The Prometheus League
Breaking News and Updates
- Abolition Of Work
- Ai
- Alt-right
- Alternative Medicine
- Antifa
- Artificial General Intelligence
- Artificial Intelligence
- Artificial Super Intelligence
- Ascension
- Astronomy
- Atheism
- Atheist
- Atlas Shrugged
- Automation
- Ayn Rand
- Bahamas
- Bankruptcy
- Basic Income Guarantee
- Big Tech
- Bitcoin
- Black Lives Matter
- Blackjack
- Boca Chica Texas
- Brexit
- Caribbean
- Casino
- Casino Affiliate
- Cbd Oil
- Censorship
- Cf
- Chess Engines
- Childfree
- Cloning
- Cloud Computing
- Conscious Evolution
- Corona Virus
- Cosmic Heaven
- Covid-19
- Cryonics
- Cryptocurrency
- Cyberpunk
- Darwinism
- Democrat
- Designer Babies
- DNA
- Donald Trump
- Eczema
- Elon Musk
- Entheogens
- Ethical Egoism
- Eugenic Concepts
- Eugenics
- Euthanasia
- Evolution
- Extropian
- Extropianism
- Extropy
- Fake News
- Federalism
- Federalist
- Fifth Amendment
- Fifth Amendment
- Financial Independence
- First Amendment
- Fiscal Freedom
- Food Supplements
- Fourth Amendment
- Fourth Amendment
- Free Speech
- Freedom
- Freedom of Speech
- Futurism
- Futurist
- Gambling
- Gene Medicine
- Genetic Engineering
- Genome
- Germ Warfare
- Golden Rule
- Government Oppression
- Hedonism
- High Seas
- History
- Hubble Telescope
- Human Genetic Engineering
- Human Genetics
- Human Immortality
- Human Longevity
- Illuminati
- Immortality
- Immortality Medicine
- Intentional Communities
- Jacinda Ardern
- Jitsi
- Jordan Peterson
- Las Vegas
- Liberal
- Libertarian
- Libertarianism
- Liberty
- Life Extension
- Macau
- Marie Byrd Land
- Mars
- Mars Colonization
- Mars Colony
- Memetics
- Micronations
- Mind Uploading
- Minerva Reefs
- Modern Satanism
- Moon Colonization
- Nanotech
- National Vanguard
- NATO
- Neo-eugenics
- Neurohacking
- Neurotechnology
- New Utopia
- New Zealand
- Nihilism
- Nootropics
- NSA
- Oceania
- Offshore
- Olympics
- Online Casino
- Online Gambling
- Pantheism
- Personal Empowerment
- Poker
- Political Correctness
- Politically Incorrect
- Polygamy
- Populism
- Post Human
- Post Humanism
- Posthuman
- Posthumanism
- Private Islands
- Progress
- Proud Boys
- Psoriasis
- Psychedelics
- Putin
- Quantum Computing
- Quantum Physics
- Rationalism
- Republican
- Resource Based Economy
- Robotics
- Rockall
- Ron Paul
- Roulette
- Russia
- Sealand
- Seasteading
- Second Amendment
- Second Amendment
- Seychelles
- Singularitarianism
- Singularity
- Socio-economic Collapse
- Space Exploration
- Space Station
- Space Travel
- Spacex
- Sports Betting
- Sportsbook
- Superintelligence
- Survivalism
- Talmud
- Technology
- Teilhard De Charden
- Terraforming Mars
- The Singularity
- Tms
- Tor Browser
- Trance
- Transhuman
- Transhuman News
- Transhumanism
- Transhumanist
- Transtopian
- Transtopianism
- Ukraine
- Uncategorized
- Vaping
- Victimless Crimes
- Virtual Reality
- Wage Slavery
- War On Drugs
- Waveland
- Ww3
- Yahoo
- Zeitgeist Movement
-
Prometheism
-
Forbidden Fruit
-
The Evolutionary Perspective
Category Archives: Transhuman News
COVID isn’t humanity’s first pandemic rodeo. How did we make it through the Spanish flu? – Cosmos
Posted: February 15, 2022 at 5:11 am
What caused the 1918 Spanish flu to cease?
I get it Miranda. Sitting, as we are, in throes of a pandemic, our interest in outbreaks of bygone eras is naturally piqued. Everythings crazy; everything changes theres no pandemic handbook, right?
Or is there?
The swings and roundabouts of the COVID pandemic feel new and strange, but its not the first time humanity has stared down a deadly viral disease. Can past pandemics offer a blueprint as to how our current mess will play out?
The US Centre for Disease Control (CDC) certainly believes theres merit to this idea.
In 2005, the CDC recreated the Spanish flu virus safely contained within tightly controlled laboratories, of course. They hoped to sequence its genome and study its secrets in order to be better prepared for future pandemics.
The Spanish flu of 1918-19 has long stood out to virologists and epidemiologists as a stark warning that devastating viruses can spring seemingly from nowhere and wreak havoc across the globe. The Spanish flu killed as many as 50 million people worldwide over a period of approximately 18 months, with an unusually high death rate among otherwise healthy young people (1535-year-olds). Around 500 million people were infected almost a third of the global population at the time.
Its easy to see the merit in trying to understand the trajectory of a virus that hit hard a century ago but no longer poses any threat. perhaps well see our own path out of the dark COVID woods. What was it that made the 1918 virus so deadly? Where did it come from? And where did it go?
Todays pandemic is mapped and documented in minute detail. We track its waves across the globe in real time, and our understanding of its internal machinery is developing rapidly. But in 1918, the study of infectious pathogens was a new field. In the wake of Spanish flu, generations of researchers and public health experts had little more than estimated death tolls and social histories with which to build a picture of the pandemic.
How could scientists understand what happened when the biggest puzzle piece, the pathogen itself, was missing?
That conundrum was initially tackled by Swedish researcher Johan Hultin in 1951. Hed heard of a mass burial site in an Alaskan village called Brevig Mission. There, the interred bodies of at least 72 Spanish flu victims lay untouched in the permafrost. Hultin with permission from village elders set to work excavating frozen corpses, hoping to find tissue harbouring traces of the 1918 virus.
Hutins efforts failed, although not for want of trying on his flight from Alaska to research facilities at the University of Iowa he tried to keep lung samples frozen using carbon dioxide from a fire extinguisher. But the technology of the time didnt allow him to properly prepare and analyse the samples.
In 1997, nearly a half century after his first attempt, Hultin returned to Brevig Mission to give it another shot. Armed with his wifes garden shears and a handful of local assistants, Hultin dug the frozen burial grounds once more.
Tech advancements had allowed other researchers to partially sequence the viral genome of a lung sample from a US serviceman who died in 1918. Hultin reset his sights on obtaining the best possible tissue samples for these scientists to work with and this time his efforts paid off.
He unearthed and preserved the perfectly frozen lungs of an Inuit woman whod died of Spanish flu complications. Just days after the lungs reached the researchers, Hultin had the news: positive genetic material had been obtained from the frozen samples.
By 1999, and largely thanks to Hultins samples, the entire code of the 1918 flu was sequenced, laying the foundations for the ultimate goal reconstructing a live version of the lost virus.
The effort began in earnest in 2005.
Using a pioneering technique called reverse genetics, CDC researchers took small, circular strands of DNA called plasmids for each of the 1918 viruss eight gene segments and inserted them into human kidney cells. The plasmids then instructed the cells to reconstruct the RNA of the complete 1918 virus. This zombie-like reanimation of the Spanish flu was then tested on mice, allowing researchers to document for the first time the mechanisms of its virulence and spread.
The results were terrifying. Four days after infection, the amount of Spanish flu virus found in the lung tissue of infected mice was 39,000 times higher than that produced by similar strains in the flu family.
By watching the virus in action, researchers were finally able to link its genetic structure with the patterns of disease it produced. Experiments showed that it was not any single component of the 1918 virus but instead, as researchers wrote at the time, the constellation of all eight genes together that made the Spanish flu an exceptionally virulent virus.
The CDC team also tested theories on the viruss origins. After much experimentation, it was ultimately a chicken egg that held the answer. When 10-day-old fertilised eggs were inoculated with the virus, the results were lethal for the nascent chicken embryos. Paired with gene sequencing evidence suggesting a close link between the Spanish flu virus and other avian influenza viruses, this insight was enough for researchers to declare that the 1918 virus first arose in birds before making the leap to humans.
So that covers off the first two big questions about the Spanish flu where it came from, and why it was so deadly.
Get an update of science stories delivered straight to your inbox.
But what about the biggest how did the pandemic end?
This first requires a little clarification.
The Spanish flu pandemic certainly ended, but the virus that kicked it off did not.
Instead, it gradually grew milder, morphing from its lethal beginnings into a much more placid sniffle.
This is a common natural progression for viruses, partly because the best evolutionary pathway optimises spread but leaves virulence primarily to random chance. This means that while theres active selection for increased transmissibility, theres a good chance that lethality will fade. But there is no concrete rule that dictates diminishing virulence, and there can certainly be surprising spikes along the way.
This was true of the Spanish flu, which ebbed and flowed in waves much like COVIDs peaks and troughs. While the general trajectory was towards becoming milder, the path wasnt linear some waves were significantly deadlier than their predecessors, just as in the current crisis, the Delta and Omicron variants have posed challenges that the first viral waves did not.
But if the Spanish flus overarching pattern is followed by SARS-CoV-2, as many experts believe it will be, we can expect that while well never eradicate the virus, it wont always be a major public health threat.
The scarily virulent 1918 form of Spanish flu that researchers reanimated is long gone, but its descendants still circulate as part of our seasonal flus. Some genetic aspects of the virus have been implicated in the lesser pandemics of 1957 and 1968; its likely that people who lived through the Spanish flu years had a degree of protection from these genetic cousins.
Here again is a lesson for current times: immunity is key. Each wave of the Spanish flu added layers of immunity to the surviving global population, until gradually the threat abated. With COVID, we know that whilst our immunity fades over a period of months following vaccination or infection, we retain an increasing ability to ward off severe disease with each exposure.
But as we wait for global immunity to build, our key public health measures remain vital in protecting us from the worst effects of the virus.
The importance of these measures was laid bare in the 1918-19 pandemic. As the flu raged a century ago, people were asked to wear masks and adopt social distancing measures, just as we are today. And, just as today, a number of anti-mask advocates opposed these impositions. But experts agree that these measures significantly dampened the death toll while immunity grew, and that they remain vital for us in the current pandemic as we wait for its severity to diminish.
When CDC researchers resurrected and characterised the Spanish Flu, they felt theyd taken significant steps towards safeguarding humanity against future outbreaks. They described how the 1918 virus was special a uniquely deadly product of nature, evolution and the intermingling of people and animals. Sound familiar?
At the time, they believed their work would serve as a portent of natures ability to produce future pandemics and help us to begin building our defences and public health capabilities.
But were we paying enough attention?
In a summary of their work on the virus, CDC researchers noted that despite advances in medicine and public health, a severe pandemic could still be devastating to populations globally.
Amongst other things, they identified a global deficiency in surveillance capacity, infrastructure, and pandemic planning, and noted insufficient critical and clinical care capacity, especially in low-income countries. They bemoaned the fact that milestones established in 2005 in revised International Health Regulations (IHR) for countries to improve their response capacity for public health emergencies had only been met by a third of countries by 2016.
In a sense, the blueprint for our current pandemic was laid out for us in CDCs study of the Spanish flu. Though the two viruses are genetically dissimilar, the lessons regarding their management were made clear years before COVID emerged.
We will inevitably make it through this pandemic, though the length of the road ahead remains unclear. But when the next outbreak occurs, will we be any better prepared than we were this time?
This much is clear: if we fail to heed the lessons of pandemics past, COVID-19 will fade from our collective memory the way that the Spanish flu did over the course of a mere century.
Why is the sky blue? What actually is carbon capture and storage? Why does my vacuum cleaner make that noise? How does bitcoin work? And could Yodareallyforce push Palpatine?
Theres no such thing as a stupid science question, but sometimes the answers can be tricky to find.
This summer weve partnered with ACM for the Summer of science: Ask us anything!Sendus your curliest chemistry conundrum, perplexing physics problem or any science question at alland well get our journalists onto the case.
See the original post here:
COVID isn't humanity's first pandemic rodeo. How did we make it through the Spanish flu? - Cosmos
Posted in Human Genetics
Comments Off on COVID isn’t humanity’s first pandemic rodeo. How did we make it through the Spanish flu? – Cosmos
Student publications prove to be purposeful and advanced – University of Cape Town News
Posted: at 5:11 am
An initiative of the University of Cape Towns (UCT) Institute of Infectious Disease and Molecular Medicines (IDM) Transformation Committee, the Postgraduate Student Publication Competition recognises student publications. It was designed to foster a culture of research publication among postgraduate students based in IDM member groups, while lauding students who have made the intellectual and practical contributions that earn them a place as first authors in published journal articles.
In her welcome at the online celebration, IDM director, Professor Valerie Mizrahi, said that getting ones work out there is a critical part of being a scientist.
The selection panel consisted of world-famous Emeritus Professors Siamon Gordon and Wieland Gevers. Emeritus Professor Gordon is an immunologist and clinician from the University of Oxford and a UCT alumnus.
Professor Mizrahi said of Gordon: [He] has been the chair of the International Scientific Advisory Committee for the Institute, I believe, since its inception certainly for as long as Ive been the director of the institute.
