that can be used in screening. BCCs most commonly have a pearly rim or can appear somewhat eczematous (see Figure 2 and Figure 3). They often ulcerate (see Figure 2). SCCs frequently have a thick keratin top layer (see Figure 4). Both BCCs and SCCs are associated with a history of sun-damaged skin. Melanomas are characterized by asymmetry, border irregularity, color variation, a diameter of more than 6 mm, and evolution (ABCDE criteria). (Refer to What Does Melanoma Look Like? on NCI's website for more information about the ABCDE criteria.) Photographs representing typical clinical presentations of these cancers are shown below.

Enlarge

Figure 2. Ulcerated basal cell carcinoma (left panel) and ulcerated basal cell carcinoma with characteristic pearly rim (right panel).

Figure 3. Superficial basal cell carcinoma (left panel) and nodular basal cell carcinoma (right panel).

Enlarge

Figure 4. Squamous cell carcinoma on the face with thick keratin top layer (left panel) and squamous cell carcinoma on the leg (right panel).

Enlarge

Figure 5. Melanomas with characteristic asymmetry, border irregularity, color variation, and large diameter.

Basal cell carcinoma (BCC) is the most common malignancy in people of European descent, with an associated lifetime risk of 30%.[1] While exposure to ultraviolet (UV) radiation is the risk factor most closely linked to the development of BCC, other environmental factors (such as ionizing radiation, chronic arsenic ingestion, and immunosuppression) and genetic factors (such as family history, skin type, and genetic syndromes) also potentially contribute to carcinogenesis. In contrast to melanoma, metastatic spread of BCC is very rare and typically arises from large tumors that have evaded medical treatment for extended periods of time. BCCs can invade tissue locally or regionally, sometimes following along nerves. A tendency for superficial necrosis has resulted in the name "rodent ulcer." With early detection, the prognosis for BCC is excellent.

This section focuses on risk factors in individuals at increased hereditary risk of developing BCC. (Refer to the PDQ summary on Skin Cancer Prevention for information about risk factors for BCC in the general population.)

Sun exposure is the major known environmental factor associated with the development of skin cancer of all types. There are different patterns of sun exposure associated with each major type of skin cancer (BCC, squamous cell carcinoma [SCC], and melanoma). (Refer to the PDQ summary on Skin Cancer Prevention for more information about sun exposure as a risk factor for skin cancer in the general population.)

The high-risk phenotype consists of individuals with the following physical characteristics:

Specifically, people with more highly pigmented skin demonstrate lower incidence of BCC than do people with lighter pigmented skin. Individuals with Fitzpatrick Type I or II skin were shown to have a twofold increased risk of BCC in a small case-control study.[2] (Refer to the Pigmentary characteristics section in the Melanoma section of this summary for a more detailed discussion of skin phenotypes based upon pigmentation.) Blond or red hair color was associated with increased risk of BCC in two large cohorts: the Nurses Health Study and the Health Professionals Follow-Up Study.[3] In women from the Nurses Health Study, there was an increased risk of BCC in women with red hair relative to those with light brown hair (adjusted relative risk [RR], 1.30; 95% confidence interval [CI], 1.201.40). In men from the Health Professionals Follow-Up Study, the risk of BCC associated with red hair was lower (RR, 1.17; 95% CI, 1.021.34) and was not significant after adjustment for melanoma family history and sunburn history.[3] Risk associated with blond hair was also increased for both men and women (RR, pooled analysis, 1.09; 95% CI, 1.021.18), and dark brown hair was protective against BCC (RR, pooled analysis, 0.89; 95% CI 0.870.92).

Individuals with BCCs and/or SCCs report a higher frequency of these cancers in their family members than do controls. The importance of this finding is unclear. Apart from defined genetic disorders with an increased risk of BCC, a positive family history of any skin cancer is a strong predictor of the development of BCC. Data from the Nurses Health Study and the Health Professionals Follow-Up Study indicate that the family history of melanoma in a first-degree relative (FDR) is associated with an increased risk of BCC in both men and women (RR, 1.31; 95% CI, 1.251.37; P <.0001).[3] A study of 376 early-onset BCC cases and 383 controls found that a family history of any type of skin cancer increased the risk of early-onset BCC (odds ratio [OR], 2.49; 95% CI, 1.803.45). This risk increased when an FDR was diagnosed with skin cancer before age 50 years (OR, 4.79; 95% CI, 2.907.90). Individuals who had a family history of both melanoma and nonmelanoma skin cancer (NMSC) had the highest risk (OR, 3.65; 95% CI, 1.797.47).[4]

A study on the heritability of cancer among 80,309 monozygotic and 123,382 dizygotic twins showed that NMSCs have a heritability of 43% (95% CI, 26%59%), suggesting that almost half of the risk of NMSC is caused by inherited factors.[5] Additionally, the cumulative risk of NMSC was 1.9-fold higher for monozygotic than for dizygotic twins (95% CI, 1.82.0).[5]

A personal history of BCC or SCC is strongly associated with subsequent BCC or SCC. There is an approximate 20% increased risk of a subsequent lesion within the first year after a skin cancer has been diagnosed. The mean age of occurrence for these NMSCs is the mid-60s.[6-11] In addition, several studies have found that individuals with a history of skin cancer have an increased risk of a subsequent diagnosis of a noncutaneous cancer;[12-15] however, other studies have contradicted this finding.[16-19] In the absence of other risk factors or evidence of a defined cancer susceptibility syndrome, as discussed below, skin cancer patients are encouraged to follow screening recommendations for the general population for sites other than the skin.

Pathogenic variants in the gene coding for the transmembrane receptor protein PTCH1, or PTCH, are associated with basal cell nevus syndrome (BCNS) and sporadic cutaneous BCCs. (Refer to the BCNS section of this summary for more information.) PTCH1, the human homolog of the Drosophila segment polarity gene patched (ptc), is an integral component of the hedgehog signaling pathway, which serves many developmental (appendage development, embryonic segmentation, neural tube differentiation) and regulatory (maintenance of stem cells) roles.

In the resting state, the transmembrane receptor protein PTCH1 acts catalytically to suppress the seven-transmembrane protein Smoothened (Smo), preventing further downstream signal transduction.[20] Binding of the hedgehog ligand to PTCH1 releases inhibition of Smo, with resultant activation of transcription factors (GLI1, GLI2), cell proliferation genes (cyclin D, cyclin E, myc), and regulators of angiogenesis.[21,22] Thus, the balance of PTCH1 (inhibition) and Smo (activation) manages the essential regulatory downstream hedgehog signal transduction pathway. Loss-of-function pathogenic variants of PTCH1 or gain-of-function variants of Smo tip this balance toward activation, a key event in potential neoplastic transformation.

Demonstration of allelic loss on chromosome 9q22 in both sporadic and familial BCCs suggested the potential presence of an associated tumor suppressor gene.[23,24] Further investigation identified a pathogenic variant in PTCH1 that localized to the area of allelic loss.[25] Up to 30% of sporadic BCCs demonstrate PTCH1 pathogenic variants.[26] In addition to BCC, medulloblastoma and rhabdomyosarcoma, along with other tumors, have been associated with PTCH1 pathogenic variants. All three malignancies are associated with BCNS, and most people with clinical features of BCNS demonstrate PTCH1 pathogenic variants, predominantly truncation in type.[27]

Truncating pathogenic variants in PTCH2, a homolog of PTCH1 mapping to chromosome 1p32.1-32.3, have been demonstrated in both BCC and medulloblastoma.[28,29] PTCH2 displays 57% homology to PTCH1.[30] While the exact role of PTCH2 remains unclear, there is evidence to support its involvement in the hedgehog signaling pathway.[28,31]

Pathogenic variants in the BAP1 gene are associated with an increased risk of a variety of cancers, including cutaneous melanoma and uveal melanoma. (Refer to the BAP1 section in the Melanoma section of this summary for more information.) Although the BCC penetrance in individuals with pathogenic variants in BAP1 is yet undescribed, there are several BAP1 families that report diagnoses of BCC.[32,33] In one study, pathogenic variant carriers from four families reported diagnoses of BCC. Tumor evaluation of BAP1 showed loss of BAP1 protein expression by immunohistochemistry in BCCs of two germline BAP1 pathogenic variant carriers but not in 53 sporadic BCCs.[32] A second report noted that four individuals from BAP1 families were diagnosed with a total of 19 BCCs. Complete loss of BAP1 nuclear expression was observed in 17 of 19 BCCs from these individuals but none of 22 control BCC specimens.[34] Loss of BAP1 nuclear expression was also reported in a series of 7 BCCs from individuals with loss of function BAP1 variants, but only in 1 of 31 sporadic BCCs.[35]

BCNS, also known as Gorlin Syndrome, Gorlin-Goltz syndrome, and nevoid BCC syndrome, is an autosomal dominant disorder with an estimated prevalence of 1 in 57,000 individuals.[36] The syndrome is notable for complete penetrance and high levels of variable expressivity, as evidenced by evaluation of individuals with identical genotypes but widely varying phenotypes.[27,37] The clinical features of BCNS differ more among families than within families.[38] BCNS is primarily associated with germline pathogenic variants in PTCH1, but families with this phenotype have also been associated with alterations in PTCH2 and SUFU.[39-41]

As detailed above, PTCH1 provides both developmental and regulatory guidance; spontaneous or inherited germline pathogenic variants of PTCH1 in BCNS may result in a wide spectrum of potentially diagnostic physical findings. The BCNS pathogenic variant has been localized to chromosome 9q22.3-q31, with a maximum logarithm of the odd (LOD) score of 3.597 and 6.457 at markers D9S12 and D9S53.[36] The resulting haploinsufficiency of PTCH1 in BCNS has been associated with structural anomalies such as odontogenic keratocysts, with evaluation of the cyst lining revealing heterozygosity for PTCH1.[42] The development of BCC and other BCNS-associated malignancies is thought to arise from the classic two-hit suppressor gene model: baseline heterozygosity secondary to germline PTCH1 pathogenic variant as the first hit, with the second hit due to mutagen exposure such as UV or ionizing radiation.[43-47] However, haploinsufficiency or dominant negative isoforms have also been implicated for the inactivation of PTCH1.[48]

The diagnosis of BCNS is typically based upon characteristic clinical and radiologic examination findings. Several sets of clinical diagnostic criteria for BCNS are in use (refer to Table 1 for a comparison of these criteria).[49-52] Although each set of criteria has advantages and disadvantages, none of the sets have a clearly superior balance of sensitivity and specificity for identifying carriers of pathogenic variants. The BCNS Colloquium Group proposed criteria in 2011 that required 1 major criterion with molecular diagnosis, two major criteria without molecular diagnosis, or one major and two minor criteria without molecular diagnosis.[52] PTCH1 pathogenic variants are found in 60% to 85% of patients who meet clinical criteria.[53,54] Most notably, BCNS is associated with the formation of both benign and malignant neoplasms. The strongest benign neoplasm association is with ovarian fibromas, diagnosed in 14% to 24% of females affected by BCNS.[46,50,55] BCNS-associated ovarian fibromas are more likely to be bilateral and calcified than sporadic ovarian fibromas.[56] Ameloblastomas, aggressive tumors of the odontogenic epithelium, have also been proposed as a diagnostic criterion for BCNS, but most groups do not include it at this time.[57]

Other associated benign neoplasms include gastric hamartomatous polyps,[58] congenital pulmonary cysts,[59] cardiac fibromas,[60] meningiomas,[61-63] craniopharyngiomas,[64] fetal rhabdomyomas,[65] leiomyomas,[66] mesenchymomas,[67] and nasal dermoid tumors. Development of meningiomas and ependymomas occurring postradiation therapy has been documented in the general pediatric population; radiation therapy for syndrome-associated intracranial processes may be partially responsible for a subset of these benign tumors in individuals with BCNS.[68-70] In addition, radiation therapy of malignant medulloblastomas in the BCNS population may result in many cutaneous BCCs in the radiation ports. Similarly, treatment of BCC of the skin with radiation therapy may result in induction of large numbers of additional BCCs.[45,46,66]

The diagnostic criteria for BCNS are described in Table 1 below.

Of greatest concern with BCNS are associated malignant neoplasms, the most common of which is BCC. BCC in individuals with BCNS may appear during childhood as small acrochordon -like lesions, while larger lesions demonstrate more classic cutaneous features.[71] Nonpigmented BCCs are more common than pigmented lesions.[72] The age at first BCC diagnosis associated with BCNS ranges from 3 to 53 years, with a mean age of 21.4 years; the vast majority of individuals are diagnosed with their first BCC before age 20 years.[50,55] Most BCCs are located on sun-exposed sites, but individuals with greater than 100 BCCs have a more uniform distribution of BCCs over the body.[72] Case series have suggested that up to 1 in 200 individuals with BCC demonstrate findings supportive of a diagnosis of BCNS.[36] BCNS has rarely been reported in individuals with darker skin pigmentation; however, significantly fewer BCCs are found in individuals of African or Mediterranean ancestry.[50,73,74] Despite the rarity of BCC in this population, reported cases document full expression of the noncutaneous manifestations of BCNS.[74] However, in individuals of African ancestry who have received radiation therapy, significant basal cell tumor burden has been reported within the radiation port distribution.[50,66] Thus, cutaneous pigmentation may protect against the mutagenic effects of UV but not against ionizing radiation.