Emeritus Professor Gevers, the second judge, conceptualised the IDM over a decade ago. He also created the facultys Department of Medical Biochemistry. [He] has held huge positions in building the science-research ecosystem of South Africa through the Academy of Science amongst many other contributions, said Mizrahi.
The 14 submissions received from students illustrated the quality of research underway at the IDM, highlighting the depth of talent of the institutes next-generation researchers.
Theres a range of publications in a range of different journals. They were of a very high standard ... Im proud to be part of the IDM family, said Gordon.
Gevers added: This gives me an opportunity to see the diverse, excellent, purposeful, advanced work that is going on [at the IDM].
2021 IDM Postgraduate Student Publication competition winners
Arash Iranzadeh, Division of Computational Biology, Department of Integrative Biomedical Sciences
The Original Research category win was shared by Arash Iranzadeh and Ryan Dinkele. Iranzadeh is based at the HIV Diversity Group.
His paper, Detection of a SARS-CoV-2 variant of concern in South Africa, published in Nature reported on and characterised a newly identified SARS-CoV-2 lineage after the first wave of the epidemic in South Africa. This Beta variant or B.1.351 has eight mutations in the spike protein which likely increase virus transmissibility.
The finding highlighted the importance of coordinated molecular surveillance systems in all parts of the world in enabling the early detection and characterisation of new lineages, as well as in informing the global response to the COVID-19 pandemic, said Iranzadeh.
It is a great honour and privilege for me to attain the prize from UCT. Im grateful to everyone who contributed to this work, he said.
Ryan Dinkele, Division of Medical Microbiology, Department of Pathology
The second recipient of the first prize for Original Research category was Ryan Dinkele, based at the Molecular Mycobacteriology Research Unit (MMRU) and the Centre of Excellence for Biomedical TB Research (CBTBR). His paper, Capture and visualization of live Mycobacterium tuberculosis bacilli from tuberculosis patient bioaerosols, was published in PLOS Pathogens.
The aim of the research was to develop a new system to detect M. tb, the bacteria that causes tuberculosis (TB), in aerosols produced by TB patients during normal breathing and coughing. This microscopy-based system provided a rapid and sensitive readout for aerosolized M. tb within 24 hours in about 90% of TB patients.
Consequently, we have a unique approach for studying TB transmission within a South African context, said Dinkele.
Referring to the accolade, he said, The recognition of this work by such distinguished scientists has provided the inspiration required to persevere in the challenging field of TB transmission.
Cheleka Mpande, Division of Immunology, Department of Pathology
Cheleka Mpande received second prize for Original Research with her paper: Immune profiling of Mycobacterium tuberculosis-specific T cells in recent and remote infection, which was published in EBioMedicine. Mpande is part of the South African Tuberculosis Vaccine Initiative.
The risk of developing TB is not uniform among individuals infected with M. tb. The risk is said to be very high in people who have recently been infected with M. tb. Current TB diagnostic tools are good at identifying people with TB disease but cannot identify people who have recently become infected with M. tb.
This formed the foundation for the studys aim: to find immunological markers that are only present during recent M. tb infection that can be used to design immune-based diagnostics. These can then be used to identify recent M. tb infection and by extension potentially infer risk of TB progression.
Referencing the research, Mpande said: This is a culmination of more than six years of work including results from some of my first successful experiments as an MSc student. It is an amazing achievement for everyone involved in the project.
Arinao Ndadza, Division of Human Genetics, Department of Pathology
Based at the Pharmacogenomics and Drug Metabolism Research Group, Arinao Ndadza was awarded third prize under the Original Research, jointly with Terry Kipkorir. Her paper, Profiling of warfarin pharmacokinetics-associated genetic variants: Black Africans portray unique genetic markers important for an African specific warfarin pharmacogenetics-dosing algorithm, was published in the Journal of Thrombosis and Haemostasis.
The papers aim was to profile and identify genetic variants that contribute to warfarin dose variability among Southern African populations. The research focused on genes with a pharmacokinetic effect meaning those involved in the disposition of the drug in terms of its absorption, transport, metabolism and excretion. This work contributes to the ongoing efforts of developing an African-specific pharmacogenetics-based warfarin dosing algorithm that can assist clinically in predicting a precise warfarin starting dose that is suitable for individuals with African ancestry.
For Ndadza this recognition validates that the research shes a part of is impactful and has the potential of being translated in the healthcare system.
This acknowledgment further proves that there is space for me to occupy and further contribute [to] pharmacogenomics and genomics research, she said.
Terry Kipkorir, Division of Medical Microbiology, Department of Pathology
Terry Kipkorir, who was also awarded third prize, had his paper, De novo cobalamin biosynthesis, transport and assimilation and cobalamin-mediated regulation of methionine biosynthesis in Mycobacterium smegmatis, published in the Journal of Bacteriology.
The papers objective is to describe the production, acquisition and utilisation of cobalamin a near-identical form of vitamin B12, in Mycobacterium smegmatis, a soil-dwelling model for M. tb. It also aimed to investigate how cobalamin-sensing via an RNA switch controls gene expression and bacterial growth.
Our findings provide key insights into the roles of cobalamin in mycobacterial physiology and pathogenicity, said Kipkorir whose study is based at the MMRU. He added that the prize affirmed the scientific rigour of the groups work and the impact of their findings in TB research.
Liam Devenish, Division of Chemical, Systems, and Synthetic Biology; Department of Integrative Biomedical Sciences
Liam Devenish, based at the Gene Expression and Biophysics Group, won first prize for his review article: Immune Regulation in Time and Space: The Role of Local- and Long-Range Genomic Interactions in Regulating Immune Responses. It was published in Frontiers in Immunology.
The review explores and frames new discoveries surrounding the phenomenon of immune priming. Immune priming is a central element of the immune system in almost all multi-cellular living organisms. It remembers previous immune challenges and becomes able to respond more appropriately and robustly in future. The paper explores this in the context of the burgeoning field of chromatin topology.
Immune priming is about discovering why we're so good at responding quickly to a staggering array of insults from the environment and from within our own bodies. Its about our immune systems learning and remembering how to be better equipped with each assault, said Devenish.
This memory spills over to various systems including resistance to developing cancers, responses to vaccines, and resilience to new organisms. The understanding that the physical shape and organisation of the genome within the nucleus informs gene regulation is one which is growing and has recently been well-applied to the field of immunology.
These genomic interactions happen at remarkable speed and with incredible accuracy to create a synchronised, powerful defence to almost any challenge we encounter, said Devenish.
He added: Sharing my thoughts and understanding has been challenging and exceptionally rewarding.
Sherazaan Ismail, Division of Medical Virology, Department of Pathology
Sherazaan Ismail straddles two groups at the IDM: the HIV/TB Immunology research group and the HIV Diversity Group. Her paper, Addressing an HIV cure in LMIC, published in Retrovirology, earned her the second prize under the Review category.
People living with HIV require lifelong antiretroviral therapy (ART) due to the persistence of HIV in a latent reservoir in infected cells. To achieve a global HIV cure it is important to understand the reservoir in different contexts, especially because viral subtypes, disease progression, and the timing of ART differs around the world.
Ismails paper discussed the latest findings in HIV cure research as well as the considerations for implementing a cure in low- and middle-income countries.
This win reaffirms that, as PhD students, we are experts in our field, and we shouldnt let impostor syndrome get in the way of fostering collaborations and making our important contributions known, she said.
Go here to read the rest:
Student publications prove to be purposeful and advanced - University of Cape Town News
Posted in Human Genetics
Comments Off on Student publications prove to be purposeful and advanced – University of Cape Town News
Lumakras (sotorasib) shows encouraging and clinically meaningful anticancer activity in patients with KRAS G12C-mutated advanced pancreatic cancer in…
Posted: at 5:11 am
DetailsCategory: Small MoleculesPublished on Tuesday, 15 February 2022 11:02Hits: 34
Centrally Confirmed Objective Response Rate of 21% and Disease Control Rate of 84%
Largest Dataset and Only Global Clinical Trial to Date to Evaluate the Efficacy and Safety of a KRAS G12C Inhibitor in Advanced Pancreatic Cancer
THOUSAND OAKS, CA, USA I February 14, 2022 I Amgen (NASDAQ: AMGN) today announced the presentation of efficacy and safety data from the CodeBreaK 100 Phase 1/2 trial in patients with KRAS G12C-mutated advanced pancreatic cancer who received LUMAKRAS (sotorasib)*. The data will be presented at the monthly American Society of Clinical Oncology (ASCO) Plenary Series on Feb. 15, 2022. Data show encouraging and clinically meaningful anticancer activity and a positive benefit:risk profile.
"Based on these exciting data, we are expanding CodeBreaK 100 to enroll more patients with pancreatic and other tumor types to better understand the efficacy and safety of LUMAKRAS in tumors outside of non-small cell lung and colorectal cancers," said David M. Reese, M.D., executive vice president of Research and Development atAmgen. "CodeBreaK is the largest and broadest global clinical trial program to date with one of the most robust, centrally reviewed datasets. As we learn more from the extensive data that we collect, we'll continue to invest in the program by expanding cohorts and exploring new combinations so that we can help as many patients as possible."
LUMAKRAS demonstrated a centrally confirmed objective response rate (ORR) of 21% and disease control rate (DCR) of 84% across 38 heavily pre-treated advanced pancreatic cancer patients. Nearly 80% of patients received LUMAKRAS as a third-line or later therapy. Eight of the 38 patients achieved a confirmed partial response (PR) performed by a blinded independent central review (BICR). Two of the eight patients with PR have ongoing responses. Median duration of response was 5.7 months with a median follow-up of 16.8 months as of the data cutoff date of Nov. 1, 2021. The results also show a median progression free survival (PFS) of 4 months and a median overall survival (OS) of almost 7 months. No new safety signals were identified with this study of patients with advanced pancreatic cancers. Treatment-related adverse events (TRAEs) of any grade occurred in 16 (42%) patients with diarrhea (5%) and fatigue (5%) as the most common grade 3 TRAEs. No TRAEs were fatal or resulted in treatment discontinuation.
"After decades of research, current treatments for patients with pancreatic cancer provide limited survival benefit, illustrating the critical need for novel, safe and effective treatment options," said John Strickler, M.D. associate professor of medicine, Duke University School of Medicine and gastrointestinal oncologist. "In the largest dataset evaluating the efficacy and safety of a KRASG12C inhibitor in heavily pretreated advanced pancreatic cancer, sotorasib achieved a centrally confirmed response rate of 21% and a disease control rate of 84%. This is clinically meaningful for patients because there is not an established standard therapy for these patients once they get to a third-line of treatment."
Cancer of the pancreas is a highly lethal malignancy. Itis the fourth leading cause of cancer-related deaths in both men and women in the U.S. with a 5-year survival rate of approximately 10%.1 There is a high unmet need for patients with advanced pancreatic cancer that has progressed after first-line treatment, where FDA-approved second-line therapy has provided survival of about six months and a response rate of 16%.2 After progression on first- and second-line chemotherapy, there are no therapies with a demonstrated survival benefit.2,3 Despite advances in treatment, few improvements have been made to improve diagnosis and treatment of pancreatic cancer.
It is estimated that approximately 90% of patients with pancreatic cancer harbor a KRAS mutation with KRAS G12C accounting for approximately 1-2% of these mutations.4-5
ASCO Plenary Series SessionASCO will host a livestream event on Tuesday, Feb. 15 at 3 p.m. ET featuring presentation of the abstract "First data for sotorasib in patients with pancreatic cancer withKRASp.G12C mutation: A phase I/II study evaluating efficacy and safety"by Dr. John Strickler from Duke University. To participate in the free and open session, participants may register and login at https://www.asco.org/meetings-education/monthly-plenary-series/program.
*LUMAKRAS is marketed as LUMYKRAS (sotorasib) in the European Union and the United Kingdom.
About LUMAKRAS/LUMYKRAS(sotorasib)Amgentook on one of the toughest challenges of the last 40 years in cancer research by developing LUMAKRAS/LUMYKRAS, a KRASG12Cinhibitor.6 LUMAKRAS/LUMYKRAS has demonstrated a positive benefit-risk profile with rapid, deep and durable anticancer activity in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC) harboring theKRASG12Cmutation with a once daily oral formulation.7
Amgenis progressing the largest and broadest global KRASG12Cinhibitor development program with unparalleled speed and exploring more than 10 sotorasib combination regimens, including triplets, with clinical trial sites spanning five continents. To date, over 4,000 patients around the world have received LUMAKRAS/LUMYKRAS through the clinical development program and commercial use.
In May 2021, LUMAKRAS was the first KRASG12C inhibitor to receive regulatory approval anywhere in the world with its approval in the U.S., under accelerated approval. LUMAKRAS/LUMYKRAS is also approved in the United Arab Emirates, the European Union, Japan and Switzerland, and in Canada and Great Britain under the FDA's Project Orbis. Through Project Orbis, Amgen also has Marketing Authorization Applications (MAAs) for sotorasib in review in Australia, Brazil, Singapore and Israel. Additionally,Amgenhas submitted MAAs in South Korea, Turkey, Taiwan, Colombia, Thailand, Mexico, Hong Kong, Saudi Arabia, Argentina, Kuwait and Qatar.
LUMAKRAS/LUMYKRAS is also being studied in multiple other solid tumors.8
About CodeBreaKThe CodeBreaK clinical development program forAmgen's drug sotorasib is designed to treat patients with an advanced solid tumor with theKRASG12Cmutation and address the longstanding unmet medical need for these cancers.
CodeBreaK 100, the Phase 1 and 2, first-in-human, open-label multicenter study, enrolled patients withKRASG12C-mutant solid tumors.7.8Eligible patients must have received a prior line of systemic anticancer therapy, consistent with their tumor type and stage of disease. The primary endpoint for the Phase 2 study was centrally assessed objective response rate. The Phase 2 trial in NSCLC enrolled 126 patients, 124 of whom had centrally evaluable lesions by RECIST at baseline.7 The Phase 2 trial in colorectal cancer (CRC) is fully enrolled and results have been published.9
CodeBreaK 200, the global Phase 3 randomized active-controlled study comparing sotorasib to docetaxel in KRASG12C-mutated NSCLC completed enrollment of 345 patients. Eligible patients had previously treated (previous treatment with both platinum doublet chemotherapy and a checkpoint inhibitor), locally-advanced and unresectable or metastatic KRASG12C-mutated NSCLC.The primary endpoint is progression-free survival and key secondary endpoints include overall survival, objective response rate, and patient-reported outcomes.
Amgenalso has several Phase 1b studies investigating sotorasib monotherapy and sotorasib combination therapy across various advanced solid tumors (CodeBreaK 101) open for enrollment. A Phase 2 randomized study will evaluate sotorasib in patients with stage IVKRASG12C-mutated NSCLC in need of first-line treatment (CodeBreaK 201).For information, please visitwww.hcp.codebreaktrials.com.
LUMAKRAS(sotorasib)U.S.IndicationLUMAKRASis indicated for the treatment of adult patients withKRASG12C-mutated locally advanced or metastatic non-small cell lung cancer (NSCLC), as determined by an FDA-approved test, who have received at least one prior systemic therapy.
This indication is approved under accelerated approval based on overall response rate (ORR) and duration of response (DOR). Continued approval for this indication may be contingent upon verification and description of clinical benefit in a confirmatory trial(s).
Please see LUMAKRASfullPrescribing Information.
AboutAmgenOncologyAtAmgenOncology, our mission to serve patients drives all that we do. That's why we're relentlessly focused on accelerating the delivery of medicines that have the potential to empower all angles of care and transform lives of people with cancer.
For the last four decades, we have been dedicated to discovering the firsts that matter in oncology and to finding ways to reduce the burden of cancer. Building on our heritage,Amgencontinues to advance the largest pipeline in the Company's history, moving with great speed to advance those innovations for the patientswhoneed them.
AtAmgen, we're advancing oncology at the speed of life.
For more information, follow us onwww.twitter.com/amgenoncology.
AboutAmgenAmgenis committed to unlocking the potential of biology for patients suffering from serious illnesses by discovering, developing, manufacturing and delivering innovative human therapeutics. This approach begins by using tools like advanced human genetics to unravel the complexities of disease and understand the fundamentals of human biology.
Amgenfocuses on areas of high unmet medical need and leverages its expertise to strive for solutions that improve health outcomes and dramatically improve people's lives. A biotechnology pioneer since 1980,Amgenhas grown to be one of the world's leading independent biotechnology companies, has reached millions of patients around the world and is developing a pipeline of medicines with breakaway potential.
Amgenis one of the 30 companies that comprise the Dow Jones Industrial Average and is also part of the Nasdaq-100 index. In 2021,Amgenwas named one of the 25 World's Best Workplaces by Fortune andGreat Placeto Work and one of the 100 most sustainable companies in the world byBarron's.
For more information, visitwww.amgen.comand follow us onwww.twitter.com/amgen.
1Siegel RL, et al. CA Cancer J Clin. 2022;72:7-33.2Tempero MA, et al. J Natl Compr Canc Netw. 2021;19:439-457.3Wang-Gillam A, et al. Eur J Cancer. 2019;108:78-87.4Nassar AH, et al. N Engl J Med. 2021;384:185-1875Cox AD, et al. Nat Rev Drug Discov. 2014;13:828-851.6Canon J, et al.Nature. 2019;575:217223.7Skoulidis F, et al.N Engl J Med.2021;384:2371-2381.8Hong DS, et al.N Engl J Med. 2020;383:1207-1217.9Fakih MG, et al, Lancet Oncol. 2022.
SOURCE: Amgen
Originally posted here:
Lumakras (sotorasib) shows encouraging and clinically meaningful anticancer activity in patients with KRAS G12C-mutated advanced pancreatic cancer in...
Posted in Human Genetics
Comments Off on Lumakras (sotorasib) shows encouraging and clinically meaningful anticancer activity in patients with KRAS G12C-mutated advanced pancreatic cancer in…
President of Tufts University to retire – The Boston Globe
Posted: at 5:11 am
This is not the same phenomenon as everybody who is throwing up their hands and saying let me out of here, said McLaughlin, who runs an annual program for new college presidents and also advises experienced presidents. These are more typical, expected tenures.
At the same time, she said, there is no question that these leaders are exhausted.
Its been a slog, McLaughlin said. They have had to spend their time making difficult decisions, many of which are controversial . . . They have been missing what presidents talk about is the joyful aspects of their jobs.
The highest positions in higher education leadership typically turn over every 10 to 15 years, but this transition comes at an inflection point for the industry. The new generation of presidents who fill these openings will have to tackle a host of issues, including the use of online education, the changing demographics of students, the increasingly unaffordable cost of a degree, the public perception of higher education, and questions about diversity and inclusion, tenure and academic freedom.
It is a time when new leadership will be grappling with what could be called new questions or maybe persistent questions, but which are being asked simultaneously with more urgency than before, McLaughlin said.
The Tufts president, Anthony Monaco, 62, who played a key role in working with the Broad Institute to develop COVID-19 testing that enabled a return to in-person learning this fall, said he has no immediate plans for his next chapter after serving 12 years as the schools leader. He said he hopes to continue to work to solve societal challenges from a new post in academia or another sector.
In an e-mail to the campus Monday morning, he thanked students, faculty, and staff for what he called an incredible personal and professional journey. Now, he wrote, is the time for a new leader with a bold vision and fresh energy.
Tufts is an esteemed university that is poised to become even greater, Monaco wrote.
Monacos announcement follows that of MIT president L. Rafael Reif, who last week announced that he will retire at the end of the year after more than a decade of service. Biddy Martin, president of Amherst College, plans to step down this summer after 11 years on the job. Dartmouth College president Philip Hanlon announced last month that he will step down in June 2023 after 10 years. Laurie Leshin also recently said she will depart Worcester Polytechnic Institute this spring to become director of the Jet Propulsion Laboratory and vice president of Caltech. And Sister Janet Eisner plans to step down after serving as president of Emmanuel College for 43 years.
In an interview on Sunday, Monaco said he is proud of the work that Tufts has accomplished during his tenure in the areas of climate change, health disparities, civic engagement, and mental health.
We have been able to have a profound impact in many different areas, he said.
Monaco said he found camaraderie during the pandemic in working with other presidents from around the region who met regularly to develop COVID protocols. Monaco was a key leader during the outbreak and helped create the COVID-19 testing program with the Broad Institute that proved pivotal in allowing universities to stay open during the pandemic.
We [presidents] shared our anxieties and plans to overcome them, he said. It was really tough for a lot of leaders to manage the rising rates of positivity . . . and by being together and sharing expertise, it really helped overall in managing such a difficult situation.
The biggest challenge for any university president, Monaco said, is trying to keep the institution focused on its long-term goals while dealing with the immediate challenges that inevitably arise. The pandemic, he said, was a prime example.
Youre trying to look toward the future while the external environment is changing rapidly and often unexpectedly, he said.
A geneticist by training, Monaco ran a center for human genetics at Oxford University in the United Kingdom before becoming pro-vice-chancellor for planning and resources at Oxford in 2007.
In an e-mail to the Tufts community on Monday, Peter R. Dolan, the chairman of the universitys board of trustees, thanked Monaco on the boards behalf for his steadfast leadership of the university, for his dedication to Tufts, and for the clear moral compass and intellectual rigor that have guided him as president.
Dolan credited Monaco for doubling the universitys endowment from $1.4 billion in 2011 to $2.8 billion today and nearly doubling undergraduate applications, from 17,097 for the class entering in fall 2011 to over 34,000 for the current admissions cycle. He also praised him for the 2016 acquisition of the School of the Museum of Fine Arts in Boston.
Monaco oversaw the university during a time of profound societal upheaval. In 2019, the university removed the Sackler name from its medical school programs and facilities, an effort to distance itself from the family and its company, Purdue Pharma, which admitted to playing a key role in fueling the opioid addiction epidemic. Also in 2019, Monaco was vocal in his support for the Deferred Action for Childhood Arrivals program, or DACA, opposing former president Donald Trumps attempt to end it.
Monaco said he wishes he had made greater strides toward expanding the universitys capacity to respond to the mental health needs of students.
Im worried that the mental health rise is not abating, he said.
Monaco said he hoped Tufts next president will focus on expanding the universitys profile as a research institution. During his tenure, he said, the university developed more masters and PhD programs. The next leader, he said, should focus on attracting more funding for research.
A. David Paltiel, professor of health policy and management at Yale School of Public Health, worked with Monaco and other college leaders in the months leading up to the reopening of campuses in the fall of 2020, as they developed a testing system that involved sending massive numbers of samples from colleges across the region to the Broad Institute in Cambridge to be processed.
Monaco was among the first college administrators to recognize that frequent, routine testing of asymptomatic people was the key to keeping campuses open, Paltiel said.
Monaco, he said, was also cognizant of his surroundings, recognizing the risks posed by the fact that Tufts is located in a residential area of Medford and Somerville.
President Monaco totally understood that his responsibility extended beyond the Tufts campus, Paltiel said. His outreach efforts allayed a lot of skepticism and fear. And, thanks largely to him, the Tufts campus remained among the very safest places in Middlesex County.
Laura Krantz can be reached at laura.krantz@globe.com. Follow her on Twitter @laurakrantz and on Instagram @laurakrantz.
Continued here:
President of Tufts University to retire - The Boston Globe
Posted in Human Genetics
Comments Off on President of Tufts University to retire – The Boston Globe
Immune cells that clear away Alzheimer’s disease protein are controlled by circadian rhythms – EurekAlert
Posted: at 5:11 am
image:Image Caption: The image of a clock with brightfield macrophage images around it shows how circadian changes of cell surface heparan sulfate proteoglycans, shown in pink, impedes phagocytosis of fluorescently labeled amyloid-beta, shown in green. As time goes on, and we have a reduction in heparan sulfate proteoglycans, we see an increase in phagocytosis, demonstrated by the bright green cells shown on the left of the clock. This image was made using our fluorescent microscopy cell images with an artistic rendering of the heparan sulfate proteoglycans from the app, Wombo. The clock was made using photoshop and clip art. view more
Credit: Gretchen Clark, made using Wombo (CC-BY 4.0, https://creativecommons.org/licenses/by/4.0/)
Researchers report that the immune cells responsible for clearing away a key protein that builds up in the brains of patients with Alzheimer's disease operate according to daily circadian rhythms. The discovery, reported by Jennifer Hurley of Rensselaer Polytechnic Institute and colleagues in a new study publishing February 10th in the journal PLOS Genetics, provides a potential explanation for the link between Alzheimer's disease and disruptions to a person's sleep cycle.
Alzheimer's disease is known to be associated with disruptions in circadian rhythms, the 24-hour cycle that controls many aspects of human behavior and physiology. For example, sleep disruptions begin years before symptoms of Alzheimer's disease appear and are linked to more severe symptoms and a higher risk of developing the disease.
In the new paper, researchers investigated a molecular mechanism potentially responsible for the connection between Alzheimer's disease and circadian rhythms. They measured the activity of immune cells responsible for clearing away proteins called amyloid-beta that build up as plaques in the brains of people with Alzheimer's disease. Using cultures of these cells grown in the lab, they discovered that the immune cells clear away the amyloid-beta on an oscillating daily cycle controlled by circadian rhythms. However, when cells lost that rhythm, the daily cycle disappeared. They further established that the underlying cause of this oscillation was changes in the number of molecules of a certain protein, heparan, on the cell's surface. The protein they identified responds to circadian rhythms and previously had been shown to play a role in clearing amyloid-beta proteins.
The new findings uncover a mechanism that links the disruption of circadian rhythms to Alzheimer's disease. The study further highlights the role of immune cells in this relationship. While more studies will be necessary, the new findings present the possibility that, if the daily clearance of amyloid-beta proteins through this mechanism can be maintained, patients may be less likely to develop Alzheimer's disease and to exhibit less severe symptoms.
Hurley adds, Understanding how our circadian rhythms can regulate cell-surface heparan levels to control the build-up of amyloid-beta may lead to the development of chronotherapeutics that alleviate the symptoms of Alzheimers Disease as well as other inflammatory diseases.