Variants associated with an increased risk of BCC in the general population appear to modify the age of BCC onset in individuals with BCNS. A study of 125 individuals with BCNS found that a variant in MC1R (Arg151Cys) was associated with an early median age of onset of 27 years (95% CI, 2034), compared with individuals who did not carry the risk allele and had a median age of BCC of 34 years (95% CI, 3040) (hazard ratio [HR], 1.64; 95% CI, 1.042.58, P = .034). A variant in the TERT-CLPTM1L gene showed a similar effect, with individuals with the risk allele having a median age of BCC of 31 years (95% CI, 2837) relative to a median onset of 41 years (95% CI, 3248) in individuals who did not carry a risk allele (HR, 1.44; 95% CI, 1.081.93, P = .014).[75]

Many other malignancies have been associated with BCNS. Medulloblastoma carries the strongest association with BCNS and is diagnosed in 1% to 5% of BCNS cases. While BCNS-associated medulloblastoma is typically diagnosed between ages 2 and 3 years, sporadic medulloblastoma is usually diagnosed later in childhood, between the ages of 6 and 10 years.[46,50,55,76] A desmoplastic phenotype occurring around age 2 years is very strongly associated with BCNS and carries a more favorable prognosis than sporadic classic medulloblastoma.[77,78] Up to three times more males than females with BCNS are diagnosed with medulloblastoma.[79] As with other malignancies, treatment of medulloblastoma with ionizing radiation has resulted in numerous BCCs within the radiation field.[46,61] Other reported malignancies include ovarian carcinoma,[80] ovarian fibrosarcoma,[81,82] astrocytoma,[83] melanoma,[84] Hodgkin disease,[85,86] rhabdomyosarcoma,[87] and undifferentiated sinonasal carcinoma.[88]

Odontogenic keratocystsor keratocystic odontogenic tumors (KCOTs), as renamed by the World Health Organization working groupare one of the major features of BCNS.[89] Demonstration of clonal loss of heterozygosity (LOH) of common tumor suppressor genes, including PTCH1, supports the transition of terminology to reflect a neoplastic process.[42] Less than one-half of KCOTs from individuals with BCNS show LOH of PTCH1.[48,90] The tumors are lined with a thin squamous epithelium and a thin corrugated layer of parakeratin. Increased mitotic activity in the tumor epithelium and potential budding of the basal layer with formation of daughter cysts within the tumor wall may be responsible for the high rates of recurrence post simple enucleation.[89,91] In a recent case series of 183 consecutively excised KCOTs, 6% of individuals demonstrated an association with BCNS.[89] A study that analyzed the rate of PTCH1 pathogenic variants in BCNS-associated KCOTs found that 11 of 17 individuals carried a germline PTCH1 pathogenic variant and an additional 3 individuals had somatic pathogenic variants in this gene.[92] Individuals with germline PTCH1 pathogenic variants had an early age of KCOT presentation. KCOTs occur in 65% to 100% of individuals with BCNS,[50,93] with higher rates of occurrence in young females.[94]

Palmoplantar pits are another major finding in BCC and occur in 70% to 80% of individuals with BCNS.[55] When these pits occur together with early-onset BCC and/or KCOTs, they are considered diagnostic for BCNS.[95]

Several characteristic radiologic findings have been associated with BCNS, including lamellar calcification of falx cerebri;[96,97] fused, splayed or bifid ribs;[98] and flame-shaped lucencies or pseudocystic bone lesions of the phalanges, carpal, tarsal, long bones, pelvis, and calvaria.[54] Imaging for rib abnormalities may be useful in establishing the diagnosis in younger children, who may have not yet fully manifested a diagnostic array on physical examination.

Table 2 summarizes the frequency and median age of onset of nonmalignant findings associated with BCNS.

Individuals with PTCH2 pathogenic variants may have a milder phenotype of BCNS than those with PTCH1 variants. Characteristic features such as palmar/plantar pits, macrocephaly, falx calcification, hypertelorism, and coarse face may be absent in these individuals.[99]

A 9p22.3 microdeletion syndrome that includes the PTCH1 locus has been described in ten children.[100] All patients had facial features typical of BCNS, including a broad forehead, but they had other features variably including craniosynostosis, hydrocephalus, macrosomia, and developmental delay. At the time of the report, none had basal cell skin cancer. On the basis of their hemizygosity of the PTCH1 gene, these patients are presumably at an increased risk of basal cell skin cancer.

Germline pathogenic variants in SUFU, a major negative regulator of the hedgehog pathway, have been identified in a small number of individuals with a clinical phenotype resembling that of BCNS.[40,41] These pathogenic variants were first identified in individuals with childhood medulloblastoma,[101] and the incidence of medulloblastoma appears to be much higher in individuals with BCNS associated with SUFU pathogenic variants than in those with PTCH1 variants.[40] SUFU pathogenic variants may also be associated with an increased predisposition to meningioma.[63,102] Conversely, odontogenic jaw keratocysts appear less frequently in this population. Some clinical laboratories offer genetic testing for SUFU pathogenic variants for individuals with BCNS who do not have an identifiable PTCH1 variant.

Rombo syndrome, a very rare probably autosomal dominant genetic disorder associated with BCC, has been outlined in three case series in the literature.[103-105] The cutaneous examination is within normal limits until age 7 to 10 years, with the development of distinctive cyanotic erythema of the lips, hands, and feet and early atrophoderma vermiculatum of the cheeks, with variable involvement of the elbows and dorsal hands and feet.[103] Development of BCC occurs in the fourth decade.[103] A distinctive grainy texture to the skin, secondary to interspersed small, yellowish, follicular-based papules and follicular atrophy, has been described.[103,105] Missing, irregularly distributed, and/or misdirected eyelashes and eyebrows are another associated finding.[103,104] The genetic basis of Rombo syndrome is not known.

Bazex-Dupr-Christol syndrome, another rare genodermatosis associated with development of BCC, has more thorough documentation in the literature than Rombo syndrome. Inheritance is accomplished in an X-linked dominant fashion, with no reported male-to-male transmission.[106-108] Regional assignment of the locus of interest to chromosome Xq24-q27 is associated with a maximum LOD score of 5.26 with the DXS1192 locus.[109] Further work has narrowed the potential location to an 11.4-Mb interval on chromosome Xq25-27; however, the causative gene remains unknown.[110]

Characteristic physical findings include hypotrichosis, hypohidrosis, milia, follicular atrophoderma of the cheeks, and multiple BCC, which manifest in the late second decade to early third decade.[106] Documented hair changes with Bazex-Dupr-Christol syndrome include reduced density of scalp and body hair, decreased melanization,[111] a twisted/flattened appearance of the hair shaft on electron microscopy,[112] and increased hair shaft diameter on polarizing light microscopy.[108] The milia, which may be quite distinctive in childhood, have been reported to regress or diminish substantially at puberty.[108] Other reported findings in association with this syndrome include trichoepitheliomas; hidradenitis suppurativa; hypoplastic alae; and a prominent columella, the fleshy terminal portion of the nasal septum.[113,114]

A rare subtype of epidermolysis bullosa simplex (EBS), Dowling-Meara (EBS-DM), is primarily inherited in an autosomal dominant fashion and is associated with pathogenic variants in either keratin-5 (KRT5) or keratin-14 (KRT14).[115] EBS-DM is one of the most severe types of EBS and occasionally results in mortality in early childhood.[116] One report cites an incidence of BCC of 44% by age 55 years in this population.[117] Individuals who inherit two EBS pathogenic variants may present with a more severe phenotype.[118] Other less phenotypically severe subtypes of EBS can also be caused by pathogenic variants in either KRT5 or KRT14.[115] Approximately 75% of individuals with a clinical diagnosis of EBS (regardless of subtype) have KRT5 or KRT14 pathogenic variants.[119]

Characteristics of hereditary syndromes associated with a predisposition to BCC are described in Table 3 below.

(Refer to the Brooke-Spiegler Syndrome, Multiple Familial Trichoepithelioma, and Familial Cylindromatosis section in the Rare Skin Cancer Syndromes section of this summary for more information about Brooke-Spiegler syndrome.)

As detailed further below, the U.S. Preventive Services Task Force does not recommend regular screening for the early detection of any cutaneous malignancies, including BCC. However, once BCC is detected, the National Comprehensive Cancer Network guidelines of care for NMSCs recommends complete skin examinations every 6 to 12 months for life.[130]

The BCNS Colloquium Group has proposed guidelines for the surveillance of individuals with BCNS (see Table 4).

Level of evidence: 5

Avoidance of excessive cumulative and sporadic sun exposure is important in reducing the risk of BCC, along with other cutaneous malignancies. Scheduling activities outside of the peak hours of UV radiation, utilizing sun-protective clothing and hats, using sunscreen liberally, and strictly avoiding tanning beds are all reasonable steps towards minimizing future risk of skin cancer.[131] For patients with particular genetic susceptibility (such as BCNS), avoidance or minimization of ionizing radiation is essential to reducing future tumor burden.

Level of evidence: 2aii

The role of various systemic retinoids, including isotretinoin and acitretin, has been explored in the chemoprevention and treatment of multiple BCCs, particularly in BCNS patients. In one study of isotretinoin use in 12 patients with multiple BCCs, including 5 patients with BCNS, tumor regression was noted, with decreasing efficacy as the tumor diameter increased.[132] However, the results were insufficient to recommend use of systemic retinoids for treatment of BCC. Three additional patients, including one with BCNS, were followed long-term for evaluation of chemoprevention with isotretinoin, demonstrating significant decrease in the number of tumors per year during treatment.[132] Although the rate of tumor development tends to increase sharply upon discontinuation of systemic retinoid therapy, in some patients the rate remains lower than their pretreatment rate, allowing better management and control of their cutaneous malignancies.[132-134] In summary, the use of systemic retinoids for chemoprevention of BCC is reasonable in high-risk patients, including patients with xeroderma pigmentosum, as discussed in the Squamous Cell Carcinoma section of this summary.

A patients cumulative and evolving tumor load should be evaluated carefully in light of the potential long-term use of a medication class with cumulative and idiosyncratic side effects. Given the possible side-effect profile, systemic retinoid use is best managed by a practitioner with particular expertise and comfort with the medication class. However, for all potentially childbearing women, strict avoidance of pregnancy during the systemic retinoid courseand for 1 month after completion of isotretinoin and 3 years after completion of acitretinis essential to avoid potentially fatal and devastating fetal malformations.

Level of evidence (retinoids): 2aii

In a phase II study of 41 patients with BCNS, vismodegib (an inhibitor of the hedgehog pathway) has been shown to reduce the per-patient annual rate of new BCCs requiring surgery.[135] Existing BCCs also regressed for these patients during daily treatment with 150 mg of oral vismodegib. While patients treated had visible regression of their tumors, biopsy demonstrated residual microscopic malignancies at the site, and tumors progressed after the discontinuation of the therapy. Adverse effects included taste disturbance, muscle cramps, hair loss, and weight loss and led to discontinuation of the medication in 54% of subjects. Based on the side-effect profile and rate of disease recurrence after discontinuation of the medication, additional study regarding optimal dosing of vismodegib is ongoing.

Level of evidence (vismodegib): 1aii

A phase III, double-blind, placebo-controlled clinical trial evaluated the effects of oral nicotinamide (vitamin B3) in 386 individuals with a history of at least two NMSCs within 5 years before study enrollment.[136] After 12 months of treatment, those taking nicotinamide 500 mg twice daily had a 20% reduction in the incidence of new BCCs (95% CI, 6%39%; P = .12). The rate of new NMSCs was 23% lower in the nicotinamide group (95% CI, 438; P =.02) than in the placebo group. No clinically significant differences in adverse events were observed between the two groups, and there was no evidence of benefit after discontinuation of nicotinamide. Of note, this study was not conducted in a population with an identified genetic predisposition to BCC.

Level of evidence (nicotinamide): 1aii

Treatment of individual BCCs in BCNS is generally the same as for sporadic basal cell cancers. Due to the large number of lesions on some patients, this can present a surgical challenge. Field therapy with imiquimod or photodynamic therapy are attractive options, as they can treat multiple tumors simultaneously.[137,138] However, given the radiosensitivity of patients with BCNS, radiation as a therapeutic option for large tumors should be avoided.[50] There are no randomized trials, but the isolated case reports suggest that field therapy has similar results as in sporadic basal cell cancer, with higher success rates for superficial cancers than for nodular cancers.[137,138]

Consensus guidelines for the use of methylaminolevulinate photodynamic therapy in BCNS recommend that this modality may best be used for superficial BCC of all sizes and for nodular BCC less than 2 mm thick.[139] Monthly therapy with photodynamic therapy may be considered for these patients as clinically indicated.

Level of evidence (imiquimod and photodynamic therapy): 4

Topical treatment with LDE225, a Smoothened agonist, has also been investigated for the treatment of BCC in a small number of patients with BCNS with promising results;[140] however, this medication is not approved in this formulation by the U.S. Food and Drug Administration.

Level of evidence (LDE225): 1

In addition to its effects on the prevention of BCCs in patients with BCNS, vismodegib may also have a palliative effect on KCOTs found in this population. An initial report indicated that the use of GDC-0449, the hedgehog pathway inhibitor now known as vismodegib, resulted in resolution of KCOTs in one patient with BCNS.[141] Another small study found that four of six patients who took 150 mg of vismodegib daily had a reduction in the size of KCOTs.[142] None of the six patients in this study had new KCOTs or an increase in the size of existing KCOTs while being treated, and one patient had a sustained response that lasted 9 months after treatment was discontinued.

Level of evidence (vismodegib): 3diii

Squamous cell carcinoma (SCC) is the second most common type of skin cancer and accounts for approximately 20% of cutaneous malignancies. Although most cancer registries do not include information on the incidence of nonmelanoma skin cancer (NMSC), annual incidence estimates range from 1 million to 5.4 million cases in the United States.[1,2]

Mortality is rare from this cancer; however, the morbidity and costs associated with its treatment are considerable.

Sun exposure is the major known environmental factor associated with the development of skin cancer of all types; however, different patterns of sun exposure are associated with each major type of skin cancer.

Unlike basal cell carcinoma (BCC), SCC is associated with chronic exposure, rather than intermittent intense exposure to ultraviolet (UV) radiation. Occupational exposure is the characteristic pattern of sun exposure linked with SCC.[3] A case-control study in southern Europe showed increased risk of SCC when lifetime sun exposure exceeded 70,000 hours. People whose lifetime sun exposure equaled or exceeded 200,000 hours had an odds ratio (OR) 8 to 9 times that of the reference group.[4] A Canadian case-control study did not find an association between cumulative lifetime sun exposure and SCC; however, sun exposure in the 10 years before diagnosis and occupational exposure were found to be risk factors.[5]

In addition to environmental radiation, exposure to therapeutic radiation is another risk factor for SCC. Individuals with skin disorders treated with psoralen and ultraviolet-A radiation (PUVA) had a threefold to sixfold increase in SCC.[6] This effect appears to be dose-dependent, as only 7% of individuals who underwent fewer than 200 treatments had SCC, compared with more than 50% of those who underwent more than 400 treatments.[7] Therapeutic use of ultraviolet-B (UVB) radiation has also been shown to cause a mild increase in SCC (adjusted incidence rate ratio, 1.37).[8] Devices such as tanning beds also emit UV radiation and have been associated with increased SCC risk, with a reported OR of 2.5 (95% confidence interval [CI], 1.73.8).[9]

Investigation into the effect of ionizing radiation on SCC carcinogenesis has yielded conflicting results. One population-based case-control study found that patients who had undergone therapeutic radiation therapy had an increased risk of SCC at the site of previous radiation (OR, 2.94), compared with individuals who had not undergone radiation treatments.[10] Cohort studies of radiology technicians, atomic-bomb survivors, and survivors of childhood cancers have not shown an increased risk of SCC, although the incidence of BCC was increased in all of these populations.[11-13] For those who develop SCC at previously radiated sites that are not sun-exposed, the latent period appears to be quite long; these cancers may be diagnosed years or even decades after the radiation exposure.[14]

The effect of other types of radiation, such as cosmic radiation, is also controversial. Pilots and flight attendants have a reported incidence of SCC that ranges between 2.1 and 9.9 times what would be expected; however, the overall cancer incidence is not consistently elevated. Some attribute the high rate of NMSCs in airline flight personnel to cosmic radiation, while others suspect lifestyle factors.[15-20]

Like BCCs, SCCs appear to be associated with exposure to arsenic in drinking water and combustion products.[21,22] However, this association may hold true only for the highest levels of arsenic exposure. Individuals who had toenail concentrations of arsenic above the 97th percentile were found to have an approximately twofold increase in SCC risk.[23] For arsenic, the latency period can be lengthy; invasive SCC has been found to develop at an average of 20 years after exposure.[24]

Current or previous cigarette smoking has been associated with a 1.5-fold to 2-fold increase in SCC risk,[25-27] although one large study showed no change in risk.[28] Available evidence suggests that the effect of smoking on cancer risk seems to be greater for SCC than for BCC.