#####
In your coverage, please use this URL to provide access to the freely available article in PLOS Genetics:
http://journals.plos.org/plosgenetics/article?id=10.1371/journal.pgen.1009994
Citation: Clark GT, Yu Y, Urban CA, Fu G, Wang C, Zhang F, et al. (2022) Circadian control of heparan sulfate levels times phagocytosis of amyloid beta aggregates. PLoS Genet 18(2): e1009994. https://doi.org/10.1371/journal.pgen.1009994
Author Countries: United States, China
Funding: This work was supported by an NIH-National Institute of Biomedical Imaging and Bioengineering Grant U01EB022546 (to J.M.H) (https://www.nibib.nih.gov/), an NIH-National Institute of General Medical Sciences Grant R35GM128687 (to J.M.H.) (https://www.nigms.nih.gov/), an National Science Foundation CAREER Award 2045674 (to J.M.H.) (https://www.nsf.gov/), National Institutes of Health grants 1RF1AG069039 (to C.W.), DK111958 and CA231074 (to R.J.L.) (https://www.nih.gov/), Rensselaer Polytechnic Startup funds (to J.M.H.) (https://www.rpi.edu/), a gift from the Warren Alpert Foundation (to J.M.H.) (https://warrenalpert.org/), and a NIH-National Institute of Aging T32 Fellowship AG057464 (to G.T.C.) (https://www.nia.nih.gov/). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Experimental study
Cells
Competing interests: The authors have declared that no competing interests exist.
Disclaimer: AAAS and EurekAlert! are not responsible for the accuracy of news releases posted to EurekAlert! by contributing institutions or for the use of any information through the EurekAlert system.
Here is the original post:
Immune cells that clear away Alzheimer's disease protein are controlled by circadian rhythms - EurekAlert
Posted in Human Genetics
Comments Off on Immune cells that clear away Alzheimer’s disease protein are controlled by circadian rhythms – EurekAlert
DNA Methylation and Its Basic Function
Posted: at 5:02 am
Abstract
In the mammalian genome, DNA methylation is an epigenetic mechanism involving the transfer of a methyl group onto the C5 position of the cytosine to form 5-methylcytosine. DNA methylation regulates gene expression by recruiting proteins involved in gene repression or by inhibiting the binding of transcription factor(s) to DNA. During development, the pattern of DNA methylation in the genome changes as a result of a dynamic process involving both de novo DNA methylation and demethylation. As a consequence, differentiated cells develop a stable and unique DNA methylation pattern that regulates tissue-specific gene transcription. In this chapter, we will review the process of DNA methylation and demethylation in the nervous system. We will describe the DNA (de)methylation machinery and its association with other epigenetic mechanisms such as histone modifications and noncoding RNAs. Intriguingly, postmitotic neurons still express DNA methyltransferases and components involved in DNA demethylation. Moreover, neuronal activity can modulate their pattern of DNA methylation in response to physiological and environmental stimuli. The precise regulation of DNA methylation is essential for normal cognitive function. Indeed, when DNA methylation is altered as a result of developmental mutations or environmental risk factors, such as drug exposure and neural injury, mental impairment is a common side effect. The investigation into DNA methylation continues to show a rich and complex picture about epigenetic gene regulation in the central nervous system and provides possible therapeutic targets for the treatment of neuropsychiatric disorders.
Keywords: epigenetics, gene regulation, neuron, synaptic plasticity, demethylation
Genetics is the study of heritable changes in gene activity or function due to the direct alteration of the DNA sequence. Such alterations include point mutations, deletions, insertions, and translocation. In contrast, epigenetics is the study of heritable changes in gene activity or function that is not associated with any change of the DNA sequence itself. Although virtually all cells in an organism contain the same genetic information, not all genes are expressed simultaneously by all cell types. In a broader sense, epigenetic mechanisms mediate the diversified gene expression profiles in a variety of cells and tissues in multicellular organisms.
In this chapter, we would introduce a major epigenetic mechanism involving direct chemical modification to the DNA called DNA methylation. Historically, DNA methylation was discovered in mammals as early as DNA was identified as the genetic material (Avery et al, 1944; McCarty and Avery, 1946). In 1948, Rollin Hotchkiss first discovered modified cytosine in a preparation of calf thymus using paper chromatography. Hotchkiss (1948) hypothesized that this fraction was 5-methylcytosine (5mC) because it separated from cytosine in a manner that was similar to the way that thymine (also known as methyluracil) separated from uracil, and he further suggested that this modified cytosine existed naturally in DNA. Although many researchers proposed that DNA methylation might regulate gene expression, it was not until the 1980s that several studies demonstrated that DNA methylation was involved in gene regulation and cell differentiation (Holliday and Pugh, 1975; Compere and Palmiter, 1981). It is now well recognized that DNA methylation, in concert with other regulators, is a major epigenetic factor influencing gene activities.
DNA methylation is catalyzed by a family of DNA methyltransferases (Dnmts) that transfer a methyl group from S-adenyl methionine (SAM) to the fifth carbon of a cytosine residue to form 5mC (). Dnmt3a and Dnmt3b can establish a new methylation pattern to unmodified DNA and are thus known as de novo Dnmt (). On the other hand, Dnmt1 functions during DNA replication to copy the DNA methylation pattern from the parental DNA strand onto the newly synthesized daughter strand (). All three Dnmts are extensively involved in the development of an embryo. By the time cells reach terminal differentiation, Dnmt expression is much reduced. This would seem to suggest that the DNA methylation pattern in postmitotic cells is stable. However, postmitotic neurons in the mature mammalian brain still express substantial levels of Dnmts, raising the possibility that Dnmts and DNA methylation may play a novel role in the brain (Goto et al, 1994; Feng et al, 2005).
DNA methylation pathways. A family of DNA methyltransferases (Dnmts) catalyzes the transfer of a methyl group from S-adenyl methionine (SAM) to the fifth carbon of cytosine residue to form 5-methylcytosine (5mC). (a) Dnmt3a and Dnmt3b are the de novo Dnmts and transfer methyl groups (red) onto naked DNA. (b) Dnmt1 is the maintenance Dnmt and maintains DNA methylation pattern during replication. When DNA undergoes semiconservative replication, the parental DNA stand retains the original DNA methylation pattern (gray). Dnmt1 associates at the replication foci and precisely replicates the original DNA methylation pattern by adding methyl groups (red) onto the newly formed daughter strand (blue).
Neurons react to the environment through patterns of depolarization that both relay information and encode a response. In recent years, it has become increasingly apparent that following depolarization, alterations in gene expression are accompanied by modifications of the epigenetic landscape that include alterations in the pattern of DNA methylation (Martinowich et al, 2003; Guo et al, 2011a). In order for the DNA methylation pattern to be altered, there must be both active DNA methylation and demethylation in the neuronal genome. However, no enzymes are known to cleave the methyl group directly from 5mC. As discussed below, the recent identification of 5-hydroxymethyl-cytosine (5hmC) in postmitotic neurons suggests that 5hmC serves as an intermediate in the DNA demethylation pathway. In this review, we will discuss the basic function of DNA methylation in epigenetic gene regulation, and further highlight its role in neural development and neurological disease.
Although the brain contains some of the highest levels of DNA methylation of any tissue in the body, 5mC only accounts for 1% of nucleic acids in the human genome (Ehrlich et al, 1982). The majority of DNA methylation occurs on cytosines that precede a guanine nucleotide or CpG sites. Overall, mammalian genomes are depleted of CpG sites that may result from the mutagenic potential of 5mC that can deaminate to thymine (Coulondre et al, 1978; Bird, 1980). The remaining CpG sites are spread out across the genome where they are heavily methylated with the exception of CpG islands (Bird et al, 1985). Interestingly, there is evidence of non-CpG methylation in mouse and human embryonic stem cells, however these methylation are lost in mature tissues (Ramsahoye et al, 2000; Lister et al, 2009). More thorough analysis of the murine frontal cortex has recently revealed that although the majority of methylation occurs within CpG sites, there is a significant percentage of methylated non-CpG sites (Xie et al, 2012). Because of its recent discovery, the role of non-CpG methylation is still unclear.
DNA methylation is essential for silencing retroviral elements, regulating tissue-specific gene expression, genomic imprinting, and X chromosome inactivation. Importantly, DNA methylation in different genomic regions may exert different influences on gene activities based on the underlying genetic sequence. In the following sections, we will further elaborate upon the role of DNA methylation in different genomic regions.
Approximately 45% of the mammalian genome consists of transposable and viral elements that are silenced by bulk methylation (Schulz et al, 2006). The vast majority of these elements are inactivated by DNA methylation or by mutations acquired over time as the result of the deamination of 5mC (Walsh et al, 1998). If expressed, these elements are potentially harmful as their replication and insertion can lead to gene disruption and DNA mutation (Michaud et al, 1994; Wu et al, 1997; Kuster et al, 1997; Gwynn et al, 1998; Ukai et al, 2003). The intracisternal A particle (IAP) is one of most aggressive retroviruses in the mouse genome (Walsh et al, 1998). IAP is heavily methylated throughout life in gametogenesis, development, and adulthood (Walsh et al, 1998; Gaudet et al, 2004). Even within the embryo when the rest of the genome is relatively hypomethylated, Dnmt1 maintains the repression of IAP elements (Gaudet et al, 2004). When Dnmt1 is depleted by genetic mutations, leading to extensive hypomethylation, IAP elements are expressed (Walsh et al, 1998; Hutnick et al, 2010). This demonstrates that within intergenic regions, one of the main roles of DNA methylation is to repress the expression of potentially harmful genetic elements.
CpG islands are stretches of DNA roughly 1000 base pairs long that have a higher CpG density than the rest of the genome but often are not methylated (Bird et al, 1985). The majority of gene promoters, roughly 70%, reside within CpG islands (Saxonov et al, 2006). In particular, the promoters for housekeeping genes are often imbedded in CpG islands (Gardiner-Garden and Frommer, 1987). CpG islands, especially those associated with promoters, are highly conserved between mice and humans (Illingworth et al, 2010). The location and preservation of CpG islands throughout evolution implies that these regions possess a functional importance.
It appears that CpG islands have been evolutionarily conserved to promote gene expression by regulating the chromatin structure and transcription factor binding. DNA is regularly wrapped around histone proteins forming small, packaged sections called nucleosomes. The more tightly associated with histone proteins the DNA is, the less permissive it is for gene expression. One of the common features of CpG islands is that they contain less nucleosomes than other stretches of DNA (Tazi and Bird, 1990; Ramirez-Carrozzi et al, 2009; Choi, 2010). The few nucleosomes with which CpG islands are associated often contain histones with modifications involved in enhancing gene expression (Tazi and Bird, 1990; Mikkelsen et al, 2007). Although 50% of CpG islands contain known transcription start sites, CpG islands are often devoid of common promoter elements such as TATA boxes (Carninci et al, 2006). As many transcription factor binding sites are GC rich, CpG islands are likely to enhance binding to transcriptional start sites. Despite their lack of common promoter elements, CpG islands enhance the accessibility of DNA and promote transcription factor binding.
The methylation of CpG islands results in stable silencing of gene expression (Mohn et al, 2008). During gametogenesis and early embryonic development, CpG islands undergo differential methylation (Wutz et al, 1997; Caspary et al, 1998; Zwart et al, 2001; Kantor et al, 2004). The ability of methylation to regulate gene expression through CpG islands is particularly important for establishing imprinting (Wutz et al, 1997; Caspary et al, 1998; Zwart et al, 2001; Choi et al, 2005). Imprinted genes are expressed from only one of the two inherited parental chromosomes and their expression is determined by the parent of inheritance. Beyond imprinted genes, DNA methylation of CpG islands regulates gene expression during development and differentiation (Shen et al, 2007; Weber et al, 2007; Fouse et al, 2008; Mohn et al, 2008; Meissner et al, 2008). As CpG islands are associated with the control of gene expression, it would be expected that CpG islands might display tissue-specific patterns of DNA methylation. Although CpG islands in intragenic and gene body regions can have tissue-specific patterns of methylation, CpG islands associated with transcription start sites rarely show tissue-specific methylation patterns (Rakyan et al, 2004; Eckhardt et al, 2006; Meissner et al, 2008; Illingworth et al, 2010; Maunakea et al, 2010). Instead, regions called CpG island shores, located as far as 2kb from CpG islands, have highly conserved patterns of tissue-specific methylation (Irizarry et al, 2009). Like CpG islands, the methylation of CpG shores is highly correlated with reduced gene expression (Irizarry et al, 2009).
The role of CpG islands in regulating gene expression is still being uncovered. Methylation of CpG islands can impair transcription factor binding, recruit repressive methyl-binding proteins, and stably silence gene expression. However, CpG islands, especially those associated with gene promoters, are rarely methylated. Further studies are needed to determine to what degree DNA methylation of CpG islands regulates gene expression.
As the majority of CpG sites within the mammalian genome are methylated, the genes themselves must also contain methylation. The gene body is considered the region of the gene past the first exon because methylation of the first exon, like promoter methylation, leads to gene silencing (Brenet et al, 2011). Evidence suggests that DNA methylation of the gene body is associated with a higher level of gene expression in dividing cells (Hellman and Chess, 2007; Ball et al, 2009; Aran et al, 2011). However, in slowly dividing and nondividing cells such as the brain, gene body methylation is not associated with increased gene expression (Aran et al, 2011; Guo et al, 2011a, 2011b; Xie et al, 2012). Furthermore, in the murine frontal cortex, methylation of non-CpG sites within gene bodies is negatively correlated with gene expression (Xie et al, 2012). How DNA methylation of the gene body contributes to gene regulation is still unclear.
The enzymes that establish, recognize, and remove DNA methylation are broken into three classes: writers, erasers, and readers. Writers are the enzymes that catalyze the addition of methyl groups onto cytosine residues. Erasers modify and remove the methyl group. Readers recognize and bind to methyl groups to ultimately influence gene expression. Thanks to the many years of research devoted to understanding how the epigenetic landscape is erased and reshaped during embryonic development, many of the proteins and mechanisms involved in DNA methylation have already been identified.
Three members of the Dnmt family directly catalyze the addition of methyl groups onto DNA: Dnmt1, Dnmt3a, and Dnmt3b. Although these enzymes share a similar structure with a large N-terminal regulatory domain and a C-terminal catalytic domain, they have unique functions and expression patterns (Yen et al, 1992; Xie et al, 1999). Probably the best studied Dnmt, especially in the nervous system, is Dnmt1, which is highly expressed in mammalian tissues including the brain (Goto et al, 1994). Unlike the other Dnmts, Dnmt1 preferentially methylates hemimethylated DNA (Pradhan et al, 1999; Ramsahoye et al, 2000). During DNA replication, Dnmt1 localizes to the replication fork where newly synthesized hemimethylated DNA is formed (Leonhardt et al, 1992). Dnmt1 binds to the newly synthesized DNA and methylates it to precisely mimic the original methylation pattern present before DNA replication (Hermann et al, 2004) (). Additionally, Dnmt1 also has the ability to repair DNA methylation (Mortusewicz et al, 2005). For this reason, Dnmt1 is called the maintenance Dnmt because it maintains the original pattern of DNA methylation in a cell lineage. Knockout of Dnmt1 in mice results in embryonic lethality between E8.0 and E10.5 (Li et al, 1992). At this time, knockout embryos exhibit a two-thirds loss of DNA methylation, in addition to numerous apoptotic cells in a variety of developing tissues including the brain. Interestingly, mouse embryonic stem cells lacking Dnmt1 remain viable (Chen et al, 1998). However, in vitro differentiation results in massive cell death, recapitulating the phenotype observed in knockout embryos (Jackson-Grusby et al, 2001). These findings firmly establish that Dnmt1 plays a critical role in cellular differentiation as well as in dividing cells.
Dnmt3a and Dnmt3b are extremely similar in structure and function. Unlike Dnmt1, both Dnmt3a and Dnmt3b when overexpressed are capable of methylating both native and synthetic DNA with no preference for hemimethylated DNA (Okano et al, 1999). For this reason, Dnmt3a and Dnmt3b are referred to as de novo Dnmt because they can introduce methylation into naked DNA (). What primarily distinguishes Dnmt3a from Dnmt3b is its gene expression pattern. Although Dnmt3a is expressed relatively ubiquitously, Dnmt3b is poorly expressed by the majority of differentiated tissues with the exception of the thyroid, testes, and bone marrow (Xie et al, 1999). Similar to Dnmt1, the knockout of Dnmt3b in mice is embryonic lethal (Okano et al, 1999). On the other hand, Dnmt3a knockout mice are runted but survive to 4 weeks after birth (Okano et al, 1999). From these results it appears that Dnmt3b is required during early development, whereas Dnmt3a is required for normal cellular differentiation.
A final member of the Dnmt family is Dnmt3L, a protein that lacks the catalytic domain present in other Dnmt enzymes (Aapola et al, 2000; Hata et al, 2002). Dnmt3L is mainly expressed in early development and is restricted to the germ cells and thymus in adulthood (Aapola et al, 2000, 2001). Although Dnmt3L has no catalytic function of its own, it associates with the Dnmt3a and Dnmt3b and stimulates their methyltransferase activity (Hata et al, 2002; Suetake et al, 2004; Jia et al, 2007). Consistent with its presence in early development and in germ cells, in mice, Dnmt3L is required for establishing both maternal and paternal genomic imprinting, for methylating retrotransposons, and for compaction of the X chromosome (Bourc'his et al, 2001; Hata et al, 2002; Kaneda et al, 2004; Bourc'his and Bestor, 2004; Webster et al, 2005; La Salle et al, 2007; Zamudio et al, 2011). Although Dnmt3L is expressed in the developing brain, Dnmt3L is downregulated during neuronal differentiation and is not observed in the brain postnatally (Lee et al, 2006; Kovacheva et al, 2007).
(How the de novo Dnmts target specific genetic regions is still unclear). However, several mechanisms have been proposed. Dnmt3a and Dnmt3b can bind to DNA via a conserved PWWP domain (Ge et al, 2004); however, it is unclear how Dnmt3a and Dnmt3b target specific DNA sequences. One hypothesis suggests that RNA interference (RNAi) mechanisms target Dnmts to silence specific sequences of DNA (Morris et al, 2004). Although RNAi is clearly involved in DNA methylation in plant cells, the existing evidence is still very weak for a role of RNAi in DNA methylation in mammalian cells. The other theory is that transcription factors regulate de novo DNA methylation. Transcription factors can regulate DNA methylation by binding to specific DNA sequence to either recruit Dnmts for methylation or protect from DNA methylation. In some cases Dnmts bind to transcription factors or components of repressor complexes to target methylation to DNA (Brenner et al, 2005). In other cases, regardless of whether the gene is expressed, the transcription factor binding can help protect CpG sites from de novo methylation (Straussman et al, 2009; Gebhard et al, 2010; Lienert et al, 2011). CpG islands appear to primarily be protected from methylation by transcription factor binding (Brandeis et al, 1994; Macleod et al, 1994; Straussman et al, 2009; Gebhard et al, 2010). When transcription factor binding sites are mutated, CpG islands are unable to retain their unmethylated state (Brandeis et al, 1994; Macleod et al, 1994). Similarly, as differentiation induces the downregulation of transcription factors that bind to specific gene promoters, the now-exposed CpG sites can be targeted for DNA methylation (Lienert et al, 2011). These studies describe two mechanisms that likely function together to establish de novo DNA methylation. Dnmt3a and Dnmt3b can either be recruited to promoters by specific transcription factors or the de novo Dnmt may simply methylate all CpG sites across the genome that are not protected by a bound transcription factor.
DNA demethylation is characterized as either passive or active. Passive DNA demethylation occurs in dividing cells. As Dnmt1 actively maintains DNA methylation during cell replication, its inhibition or dysfunction allows newly incorporated cytosine to remain unmethylated and consequently reduces the overall methylation level following each cell division. Active DNA demethylation can occur in both dividing and nondividing cells but the process requires enzymatic reactions to process the 5mC in order to revert it back to a naked cytosine (Mayer et al, 2000; Oswald et al, 2000; Paroush et al, 1990; Zhang et al, 2007). As of yet, there is no known mechanism in mammalian cells that can cleave the strong covalent carbon-to-carbon bond that connects cytosine to a methyl group. Instead, demethylation occurs through a series of chemical reactions that further modify 5mC, by deamination and/or oxidation reactions to a product that is recognized by the base excision repair (BER) pathway to replace the modified base with naked cytosine. Although it is generally agreed upon that the BER pathway is the final step in DNA demethylation, the specific enzymes and the chemical intermediates that are formed during DNA demethylation are still debated (Bhutani et al, 2011).
Several mechanisms of active DNA demethylation have been proposed (). 5mC can be chemically modified at two sites, the amine group and the methyl group. Deamination of the amine to a carbonyl group by AID/APOBEC (activation-induced cytidine deaminase/apolipoprotein B mRNA-editing enzyme complex) effectively converts 5mC into thymine, thus creating a G/T mismatch and inducing the BER pathway to correct the base. Overexpression of AID/APOBEC promotes DNA demethylation in zebrafish (Rai et al, 2008), whereas knockdown or knockout inhibits the DNA demethylation of various genes necessary for cellular reprogramming and development (Bhutani et al, 2010; Muramatsu et al, 2000; Popp et al, 2010). Unlike the various Dnmt knockout mice, knockout AID mice are viable and fertile. If global DNA demethylation is as critical as DNA methylation in early development, then the knockout AID mice study raises the possibility that multiple mechanisms for active DNA demethylation exist and can compensate for one another.
Active DNA demethylation pathways. 5-Methylcytosine (5mC) can be chemically modified at two sites: the amine group and the methyl group. The amine group of 5mC can be deaminated (green) by AID/APOBEC, converting 5mC into thymine (Thy). The methyl group of 5mC can be modified by the addition of a hydroxyl group mediated by Tet enzymes to generate 5-hydroxymethyl-cytosine (5hmC). 5hmC can also be chemically modified at two sites: the amine group and the hydroxymethyl group. AID/APOBEC can deaminate (green) 5hmC to produce 5-hydroxymethyl-uracil (5hmU). In another chemical pathway for 5hmC is that Tet can further oxidize (yellow) 5hmC to form 5-formyl-cytosine (5fC) and then 5-carboxy-cytosine (5caC). Eventually, the products of each pathwayThy, 5hmU, 5fC, and 5caCare recognized and cleaved off to replace with a naked cytosine mediated by TDG and/or SMUG1, both components of the base excision repair pathway (red).
In line with the multiple mechanisms hypothesis, another active DNA demethylation mechanism is found to be mediated by the teneleven translocation (Tet) enzymes Tet1, Tet2, and Tet3. Tet enzymes add a hydroxyl group onto the methyl group of 5mC to form 5hmC (Tahiliani et al, 2009; Ito et al, 2010). The developed brain contains significant 5hmC levels in multiple regions, ranging from 0.3 to 0.7%, which is approximately tenfold lower than the average abundance of 5mC (Kriaucionis and Heintz, 2009; Globisch et al, 2010). Once 5hmC is formed, two separate mechanisms can convert 5hmC back into cytosine in mammals. In the first, iterative oxidation by Tet enzymes continues to oxidize 5hmC first to 5-formyl-cytosine and then to 5-carboxy-cytosine (Ito et al, 2011). In the second, 5hmC is deaminated by AID/APOBEC to form 5-hydroxymethyl-uracil (Guo et al, 2011b). Consistent with the role of Tet in converting 5mC into 5hmC in vivo, Tet1 knockout mouse embryonic stem cells have reduced levels of 5hmC that is accompanied by a subtle increase in 5mC at a global level (Dawlaty et al, 2011).
Whether 5hmC functions only as an intermediate in DNA demethylation is still unclear. Like methylation, 5hmC may regulate gene expression. In support of this theory, the conversion of 5mC to 5hmC impairs the binding of the repressive methyl-binding protein MeCP2 (Valinluck et al, 2004). But what is clear at this time is that 5hmC is found in vivo in mammalian tissue and may play an important role in regulating DNA demethylation and gene expression.
In all the mentioned mechanisms of active DNA demethylation, the BER pathway uses thymine DNA glycosylase (TDG) to cleave off the modified residuethymine, 5-hydroxymethyl-uracil, 5-formyl-cytosine, and 5-carboxy-cytosineand replace it with a naked cytosine (Cortellino et al, 2011; He et al, 2011). TDG is essential for DNA demethylation and is required for normal development. Knockout or inactivation of TDG leads to embryonic lethality in mice. Moreover, these mutant embryos exhibit hypermethylation, particularly in imprinting genes such as Igf2 and H19, suggesting that active demethylation by TDG protects imprinted genes from spontaneous de novo methylation (Cortellino et al, 2011). Single-strand-selective monofunctional uracil-DNA glycosylase 1 (SMUG1), another BER enzyme from the same uracil DNA glycosylase family as TDG, is also found to be involved in DNA demethylation (Cortellino et al, 2011; Guo et al, 2011a, 2011b). In summary, active DNA demethylation arises from multiple pathways involving multiple enzymes and this complexity has likely contributed to much of the current scientific debate.
Whereas DNA methylation may itself reduce gene expression by impairing the binding of transcriptional activators, a second class of proteins with a high affinity for 5mC inhibits transcription factor binding. DNA methylation is recognized by three separate families of proteins: the MBD proteins, the UHRF proteins, and the zinc-finger proteins. Of these families, the MBD was the first to be identified. MBD proteins contain a conserved methyl-CpG-binding domain (MBD) that confers a higher affinity for single methylated CpG sites (Nan et al, 1993). This family includes MeCP2, the first identified methyl-binding protein, along with MBD1, MBD2, MBD3, and MBD4 (Meehan et al, 1989; Lewis et al, 1992; Hendrich and Bird, 1998). MBDs are more highly expressed in the brain than in any other tissue, and many MBDs are important for normal neuronal development and function (Amir et al, 1999). Of the MBD family, MBD3 and MBD4 are unusual. For example, MBD3 is incapable of directly binding to DNA due to a mutation in its MBD domain (Hendrich and Bird, 1998). Although MBD4 binds to DNA normally, it preferentially recognizes when guanine is mismatched with a thymine, uracil, or 5-fluorouracil and associates with proteins involved in DNA mismatch repair (Bellacosa et al, 1999; Hendrich et al, 1999; Petronzelli et al, 2000; Millar et al, 2002; Wong et al, 2002). The remaining members of the MBD family have the ability to directly bind to methylated DNA and contain a transcriptional repression domain (TRD) that allows MBD proteins to bind to a variety of repressor complexes (Nan et al, 1998; Ng et al, 1999; Sarraf and Stancheva, 2004). In addition to its role as a transcriptional repressor, MeCP2 appears to have a unique role in the maintenance of DNA methylation. MeCP2 binds to Dnmt1 via its TRD and can recruit Dnmt1 to hemimethylated DNA to perform maintenance methylation (Kimura and Shiota, 2003). Although MBDs are the best studied class of methyl-binding proteins, they are not the only one.