Additional reports have suggested weak associations between SCC and exposure to insecticides, herbicides, or fungicides.[29]

Like melanoma and BCC, SCC occurs more frequently in individuals with lighter skin than in those with darker skin.[3,30] A case-control study of 415 cases and 415 controls showed similar findings; relative to Fitzpatrick Type I skin, individuals with increasingly darker skin had decreased risks of skin cancer (ORs, 0.6, 0.3, and 0.1, for Fitzpatrick Types II, III, and IV, respectively).[31] (Refer to the Pigmentary characteristics section in the Melanoma section of this summary for a more detailed discussion of skin phenotypes based upon pigmentation.) The same study found that blue eyes and blond/red hair were also associated with increased risks of SCC, with crude ORs of 1.7 (95% CI, 1.22.3) for blue eyes, 1.5 (95% CI, 1.12.1) for blond hair, and 2.2 (95% CI, 1.53.3) for red hair.

However, SCC can also occur in individuals with darker skin. An Asian registry based in Singapore reported an increase in skin cancer in that geographic area, with an incidence rate of 8.9 per 100,000 person-years. Incidence of SCC, however, was shown to be on the decline.[30] SCC is the most common form of skin cancer in black individuals in the United States and in certain parts of Africa; the mortality rate for this disease is relatively high in these populations.[32,33] Epidemiologic characteristics of, and prevention strategies for, SCC in those individuals with darker skin remain areas of investigation.

Freckling of the skin and reaction of the skin to sun exposure have been identified as other risk factors for SCC.[34] Individuals with heavy freckling on the forearm were found to have a 14-fold increase in SCC risk if freckling was present in adulthood, and an almost threefold risk if freckling was present in childhood.[34,35] The degree of SCC risk corresponded to the amount of freckling. In this study, the inability of the skin to tan and its propensity to burn were also significantly associated with risk of SCC (OR of 2.9 for severe burn and 3.5 for no tan).

The presence of scars on the skin can also increase the risk of SCC, although the process of carcinogenesis in this setting may take years or even decades. SCCs arising in chronic wounds are referred to as Marjolins ulcers. The mean time for development of carcinoma in these wounds is estimated at 26 years.[36] One case report documents the occurrence of cancer in a wound that was incurred 59 years earlier.[37]

Immunosuppression also contributes to the formation of NMSCs. Among solid-organ transplant recipients, the risk of SCC is 65 to 250 times higher, and the risk of BCC is 10 times higher than that observed in the general population, although the risks vary with transplant type.[38-41] NMSCs in high-risk patients (solid-organ transplant recipients and chronic lymphocytic leukemia patients) occur at a younger age, are more common and more aggressive, and have a higher risk of recurrence and metastatic spread than these cancers do in the general population.[42,43] Additionally, there is a high risk of second SCCs.[44,45] In one study, over 65% of kidney transplant recipients developed subsequent SCCs after their first diagnosis.[44] Among patients with an intact immune system, BCCs outnumber SCCs by a 4:1 ratio; in transplant patients, SCCs outnumber BCCs by a 2:1 ratio.

This increased risk has been linked to an interaction between the level of immunosuppression and UV radiation exposure. As the duration and dosage of immunosuppressive agents increase, so does the risk of cutaneous malignancy; this effect is reversed with decreasing the dosage of, or taking a break from, immunosuppressive agents. Heart transplant recipients, requiring the highest rates of immunosuppression, are at much higher risk of cutaneous malignancy than liver transplant recipients, in whom much lower levels of immunosuppression are needed to avoid rejection.[38,46,47] The risk appears to be highest in geographic areas with high UV exposure.[47] When comparing Australian and Dutch organ transplant populations, the Australian patients carried a fourfold increased risk of developing SCC and a fivefold increased risk of developing BCC.[48] This finding underlines the importance of rigorous sun avoidance, particularly among high-risk immunosuppressed individuals.

Link:
Genetics of Skin Cancer (PDQ)Health Professional Version ...

Read More...

Genetics | Answers in Genesis

Monday, December 5th, 2016

Scientists have discovered an unmistakable language within all living things. Like a miniature library, DNA stores piles of information in extraordinary molecules that specify the details of everything from the shape of flower petals to the color of your eyes. A supremely intelligent Author and Life-Giver left His indelible message in every living thing.

The species on earth today descend from the original created kinds of Genesis 1. The many inter-species breedings that are possible today (e.g., zonkeys, wholphins), as well as the close similarities within biological groups (e.g., the canine group) that are distinct from one another, remind us of this fact. But exactly why the created kinds have fractured into many incompatible species has only been answered indirectly by creationists.

Successful evolution requires the addition of new information and new genes that produce new proteins that are found in new organs and systems. Losing structures, or misplacing their development, should not be equated with the increased information that is needed to form novel structures and cellular systems.

Minimal genomes is the number of genes considered essential for a bacterium to survive in a nutrient-rich, stress-free and competitor-free environment in the lab. Evolutionists believe if the genes universal to all life can be determined then its just a matter of tinkering with the existing genetic information via mutations to go from goo to you.

Continue reading here:
Genetics | Answers in Genesis

Read More...

Genetics – Advocate Health Care

Monday, December 5th, 2016

The Division of Genetics at Advocate Medical Group offers a team of genetic specialists to help individuals and families navigate the complex arena of genetics and genomics. We are committed to the diagnosis, management, and treatment of patients with genetic disorders.

Our specially trained clinical geneticists and genetic counselors provide a full range of services including:

We guide families facing hereditary and genetic disorders through complicated genetic issues in an easy-to-understand manner and provide educational resources helpful to your understanding of a genetic disorder. Our specialists can also help you identify support groups and social services, and coordinate and refer you to appropriate specialty providers based on your diagnosis.

The Division of Genetics offers comprehensive care that extends beyond genetic counseling and diagnosis. Our patients have access to multidisciplinary clinics that offer exceptional, compassionate care to children and adults with a variety of genetic disorders. Individuals in these multidisciplinary clinics have the opportunity to be evaluated by an experienced treatment team which includes multiple specialists from different healthcare disciplines.

Our specialists will determine which genetic tests are most appropriate for your particular situation. We discuss the risks, benefits, and limitations of genetic testing, as well as the emotional issues of a diagnosis and knowing your risk.

The Advocate Medical Group Genetics offers a range of services.

Follow this link:
Genetics - Advocate Health Care

Read More...

Saccharomyces Genome Database

Monday, November 14th, 2016

Rap1-GFP and Calcofluor White staining of stationary phase cells. Image courtesy of M. Guidi, M. Ruault and A. Taddei, Institut Curie (Paris).

Pma1-mCherry and Vma1-GFP localization in mitotic cells. Image courtesy of M. Eastwood, Fred Hutch and M. Meneghini, University of Toronto.

CCCP-induced decrease of mitochondrial membrane potential (below) or control treatment (above) as measured by MitoLoc. Image courtesy of Dr. Jakob Vowinckel, Ralser Lab, University of Cambridge.

Redistribution of Msn5 pools from the nucleus to the cytoplasm upon glucose deprivation. Image courtesy of H. Huang and A. Hopper, Ohio State University.

Floccule of yeast rho0 cells expressing PTS1-GFP as a peroxisomal marker, stained with calcofluor white. Image courtesy of Dr. Jakob Vowinckel, University of Cambridge

S. cerevisiae membrane proteins visualized by RFP and GFP. Image courtesy of Masur. Wikimedia Commons.

Peroxisome (red) and mitochondrial (green) fission defects in vps1 fis1 double deletion strain transformed with FIS1. Image courtesy of S. Lefevre, S. Kumar and I. van der Klei, University of Groningen.

Yeast cells expressing TRK1/GFP. Image courtesy of V. Zayats and J. Ludwig, Center of Nanobiology and Structural Biology, AV CR.

The distribution of mtDNA (green) within the mitochondrial network (red). Image courtesy of Christof Osman and Peter Walter, University of California, San Francisco

The distribution of ER exit sites (ERES, green) within the ER (red). Image courtesy of A. Nakano and K. Kurokawa, RIKEN.

Cell, actin and nuclear morphology of yeast cells treated with DMSO (left) and poacic acid (right). Images courtesy of Hiroki Okada and Yoshikazu Ohya, University of Tokyo.

Localization of active Ras in a wild type strain Image courtesy of S. Colombo and E. Martegani, University Milano Bicocca

Sectored colonies showing loss of silencing at the HML locus Image courtesy of Anne Dodson, UC Berkeley

Pma1p imaged using the RITE tagging system in mother (green) and daughter cells (red) Image courtesy of Dan Gottschling Ph.D., Fred Hutchinson Cancer Research Center

Lipid droplets in fld1 mutant images by CARS Image courtesy of Heimo Wolinski, Ph.D. and Sepp D. Kohlwein, Ph.D., University of Graz, Austria

Fpr3p accumulation in the nucleolus of S. cerevisiae Image courtesy of Amy MacQueen, Ph.D., Wesleyan University anti-Fpr3 antibody courtesy of Jeremy Thorner, Ph.D., UC Berkeley

San1 strain visualized with FUN and calcofluor white Image courtesy of the Bruschi lab, ICGEB, Trieste, Italy

Single MDN1 mRNAs detected by FISH Image courtesy of the Zenklusen Lab, Universit de Montral

Localization of Ace2-GFP to daughter cell nuclei Image courtesy of Eric Weiss, Ph.D. Northwestern University

Read more from the original source:
Saccharomyces Genome Database

Read More...

genetics facts, information, pictures | Encyclopedia.com …

Thursday, November 3rd, 2016

I. Genetics And BehaviorP. L. Broadhurst

BIBLIOGRAPHY

II. Demography and Population GeneticsJean Sutter

BIBLIOGRAPHY

III. Race and GeneticsJ. N. Spuhler

BIBLIOGRAPHY

Behavior genetics is a relatively new cross-disciplinary specialization between genetics and Psychology. It is so new that it hardly knows what to call itself. The term behavior genetics is gaining currency in the United States; but in some quarters there, and certainly elsewhere, the term psycho-genetics is favored. Logically, the best name would be genetical psychology, since the emphasis is on the use of the techniques of genetics in the analysis of behavior rather than vice versa; but the in evitable ambiguity of that term is apparent. Psy chologists generally use the terms genetic or genetical in two senses: in the first and older sense of developmental, or ontogenetic; and in the second, more recent usage relating to the analysis of inheritance. The psychologist G. Stanley Hall coined the term genetic before the turn of the century to denote developmental studies (witness the Journal of Genetic Psychology), and Alfred Binet even used the term psychogenetic in this sense. But with the rapid rise of the discipline now known as genetics after the rediscovery of the Mendelian laws in 1900, William Bateson, one of the founders of this new science, pre-empted the term genetic in naming it, thereby investing genetic with the double meaning that causes the current confusion. Psychological genetics, with its obvious abbreviation, psychogenetics, is probably the best escape from the dilemma.

Importance of genetics in behavior. The importance of psychogenetics lies in the fundamental nature of the biological processes in our understanding of human social behavior. The social sciences, and psychology in particular, have long concentrated on environmental determinants of behavior and neglected hereditary ones. But it is clear that in many psychological functions a substantial portion of the observed variation, roughly of the order of 50 per cent for many traits, can be ascribed to hereditary causation. To ignore this hereditary contribution is to impede both action and thought in this area.

This manifold contribution to behavioral variation is not a static affair. Heredity and environment interact, and behavior is the product, rather than the sum, of their respective contributions. The number of sources of variability in both he redity and environment is large, and the consequent number of such possible products even larger. Nevertheless, these outcomes are not incalculable, and experimental and other analyses of their limits are of immense potential interest to the behavioral scientist. The chief theoretical interest lies in the analysis of the evolution of behavior; and the chief practical significance, so far as can be envisaged at present, lies in the possibilities psychogenetics has for the optimization of genetic potential by manipulation of the environmental expression of it.

Major current approaches. The major approaches to the study of psychogenetics can be characterized as the direct, or experimental, and the indirect, or observational. The former derive principally from the genetical parent of this hybrid discipline and involve the manipulation of the heredity of experimental subjects, usually by restricting the choice of mates in some specially defined way. Since such techniques are not possible with human subjects a second major approach exists, the indirect or observational, with its techniques derived largely from psychology and sociology. The two approaches are largely complementary in the case of natural genetic experiments in human populations, such as twinning or cousin marriages. Thus, the distinction between the two is based on the practicability of controlling in some way the essentially immutable genetic endowmentin a word, the genotypeof the individuals subject to investigation. With typical experimental animals (rats, mice, etc.) and other organisms used by the geneticist, such as the fruit fly and many microorganisms, the genotype can often be specified in advance and populations constructed by the hybridization of suitable strains to meet this specification with a high degree of accuracy. Not so with humans, where the genotype must remain as given, and indeed where its details can rarely be specified with any degree of accuracy except for certain physical characteristics, such as blood groups. Observational, demographic, and similar techniques are therefore all that are available here. The human field has another disadvantage in rigorous psychogenetic work: the impossibility of radically manipulating the environmentfor example, by rearing humans in experimental environ ments from birth in the way that can easily be done with animals in the laboratory. Since in psychogenetics, as in all branches of genetics, one deals with a phenotypein this case, behavior and since the phenotype is the end product of the action, or better still, interaction of genotype and environment, human psychogenetics is fraught with double difficulty. Analytical techniques to be mentioned later can assist in resolving some of these difficulties.

Definition. To define psychogenetics as the study of the inheritance of behavior is to adopt a misleadingly narrow definition of the area of study, and one which is unduly restrictive in its emphasis on the hereditarian point of view. Just as the parent discipline of genetics is the analysis not only of the similarities between individuals but also of the differences between them, so psychogenetics seeks to understand the basis of individual differences in behavior. Any psychogenetic analysis must therefore be concerned with the environmental determinants of behavior (conventionally implicated in the genesis of differences) in addition to the hereditary ones (the classic source of resemblances). But manifestly this dichotomy does not always operate, so that for this reason alone the analysis of environmental effects must go hand in hand with the search for genetic causation. This is true even if the intention is merely to exclude the influence of the one the better to study the other; but the approach advocated here is to study the two in tandem, as it were, and to determine the extent to which the one interacts with the other. Psychogenetics is best viewed as that specialization which concerns itself with the interaction of heredity and environment, insofar as they affect behavior. To attempt greater precision is to become involved in subtle semantic problems about the meanings of terms.

At first sight many would tend to restrict environmental effects to those operating after the birth of the organism, but to do so would be to exclude prenatal environmental effects that have been shown to be influential in later behavior. On the other hand, to broaden the concept of environment to include all influences after fertilization the point in time at which the genotype is fixed permits consideration of the reciprocal influence of parts of the genotype upon each other. Can environment include the rest of the genotype, other than that part which is more or less directly concerned with the phenotype under consideration? This point assumes some importance since there are characteristics, not behavioralat least, none that are behavioral have so far been reported whose expression depends on the nature of the other genes present in the organism. In the absence of some of them, or rather certain alleles of the gene pairs, the value phenotypically observed would be different from what it would be if they were present. That is, different components of the genotype, in interplay with one another, modify phenotypic expression of the characteristic they in fluence. Can such indirect action, which recalls that of a chemical catalyst, best be considered as environmental or innate? It would be preferable to many to regard this mechanism as a genetic effect rather than an environmental one in the usually accepted sense. Hence, the definition of the area of study as one involving the interaction of heredity and environment, while apparently adding complexity, in fact serves to reduce confusion.

It must be conceded that this view has not as yet gained general acceptance. In some of the work reviewed in the necessarily brief survey of the major findings in this area, attempts have been made to retain a rather rigid dichotomy between heredity and environmentnature versus nurture in fact, an either/or proposition that the facts do not warrant. The excesses of both sides in the controversies of the 1920sfor example, the famous debate between Watson and McDougall over the relative importance of learned (environmental) and instinctive (genetic) determinants of behavior show the fallacies that extreme protagonists on either side can entertain if the importance of the interaction effect is ignored.

Gene action. The nature of gene action as such is essentially conducive to interaction with the environment, since the behavioral phenotype we observe is the end product of a long chain of action, principally biochemical, originating in the chromosome within the individual cell. A chromosome has a complex structure, involving DNA (deoxyribonucleic acid) and the connections of DNA with various proteins, and may be influenced in turn by another nucleic acid, RNA (ribonucleic acid), also within the cell but external to the nucleus. There are complex structures and sequences of processes, anatomical, physiological, and hormonal, which underlie normal development and differentiation of structure and function in the growth, development, and maturation of the organism. Much of this influence is determined genetically in the sense that the genotype of the organism, fixed at conception, determines how it proceeds under normal environmental circumstances. But it would be a mistake to regard any such sequence as rigid or immutable, as we shall see.

The state of affairs that arises when a number of genetically determined biochemical abnormalities affect behavior is illustrative of the argument. Many of these biochemical deficiencies or inborn errors of metabolism in humans are the outcome of a chain of causation starting with genie structures, some of them having known chromosomal locations. Their effects on the total personalitythat is, the sum total of behavorial variation that makes the individual uniquecan range from the trivial to the intense. The facility with which people can taste a solution of phenylthiocarbamide (PTC), a synthetic substance not found in nature, varies in a relatively simple genetical way: people are either tasters or nontasters in certain rather well-defined proportions, with a pattern of inheritance determined probably by one gene of major effect. But being taste blind or not is a relatively unimportant piece of behavior, since one is never likely to encounter it outside a genetical experiment. (It should perhaps be added that there is some evidence that the ability to taste PTC may be linked with other characteristics of some importance, such as susceptibility to thyroid disease.) Nevertheless, this example is insignificant compared with the psychological effect of the absence of a biochemical link in patients suffering from phenylketonuria. They are unable to metabolize phenylalanine to tyrosine in the liver, with the result that the phenylalanine accumulates and the patient suffers multiple defects, among which is usually gross intellectual defect, with an IQ typically on the order of 30. This gross biochemical failure is mediated by a single recessive gene that may be passed on in a family unnoticed in heterozygoussingle doseform but becomes painfully apparent in the unfortunate individual who happens to receive a double dose and consequently is homozygous for the defect.

Alternatively, a normal dominant gene may mutate to the recessive form and so give rise to the trouble. While mutation is a relatively rare event individually, the number of genes in each individualprobably on the order of ten thousand and the number of individuals in a population make it statistically a factor to be reckoned with. One of the best documented cases of a deleterious mutation of this kind giving rise to a major defect relates to the hemophilia transmitted, with certain important political consequences, to some of the descendants of Queen Victoria of England. The dependence of the last tsarina of Russia on the monk Rasputin was said to be based in part on the beneficial therapeutic effect of his hypnotic techniques on the uncontrollable bleeding of the Tsarevitch Alexis. Victoria was almost certainly heterozygous for hemophilia and, in view of the absence of any previous record of the defect in the Hanoverian dynasty, it seems likely that the origin of the trouble was a mutation in one of the germ cells in a testicle of Victorias father, the duke of Kent, before Victoria was conceived in August 1818.

But however it comes about, a defect such as phenylketonuria can be crippling. Fortunately, its presence can be diagnosed in very early life by a simple urine test for phenyl derivatives. The dependence of the expression of the genetic defect on the environmental circumstances is such that its effect can be mitigated by feeding the afflicted infant with a specially composed diet low in the phenylalanine with which the patients biochemical make-up cannot cope. Here again, therefore, one sees the interaction of genotype and environment in this case the type of food eaten. Many of the human biochemical defects that have been brought to light in recent years are rather simply determined genetically, in contrast with the prevailing beliefs about the bases of many behavioral characteristics including intelligence, personality, and most psychotic and neurotic disorders. This is also true of several chromosomal aberrations that have been much studied recently and that are now known to be implicated in various conditions of profound behavioral importance. Prominent among these is Downs syndrome (mongolism) with, again, effects including impairment of cognitive power. [SeeIntelligence and Intelligence Testing; Mental Disorders, articles onBiological AspectsandGenetic Aspects.]

Sex as a genetic characteristic. The sex difference is perhaps the most striking genetically determined difference in behavior and the one that is most often ignored in this connection. Primary sex is completely determined genetically at the moment of fertilization of the ovum; in mammals sex depends on whether the spermatozoon effecting fertilization bears an X or a Y chromosome to combine with the X chromosome inevitably contributed by the ovum. The resulting gamete then has the form of an XX (female) or an XY (male) individual. This difference penetrates every cell of every tissue of the resulting individual and in turn is responsible for the observable gross differences in morphology. These, in turn, subserve differences of physiological function, metabolism, and endocrine function which profoundly influence not only those aspects of behavior relating to sexual behav ior and reproductive function in the two sexes but many other aspects as well. But behavior is also influenced by social and cultural pressures, so that the resulting sex differences in behavior as observed by the psychologist are especially good examples of a phenotype that must be the and product of both genetic and environmental forces. There is a large literature on sex differences in human behavior and a sizable one on such differ ences in animal behavior, but there has been little attempt to assess this pervasive variation in terms of the relative contribution of genetic and environmental determinants. This is partly because of the technical difficulties of the problem, in the sense that all subjects must be of either one sex or the othercrossing males with females will always result in the same groups as those one started with, either males or femalesthere being, in general, no genetically intermediate sex against which to evaluate either and identical twins being inevitably of like sex. It is also partly because the potential of genetic analyses that do not involve direct experi mentation has not been realized. This is especially so since the causal routes whereby genetic determinants of sex influence many of the behavioral phenotypes observed are often better understood than in other cases where the genetic determinants underlying individual differences manifest in a population are not so clear-cut. [SeeIndividualDifferences, article onSex Differences.]

Sex linkage. There is one exception to the general lack of interest in the biometrical analysis of sex differences having behavioral connotations: sex-linked conditions. That is to say, it is demonstrated or postulated that the gene or genes responsible for the behavioroften a defect, as in the case of color blindness, which has a significantly greater incidence in males than in femalesare linked with the sex difference by virtue of their location on the sex chromosome determining genetic sex. Thus it is that sex can be thought of as a chromosomal difference of regular occurrence, as opposed to aberrations of the sort which give rise to pathological conditions, such as Downs syndrome. Indeed there are also various anomalies of genetic sex that give rise to problems of sexual identity, in which the psychological and overt be havioral consequences can be of major importance for the individual. While the evidence in such cases of environmental modification of the causative genetic conditions is less dramatic than in phenylketonuria, interaction undoubtedly exists, since these chromosomal defects of sex differentiation can in some cases be alleviated by appropriate surgical and hormonal treatment. [SeeSexual BEHavior, article onSexual Deviation: Psychological Aspects; andVision, article onColor Vision and Color Blindness.]

Human psychogenetics. It is abundantly clear that most of the phenotypes the behavioral scientist is interested in are multidetermined, both environmentally and genetically. The previous examples, however, are the exception rather than the rule, and their prominence bears witness that our understanding of genetics and behavior is as yet so little advanced that the simpler modes of genetic expression have been the first to be explored. In genetics itself, the striking differences in seed configuration used by Mendel in his classic crosses of sweet peas are determined by major genes with full dominance acting simply. But such clear-cut expression, especially of dominance, is unusual in human psychogenetics, and more complex statistical techniques are necessary to evaluate multiple genetic and environmental effects acting to produce the observed phenotype.

Whatever the analysis applied to the data gathered in other fields, in human psychogenetics the method employed cannot be the straightforward Mendelian one of crossbreeding which, in various elaborations, remains the basic tool of the geneticist today. Neither can it be the method of selection artificial, as opposed to naturalthat is other wise known as selective breeding. Indeed, none of the experimental techniques that can be applied to any other organism, whatever the phenotype being measured, is applicable to man, since experimental mating is effectively ruled out as a permissible technique in current cultures. It may be remarked in passing that such has not always been the case. The experiment of the Mogul emperor, Akbar, who reared children in isolation to determine their natural religion (and merely produced mutes) and the eugenics program of J. H. Noyes at the Oneida Community in New York State in the nineteenth century are cases in point. The apparent inbreeding of brother with sister among the rulers of ancient Egypt in the eighteenth dynasty (sixteenth to fourteenth century B.C.), which is often quoted as an example of the absence in humans of the deleterious effects of inbreeding (inbreeding depression), may not be all it seems. It is likely that the definition of sister and brother in this context did not necessarily have the same biological relevance that it has today but was rather a cultural role that could be defined, at least in this case, at will.

Twin study. In the absence of the possibility of an experimental approach, contemporary re search in human psychogenetics must rely on natural genetic experiments. Of these, the one most widely used and most industriously studied is the phenomenon of human twinning. Credit for the recognition of the value of observations on twins can be given to the nineteenth-century English scientist entist Francis Galton, who pioneered many fields of inquiry. He may be justly regarded as the father of psychogenetics for the practical methods he introduced into this field, such as the method of twin study, as well as for his influence which extended, although indirectly, even to the American experimenters in psychogenetics during the early decades of the present century.

Twin births are relatively rare in humans and vary in frequency with the ethnic group. However, the extent to which such ethnic groups differ among themselves behaviorally as a result of the undoubted genetic differences, of which incidence of multiple births is but one example, is controversial. As is well known, there are two types of twins: the monozygotic or so-called identical twins, derived from a single fertilized ovum that has split into two at an early stage in development, and the dizygotic or so-called fraternal twins, developed from two separate ova fertilized by different spermatozoa. These two physical types are not always easy to differentiate, although this difficulty is relatively miner in twin study. Nonetheless, they have led to two kinds of investigation. The first relates to differences in monozygotic twins who have identical hereditary make-up but who have been reared apart and thus subjected to different environmental influences during childhood; and the second relates to the comparison of the two types of twins usually restricted to like-sex pairs, since fraternal twins can differ in sex. The latter method supposes all differences between monozygotic pairs to be due to environmental origin, whereas the (greater) difference between dizygotic pairs is of environmental plus genetic origin. Thus, the relative contribution of the two sources of variation can be evaluated.

Findings obtained from either method have not been especially clear-cut, both because of intractable problems regarding the relative weight to be placed upon differences in the environment in which the twins have been reared and because of the sampling difficulties, which are likely to be formidable in any twin study. Nevertheless, interesting inferences can be drawn from twin study. The investigation of separated monozygotic twins has shown that while even with their identical heredity they can differ quite widely, there exists a significant resemblance in basic aspects of personality including intelligence, introversion, and neurotic tendencies, and that these resemblances can persist despite widely different environments in which the members of a pair are reared. Such findings emphasize the need to consider the contribution of genotype and environment in an inter active senseclearly some genotypes represented in the personality of monozygotic twin pairs are sensitive to environmentally induced variation, whereas others are resistant to it.

Comparisons between monozygotic and dizygotic twins reared together suggest that monozygotic twins more closely resemble each other in many aspects of personality, especially those defining psychological factors such as neuroticism and introversion-extroversion. The increase in the differences between the two types of twins when factor measures are usedas opposed to simple test scoressuggests that a more basic biological stratum is tapped by factor techniques, since the genetic determination seems greater than where individual tests are employed. Here again, the de gree to which any phenotype is shown to be hereditary in origin is valid only for the environment in which it developed and is measured; different environments may well yield different results. The problems of environmental control in human samples are so intractable that some students of the subject have questioned whether the effort and undoubted skill devoted to twin study have been well invested, in view of the inherent and persisting equivocality of the outcome.