The UHRF (ubiquitin-like, containing PHD and RING finger domain) proteins, including UHRF1 and UHRF2, are multidomain proteins that flip out and bind methylated cytosines via a SET- and RING-associated DNA-binding domain (Hashimoto et al, 2008, 2009). Unlike most methyl-binding proteins, the primary function of UHRF proteins is not to bind to DNA and repress transcription. The UHRF protein family first binds to Dnmt1 and then targets it to hemimethylated DNA in order to maintain DNA methylation, especially during DNA replication (Sharif et al, 2007; Bostick et al, 2007; Achour et al, 2008). UHRF1 appears to interact so closely with Dnmt1 that its deletion, like the deletion of Dnmt1, leads to embryonic lethality (Muto et al, 2002).
The last family of methyl-binding proteins binds to methylated DNA by a zinc-finger domain and is composed of Kaiso, ZBTB4, and ZBTB38 (Prokhortchouk et al, 2001; Filion et al, 2006). Although ZBTB4 and ZBTB38 have distinct tissue expression patterns, both are highly expressed in the brain and can bind to a single methylated CpG. The zinc-finger domain proteins are unusual. Despite their ability to recognize methylcytosine, both Kaiso and ZBTB4 have preferential binding for a sequence motif lacking a methylcytosine (Daniel et al, 2002; Sasai et al, 2010). Unlike other methyl-binding proteins, Kaiso preferentially binds to two consecutively methylated CpG sites (Daniel et al, 2002). Yet despite their differences, zinc-finger domain proteins, similar to the MBD family, repress transcription in a DNA methylation-dependent manner (Prokhortchouk et al, 2001; Yoon et al, 2003; Filion et al, 2006; Lopes et al, 2008).
DNA methylation works with histone modifications and microRNA (miRNA) to regulate transcription (). In eukaryotes, DNA is associated with histone proteins that help to package the long strands of DNA into the small nuclear compartment. Chemical modifications that include methylation, acetylation, ubiquitination, and phosphorylation are added to three specific amino acids on the N-terminal histone tails. These modifications influence not only how DNA strands are packaged but also their transcriptional activity. Histone modifications that loosen DNA association with histones generally provide a permissive environment for transcription, whereas histone modifications that tightly package DNA and histones repress gene expression. Dnmts directly interact with enzymes that regulate histone modifications typically involved in gene repression (). Both Dnmt1 and Dnmt3a are known to bind to the histone methyltransferase SUV39H1 that restricts gene expression by methylation on H3K9 (Fuks et al, 2003). Furthermore, Dnmt1 and Dnmt3b can both bind to histone deacetylases that remove acetylation from histones to make DNA pack more tightly and restrict access for transcription (Fuks et al, 2000; Geiman et al, 2004). In general, Dnmts cooperate with histone-modifying enzymes involved in adding and/or stripping histone markers in order to impose a repressive state on a gene region.
Epigenetic crosstalk. Transcription is ultimately regulated by the interaction of multiple epigenetic mechanisms that cooperate to activate or silence gene expression. Methylation is regulated by proteins such as Dnmt and Tet (purple) that are involved in the active addition or chemical modification (such as hydroxymethylation in red) of DNA methylation. To suppress gene expression, Dnmts target CpG sites and actively methylate DNA. For some Dnmts, their catalytic activity is enhanced by association with histone tails and with Dnmt3L. DNA methylation is recognized by methyl-binding proteins such as MBDs (yellow) that along with Dnmts recruit enzymes that modify the histone tails (orange) including histone deacetylases (HDACs), which remove acetylation (red), and histone methyltransferases (HMTs), which methylate histones (green) and in conjunction with DNA methylation serve to further repress gene expression. In regions of DNA with activate transcription, Tet removes DNA methylation, and histone tails in this region often contain H3K4me3 that inhibits Dnmt binding to unmethylated CpG sites and maintains a permissive environment for transcription.
Histone modifications can also influence the DNA methylation pattern (). Dnmt3L binds to H3 histone tails and recruits Dnmt3a and Dnmt3b to methylate DNA (Ooi et al, 2007). The direct binding of Dnmt3a to the H3 histone tail, sometimes facilitated by H3K36 trimethylation, a repressive histone mark, also stimulates its methyltransferase activity (Dhayalan et al, 2010; Li et al, 2011a). However, the presence of the active histone modification H3K4 trimethylation (H3K4me3) impairs the binding of Dnmt3a, Dnmt3b, and Dnmt3L to H3 histone tails and prevents methylation (Ooi et al, 2007; Zhang et al, 2010). CpG islands contain particularly high levels of H3K4me3 (Mikkelsen et al, 2007). Cfp1 is a component of the H3K4 methyltransferase complex that targets unmethylated CpG sites often found at murine CpG islands and may play a role in maintaining their hypomethylation (Lee and Skalnik, 2005; Thomson et al, 2010). Little is known regarding how the DNA demethylation machinery interacts with histone modifications, yet there is still evidence to suggest that they cooperate. For instance, elevated histone acetylation can trigger DNA demethylation (Cervoni and Szyf, 2001; D'Alessio et al, 2007). Tet1 contains a DNA-binding motif similar to Ctf1, suggesting that both proteins target similar sites, in this case CpG islands, to maintain DNA demethylation (Tahiliani et al, 2009). Although a direct connection between the two has yet to been shown, Tet1 does indeed localize to CpG islands and its depletion results in an increase in methylation within those CpG islands in mouse embryonic stem cell studies (Ficz et al, 2011; Wu and Zhang, 2011). Future studies are needed to further probe the interaction of Tet with histone modifications.
Methyl-binding proteins serve as the strongest link between DNA methylation and histone modification. Both the MBDs and the UHRF proteins interact with methylated DNA and histones to enhance gene repression () (Nan et al, 1998; Ng et al, 1999; Sarraf and Stancheva, 2004; Citterio et al, 2004; Karagianni et al, 2008). MeCP2 recruits histone deacetylases to remove active histone modifications and repress gene transcription (Jones et al, 1998; Nan et al, 1998; Fuks et al, 2003). Furthermore, MeCP2 enhances the repressive chromatin state by recruiting histone methyltransferases that add repressive H3K9 methylation (Fuks et al, 2003). Overall, DNA methylation and histone modifications work closely together to regulate gene expression.
Recently, miRNAs have emerged as another important epigenetic mechanism that influences gene expression. Precursor miRNA forms a double-stranded RNA connected by a hairpin loop. Once transported into the cytoplasm precursor, miRNA is processed by Dicer to generate a 2223 nucleotide miRNA:miRNA* duplex. Mature miRNAs associate with a miRNA-induced silencing complex (miRISC) that can bind to their target mRNA and repress gene expression by inhibiting translation or inducing RNA degradation (Berezikov, 2011). Like other sequences within the genome, DNA methylation can regulate the expression of miRNAs (Han et al, 2007; Lujambio et al, 2008). The loss of both Dnmt1 and Dnmt3b in a colon cancer cell line revealed that 10% of detected miRNAs are regulated by DNA methylation (Han et al, 2007). When Dnmts are inhibited, cancer cells reactivate some miRNAs that are initially silenced by hypermethylation of their CpG islands (Lujambio et al, 2008). Understood together, these studies demonstrate that DNA methylation regulates miRNA expression.
Conversely, miRNAs can also regulate histone modifications and Dnmt expression and, in so doing, regulate DNA methylation (Benetti et al, 2008; Sinkkonen et al, 2008). Knockout of Dicer in mouse embryonic stem cells results in depletion of miRNAs, one of which is miRNA-290, which indirectly regulates Dnmt3a and Dnmt3b expression (Benetti et al, 2008; Sinkkonen et al, 2008). This leads to a loss of DNA methylation and an increase in repressive histone methylation at H3K9. These studies provided evidence of a bidirectional influence between miRNA and DNA methylation.
The precise temporal regulation of de novo methylation and demethylation is particularly important for the differentiation and maturation of the mammalian central nervous system (CNS). Multipotent neural progenitor cells (NPCs) sequentially undergo neurogenesis and astrogliogenesis (Qian et al, 2000; Sauvageot and Stiles, 2002). In particular, the differentiation switch of NPCs from neurogenesis to astrogliogenesis coincides with DNA methylation and demethylation events on the glial fibrillary acidic protein (Gfap) gene promoter region (Teter et al, 1994). Early in neurogenesis at E11.5, DNA methylation of the Gfap promoter represses its expression (Teter et al, 1996; Takizawa et al, 2001). The continual expression of Dnmt1 in NPCs has been found to be important for the maintenance of the methylation pattern on the Gfap promoter through subsequent cell divisions (Fan et al, 2005). Interestingly, neurogenesis from E11.5 to E14.5 is the only time during neural development that Dnmt3b is strongly expressed before declining to nearly undetectable levels in the CNS (Fan et al, 2005). At E14.5, the Gfap promoter undergoes DNA demethylation to coincide with the differentiation of the astrocytic lineage (Teter et al, 1996). As development further progresses, the decline of Dnmt3b and the peak expression of Dnmt3a at 3 weeks postnatally coincide with remethylation and reduced transcription of the Gfap promoter (Fan et al, 2005; Nguyen et al, 2007). The coordinated expression of Dnmts and their ability to regulate the methylation pattern of the Gfap promoter organize and regulate neuronal development.
The importance of these coordinated events is highlighted by conditional knockout models of Dnmts during neural development (Fan et al, 2001, 2005; Golshani et al, 2005; Nguyen et al, 2007; Hutnick et al, 2009; Feng et al, 2010). Conditional knockout of Dnmt1 between E8.5 and E13.5, a time period that coincides with neurogenesis, leads to hypomethylation of differentiating neurons and demethylation of the Gfap promoter in neural precursor cells, thus accelerating astrogliosis (Fan et al, 2001, 2005). Hypomethylated neurons are characterized by multiple maturation defects including dendritic arborization and impaired neuronal excitability (Fan et al, 2001; Golshani et al, 2005; Hutnick et al, 2009). These results are consistent with the conclusion that Dnmt1 has a critical role in neuronal differentiation and in maintaining the methylation of the Gfap promoter. Furthermore, these results suggest that DNA methylation is essential for neuronal maturation. If Dnmt3a is knocked out instead in the neural precursor cells, the majority of cortical neurons develop normally (Nguyen et al, 2007). In this case, the Gfap promoter that is normally remethylated in postnatal astrocytes remains hypomethylated in 50% of cortical tissue, likely corresponding to the glial population (Nguyen et al, 2007). This is consistent with the idea that Dnmt3a is not required during differentiation or maturation by the majority of cortical neurons. Together, the knockout data confirm that precise regulation of DNA methylation is essential for differentiation and maturation of the CNS.
Like Dnmts, methyl-binding proteins are expressed in embryonic stem cells and in neural precursors, but unlike Dnmts their expression has little effect on neuronal or glial differentiation (Kishi and Macklis, 2004; Martin Caballero et al, 2009). Of the MBD proteins, MeCP2 is the best studied in the CNS because its mutation results in Rett Syndrome, one of the most common forms of mental retardation in females (Amir et al, 1999). During development, MeCP2 is first expressed in the brainstem and thalamus, the most ancient regions of the brain, followed by a rostral progression of expression (LaSalle et al, 2001; Shahbazian et al, 2002). Like other methyl-binding proteins, MeCP2 associates with a variety of transcriptional repressors, including Dnmt1, and influences gene expression (Nan et al, 1998; Ng et al, 1999; Kimura and Shiota, 2003; Sarraf and Stancheva, 2004). Neural activity leads to phosphorylation of MeCP2, altering its ability to bind gene promoters and silence gene expression (Zhou et al, 2006; Tao et al, 2009). MeCP2 is required for normal neuronal maturation and its loss or the loss of its ability to be phosphorylated results in aberrant dendritic arborization, synaptic function, and plasticity (Chen et al, 2001; Moretti et al, 2006; Asaka et al, 2006; Nelson et al, 2006; Cohen et al, 2011; Li et al, 2011b).
At the conclusion of neural development, the brain primarily consists of postmitotic neurons and glial cells with little proliferation potential. Although the expression of Dnmts normally declines in terminally differentiated cells, the brain appears to be an exception. Both Dnmt1 and Dnmt3a are expressed by postmitotic neurons, whereas Dnmt3b expression is either low or virtually undetectable (Goto et al, 1994; Inano et al, 2000; Fan et al, 2005). This surprising discovery led researchers to investigate the role of active DNA methylation in postmitotic neurons of the adult brain.
To date, the majority of the DNA methylation mechanism has been characterized in embryonic stem cells. Although this in vitro model may predict the function of DNA methylation in a dividing cell, embryonic stem cells are an inadequate model for studying DNA methylation in a postmitotic cell. The fact that Dnmts are required for normal neuronal differentiation and maturation hinders the study of DNA methylation solely in postmitotic neurons. Despite these limitations, two models have emerged to study DNA methylation in postmitotic neurons.