Multivariate methods. Methods of twin study, introduced largely to improve upon the earlier methods of familial correlation (parents with off spring, sib with sib, etc.), have been combined with them. Familial correlation methods them selves have not been dealt with here, since within-family environments are bound to be even greater contaminants in determining the observed behavior than environments in twin study methods. Never theless, used on a large scale and in conjunction with twin study and with control subjects selected at random from a population, multivariate methods show promise for defining the limits of environmental and genotypic interaction. So far, the solutions to the problems of biometrical analysis posed by this type of investigation have been only partial, and the sheer weight of effort involved in locating and testing the requisite numbers of subjects standing in the required relationships has deterred all but a few pioneers. Despite the undoubtedly useful part such investigations have played in defining the problems involved, the absence of the possibility of experimental breeding has proved a drawback in the provision of socially useful data.

Animal psychogenetics. Recourse has often been had to nonhuman subjects. The additional problem thereby incurred of the relevance of comparative data to human behavior is probably balanced by the double refinements of the control of both the heredity and the environment of the experimental subjects. Two major methods of genetics have been employed, both intended to produce subjects of predetermined genotype: the crossbreeding of strains of animals of known genotype; and phenotypic selection, the mating of like with like to increase a given characteristic in a population.

Selection. Behavioral phenotypes of interest have been studied by the above methods, often using laboratory rodents. For example, attributes such as intelligence, activity, speed of conditioning, and emotionality have been selectively bred in rats.

Selection for emotional behavior in the rat will serve as an example of the techniques used and the results achieved. Rats, in common with many other mammalian species, defecate when afraid. A technique of measuring individual differences in emotional arousal is based on this propensity. The animal under test is exposed to mildly stressful noise and light stimulation in an open field or arena. The number of fecal pellets deposited serves as an index of disturbance, and in this way the extremes among a large group of rats can be characterized as high or low in emotional responsiveness. Continued selection from within the high and low groups will in time produce two distinct strains. Control of environmental variables is achieved by a rigid standardization of the conditions under which the animals are reared before being subjected to the test as adults. Careful checks on maternal effects, both prenatal and postnatal, reveal these effects to be minimal.

Such an experiment does little beyond establishing the importance of the genetic effect on the given strains in the given environment. While there are techniques for assessing the relative importance of the genetic and environmental contributions to the variation observed under selection, they are better suited to the analysis of the outcome of experiments using the alternative major genetical method, that of crossbreeding of inbred strains.

Crossbreeding. Strains used in crossbreeding experiments have usually been inbred for a phenotypic character of interest, although not usually a behavioral one. However, this does not preclude the use of these inbred strains for behavioral studies, since linkage relationships among genes ensure that selection for factors multidetermined genetically often involves multiple changes in characteristics other than those selected for, and behavior is no exception to this rule. Moreover, the existence of such inbred strains constitutes perhaps the most important single advantage of animals as subjects, since it enables simplifying assumptions regarding the homozygosity or genetic uniformity of such strains to be made in analysis of the outcome of crosses. Members of inbred strains are theoretically as alike as monozygotic twin pairs, so that genetic relationshipswhich in human populations can be investigated only after widespread efforts to find themcan be multiplied at will in laboratory animals.

This approach allows a more sensitive analysis of the determinants, both environmental and genetic, of the behavioral phenotype under observation. In addition, the nature of the genetic forces can be further differentiated into considerations of the average dominance effects of the genes in volved, the extent to which they tend to increase or decrease the metrical expression of the behavioral phenotype, and the extent to which the different strains involved possess such increasers or de creases. Finally, rough estimates of the number of these genes can be given. But the analysis depends upon meeting requirements regarding the scaling of the metric upon which the behavior is measured and is essentially a statistical one. That is, only average effects of cumulative action of the relatively large number of genes postulated as in volved can be detected. Gone are the elegantly simple statistics derived from the classical Men-delian analyses of genes of major effect, often displaying dominance, like those encountered incertain human inborn errors of metabolism. There is little evidence of the existence of comparable genes of major effect mediating behavior in laboratory animals, although some have been studied in in sects, especially the fruit fly.

A typical investigation of a behavioral phenotype might take the form of identifying two inbred strains known to differ in a behavioral trait, measuring individuals from these strains, and then systematically crossing them and measuring all offspring. When this was done for the runway performance of mice, an attribute related to their temperamental wildness, the results, analyzed by the techniques of biometrical genetics, showed that the behavior was controlled by at least three groups of genes (a probable underestimate). The contributions of these groups were additive to each other and independent of the environment when measured on a logarithmic scale but interacted with each other and with the environment on a linear scale. These genes showed a significant average dominance effect, and there was a preponderance of dominant genes in the direction of greater wildness. The heritability ratio of the contributions of nature and nurture was around seven to three.

The use of inbred lines may be restricted to first filial crosses if a number of such crosses are made from several different lines. This increases precision of analysis in addition to allowing a proportionate decrease in the amount of laboratory work. One investigation examined the exploratory behavior of six different strains of rats in an open field of the kind used for the selection mentioned above. On a linear scale there were no untoward environmental effects, including specifically prenatal maternal ones. The heritability ratio was high, around nine to one; and while there was a significant average dominance component among the genes determining exploration, there was no preponderance of dominants or recessively acting genes among increasers or decreasers. The relative standing in this respect of the parental strains could be established with some precision.

Limitations. While the methods described above have allowed the emergence of results that ultimately may assist our understanding of the mechanisms of behavioral inheritance, it cannot be said that much substantial progress has yet been made. Until experiments explore the effect of a range of different genotypes interacting with a range of environments of psychological interest and consequence, little more can be expected. Manipulating heredity in a single standard environment or manipulating the environment of a single standard genotype can only provide conclusions so limited to both the genotypes and conditions employed that they have little usefulness in a wider context. When better experiments are performed, as seems likely in the next few decades, then problems of some sociological importance and interest will arise in the application of these experiments to the tasks of maximizing genetic potential and perfecting environmental control for the purpose of so doing. A new eugenics may well develop, but grappling with the problems of its impact on contemporary society had best be left to future generations.

P. L. Broadhurst

[Directly related are the entriesEugenics; Evolution; Mental Disorders, article onGenetic Aspects. Other relevant material may be found inIndividual Differences, article onSex Differences; Instinct; Intelligence and Intelligence Testing; Mental Ertardation; Psychology, article onConstitutional Psychology.]

Broadhurst, P. L. 1960 Experiments in Psychogenetics: Applications of Biometrical Genetics to the Inheritance of Behavior. Pages 1-102 in Hans J. Eysenck (editor), Experiments in Personality. Volume 1: Psychogenetics and Psychopharmacology. London: Routledge. Selection and crossbreeding methods applied to laboratory rats.

Catteix, RaymondB.; Stice, GlenF.; and Kristy, Nor TonF. 1957 A First Approximation to Nature-Nurture Ratios for Eleven Primary Personality Factors in Objective Tests. Journal of Abnormal and Social Psychology 54:143159. Pioneer multivariate analysis combining twin study and familial correlations.

Fuller, JohnL.; and Thompson, W. Robert 1960 Be havior Genetics. New York: Wiley. A comprehen sive review of the field.

Mather, Kenneth1949 Biometrical Genetics: The Study of Continuous Variation. New York: Dover. The classic work on the analysis of quantitative char acteristics.

Shields, James1962 Monozygotic Twins Brought Up Apart and Brought Up Together: An Investigation Into the Genetic and Environmental Causes of Variation in Personality. Oxford Univ. Press.

The best available definition of population genetics is doubtless that of Malcot: It is the totality of mathematical models that can be constructed to represent the evolution of the structure of a population classified according to the distribution of its Mendelian genes (1955, p. 240). This definition, by a probabilist mathematician, gives a correct idea of the constructed and abstract side of this branch of genetics; it also makes intelligible the rapid development of population genetics since the advent of Mendelism.

In its formal aspect this branch of genetics might even seem to be a science that is almost played out. Indeed, it is not unthinkable that mathematicians have exhausted all the structural possibilities for building models, both within the context of general genetics and within that of the hypothesesmore or less complex and abstractthat enable us to characterize the state of a population.

Two major categories of models can be distinguished: determinist models are those in which variations in population composition over time are rigorously determined by (a) a known initial state of the population; (b) a known number of forces or pressures operating, in the course of generations, in an unambiguously defined fashion (Male-cot 1955, p. 240). These pressures involve mutation, selection, and preferential marriages (by consanguinity, for instance). Determinist models, based on ratios that have been exactly ascertained from preceding phenomena, can be expressed only in terms of populations that are infinite in the mathematical sense. In fact, it is only in this type of population that statistical regularities can emerge (Malecot 1955). In these models the composition of each generation is perfectly defined by the composition of the preceding generation.

Stochastic models, in contrast to determinist ones, involve only finite populations, in which the gametes that, beginning with the first generation, are actually going to give birth to the new generation represent only a finite number among all possible gametes. The result is that among these active, or useful, gametes (Malecot 1959), male or female, the actual frequency of a gene will differ from the probability that each gamete had of carrying it at the outset.

The effect of chance will play a prime role, and the frequencies of the genes will be able to drift from one generation to the other. The effects of random drift and of genetic drift become, under these conditions, the focal points for research.

The body of research completed on these assumptions does indeed form a coherent whole, but these results, in spite of their brilliance, are marked by a very noticeable formalism. In reality, the models, although of great importance at the conceptual level, are often too far removed from the facts. In the study of man, particularly, the problems posed are often too complex for the solutions taken directly from the models to describe concrete reality.

Not all these models, however, are the result of purely abstract speculation; construction of some of them has been facilitated by experimental data. To illustrate this definition of population genetics and the problems that it raises, this article will limit itself to explaining one determinist model, both because it is one of the oldest and simplest to under stand and because it is one of those most often verified by observation.

A determinist model. Let us take the case of a particular human population: the inhabitants of an island cut off from outside contacts. It is obvious that great variability exists among the genes carried by the different inhabitants of this island. The genotypes differ materially from one another; in other words, there is a certain polymorphism in the populationpolymorphism that we can define in genetic terms with the help of a simple example.

Let us take the case of autosome (not connected with sex) gene a, transmitting itself in a mono-hybrid diallely. In relation to it individuals can be classified in three categories: homozygotes whose two alleles are a (a/a); heterozygotes, carriers of a and its allele a (a/a); and the homozygotes who are noncarriers of a (a/a). At any given moment or during any given generation, these three categories of individuals exist within the population in certain proportions relative to each other.

Now, according to Mendels second law (the law of segregation), the population born out of a cross between an individual who is homozygote for a (a/a) and an individual who is homozygote for a (a/a) will include individuals a/a, a/a, and a/a in the following proportions: one-fourth a/a, one-half a/a, and one-fourth a/a. In this popu lation the alleles a and a have the same frequency, one-half, and each sex produces half a and half a. If these individuals are mated randomly, a simple algebraic calculation quickly demonstrates that individuals of the generation following will be quan titatively distributed in the same fashion: one-fourth a/a, one-half a/a, and one-fourth a/a. It will be the same for succeeding generations.

It can therefore be stated that the genetic structure of such a population does not vary from one generation to the other. If we designate by p the initial proportion of a/a individuals and by q that of a/a individuals, we get p + q = 1, or the totality of the population. Applying this system of symbols to the preceding facts, it can be easily shown that the proportion of individuals of all three categories in the first generation born from a/a and a/a equals p2, 2pq, q2. In the second and third generation the frequency of individuals will always be similar: p2, 2pq, q2.

Until this point, we have remained at the individual level. If we proceed to that of the gametes carrying a or a and to that of genes a and a, we observe that their frequencies intermingle. In the type of population discussed above, the formula p2, 2pq, q2 still applies perfectly, therefore, to the gametes and genes. This model, which can be regarded as a formalization of the Hardy-Weinberg law, has other properties, but our study of it will stop here. (For a discussion of the study of isolated populations, see Sutter & Tabah 1951.)

Model construction and demographic reality. The Hardy-Weinberg law has been verified by numerous studies, involving both vegetable and animal species. The findings in the field of human blood groups have also been studied for a long time from a viewpoint derived implicitly from this law, especially in connection with their geographic distribution. Under the system of reproduction by sexes, a generation renews itself as a result of the encounter of the sexual cells (gametes) produced by individuals of both sexes belonging to the living generation. In the human species it can be said that this encounter takes place at random. One can imagine the advantage that formal population genetics can take of this circumstance, which can be compared to drawing marked balls by lot from two different urns. Model construction, already favored by these circumstances, is favored even further if the characteristics of the population utilized are artificially defined with the help of a certain number of hypotheses, of which the following is a summary description:

(1) Fertility is identical for all couples; there is no differential fertility.

(2) The population is closed; it cannot, there fore, be the locus of migrations (whether immigration or emigration).

(3) Marriages take place at random; there is no assortative mating.

(4) There are no systematic preferential marriages (for instance, because of consanguinity).

(5) Possible mutations are not taken into consideration.

(6) The size of the population is clearly denned. On the basis of these working hypotheses, the whole of which constitutes panmixia, it was possible, not long after the rediscovery of Mendels laws, to construct the first mathematical models. Thus, population genetics took its first steps forward, one of which was undoubtedly the Hardy-Weinberg law.

Mere inspection of the preceding hypotheses will enable the reader to judge how, taken one by one, they conflict with reality. In fact, no human population can be panmictic in the way the models are.

The following evidence can be cited in favor of this conclusion:

(1) Fertility is never the same with all couples. In fact, differential fertility is the rule in human populations. There is always a far from negligible sterility rate of about 18 per cent among the large populations of Western civilization. On the other hand, the part played by large families in keeping up the numbers of these populations is extremely important; we can therefore generalize by emphasizing that for one or another reason individ uals carrying a certain assortment of genes reproduce themselves more or less than the average number of couples. That is what makes for the fact that in each population there is always a certain degree of selection. Hypothesis (1) above, essential to the construction of models, is therefore very far removed from reality.

(2) Closed populations are extremely rare. Even among the most primitive peoples there is always a minimum of emigration or immigration. The only cases where one could hope to see this condition fulfilled at the present time would be those of island populations that have remained extremely primitive.

(3) With assortative mating we touch on a point that is still obscure; but even if these phe nomena remain poorly understood, it can nevertheless be said that they appear to be crucial in determining the genetic composition of populations. This choice can be positive: the carriers of a given characteristic marry among each other more often than chance would warrant. The fact was demonstrated in England by Pearson and Lee (1903): very tall individuals have a tendency to marry each other, and so do very short ones. Willoughby (1933) has reported on this question with respect to a great number of somatic characteristics other than heightfor example, coloring of hair, eyes and skin, intelligence quotient, and so forth. Inversely, negative choice makes individuals with the same characteristics avoid marrying one another. This mechanism is much less well known than the above. The example of persons of violent nature (Dahlberg 1943) and of red-headed individuals has been cited many times, although it has not been possible to establish valid statistics to support it.

(4) The case of preferential marriages is not at all negligible. There are still numerous areas where marriages between relatives (consanguineous marriages) occur much more frequently than they would as the result of simple random encounters. In addition, recent studies on the structures of kinship have shown that numerous populations that do not do so today used to practice preferential marriagemost often in a matrilinear sense. These social phenomena have a wide repercussion on the genetic structure of populations and are capable of modifying them considerably from one generation to the other.

(5) Although we do not know exactly what the real rates of mutation are, it can be admitted that their frequency is not negligible. If one or several genes mutate at a given moment in one or several individuals, the nature of the gene or genes is in this way modified; its stability in the population undergoes a disturbance that can considerably transform the composition of that population.

(6) The size of the population and its limits have to be taken into account. We have seen that this is one of the essential characteristics important in differentiating two large categories of models.

The above examination brings us into contact with the realities of population: fertility, fecundity, nuptiality, mortality, migration, and size are the elements that are the concern of demography and are studied not only by this science but also very often as part of administrative routine. Leaving aside the influence of size, which by definition is of prime importance in the technique of the models, there remain five factors to be examined from the demographic point of view. Mutation can be ruled out of consideration, because, although its importance is great, it is felt only after the passage of a certain number of generations. It can therefore be admitted that it is not of immediate interest.

We can also set aside choice of a mate, because the importance of this factor in practice is still unknown. Accordingly, there remain three factors of prime importance: fertility, migration, and preferential marriage. Over the last decade the progressive disappearance of consanguineous marriage has been noted everywhere but in Asia. In many civilized countries marriage between cousins has practically disappeared. It can be stated, therefore, that this factor has in recent years become considerably less important.

Migrations remain very important on the genetic level, but, unfortunately, precise demographic data about them are rare, and most of the data are of doubtful validity. For instance, it is hard to judge how their influence on a population of Western culture could be estimated.

The only remaining factor, fertility (which to geneticists seems essential), has fortunately been studied in satisfactory fashion by demographers. To show the importance of differential fertility in human populations, let us recall a well-known cal culation made by Karl Pearson in connection with Denmark. In 1830, 50 per cent of the children in that country were born of 25 per cent of the parents. If that fertility had been maintained at the same rate, 73 per cent of the second-generation Danes and 97 per cent of the third generation would have been descended from the first 25 per cent. Similarly, before World War I, Charles B. Davenport calculated, on the basis of differential fertility, that 1,000 Harvard graduates would have only 50 descendants after two centuries, while 1,000 Rumanian emigrants living in Boston would have become 100,000.

Human reproduction involves both fecundity (capacity for reproduction) and fertility (actual reproductive performance). These can be estimated for males, females, and married couples treated as a reproductive unit. Let us rapidly review the measurements that demography provides for geneticists in this domain.

Crude birth rate. The number of living births in a calendar year per thousand of the average population in the same year is known as the crude birth rate. The rate does not seem a very useful one for geneticists: there are too many different groups of childbearing age; marriage rates are too variable from one population to another; birth control is not uniformly diffused, and so forth.

General fertility rate. The ratio of the number of live births in one year to the average number of women capable of bearing children (usually defined as those women aged 15 to 49) is known as the general fertility rate. Its genetic usefulness is no greater than that of the preceding figure. Moreover, experience shows that this figure is not very different from the crude birth rate.

Age-specific fertility rates. Fertility rates according to the age reached by the mother during the year under consideration are known as age-specific fertility rates. Demographic experience shows that great differences are observed here, depending on whether or not the populations are Malthusianin other words, whether they practice birth control or not. In the case of a population where the fertility is natural, knowledge of the mothers age is sufficient. In cases where the population is Malthusian, the figure becomes interesting when it is calculated both by age and by age group of the mothers at time of marriage, thus combining the mothers age at the birth of her child and her age at marriage. This is generally known as the age-specific marital fertility rate. If we are dealing with a Malthusian population, it is preferable, in choosing the sample to be studied, to take into consideration the age at marriage rather than the age at the childs birth. Thus, while the age at birth is sufficient for natural populations, these techniques cannot be applied indiscriminately to all populations.

Family histories. Fertility rates can also be calculated on the basis of family histories, which can be reconstructed from such sources as parish registries (Fleury & Henry 1965) or, in some countries, from systematic family registrations (for instance, the Japanese koseki or honseki). The method for computing the fertility rate for, say, the 25-29-year-old age group from this kind of data is first to determine the number of legitimate births in the group. It is then necessary to make a rigorous count of the number of years lived in wedlock between their 25th and 30th birthdays by all the women in the group; this quantity is known as the groups total woman-years. The number of births is then divided by the number of woman-years to obtain the groups fertility rate. This method is very useful in the study of historical problems in genetics, since it is often the only one that can be applied to the available data.

Let us leavefer tility rates in order to examine rates of reproduction. Here we return to more purely genetic considerations, since we are looking for the mechanism whereby one generation is replaced by the one that follows it. Starting with a series of fertility rates by age groups, a gross reproduction rate can be calculated that gives the average number of female progeny that would be born to an age cohort of women, all of whom live through their entire reproductive period and continue to give birth at the rates prevalent when they themselves were born. The gross reproduction rate obtaining for a population at any one time can be derived by combining the rates for the different age cohorts.

A gross reproduction rate for a real generation can also be determined by calculating the average number of live female children ever born to women of fifty or over. As explained above, this rate is higher for non-Malthusian than for Malthusian populations and can be refined by taking into consideration the length of marriage.

We have seen that in order to be correct, it is necessary for the description of fertility in Malthusian populations to be closely related to the date of marriage. Actually, when a family reaches the size that the parents prefer, fertility tends to approach zero. The preferred size is evidently related to length of marriage in such a manner that fertility is more closely linked with length of marriage than with age at marriage. In recent years great progress has been made in the demographic analysis of fertility, based on this kind of data. This should en ablegeneticists to be more circumspect in their choice of sections of the population to be studied.

Americans talk of cohort analysis, the French of analysis by promotion (a term meaning year or class, as we might speak of the class of 1955). A cohort, or promotion, includes all women born within a 12-month period; to estimate fertility or mortality, it is supposed that these women are all born at the same moment on the first of January of that year. Thus, women born between January 1, 1900, and January 1, 1901, are considered to be exactly 15 years old on January 1, 1915; exactly 47 years old on January 1, 1947; and so forth.

The research done along these lines has issued in the construction of tables that are extremely useful in estimating fertility in a human population. As we have seen, it is more useful to draw up cohorts based on age at marriage than on age at birth. A fertility table set up in this way gives for each cohort the cumulative birth rate, by order of birth and single age of mother, for every woman surviving at each age, from 15 to 49. The progress that population genetics could make in knowing real genie frequencies can be imagined, if it could concentrate its research on any particular cohort and its descendants.

This rapid examination of the facts that demography can now provide in connection with fertility clearly reveals the variables that population genetics can use to make its models coincide with reality. The models retain their validity for genetics because they are still derived from basic genetic concepts; their application to actual problems, however, should be based on the kind of data mentioned above. We have voluntarily limited ourselves to the problem of fertility, since it is the most important factor in genetics research.

The close relationship between demography and population genetics that now appears can be illustrated by the field of research into blood groups. Although researchers concede that blood groups are independent of both age and sex, they do not explore the full consequences of this, since their measures are applied to samples of the population that are representative only in a demographic sense. We must deplore the fact that this method has spread to the other branches of genetics, since it is open to criticism not only from the demographic but from the genetic point of view. By proceeding in this way, a most important factor is overlookedthat of genie frequencies.

Let us admit that the choice of a blood group to be studied is of little impor tance when the characteristic is widely distributed throughout the populationfor instance, if each individual is the carrier of a gene taken into account in the system being studied (e.g., a system made up of groups A, B, and O). But this is no longer the case if the gene is carried only by a few individualsin other words, if its frequency attains 0.1 per cent or less. In this case (and cases like this are common in human genetics) the structure of the sample examined begins to take on prime importance.

A brief example must serve to illustrate this cardinal point. We have seen that in the case of rare recessive genes the importance of consan guineous marriages is considerable. The scarcer that carriers of recessive genes become in the pop ulation as a whole, the greater the proportion of such carriers produced by consanguineous marriages. Thus if as many as 25 per cent of all individuals in a population are carriers of recessive genes, and if one per cent of all marriages in that population are marriages between first cousins, then this one per cent of consanguineous unions will produce 1.12 times as many carriers of recessive genes as will be produced by all the unions of persons not so related. But if recessive genes are carried by only one per cent of the total population, then the same proportion of marriages between first cousins will produce 2.13 times as many carriers as will be produced by all other marriages. This production ratio increases to 4.9 if the total frequency of carriers is .01 per cent, to 20.2 if it is .005 per cent, and to 226 if it is .0001 per cent. Under these conditions, one can see the importance of the sampling method used to estimate the frequency within a population, not only of the individuals who are carriers but of the gametes and genes themselves.

Genealogical method. It should be emphasized that genetic studies based on genealogies remain the least controversial. Studying a population where the degrees of relationship connecting individuals are known presents an obvious interest. Knowing one or several characteristics of certain parents, we can follow what becomes of these in the descendants. Their evolution can also be considered from the point of view of such properties of genes as dominance, recessiveness, expressivity, and penetrance. But above all, we can follow the evolution of these characteristics in the population over time and thus observe the effects of differential fertility. Until now the genealogical method was applicable only to a numerically sparse population, but progress in electronic methods of data processing permits us to anticipate its application to much larger populations (Sutter & Tabah 1956).

Dynamic studies. In very large modern populations it would appear that internal analysis of cohorts and their descendants will bring in the future a large measure of certainty to research in population genetics. In any case, it is a sure way to a dynamic genetics based on demographic reality. For instance, it has been recommended that blood groups should be studied according to age groups; but if we proceed to do so without regard for demographic factors, we cannot make our observations dynamic. Thus, a study that limits itself to, let us say, the fifty- to sixty-year-old age group will have to deal with a universe that includes certain genetically dead elements, such as unmarried and sterile persons, which have no meaning from the dynamic point of view. But if a study is made of this same fifty- to sixty-year-old age group and then of the twenty- to thirty-year-old age group, and if in the older group only those individuals are considered who have descendants in the younger group, the dynamic potential of the data is maximized. It is quite possible to subject demographic cohorts to this sort of interpretation, because in many countries demographic statistics supply series of individuals classified according to the mothers age at their birth.

This discussion would not be complete if we did not stress another aspect of the genetic importance of certain demographic factors, revealed by modern techniques, which have truly created a demographic biology. Particularly worthy of note are the mothers age, order of birth, spacing between births, and size of family.

The mothers age is a great influence on fecundity. A certain number of couples become in capable of having a second child after the birth of the first child; a third child after the second; a fourth after the third; and so forth. This sterility increases with the length of a marriage and especially after the age of 35. It is very important to realize this when, for instance, natural selection and its effects are being studied.

The mothers age also strongly influences the frequency of twin births (monozygotic or dizygotic), spontaneous abortions, stillborn or abnormal births, and so on. Many examples can also be given of the influence of the order of birth, the interval between births, and the size of the family to illustrate their effect on such things as fertility, mortality, morbidity, and malformations.

It has been demonstrated above how seriously demographic factors must be taken into consideration when we wish to study the influence of the genetic structure of populations. We will leave aside the possible environmental influences, such as social class and marital status, since they have previously been codified by Osborn (1956/1957) and Larsson (19561957), among others. At the practical level, however, the continuing efforts to utilize vital statistics for genetic purposes should be pointed out. In this connection, the research of H. B. Newcombe and his colleagues (1965), who are attempting to organize Canadian national statistics for use in genetics, cannot be too highly praised. The United Nations itself posed the problem on the world level at a seminar organized in Geneva in 1960. The question of the relation between demography and genetics is therefore being posed in an acute form.

These problems also impinge in an important way on more general philosophical issues, as has been demonstrated by Haldane (1932), Fisher (1930), and Wright (1951). It must be recognized, however, that their form of Neo-Darwinism, although it is based on Mendelian genetics, too often neglects demographic considerations. In the future these seminal developments should be renewed in full confrontation with demographic reality.

Jean Sutter

[Directly related are the entriesCohort Analysis; Fertility; Fertility Control. Other relevant ma terial may be found inNuptiality; Race; SocialBehavior, Animal, article onThe Regulation of Animal Populations.]

Barclay, George W. 1958 Techniques of Population Analysis. New York: Wiley.

Dahlberg, Gunnar(1943)1948 Mathematical Methods for Population Genetics. New York and London: Inter-science. First published in German.

Dunn, Leslie C. (editor) 1951 Genetics in the Twentieth Century: Essays on the Progress of Genetics During Its First Fifty Years. New York: Macmillan.

Fisher, R. A. (1930) 1958 The Genetical Theory of Natural Selection. 2d ed., rev. New York: Dover.

See original here:
genetics facts, information, pictures | Encyclopedia.com ...

Read More...

Annual Reviews – Home

Sunday, October 30th, 2016

This site uses cookies to improve performance. If your browser does not accept cookies, you cannot view this site.

There are many reasons why a cookie could not be set correctly. Below are the most common reasons:

This site uses cookies to improve performance by remembering that you are logged in when you go from page to page. To provide access without cookies would require the site to create a new session for every page you visit, which slows the system down to an unacceptable level.

This site stores nothing other than an automatically generated session ID in the cookie; no other information is captured.

In general, only the information that you provide, or the choices you make while visiting a web site, can be stored in a cookie. For example, the site cannot determine your email name unless you choose to type it. Allowing a website to create a cookie does not give that or any other site access to the rest of your computer, and only the site that created the cookie can read it.

Read more:
Annual Reviews - Home

Read More...

Genetics – Wikipedia

Thursday, October 20th, 2016

This article is about the general scientific term. For the scientific journal, see Genetics (journal).

Genetics is the study of genes, genetic variation, and heredity in living organisms.[1][2] It is generally considered a field of biology, but it intersects frequently with many of the life sciences and is strongly linked with the study of information systems.

The father of genetics is Gregor Mendel, a late 19th-century scientist and Augustinian friar. Mendel studied 'trait inheritance', patterns in the way traits were 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.