The first model utilizes pharmacological inhibitors such as 5-aza-2-deoxycytadine, zebularine, or RG108 injected into the brain to impair Dnmt activity (Levenson et al, 2006; Miller and Sweatt, 2007; Miller et al, 2010). Although pharmalogical inhibition of Dnmts has the advantage of inhibiting DNA methylation at the designated time point, the mechanism of action of Dnmt inhibitors in postmitotic neurons is still debated. Nucleoside inhibitors such as 5-aza-2-deoxycytadine and zebularine must be first incorporated into DNA in order to inhibit Dnmts (Creusot et al, 1982). Once incorporated into the DNA, the nucleoside inhibitors trap Dnmts on the chromosome, which can lead to substantial cellular toxicity (Jttermann et al, 1994; Zhou et al, 2002). Although it is possible that the BER pathway may be the source of nucleoside incorporation, it is still unclear how nucleoside inhibitors are incorporated into the DNA of a nondividing cell, like a postmitotic neuron. Because of the concerns surrounding the use of nucleoside inhibitors, new-generation Dnmt inhibitors, which include RG108, have been developed. After the discovery of the structure of the catalytic domain of Dnmt1, RG108 was identified in an in silico screen as a small molecule that could inhibit Dnmt1 without being incorporated into the DNA (Brueckner et al, 2005; Stresemann et al, 2006). RG108 has emerged as a promising Dnmt1 inhibitor with less cytotoxicity. In cell-free assays, RG108 is capable of impairing Dnmts with catalytic domains similar to Dnmt1. Although Dnmt3a and Dnmt3b share a highly conserved catalytic domain to Dnmt1, it is still unclear whether RG108 inhibits these methyltransferases (Brueckner et al, 2005).
To alleviate concerns of off-target effects and incomplete Dnmt inhibition, a second experimental model has emerged. In this model, Dnmts are conditionally knocked out using the cre/loxP system, with cre expressed by a brain-specific promoter (Fan et al, 2001; Golshani et al, 2005; Nguyen et al, 2007; Hutnick et al, 2009; Feng et al, 2010). Unlike pharmacological inhibition, this second method allows investigators to study the role of a specific Dnmt in subpopulations of neurons. Dnmts are required for normal neuronal differentiation. Therefore, to study the role of Dnmts in the adult brain, cre must be expressed by postmitotic brain-specific promoters such as CamKII (Fan et al, 2001; Golshani et al, 2005; Nguyen et al, 2007; Hutnick et al, 2009; Feng et al, 2010).
In postmitotic neurons, Dnmt1 and Dnmt3a appear to have overlapping roles. Neither knockout of Dnmt1 nor of Dnmt3a in forebrain postmitotic neurons leads to any observable change in DNA methylation, gene expression, synaptic plasticity, or behavior (Feng et al, 2010). However, double knockouts have reduced DNA methylation that leads to deficits in synaptic plasticity in addition to learning and memory. Although research has yet to tease apart the role of Dnmt1 vs Dnmt3a in postmitotic neurons, DNA methylation has repeatedly been shown to play a role in learning and memory in the adult brain.
When neuronal activity is inhibited during fear conditioning, not only is memory formation prevented but so are changes in DNA methylation (Lubin et al, 2008). Early studies demonstrated that in vitro neuronal activity regulated the expression of Bdnf in an activity-dependent manner (Martinowich et al, 2003). Neuronal depolarization demethylates the Bdnf promoter, releasing the MeCP2 repressor complex from the promoter and increasing Bdnf expression (Martinowich et al, 2003). Persistent activity in neurons, as occurs during electroconvulsive stimulation or exercise, leads to active DNA methylation and demethylation across several genes within the brain. However, alterations in DNA methylation do not always correlate with the alterations in gene expression observed after heightened activity (Guo et al, 2011a). Hence, although both DNA methylation and demethylation are altered by neuronal activity, DNA methylation functions alongside other regulatory proteins and epigenetic mechanisms that determine gene expression.
Another class of proteins that work with DNA methylation to regulate gene expression in the CNS is the class of methyl-binding proteins. Methyl-binding proteins are continually expressed in the adult CNS and often act as repressors that recognize and bind to methylated cytosines (Nan et al, 1998; Ng et al, 1999; Sarraf and Stancheva, 2004). Hence, when methylation is removed as a result of neuronal activity, it is not surprising that MBDs are often released from promoters (Martinowich et al, 2003). However, the role of methyl-binding proteins is not this simple. Some MBDs like MeCP2 undergo posttranslational modifications that alter their ability to bind to DNA (Zhou et al, 2006; Tao et al, 2009). Phosphorylation of MeCP2 is induced by neuronal activity and results in altered gene expression. When phosphorylation of MeCP2 is inhibited, synapse formation, synaptic plasticity, and learning and memory behavior are all affected (Cohen et al, 2011; Li et al, 2011a, 2011b). As phosphorylation is normally a short-term modification, activity-dependent phosphorylation may temporarily release MeCP2 from promoters, making the gene sequence accessible for demethylation. On the other hand, DNA methylation and demethylation may be responsible for long-term changes in gene expression that regulate synaptic plasticity as well as learning and memory.
The pattern of DNA methylation established during development can be modulated by neural activity in order to encode learning and memory. When the mechanisms that establish and recognize the DNA methylation pattern are dysfunctional, problems with learning and memory frequently result. One of the most common forms of mental retardation, Rett Syndrome, is frequently caused by a mutation to the methyl-binding protein MeCP2 (Amir et al, 1999). The onset of symptoms at 618 months of age coincides with a time in early development when sensory experience is driving dendritic pruning and shaping connections in the brain (Samaco and Neul, 2011). Although MeCP2 is expressed by the majority of cells, it is particularly important for normal neuronal function. In mice, loss of MeCP2 in neurons is sufficient to recapitulate the majority of Rett symptoms (Chen et al, 2001; Guy et al, 2001). The phenotype of MeCP2 mutant mice can be reversed by restoration of the MeCP2 gene in postmitotic neurons (Luikenhuis et al, 2004; Giacometti et al, 2007; Guy et al, 2007). As previously mentioned, MeCP2 is regulated by neuronal activity and in turn regulates the expression of BDNF, which has enhanced expression following depolarization (Martinowich et al, 2003). The overexpression of BDNF in postmitotic neurons of MeCP2 mutant mice ameliorates their phenotype, suggesting that MeCP2 is critical for regulating the expression of genes like BDNF that are regulated by neuronal activity and essential for normal cognitive function (Chang et al, 2006). The role of MeCP2 in Rett Syndrome will be further discussed in later chapters.
One extremely rare neurodegenerative disease illustrates the importance of proper DNMT activity in the adult brain. Patients with hereditary sensory and autonomic neuropathy type 1 (HSAN1) develop dementia and hearing loss in adulthood that result from an autosomal-dominant mutation in the N-terminal regulatory domain of DNMT1 (Klein et al, 2011). This mutation results in misfolding, impaired nuclear localization, and early degradation of DNMT1. However, the mutation does not affect the targeting of DNMT1 to the replication foci during cellular replication, but the DNMT1 association with heterochromatin beyond S phase is disrupted. This association may affect the maintenance of DNA methylation within these regions. Although there is only a modest 8% reduction of global DNA methylation level, neurodegeneration does result. The involvement of DNMT1 in the pathogenic mechanism of HSAN1 supports the necessity of DNMT1 in the adult brain.
Improper methylation of a single gene or a single allele can have drastic consequences within the brain. Fragile X Syndrome is caused by abnormal methylation of a trinucleotide repeat expansion in the FMR1 gene on the X chromosome and is a common form of mental retardation (Verkerk et al, 1991; Turner et al, 1996). The hypermethylation in the repeat expansion of FMR1 results in transcriptional silencing (Devys et al, 1993). Translation of the FMR1 gene is regulated by neuronal activity (Weiler et al, 1997) and its protein product, FMRP, is involved in protein synthesis at the synapses following depolarization (for review, see Fatemi and Folsom, 2011). Similarly, improper methylation of a single imprinted allele, found in some disorders such as PraderWilli Syndrome and Angelman Syndrome, can cause significant mental impairments (for review, see Buiting, 2010). As incorrect expression or loss of function of a single gene can have a dramatic effect in the brain, it is important to understand the mechanism of how DNA methylation affects gene expression.
DNA methylation can also be altered by repeated modulation of the microenvironment of the brain. In the case of recurrent seizures this microenvironment is repeatedly subject to unusual, synchronized neuronal activity. One way to mimic this unusual neural activity is by electric convulsive stimulation, which was found to result in genome-wide changes in the DNA methylation pattern (Ma et al, 2009; Guo et al, 2011a). Similarly, repeated drug usage modulates neuronal function as in the case of cocaine. Cocaine usage modulates Dnmt3a expression within the nucleus accumbens and enhances spine formation (LaPlant et al, 2010). Also, repeated cocaine usage increases MeCP2 that, in turn, increases Bdnf expression (Im et al, 2010). Sometimes, drug exposure, like neural activity, can add posttranslational modifications to components of the methylation machinery such as MeCP2 (Deng et al, 2010; Hutchinson et al, 2012).
Although DNA methylation is clearly altered in the above disorders stemming from mutations, inappropriate methylation, or repeated modulation of the microenvironment, the role of DNA methylation in most psychiatric disorders is less clear. Yet, there is mounting evidence that altered patterns of DNA methylation are associated with many psychiatric disorders. For example, early-life stress in the form of maternal neglect was sufficient to alter DNA methylation in the brain of a rodent model (Weaver et al, 2004). Maternal neglect increased methylation within the promoter of the glucocorticoid receptor, thus reducing its expression. Surprisingly, this alteration in the DNA methylation pattern was retained into adulthood, leading to a heightened stress response. Similarly, in humans, childhood abuse results in increased methylation of the promoter for the glucocorticoid receptor and a decrease in its expression, recapitulating the rodent model (McGowan et al, 2009). Furthermore, altered patterns of DNA methylation are observed in psychiatric patients diagnosed with schizophrenia and bipolar disorder (Mill et al, 2008).
DNA methylation varies not only between tissues but also between brain regions, between gray matter and white matter, and possibly even between cells (Ladd-Acosta et al, 2007; Ghosh et al, 2010). Although current technology limits our ability to distinguish cell-specific methylation patterns, the advent of next-generation DNA sequencing has provided powerful tools to examine the genome-wide DNA methylation pattern with single-nucleotide resolution (Meissner et al, 2008; Lister et al, 2009; Popp et al, 2010). As technology improves, the cost of performing sequencing analysis will decline, thus making the technology more accessible. Recent technical developments have allowed for genome-wide DNA methylation analysis to be performed even with a sample amount as low as 150ng (Popp et al, 2010). Aberrant DNA methylation patterns are observed in a wide variety of psychiatric and neurological illnesses. With declining costs and the ability to perform genome-wide methylation analysis on limited tissue quantities, it will soon be possible to map-out genome-wide DNA methylation patterns from distinct brain regions from patients with neurological and psychiatric disorders. The analysis of neural tissue from psychiatric patients will lead to new insights into the etiology of psychiatric illness and open up new avenues of drug discovery and targeted therapies.
Although current protocols enable scientists to precisely quantify DNA methylation at single-nucleotide resolution using progressively smaller tissue quantities, many of the most commonly used methods for profiling and quantification of DNA methylation, such as bisulfite sequencing and methylation-sensitive enzyme-based assays, are unable to distinguish between 5hmC and 5mC (Tahiliani et al, 2009; Huang et al, 2010). A few protocols are capable of distinguishing 5hmC from 5mC in the genome: CpG end-labeling followed by thin-layer chromatography (Tahiliani et al, 2009) and high-performance liquid chromatography (HPLC) with either UV detection (Liutkeviciute et al, 2009) or tandem mass spectrometry (Globisch et al, 2010; Le et al, 2011). Hydroxymethylated DNA can be enriched using antibodies that bind specifically to 5hmC or by biotinylation of modified 5hmC and precipitated sequences can be identified using microarray chips or by DNA sequencing (Szwagierczak et al, 2010; Ficz et al, 2011; Jin et al, 2011; Pastor et al, 2011; Wu and Zhang, 2011). Although these methods can quantify 5hmC and identify DNA sequences with which it is associated, single base-pair resolution has not been attained. In order to clarify the genomic distribution and the epigenetic role of 5hmC in the brain, a locus-specific method of identifying 5hmC will need to be developed.
As other high-throughput techniques, including RNA and chromatin immunoprecipitation (ChIP) sequencing, become more accessible to researchers, there is a growing need to integrate high-throughput data. Currently, DNA methylation, histone modification, and miRNA are studied in relative isolation. In order to fully understand how gene expression is regulated within the nervous system, future research must consider the epigenome as a whole. By dissecting the biological mechanisms that mediate crosstalk among these biological mechanisms and integrating high-throughput data, we can begin to study the epigenome as a whole. Finally, for a complete understanding of how the epigenome regulates gene expression, future research will have to uncover the biological mechanisms that mediate activity-dependent changes in the epigenomic landscape of the mammalian brain.
Read the rest here:
DNA Methylation and Its Basic Function
Posted in DNA
Comments Off on DNA Methylation and Its Basic Function
DNA in 1995 attempted abduction case could give answers to Morgan Nick kidnapping – KARK
Posted: at 5:02 am
*Editorial note: Morgans mother, Colleen Nick, was made aware of our findings prior to the publication and broadcast of this story.
VAN BUREN, Ark. Page by page, a case file is put together.
This is the property and evidence found that we took, retired Van Buren Police Detective Kevin Johnson recalled. These are my notes right here.
It was August of 1995 when Johnson got a call to investigate an attempted kidnapping on South Fifth Street. An 11-year-old girl with her brothers was getting a soda and french fries. Records show a drunk man, driving a red Chevy pickup truck pulled up and started making sexual comments to her.