Trait inheritance and molecular inheritance mechanisms of genes are still primary principles of genetics in the 21st century, but modern genetics has expanded beyond inheritance to studying the function and behavior of genes. Gene structure and function, variation, and distribution are studied within the context of the cell, the organism (e.g. dominance) and within the context of a population. Genetics has given rise to a number of sub-fields including epigenetics and population genetics. Organisms studied within the broad field span the domain of life, including bacteria, plants, animals, and humans.

Genetic processes work in combination with an organism's environment and experiences to influence development and behavior, often referred to as nature versus nurture. The intra- or extra-cellular environment of a cell or organism may switch gene transcription on or off. A classic example is two seeds of genetically identical corn, one placed in a temperate climate and one in an arid climate. While the average height of the two corn stalks may be genetically determined to be equal, the one in the arid climate only grows to half the height of the one in the temperate climate due to lack of water and nutrients in its environment.

The word genetics stems from the Ancient Greek genetikos meaning "genitive"/"generative", which in turn derives from genesis meaning "origin".[3][4][5]

The observation that living things inherit traits from their parents has been used since prehistoric times to improve crop plants and animals through selective breeding.[6] The modern science of genetics, seeking to understand this process, began with the work of Gregor Mendel in the mid-19th century.[7]

Prior to Mendel, Imre Festetics, a Hungarian noble, who lived in Kszeg before Mendel, was the first who used the word "genetics". He described several rules of genetic inheritance in his work The genetic law of the Nature (Die genetische Gestze der Natur, 1819). His second law is the same as what Mendel published. In his third law, he developed the basic principles of mutation (he can be considered a forerunner of Hugo de Vries.)[8]

Other theories of inheritance preceded his work. A popular theory during Mendel's time was the concept of blending inheritance: the idea that individuals inherit a smooth blend of traits from their parents.[9] Mendel's work provided examples where traits were definitely not blended after hybridization, showing that traits are produced by combinations of distinct genes rather than a continuous blend. Blending of traits in the progeny is now explained by the action of multiple genes with quantitative effects. Another theory that had some support at that time was the inheritance of acquired characteristics: the belief that individuals inherit traits strengthened by their parents. This theory (commonly associated with Jean-Baptiste Lamarck) is now known to be wrongthe experiences of individuals do not affect the genes they pass to their children,[10] although evidence in the field of epigenetics has revived some aspects of Lamarck's theory.[11] Other theories included the pangenesis of Charles Darwin (which had both acquired and inherited aspects) and Francis Galton's reformulation of pangenesis as both particulate and inherited.[12]

Modern genetics started with Gregor Johann Mendel, a scientist and Augustinian friar who studied the nature of inheritance in plants. In his paper "Versuche ber Pflanzenhybriden" ("Experiments on Plant Hybridization"), presented in 1865 to the Naturforschender Verein (Society for Research in Nature) in Brnn, Mendel traced the inheritance patterns of certain traits in pea plants and described them mathematically.[13] Although this pattern of inheritance could only be observed for a few traits, Mendel's work suggested that heredity was particulate, not acquired, and that the inheritance patterns of many traits could be explained through simple rules and ratios.

The importance of Mendel's work did not gain wide understanding until the 1890s, after his death, when other scientists working on similar problems re-discovered his research. William Bateson, a proponent of Mendel's work, coined the word genetics in 1905.[14][15] (The adjective genetic, derived from the Greek word genesis, "origin", predates the noun and was first used in a biological sense in 1860.)[16] Bateson both acted as a mentor and was aided significantly by the work of women scientists from Newnham College at Cambridge, specifically the work of Becky Saunders, Nora Darwin Barlow, and Muriel Wheldale Onslow.[17] Bateson popularized the usage of the word genetics to describe the study of inheritance in his inaugural address to the Third International Conference on Plant Hybridization in London, England, in 1906.[18]

After the rediscovery of Mendel's work, scientists tried to determine which molecules in the cell were responsible for inheritance. In 1911, Thomas Hunt Morgan argued that genes are on chromosomes, based on observations of a sex-linked white eye mutation in fruit flies.[19] In 1913, his student Alfred Sturtevant used the phenomenon of genetic linkage to show that genes are arranged linearly on the chromosome.[20]

Although genes were known to exist on chromosomes, chromosomes are composed of both protein and DNA, and scientists did not know which of the two is responsible for inheritance. In 1928, Frederick Griffith discovered the phenomenon of transformation (see Griffith's experiment): dead bacteria could transfer genetic material to "transform" other still-living bacteria. Sixteen years later, in 1944, the AveryMacLeodMcCarty experiment identified DNA as the molecule responsible for transformation.[21] The role of the nucleus as the repository of genetic information in eukaryotes had been established by Hmmerling in 1943 in his work on the single celled alga Acetabularia.[22] The HersheyChase experiment in 1952 confirmed that DNA (rather than protein) is the genetic material of the viruses that infect bacteria, providing further evidence that DNA is the molecule responsible for inheritance.[23]

James Watson and Francis Crick determined the structure of DNA in 1953, using the X-ray crystallography work of Rosalind Franklin and Maurice Wilkins that indicated DNA had a helical structure (i.e., shaped like a corkscrew).[24][25] Their double-helix model had two strands of DNA with the nucleotides pointing inward, each matching a complementary nucleotide on the other strand to form what looks like rungs on a twisted ladder.[26] This structure showed that genetic information exists in the sequence of nucleotides on each strand of DNA. The structure also suggested a simple method for replication: if the strands are separated, new partner strands can be reconstructed for each based on the sequence of the old strand. This property is what gives DNA its semi-conservative nature where one strand of new DNA is from an original parent strand.[27]

Although the structure of DNA showed how inheritance works, it was still not known how DNA influences the behavior of cells. In the following years, scientists tried to understand how DNA controls the process of protein production.[28] It was discovered that the cell uses DNA as a template to create matching messenger RNA, molecules with nucleotides very similar to DNA. The nucleotide sequence of a messenger RNA is used to create an amino acid sequence in protein; this translation between nucleotide sequences and amino acid sequences is known as the genetic code.[29]

With the newfound molecular understanding of inheritance came an explosion of research.[30] A notable theory arose from Tomoko Ohta in 1973 with her amendment to the neutral theory of molecular evolution through publishing the nearly neutral theory of molecular evolution. In this theory, Ohta stressed the importance of natural selection and the environment to the rate at which genetic evolution occurs.[31] One important development was chain-termination DNA sequencing in 1977 by Frederick Sanger. This technology allows scientists to read the nucleotide sequence of a DNA molecule.[32] In 1983, Kary Banks Mullis developed the polymerase chain reaction, providing a quick way to isolate and amplify a specific section of DNA from a mixture.[33] The efforts of the Human Genome Project, Department of Energy, NIH, and parallel private efforts by Celera Genomics led to the sequencing of the human genome in 2003.[34]

At its most fundamental level, inheritance in organisms occurs by passing discrete heritable units, called genes, from parents to progeny.[35] This property was first observed by Gregor Mendel, who studied the segregation of heritable traits in pea plants.[13][36] In his experiments studying the trait for flower color, Mendel observed that the flowers of each pea plant were either purple or whitebut never an intermediate between the two colors. These different, discrete versions of the same gene are called alleles.

In the case of the pea, which is a diploid species, each individual plant has two copies of each gene, one copy inherited from each parent.[37] Many species, including humans, have this pattern of inheritance. Diploid organisms with two copies of the same allele of a given gene are called homozygous at that gene locus, while organisms with two different alleles of a given gene are called heterozygous.

The set of alleles for a given organism is called its genotype, while the observable traits of the organism are called its phenotype. When organisms are heterozygous at a gene, often one allele is called dominant as its qualities dominate the phenotype of the organism, while the other allele is called recessive as its qualities recede and are not observed. Some alleles do not have complete dominance and instead have incomplete dominance by expressing an intermediate phenotype, or codominance by expressing both alleles at once.[38]

When a pair of organisms reproduce sexually, their offspring randomly inherit one of the two alleles from each parent. These observations of discrete inheritance and the segregation of alleles are collectively known as Mendel's first law or the Law of Segregation.

Geneticists use diagrams and symbols to describe inheritance. A gene is represented by one or a few letters. Often a "+" symbol is used to mark the usual, non-mutant allele for a gene.[39]

In fertilization and breeding experiments (and especially when discussing Mendel's laws) the parents are referred to as the "P" generation and the offspring as the "F1" (first filial) generation. When the F1 offspring mate with each other, the offspring are called the "F2" (second filial) generation. One of the common diagrams used to predict the result of cross-breeding is the Punnett square.

When studying human genetic diseases, geneticists often use pedigree charts to represent the inheritance of traits.[40] These charts map the inheritance of a trait in a family tree.

Organisms have thousands of genes, and in sexually reproducing organisms these genes generally assort independently of each other. This means that the inheritance of an allele for yellow or green pea color is unrelated to the inheritance of alleles for white or purple flowers. This phenomenon, known as "Mendel's second law" or the "Law of independent assortment", means that the alleles of different genes get shuffled between parents to form offspring with many different combinations. (Some genes do not assort independently, demonstrating genetic linkage, a topic discussed later in this article.)

Often different genes can interact in a way that influences the same trait. In the Blue-eyed Mary (Omphalodes verna), for example, there exists a gene with alleles that determine the color of flowers: blue or magenta. Another gene, however, controls whether the flowers have color at all or are white. When a plant has two copies of this white allele, its flowers are whiteregardless of whether the first gene has blue or magenta alleles. This interaction between genes is called epistasis, with the second gene epistatic to the first.[41]

Many traits are not discrete features (e.g. purple or white flowers) but are instead continuous features (e.g. human height and skin color). These complex traits are products of many genes.[42] The influence of these genes is mediated, to varying degrees, by the environment an organism has experienced. The degree to which an organism's genes contribute to a complex trait is called heritability.[43] Measurement of the heritability of a trait is relativein a more variable environment, the environment has a bigger influence on the total variation of the trait. For example, human height is a trait with complex causes. It has a heritability of 89% in the United States. In Nigeria, however, where people experience a more variable access to good nutrition and health care, height has a heritability of only 62%.[44]

The molecular basis for genes is deoxyribonucleic acid (DNA). DNA is composed of a chain of nucleotides, of which there are four types: adenine (A), cytosine (C), guanine (G), and thymine (T). Genetic information exists in the sequence of these nucleotides, and genes exist as stretches of sequence along the DNA chain.[45]Viruses are the only exception to this rulesometimes viruses use the very similar molecule, RNA, instead of DNA as their genetic material.[46] Viruses cannot reproduce without a host and are unaffected by many genetic processes, so tend not to be considered living organisms.

DNA normally exists as a double-stranded molecule, coiled into the shape of a double helix. Each nucleotide in DNA preferentially pairs with its partner nucleotide on the opposite strand: A pairs with T, and C pairs with G. Thus, in its two-stranded form, each strand effectively contains all necessary information, redundant with its partner strand. This structure of DNA is the physical basis for inheritance: DNA replication duplicates the genetic information by splitting the strands and using each strand as a template for synthesis of a new partner strand.[47]

Genes are arranged linearly along long chains of DNA base-pair sequences. In bacteria, each cell usually contains a single circular genophore, while eukaryotic organisms (such as plants and animals) have their DNA arranged in multiple linear chromosomes. These DNA strands are often extremely long; the largest human chromosome, for example, is about 247 million base pairs in length.[48] The DNA of a chromosome is associated with structural proteins that organize, compact and control access to the DNA, forming a material called chromatin; in eukaryotes, chromatin is usually composed of nucleosomes, segments of DNA wound around cores of histone proteins.[49] The full set of hereditary material in an organism (usually the combined DNA sequences of all chromosomes) is called the genome.

While haploid organisms have only one copy of each chromosome, most animals and many plants are diploid, containing two of each chromosome and thus two copies of every gene.[37] The two alleles for a gene are located on identical loci of the two homologous chromosomes, each allele inherited from a different parent.

Many species have so-called sex chromosomes that determine the gender of each organism.[50] In humans and many other animals, the Y chromosome contains the gene that triggers the development of the specifically male characteristics. In evolution, this chromosome has lost most of its content and also most of its genes, while the X chromosome is similar to the other chromosomes and contains many genes. The X and Y chromosomes form a strongly heterogeneous pair.

When cells divide, their full genome is copied and each daughter cell inherits one copy. This process, called mitosis, is the simplest form of reproduction and is the basis for asexual reproduction. Asexual reproduction can also occur in multicellular organisms, producing offspring that inherit their genome from a single parent. Offspring that are genetically identical to their parents are called clones.

Eukaryotic organisms often use sexual reproduction to generate offspring that contain a mixture of genetic material inherited from two different parents. The process of sexual reproduction alternates between forms that contain single copies of the genome (haploid) and double copies (diploid).[37] Haploid cells fuse and combine genetic material to create a diploid cell with paired chromosomes. Diploid organisms form haploids by dividing, without replicating their DNA, to create daughter cells that randomly inherit one of each pair of chromosomes. Most animals and many plants are diploid for most of their lifespan, with the haploid form reduced to single cell gametes such as sperm or eggs.

Although they do not use the haploid/diploid method of sexual reproduction, bacteria have many methods of acquiring new genetic information. Some bacteria can undergo conjugation, transferring a small circular piece of DNA to another bacterium.[51] Bacteria can also take up raw DNA fragments found in the environment and integrate them into their genomes, a phenomenon known as transformation.[52] These processes result in horizontal gene transfer, transmitting fragments of genetic information between organisms that would be otherwise unrelated.

The diploid nature of chromosomes allows for genes on different chromosomes to assort independently or be separated from their homologous pair during sexual reproduction wherein haploid gametes are formed. In this way new combinations of genes can occur in the offspring of a mating pair. Genes on the same chromosome would theoretically never recombine. However, they do via the cellular process of chromosomal crossover. During crossover, chromosomes exchange stretches of DNA, effectively shuffling the gene alleles between the chromosomes.[53] This process of chromosomal crossover generally occurs during meiosis, a series of cell divisions that creates haploid cells.

The first cytological demonstration of crossing over was performed by Harriet Creighton and Barbara McClintock in 1931. Their research and experiments on corn provided cytological evidence for the genetic theory that linked genes on paired chromosomes do in fact exchange places from one homolog to the other.