The case file reveals the man started waving money around like a fan. He told the 11-year-old that she could have the money if she would go to his house. Officers report the girl started to run and scream. The truck took off and swiped a utility pole along the way.
Johnson arrived on scene with heightened senses because a town over, investigators were entering their third month into the search for Morgan Nick.
Your senses are more heightened in whats going on right now. Those dont happen. Theyre very rare, Johnson said.
A key witness reported getting the trucks license plate. They told officers it was WJA 385, but that plate didnt hit. Officers then transposed the numbers to WJA 835, which was a hit. The license plate search returned to a red truck, registered down the road to Billy Jack Lincks.
Johnson knew he had the suspect.
Once we went to his place of residence [and] found the pickup, found the mark on his truck, he was intoxicated, Johnson recalled. Yeah, we knew pretty quick.
Lincks was interviewed by Johnson at Van Buren Police and then charged and convicted of sexual solicitation of a child.
When asked if Morgan Nick ever came up in the search, Johnson noted that his agency reached out to state and federal authorities.
Thats the very reason we did turn that over to state police and FBI, he said.
Johnsons 1995 memo reveals Lincks was released to the Arkansas State Police and the FBI for further interviews.
We know what that interview was. That was the Morgan Nick interview. My interview was over, he said.
More than a week later and with a search warrant in hand, records reveal the ASP searched Lincks red pickup truck.
I think [it was] specific for Morgan Nick, Johnson said.
Documents show, investigators found hair fibers on the seat and floorboard, duct tape, a tarp, rope, machete and blood in one of the seats.
Any sort of DNA or blood evidence is always one of the most important aspects of any investigation, former prosecutor and partner at Kamps & Ward Law Firm Kelly Ward explained. He lived about 13 minutes from where Morgan disappeared.
DNA technology started coming online in Arkansas in 1995, about the same time the Crime Lab confirmed blood on Lincks truck seat.
A state laboratory document shows the blood was retained for consideration of possible future analysis. So where is the blood, and could it offer any insight into Morgans case?
ASP spokesman Bill Sadler provided a written statement that the sample collected from Lincks truck was last known to be potentially used in a separate criminal investigation.
On September 20, 1995, twenty-two days after the investigation began, an Arkansas State Police CID investigator discussed with crime laboratory personnel the possibility of submitting one particular evidence sample from the Van Buren case, along with other evidence, to the FBI Laboratory for analysis in connection with another case, Sadler wrote.
There was no mention of Morgan Nick, though.
At this juncture, it would be inappropriate for the Arkansas State Police to offer any further narrative regarding the evidence as it may relate to the case presently under the jurisdiction of another law enforcement agency, Sadler said.
Sources said the blood sample is not at the states Crime Lab. Alma Police Chief Jeff Pointer confirms the blood evidence is not at his department, either.
So, could it be with the feds? FBI Little Rock only had two words: No comment.
You need to analyze that DNA. You need to compare that DNA to a relative of Morgan Nick, Ward said. If it was a match, what it would tell us is, Morgan Nick was in that truck at one time.
Johnson said he did not collect the blood or hair fibers from Lincks truck. In 1995, Arkansas law did not require agencies to keep evidence from some cases, such as attempted kidnappings. Were told that changed in 2011.
I wish we could go back and look at all the evidence and take it now underneath the microscope that we have now in 2022, Johnson said.
The former detective recalled Lincks being cooperative with him, but documents show that when ASP and the FBI started their interview with Lincks he lawyered up.
Detectives went home-to-home in 1995 to speak with Lincks neighbors. One neighbor told an investigator that he thought Lincks had a camper shell on the red pickup truck about two months prior. Last year, Alma Police released a photo of a red pickup truck with a camper shell on it. Officers believe the driver is Morgans abductor.
Johnson said some characteristics of the truck and of Lincks may fit with what happened in Alma, but he still has a hard time wrapping his mind around it.
I did not think he was the type that would do that, but that was me personally, he said. I had dealt with Mr. Lincks since 1983. He had been arrested several times for DWI.
The former detective said one day the right pieces of the puzzle will come together and the case will eventually be solved.
Someone will say the right thing at the right time, he said. The FBI, I know they have my field notes from the Morgan Nick case.
At this moment, no one has confirmed where the blood evidence is.
Records show in 1992, Lincks was placed on a suspended sentence for sexual abuse of a young girl. Lincks died in 2000.
Last November, the FBI Little Rock named Lincks a person of interest in Morgans abduction. Federal authorities asked anyone who knew him to call the FBI.
Mr. Lincks was born and raised in Crawford County, Arkansas. He served with the U.S. Army during World War II and then worked at Braniff Airlines in Dallas, Texas, from 1962 to 1974. He returned to Van Buren, Arkansas, sometime in the late 1970s, the FBI announced at the time.
If you have any information about Lincks, you are asked to call the FBI at: 1-800-CALL-FBI.
Every piece of information about Lincks life is important-no detail is too small or insignificant, said FBI agents.
Have a story you need Working 4 You to investigate? Call (501) 340-4448.
Originally posted here:
DNA in 1995 attempted abduction case could give answers to Morgan Nick kidnapping - KARK
Posted in DNA
Comments Off on DNA in 1995 attempted abduction case could give answers to Morgan Nick kidnapping – KARK
Mawi DNA Technologies Awarded Two ISO Certifications – Business Wire
Posted: at 5:02 am
HAYWARD, Calif.--(BUSINESS WIRE)--Mawi DNA Technologies (Mawi), a leading biosampling collection company, announces it has been awarded the ISO 9001:2015 Quality Management Systems and ISO 13485:2016 Medical Devices Quality Management Systems certifications. Mawi developed and commercialized the iSWAB technology, an innovative system for collection of biosamples.
This internationally recognized standard signifies that Mawi has developed, maintains and continuously improves its business processes. This is accomplished by measuring and comparing business performance to appropriate quality standards. The ISO certificate helps to improve business credibility and authority, as well as the overall efficiency of the business.
The ISO certification reinforces our quality structure as a company and as a supplier for our valued customers, said Hanzel Lawas, Mawis QMS and Operations Manager. He added that Mawi celebrates this certification as it supports many COVID-19 projects by maintaining an uninterrupted supply of 100% USA-made innovative sample collection technologies.
Our company has and will continue to use the ISO standard in all processes and in the design and development of new products, Jerome David, Mawis VP of Sales and Marketing, added. The ISO certification ensures that our company meets the Federal Food and Drug Administrations requirements and is the first major step to complying with European (EU) regulatory standards for obtaining CE Mark certification, which we expect to have by Q2 2022. Many products require CE marking before they can be sold in the EU.
Mawis mission is to uphold sample integrity at room temperature from anywhere in the world enabling true sample diversity across any geography or population segment. At Mawi, The Future of Biosampling is Here. For more information, visit http://www.mawidna.com.
See original here:
Mawi DNA Technologies Awarded Two ISO Certifications - Business Wire
Posted in DNA
Comments Off on Mawi DNA Technologies Awarded Two ISO Certifications – Business Wire
GoFundMe’s Sordid History of Censorship of Conservative Causes – Daily Signal
Posted: February 11, 2022 at 7:10 am
The popular crowdfunding site GoFundMe has taken down yet another conservative donation campaign.
As donations for the Freedom Convoy trucker protest in Ottawa, Ontario, reached $10 million on Feb. 4, GoFundMe pulled the plug on it and took the page down, alleging violence and unlawful activity as an excuse.
To add insult to injury, the company wasnt originally planning on returning the donated funds from the canceled page unless asked. Instead, GoFundMe claimed it intended to distribute the money to charities the truckers selected. After massive public pushbackespecially from a number of states attorneys generalthe platform relented and said it would automatically return the funds.
GoFundMes excuse for closing the donations page down doesnt pass the smell test, because it has a long history of deplatforming conservative causes while conspicuously leaving leftist crowdfunding efforts alone.
Consider a case from back in November 2020, when the site canceled a campaign to erect a billboard in California advertising a book critical of puberty blockers for kids. Local parents tried to get a sign erected that would have read Puberty is not a medical condition, along with a photo of the book Irreversible Damage: The Transgender Craze Seducing Our Daughters by Abigail Shrier.
GoFundMe offered no explanation beyond claiming the campaign went against the sites user rules.
In March of last year, it censored another cause for purely political reasons: A womens rape crisis center in Vancouver, British Columbia, had crowdfunded $7,000 before GoFundMe killed the page. The page was started by a short-lived internet community called Super Straight that wanted to support the one rape crisis center in the city explicitly for biological women. That ran afoul of GoFundMes liberal sensibilities regarding transgender people, so the page had to go.
While GoFundMe has ramped up its efforts against conservatives in recent years, the practice tracks back to the sites early days.
In April 2015, The Daily Signal reported a pair of stories about bakers Aaron and Melissa Klein and florist Barronelle Stutzman, who had their GoFundMe campaigns stripped from the platform after they refused to provide service for gay weddings. The Kleins story is notable insofar as it ushered in a change in policy directly responsible for todays problems.
Prior to the Kleins, GoFundMes terms of service prohibited campaigns in defense of formal charges of heinous crimes, including violent, hateful, or sexual acts. After they removed the Kleins page, the terms were updated to include discriminatory acts.
To the leftists in charge at GoFundMe, anything contrary to their narrative can be construed as hateful or discriminatory, and therefore, ripe for takedown.
GoFundMes bias in deciding what stays and what goes on its platform becomes even more evident when one looks at the types of violent leftist content thats allowed to remain.
Numerous funding campaigns for violent Antifa and Black Lives Matter rioters have been allowed to remain up on the site while conservatives who have done nothing wrong except go against leftist dogma are frequently deleted.
The double standard employed by platforms like GoFundMe over what is acceptable is dangerous. American democracy cannot survive a system where only one political ideology can support causes it likes.
Thankfully, however, there are burgeoning alternatives. As of this writing, the Canadian truckers have raised more than $6 million on the Christian crowdfunding site GiveSendGo. While it would be preferable if GoFundMe would drop its ideological bias and accept donations to non-leftist causes, in the meantime conservatives should financially support causes they agree with through whatever means necessary.
The left might try and stop us, but well just keep on truckin.
Have an opinion about this article? To sound off, please email[emailprotected]and well consider publishing your edited remarks in our regular We Hear You feature. Remember to include the url or headline of the article plus your name and town and/or state.
See the original post:
GoFundMe's Sordid History of Censorship of Conservative Causes - Daily Signal
Posted in Censorship
Comments Off on GoFundMe’s Sordid History of Censorship of Conservative Causes – Daily Signal
MidPoint: The Age of Censorship with Lois Lowry and Colette Bancroft – WMNF
Posted: at 7:10 am
February 9, 2022 by Rayna Kanas&Shelley Reback and filed under African American, American History, Education, LGBT, Literature, News and Public Affairs, State Government, Youth.
The banning of books and the prohibition against teaching real, accurate history used to be the plot of dystopian novels and Nazi policy in Germany in 1933, but now its part of our reality. WMNF MidPoint host Shelley Reback spoke with award-winning author Lois Lowry, author of The Giver and other books for middle readers, and Colette Bancroft, book editor of the Tampa Bay Times, on Feb. 9 about this increasing threat in our communities.
Listen to the full episode here:
As new anti-woke legislation continues to pass through the Florida legislature, Polk County decided to skip the wait and take matters into their own hands. In Polk County, 16 books have already been pulled from school shelves after a bible-based group lobbied for their removal on the grounds that they allegedly violated Florida obscenity laws by containing explicit sexual material distributed to children. The books, which include award-winners like Beloved and The Kite Runner, largely center around racism or LGBTQ+ issues. The new Stop Woke legislation pending now in Tallahassee and supported by Governor DeSantis, would not only require they be removed for making adolescents uncomfortable,but would also give parents the right to sue the school system and teachers for making them available.
Reback questioned Lowry about writing books for young people about difficult subjects that may make them uncomfortable. Thats the purpose of many of the books kids read today, Lowry told WMNF. Growing up is a lonely existence The thing that alleviates that loneliness is the companionship of a book.
Since its publication in 1993, Lowrys Newbery-Award-winning novel The Giver has- ironically- been both widely assigned in middle schools and is often the subject of censorship. The novel takes place in a dystopian society that celebrates Sameness and which has censored all sources of negativity: history, memories, color and even human emotion have all been suppressed. The plot centers around the quest of the only two citizens who retain memories of the past to return their society to one that accepts the full reality of existence: its not always positive, but with knowledge and feelings it is richer and deeper and makes life worth living. Reback noted that many of these themes in The Giver are at issue in Florida in 2022 as the Legislature debates these new censorship and surveillance bills.
Bancroft noted that these calls for censorship are not new, though the recent influx of censorship legislation could be prompted by an increase in diversity among content producers. As more minority groups find their voice and speak up, the white-majority lens of history is shed, but the white Republican majority that controls our politics may feel threatened.
The reality of history and its effects on the current world is uncomfortable. Learning about it is undeniably difficult. But Reback, Lowry and Bancroft agreed on the philosophy proposed by George Santayana:
Those who cannot remember the past are condemned to repeat it.
Tags: books, censorship, Florida Legislature, literature, MidPoint, wmnf, WMNF News
See original here:
MidPoint: The Age of Censorship with Lois Lowry and Colette Bancroft - WMNF
Posted in Censorship
Comments Off on MidPoint: The Age of Censorship with Lois Lowry and Colette Bancroft – WMNF