The probability of chromosomal crossover occurring between two given points on the chromosome is related to the distance between the points. For an arbitrarily long distance, the probability of crossover is high enough that the inheritance of the genes is effectively uncorrelated.[54] For genes that are closer together, however, the lower probability of crossover means that the genes demonstrate genetic linkage; alleles for the two genes tend to be inherited together. The amounts of linkage between a series of genes can be combined to form a linear linkage map that roughly describes the arrangement of the genes along the chromosome.[55]

Genes generally express their functional effect through the production of proteins, which are complex molecules responsible for most functions in the cell. Proteins are made up of one or more polypeptide chains, each of which is composed of a sequence of amino acids, and the DNA sequence of a gene (through an RNA intermediate) is used to produce a specific amino acid sequence. This process begins with the production of an RNA molecule with a sequence matching the gene's DNA sequence, a process called transcription.

This messenger RNA molecule is then used to produce a corresponding amino acid sequence through a process called translation. Each group of three nucleotides in the sequence, called a codon, corresponds either to one of the twenty possible amino acids in a protein or an instruction to end the amino acid sequence; this correspondence is called the genetic code.[56] The flow of information is unidirectional: information is transferred from nucleotide sequences into the amino acid sequence of proteins, but it never transfers from protein back into the sequence of DNAa phenomenon Francis Crick called the central dogma of molecular biology.[57]

The specific sequence of amino acids results in a unique three-dimensional structure for that protein, and the three-dimensional structures of proteins are related to their functions.[58][59] Some are simple structural molecules, like the fibers formed by the protein collagen. Proteins can bind to other proteins and simple molecules, sometimes acting as enzymes by facilitating chemical reactions within the bound molecules (without changing the structure of the protein itself). Protein structure is dynamic; the protein hemoglobin bends into slightly different forms as it facilitates the capture, transport, and release of oxygen molecules within mammalian blood.

A single nucleotide difference within DNA can cause a change in the amino acid sequence of a protein. Because protein structures are the result of their amino acid sequences, some changes can dramatically change the properties of a protein by destabilizing the structure or changing the surface of the protein in a way that changes its interaction with other proteins and molecules. For example, sickle-cell anemia is a human genetic disease that results from a single base difference within the coding region for the -globin section of hemoglobin, causing a single amino acid change that changes hemoglobin's physical properties.[60] Sickle-cell versions of hemoglobin stick to themselves, stacking to form fibers that distort the shape of red blood cells carrying the protein. These sickle-shaped cells no longer flow smoothly through blood vessels, having a tendency to clog or degrade, causing the medical problems associated with this disease.

Some DNA sequences are transcribed into RNA but are not translated into protein productssuch RNA molecules are called non-coding RNA. In some cases, these products fold into structures which are involved in critical cell functions (e.g. ribosomal RNA and transfer RNA). RNA can also have regulatory effects through hybridization interactions with other RNA molecules (e.g. microRNA).

Although genes contain all the information an organism uses to function, the environment plays an important role in determining the ultimate phenotypes an organism displays. This is the complementary relationship often referred to as "nature and nurture". The phenotype of an organism depends on the interaction of genes and the environment. An interesting example is the coat coloration of the Siamese cat. In this case, the body temperature of the cat plays the role of the environment. The cat's genes code for dark hair, thus the hair-producing cells in the cat make cellular proteins resulting in dark hair. But these dark hair-producing proteins are sensitive to temperature (i.e. have a mutation causing temperature-sensitivity) and denature in higher-temperature environments, failing to produce dark-hair pigment in areas where the cat has a higher body temperature. In a low-temperature environment, however, the protein's structure is stable and produces dark-hair pigment normally. The protein remains functional in areas of skin that are colder such as its legs, ears, tail and face so the cat has dark-hair at its extremities.[61]

Environment plays a major role in effects of the human genetic disease phenylketonuria.[62] The mutation that causes phenylketonuria disrupts the ability of the body to break down the amino acid phenylalanine, causing a toxic build-up of an intermediate molecule that, in turn, causes severe symptoms of progressive mental retardation and seizures. However, if someone with the phenylketonuria mutation follows a strict diet that avoids this amino acid, they remain normal and healthy.

A popular method for determining how genes and environment ("nature and nurture") contribute to a phenotype involves studying identical and fraternal twins, or other siblings of multiple births.[63] Because identical siblings come from the same zygote, they are genetically the same. Fraternal twins are as genetically different from one another as normal siblings. By comparing how often a certain disorder occurs in a pair of identical twins to how often it occurs in a pair of fraternal twins, scientists can determine whether that disorder is caused by genetic or postnatal environmental factors whether it has "nature" or "nurture" causes. One famous example is the multiple birth study of the Genain quadruplets, who were identical quadruplets all diagnosed with schizophrenia.[64] However such tests cannot separate genetic factors from environmental factors affecting fetal development.

The genome of a given organism contains thousands of genes, but not all these genes need to be active at any given moment. A gene is expressed when it is being transcribed into mRNA and there exist many cellular methods of controlling the expression of genes such that proteins are produced only when needed by the cell. Transcription factors are regulatory proteins that bind to DNA, either promoting or inhibiting the transcription of a gene.[65] Within the genome of Escherichia coli bacteria, for example, there exists a series of genes necessary for the synthesis of the amino acid tryptophan. However, when tryptophan is already available to the cell, these genes for tryptophan synthesis are no longer needed. The presence of tryptophan directly affects the activity of the genestryptophan molecules bind to the tryptophan repressor (a transcription factor), changing the repressor's structure such that the repressor binds to the genes. The tryptophan repressor blocks the transcription and expression of the genes, thereby creating negative feedback regulation of the tryptophan synthesis process.[66]

Differences in gene expression are especially clear within multicellular organisms, where cells all contain the same genome but have very different structures and behaviors due to the expression of different sets of genes. All the cells in a multicellular organism derive from a single cell, differentiating into variant cell types in response to external and intercellular signals and gradually establishing different patterns of gene expression to create different behaviors. As no single gene is responsible for the development of structures within multicellular organisms, these patterns arise from the complex interactions between many cells.

Within eukaryotes, there exist structural features of chromatin that influence the transcription of genes, often in the form of modifications to DNA and chromatin that are stably inherited by daughter cells.[67] These features are called "epigenetic" because they exist "on top" of the DNA sequence and retain inheritance from one cell generation to the next. Because of epigenetic features, different cell types grown within the same medium can retain very different properties. Although epigenetic features are generally dynamic over the course of development, some, like the phenomenon of paramutation, have multigenerational inheritance and exist as rare exceptions to the general rule of DNA as the basis for inheritance.[68]

During the process of DNA replication, errors occasionally occur in the polymerization of the second strand. These errors, called mutations, can affect the phenotype of an organism, especially if they occur within the protein coding sequence of a gene. Error rates are usually very low1 error in every 10100million basesdue to the "proofreading" ability of DNA polymerases.[69][70] Processes that increase the rate of changes in DNA are called mutagenic: mutagenic chemicals promote errors in DNA replication, often by interfering with the structure of base-pairing, while UV radiation induces mutations by causing damage to the DNA structure.[71] Chemical damage to DNA occurs naturally as well and cells use DNA repair mechanisms to repair mismatches and breaks. The repair does not, however, always restore the original sequence.

In organisms that use chromosomal crossover to exchange DNA and recombine genes, errors in alignment during meiosis can also cause mutations.[72] Errors in crossover are especially likely when similar sequences cause partner chromosomes to adopt a mistaken alignment; this makes some regions in genomes more prone to mutating in this way. These errors create large structural changes in DNA sequence duplications, inversions, deletions of entire regions or the accidental exchange of whole parts of sequences between different chromosomes (chromosomal translocation).

Mutations alter an organism's genotype and occasionally this causes different phenotypes to appear. Most mutations have little effect on an organism's phenotype, health, or reproductive fitness.[73] Mutations that do have an effect are usually deleterious, but occasionally some can be beneficial.[74] Studies in the fly Drosophila melanogaster suggest that if a mutation changes a protein produced by a gene, about 70 percent of these mutations will be harmful with the remainder being either neutral or weakly beneficial.[75]

Population genetics studies the distribution of genetic differences within populations and how these distributions change over time.[76] Changes in the frequency of an allele in a population are mainly influenced by natural selection, where a given allele provides a selective or reproductive advantage to the organism,[77] as well as other factors such as mutation, genetic drift, genetic draft,[78]artificial selection and migration.[79]

Over many generations, the genomes of organisms can change significantly, resulting in evolution. In the process called adaptation, selection for beneficial mutations can cause a species to evolve into forms better able to survive in their environment.[80] New species are formed through the process of speciation, often caused by geographical separations that prevent populations from exchanging genes with each other.[81] The application of genetic principles to the study of population biology and evolution is known as the "modern synthesis".

By comparing the homology between different species' genomes, it is possible to calculate the evolutionary distance between them and when they may have diverged. Genetic comparisons are generally considered a more accurate method of characterizing the relatedness between species than the comparison of phenotypic characteristics. The evolutionary distances between species can be used to form evolutionary trees; these trees represent the common descent and divergence of species over time, although they do not show the transfer of genetic material between unrelated species (known as horizontal gene transfer and most common in bacteria).[82]

Although geneticists originally studied inheritance in a wide range of organisms, researchers began to specialize in studying the genetics of a particular subset of organisms. The fact that significant research already existed for a given organism would encourage new researchers to choose it for further study, and so eventually a few model organisms became the basis for most genetics research.[83] Common research topics in model organism genetics include the study of gene regulation and the involvement of genes in development and cancer.

Organisms were chosen, in part, for convenienceshort generation times and easy genetic manipulation made some organisms popular genetics research tools. Widely used model organisms include the gut bacterium Escherichia coli, the plant Arabidopsis thaliana, baker's yeast (Saccharomyces cerevisiae), the nematode Caenorhabditis elegans, the common fruit fly (Drosophila melanogaster), and the common house mouse (Mus musculus).

Medical genetics seeks to understand how genetic variation relates to human health and disease.[84] When searching for an unknown gene that may be involved in a disease, researchers commonly use genetic linkage and genetic pedigree charts to find the location on the genome associated with the disease. At the population level, researchers take advantage of Mendelian randomization to look for locations in the genome that are associated with diseases, a method especially useful for multigenic traits not clearly defined by a single gene.[85] Once a candidate gene is found, further research is often done on the corresponding gene the orthologous gene in model organisms. In addition to studying genetic diseases, the increased availability of genotyping methods has led to the field of pharmacogenetics: the study of how genotype can affect drug responses.[86]

Individuals differ in their inherited tendency to develop cancer,[87] and cancer is a genetic disease.[88] The process of cancer development in the body is a combination of events. Mutations occasionally occur within cells in the body as they divide. Although these mutations will not be inherited by any offspring, they can affect the behavior of cells, sometimes causing them to grow and divide more frequently. There are biological mechanisms that attempt to stop this process; signals are given to inappropriately dividing cells that should trigger cell death, but sometimes additional mutations occur that cause cells to ignore these messages. An internal process of natural selection occurs within the body and eventually mutations accumulate within cells to promote their own growth, creating a cancerous tumor that grows and invades various tissues of the body.

Normally, a cell divides only in response to signals called growth factors and stops growing once in contact with surrounding cells and in response to growth-inhibitory signals. It usually then divides a limited number of times and dies, staying within the epithelium where it is unable to migrate to other organs. To become a cancer cell, a cell has to accumulate mutations in a number of genes (37) that allow it to bypass this regulation: it no longer needs growth factors to divide, it continues growing when making contact to neighbor cells, and ignores inhibitory signals, it will keep growing indefinitely and is immortal, it will escape from the epithelium and ultimately may be able to escape from the primary tumor, cross the endothelium of a blood vessel, be transported by the bloodstream and will colonize a new organ, forming deadly metastasis. Although there are some genetic predispositions in a small fraction of cancers, the major fraction is due to a set of new genetic mutations that originally appear and accumulate in one or a small number of cells that will divide to form the tumor and are not transmitted to the progeny (somatic mutations). The most frequent mutations are a loss of function of p53 protein, a tumor suppressor, or in the p53 pathway, and gain of function mutations in the ras proteins, or in other oncogenes.

DNA can be manipulated in the laboratory. Restriction enzymes are commonly used enzymes that cut DNA at specific sequences, producing predictable fragments of DNA.[89] DNA fragments can be visualized through use of gel electrophoresis, which separates fragments according to their length.

The use of ligation enzymes allows DNA fragments to be connected. By binding ("ligating") fragments of DNA together from different sources, researchers can create recombinant DNA, the DNA often associated with genetically modified organisms. Recombinant DNA is commonly used in the context of plasmids: short circular DNA molecules with a few genes on them. In the process known as molecular cloning, researchers can amplify the DNA fragments by inserting plasmids into bacteria and then culturing them on plates of agar (to isolate clones of bacteria cells). ("Cloning" can also refer to the various means of creating cloned ("clonal") organisms.)

DNA can also be amplified using a procedure called the polymerase chain reaction (PCR).[90] By using specific short sequences of DNA, PCR can isolate and exponentially amplify a targeted region of DNA. Because it can amplify from extremely small amounts of DNA, PCR is also often used to detect the presence of specific DNA sequences.

DNA sequencing, one of the most fundamental technologies developed to study genetics, allows researchers to determine the sequence of nucleotides in DNA fragments. The technique of chain-termination sequencing, developed in 1977 by a team led by Frederick Sanger, is still routinely used to sequence DNA fragments.[91] Using this technology, researchers have been able to study the molecular sequences associated with many human diseases.

As sequencing has become less expensive, researchers have sequenced the genomes of many organisms, using a process called genome assembly, which utilizes computational tools to stitch together sequences from many different fragments.[92] These technologies were used to sequence the human genome in the Human Genome Project completed in 2003.[34] New high-throughput sequencing technologies are dramatically lowering the cost of DNA sequencing, with many researchers hoping to bring the cost of resequencing a human genome down to a thousand dollars.[93]

Next generation sequencing (or high-throughput sequencing) came about due to the ever-increasing demand for low-cost sequencing. These sequencing technologies allow the production of potentially millions of sequences concurrently.[94][95] The large amount of sequence data available has created the field of genomics, research that uses computational tools to search for and analyze patterns in the full genomes of organisms. Genomics can also be considered a subfield of bioinformatics, which uses computational approaches to analyze large sets of biological data. A common problem to these fields of research is how to manage and share data that deals with human subject and personally identifiable information. See also genomics data sharing.

On 19 March 2015, a leading group of biologists urged a worldwide ban on clinical use of methods, particularly the use of CRISPR and zinc finger, to edit the human genome in a way that can be inherited.[96][97][98][99] In April 2015, Chinese researchers reported results of basic research to edit the DNA of non-viable human embryos using CRISPR.[100][101]

See the original post here:
Genetics - Wikipedia

Read More...

Page 37«..1020..36373839..»


2025 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick