First mice engineered to survive COVID-19 like young, healthy … – Science Daily

Researchers have genetically engineered the first mice that get a human-like form of COVID-19, according to a study published online November 1in Nature.

Led by researchers from NYU Grossman School of Medicine, the new work created lab mice with human genetic material for ACE2 -- a protein snagged by the pandemic virus so it can attach to human cells as part of the infection. The mice with this genetic change developed symptoms similar to young humans infected with the virus causing COVID-19, instead of dying upon infection as had occurred with prior mouse models.

"That these mice survive creates the first animal model that mimics the form of COVID-19 seen in most people -- down to the immune system cells activated and comparable symptoms," said senior study author Jef Boeke, the Sol and Judith Bergstein Director of the Institute for Systems Genetics at NYU Langone Health. "This has been a major missing piece in efforts to develop new drugs against this virus."

"Given that mice have been the lead genetic model for decades," added Boeke, "there are thousands of existing mouse lines that can now be crossbred with our humanized ACE2 mice to study how the body reacts differently to the virus in patients with diabetes or obesity, or as people age."

Problem of Large DNA

The new study revolves around a new method to edit DNA, the 3 billion "letters" of the genetic code that serve as instructions for building our cells and bodies.

While famous techniques like CRISPR enable the editing of DNA editing just one or a few letters at a time, some challenges require changes throughout genes that can be up to 2 million letters long. In such cases, it may be more efficient to build DNA from scratch, with far-flung changes made in large swaths of code pre-assembled and then swapped into a cell in place of its natural counterpart. Because human genes are so complex, Boeke's lab first developed its "genome writing" approach in yeast, one-celled fungi that share many features with human cells but that are simpler and easier to study.

More recently, Boeke's team adapted their yeast techniques to the mammalian genetic code, which is made up of not just of genes that encode proteins, but also of many switches that turn on different genes at different levels in different cell types. By studying this poorly understood "dark matter" that regulates genes, the research team was able to design living mice with cells that had more human-like levels of ACE gene activity for the first time. The study authors used yeast cells to assemble DNA sequences of up to 200,000 letters in a single step, and then delivered these "naked" DNAs into mouse embryonic stem cells using their new delivery method, mSwAP-In.

Overcoming the size limits of past methods, mSwAP-In delivered a humanized mouse model of COVID-19 pathology by "overwriting" 72 kilobases (kb) of mouse Ace2 code with 180 kb of the human ACE2 gene and its regulatory DNA.

To accomplish this cross-species swap, the study method cut into a key spot in the DNA code around the natural gene, swapped in a synthetic counterpart in steps, and with each addition, added a quality control mechanism so that only cells with the synthetic gene survived. The research team then worked with Sang Yong Kim at NYU's Rodent Genome Engineering Lab using a stem cell technique called "tetraploid complementation" to create a living mouse whose cells included the overwritten genes.

In addition, the researchers had previously designed a synthetic version of the gene Trp53, the mouse version of the human gene TP53, and swapped it into mouse cells. The protein encoded by this gene coordinates the cell's response to damaged DNA, and can even instruct cells containing it to die to prevent the build-up of cancerous cells. When this "guardian of the genome" itself becomes faulty, it is a major contributor to human cancers.

Whereas the ACE2 experiments had swapped in an unchanged version of a human gene, the synthetic, swapped-in Trp53 gene had been designed to no longer include a combination of molecular code letters -- cytosine (C) next to guanine (G) -- known to be vulnerable to random, cancer-causing changes. The researchers overwrote key CG "hotspots" with code containing a different DNA letter in adenine (A).

"The AG switch left the gene's function intact, but lessened its vulnerability to mutation, with the swap predicted to lead to a 10-to-50 fold lower mutation rate," said first author Weimin Zhang, PhD, a post-doctoral scholar in Boeke's lab. "Our goal is to demonstrate in a living test animal that this swap leads to fewer mutations and fewer resulting tumors, and those experiments are being planned."

The work was funded by National Institutes of Health CEGS grant 1RM1HG009491 and Perlmutter Cancer Center Support Grant P30CA016087. Boeke is a founder of CDI Labs, Inc., a founder of Neochromosome, Inc.; a founder of ReOpen Diagnostics, LLC, and serves or has served on the scientific advisory boards of Logomix Inc., Modern Meadow, Inc., Rome Therapeutics, Inc., Sample6, Inc., Sangamo, Inc., Tessera Therapeutics, Inc. and the Wyss Institute. Boeke also receives consulting fees and royalties from OpenTrons, and holds equity in the company. These relationships are managed in accordance with the policies of NYU.

Follow this link:
First mice engineered to survive COVID-19 like young, healthy ... - Science Daily

First Mice Engineered to Survive COVID-19 Are Like Young, Healthy … – NYU Langone Health

Researchers have genetically engineered the first mice that get a humanlike form of COVID-19, according to a study published online November 1 in Nature.

Led by researchers from NYU Grossman School of Medicine, the new work created lab mice with human genetic material for ACE2a protein snagged by the pandemic virus so that it can attach to human cells as part of the infection. The mice with this genetic change developed symptoms similar to those of young humans infected with the virus causing COVID-19, instead of dying upon infection, as had occurred with prior mouse models.

That these mice survive creates the first animal model that mimics the form of COVID-19 seen in most peopledown to the immune system cells activated and comparable symptoms, said senior study author Jef D. Boeke, PhD, the Sol and Judith Bergstein Director of the Institute for Systems Genetics at NYU Langone Health. This has been a major missing piece in efforts to develop new drugs against this virus.

Given that mice have been the lead genetic model for decades, added Dr. Boeke, there are thousands of existing mouse lines that can now be crossbred with our humanized ACE2 mice to study how the body reacts differently to the virus in patients with diabetes or obesity, or as people age.

The new study revolves around a new method to edit DNA, the 3 billion letters of the genetic code that serve as instructions for building our cells and bodies.

While famous techniques like CRISPR enable editing DNA just one or a few letters at a time, some challenges require changes throughout genes that can be up to 2 million letters long. In such cases, it may be more efficient to build DNA from scratch, with far-flung changes made in large swaths of code preassembled and then swapped into a cell in place of its natural counterpart. Because human genes are so complex, Dr. Boekes lab first developed its genome writing approach in yeast, one-celled fungi that share many features with human cells but that are simpler and easier to study.

More recently, Dr. Boekes team adapted their yeast techniques to the mammalian genetic code, which is made up not only of genes that encode proteins but also of many switches that turn on different genes at different levels in different cell types. By studying this poorly understood dark matter that regulates genes, the research team was able to design for the first time living mice with cells that had more humanlike levels of ACE gene activity. The study authors used yeast cells to assemble DNA sequences of up to 200,000 letters in a single step, and then delivered these naked DNAs into mouse embryonic stem cells using their new delivery method, mSwAP-In.

Overcoming the size limits of past methods, mSwAP-In delivered a humanized mouse model of COVID-19 pathology by overwriting 72 kilobases (kb) of mouse Ace2 code with 180 kb of the human ACE2 gene and its regulatory DNA. To accomplish this cross-species swap, the study method cut into a key spot in the DNA code around the natural gene, swapped in a synthetic counterpart in steps, and with each addition, added a quality control mechanism so that only cells with the synthetic gene survived. The research team then worked with Sang Y. Kim, PhD, at NYU Langones Rodent Genetic Engineering Laboratory, using a stem cell technique called tetraploid complementation to create a living mouse whose cells included the overwritten genes.

In addition, the researchers had previously designed a synthetic version of the gene Trp53, the mouse version of the human gene TP53, and swapped it into mouse cells. The protein encoded by this gene coordinates the cells response to damaged DNA, and it can even instruct cells containing it to die to prevent the buildup of cancerous cells. When this guardian of the genome itself becomes faulty, it turns into a major contributor to human cancers.

Whereas the ACE2 experiments had swapped in an unchanged version of a human gene, the synthetic, swapped-in Trp53 gene had been designed to no longer include a combination of molecular code letterscytosine (C) next to guanine (G)known to be vulnerable to random cancer-causing changes. The researchers overwrote key CG hot spots with code containing a different DNA letter, adenine (A).

The AG switch left the genes function intact, but lessened its vulnerability to mutation, with the swap predicted to lead to a ten- to fiftyfold lower mutation rate, said first author Weimin Zhang, PhD, a postdoctoral scholar in Dr. Boekes lab. Our goal is to demonstrate in a living test animal that this swap leads to fewer mutations and fewer resulting tumors, and those experiments are being planned.

Along with Dr. Boeke and Dr. Zhang, NYU Langone study authors were Ran Brosh, PhD; Aleksandra Wudzinska, MPhil; Yinan Zhu; Noor Chalhoub; Emily Huang; and Hannah Ashe in the Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology; Ilona Golynker; Luca Carrau, PhD; Payal Damani-Yokota, PhD; Camille Khairallah, PhD; Kamal M. Khanna, PhD; and Benjamin tenOever, PhD; in the Department of Microbiology; and Matthew T. Maurano and Dr. Kim in the Department of Pathology.

The work was funded by National Institutes of Health CEGS grant 1RM1HG009491 and Perlmutter Cancer Center Support Grant P30CA016087. Dr. Boeke is a founder of CDI Labs Inc., a founder of Neochromosome Inc., a founder of ReOpen Diagnostics LLC, and serves or has served on the scientific advisory boards of Logomix Inc., Modern Meadow, Rome Therapeutics, Sample6, Sangamo Therapeutics, Tessera Therapeutics, and the Wyss Institute. Dr. Boeke also receives consulting fees and royalties from Opentrons and holds equity in the company. These relationships are managed in accordance with the policies of NYU Langone Health.

Greg Williams Phone: 212-404-3500 Gregory.Williams@NYULangone.org

Read the rest here:
First Mice Engineered to Survive COVID-19 Are Like Young, Healthy ... - NYU Langone Health

A play-by-play of the FDA’s meeting on Vertex, CRISPR’s sickle cell … – BioPharma Dive

A decade ago, scientists outlined the gene editing potential of CRISPR, turning the vestiges of a bacterial immune system into one of the biotechnology industrys most powerful tools.

On Tuesday, a group of advisers to the Food and Drug Administration met to discuss the merits of what could be the first CRISPR medicine approved by the agency: a treatment for sickle cell disease from partners Vertex Pharmaceuticals and CRISPR Therapeutics.

Documents published last week show that FDA scientists are focused on the technical aspects of how CRISPR does its DNA-editing work. They seem relatively convinced the treatment, known as exa-cel, is effective.

BioPharma Dive tracked the daylong meeting and reported on the discussion here. The most recent entries are listed first.

An early wrap

About an hour before the meeting was scheduled to end, OTP Director Verdun thanked public speakers, Vertex and FDA staff as she drew the days discussions to a close.

An important part of our mission is not just evaluating efficacy, but safety, both short- and long-term, and doing what we can to evaluate both known and unknown risk of therapies, Verdun said.

She noted that the FDA will take the committees discussions and recommendations into consideration as it completes its review of exa-cel.

While the FDA doesnt have to follow the advice of its advisory panels, it often does. The agency is set to decide on exa-cels approval by Dec. 8. Gwendolyn Wu

The committee's view

Committee chair Ahsan closed the discussion by summarizing the panelists views. They agreed, she said, that Vertexs analysis of off-target risks was reasonably detailed, but indicated further study could still be useful. So then the question becomes, when have we done enough theoretical analysis to allow us to move forward? she said.

Ahsan added that, to better understand if and how off-target edits emerge, Vertex needs to continue to monitor patients. Yet she acknowledged that approach might have some limitations, however.

It would be nice to see some evaluation of monitoring the edits over real time, looking at clonal expansion, Ahsan said. But it's unsure the technology that would be used to do that, whether whole genome sequencing [or something else], would actually have the detection levels to give us meaningful information there. Jonathan Gardner

Enemy of the good

The committee has ended its cross-examination of Vertex and moved on to the broader discussion. Scot Wolfe, professor of molecular, cell, and cancer biology at the University of Massachusetts medical school, summarized Vertexs methods as pretty detailed, and quibbled only with the depth of the analysis.

We want to be careful to not let the perfect be the enemy of the good, Wolfe said. You want to do as good a job as you possibly can, but at some point, you have to try things in patients. I think in this case, there is a huge unmet need.

That was echoed by Alexis Komor, an assistant professor of chemistry and biochemistry at University of California, San Diego. Do we have the technology to sequence every single patient and do an expansive individualized on-target analysis on each one? Probably, but is that reasonable to expect from them at this point? I dont know, she said. Jonathan Gardner

Cellular sampling

FDA panelists asked a number of questions to agency reviewers and to Vertex. Joseph Wu, one of the panelists and director of the Stanford Cardiovascular Institute, noted how Vertexs cellular off-target analysis used donor cells from only three sickle cell patients, while the company has treated dozens more in the exa-cel trial.

You've had several years with these samples. Why not just do the actual analysis rather than the in silico modeling?" Wu said.

The sickle cell patient samples Vertex used were among 14 donor cell samples that the company has tested, and there's no data to suggest that the result would be different for patients with sickle cell than the other possibilities, Altshuler responded.

Whether we were to do the testing, Altshuler said, we'd end up at the same place, I believe, which is the risk assessment.

Wus line of questioning, as well as the concerns raised by Singh in her analysis, return to a similar theme: how much off-target analysis is enough?

I suspect if there were hard guidelines we wouldn't be having this meeting, committee chair Ahsan said. Gwendolyn Wu

The regulator's view

FDA staff just finished outlining the agencys view of exa-cel. The first presentation, by reviewer Karl Kasamon, gave a clinical overview of the therapy and largely lined up with Vertexs assessment of its efficacy. The second, by bioinformatics expert Komudi Singh, was a detailed review of the days main issue: off-target CRISPR editing.

Singh described how off-target edits, while potentially inconsequential, can be harmful if they occur in regions of the genome that have regulatory functions or code for a protein. There are a number of tools available to assess this likelihood and Vertex took several approaches, including using computer algorithms to identify possible off-target sites and sequencing donor cells that were edited.

Singh raised a number of concerns with Vertexs methods. For example, the database used by the company contains only a small amount of sequencing data from the intended population for exa-cel treatment. It also may not be representative of genetic variants that present higher risk of off-target edits. Vertexs cellular analysis, meanwhile, used a small sample of donor cells.

Singh ended by asking the committee to provide recommendations, setting up the days concluding discussion on whether other studies are needed to gauge exa-cels safety. Ned Pagliarulo

Victoria and Jimi

The open public hearing began with Victoria Gray, a 38-year-old woman who was the first patient to be treated with exa-cel. She spoke of sickle cells pain, which she compared to being simultaneously hit by a truck and lightning, and the diseases effects on her life and family.

She met with physician Haydar Frangoul, who offered her a spot in the exa-cel trial. I said yes without hesitation, knowing that I would be the first person but this was my opportunity to fight, Gray recounted.

She no longer has pain crises after receiving exa-cel, nor does she need blood transfusions. I now work full time and I contribute to my household and my community.

Her experience was similar to that of Jimi Olaghere, who participated in the exa-cel study about three years ago.

Gene therapy has given me the ability to take full control of my life, said Olaghere. In a world where the deck was stacked against me, gene therapy has been a winning hand. While I recognize gene editing wont be the solution for everyone I strongly recommend [sickle cell] warriors to consider this one-time therapy. Ned Pagliarulo

Lunch break

The panel is now on break until 12:35 p.m., when we'll hear from patients and advocates during the open public hearing. Ned Pagliarulo

Like a hammer hitting a wall

The first two patients who entered the exa-cel trial have seen notable improvements since they were treated, said Haydar Frangoul, hematology and oncology medical director at Tristar Centennial Medical Group in Nashville and lead investigator of the exa-cel studies.

One, a 33-year-old woman who couldn't walk or even pull up a spoon to feed herself during pain crises, has been a
ble to take up full-time work and become more active in caring for four children since receiving the therapy. Another, a 13-year-old girl who was hospitalized several times each year because of pain crises, can attend school regularly because she no longer needs as much medical care.

Frangoul also urged exa-cels use early in the disease course because of the cumulative effects of the disease on organs and joints.

Sickle cell disease is like a hammer hitting a wall, he said. I can take away the hammer. But we cannot reverse the damage. We cannot fix the wall. Jonathan Gardner

Safety follow-up

To track exa-cel safety over the long term, Vertex plans to rely on two 15-year studies, including a registry-based study that would begin post-approval and involve sickle cell patients treated with exa-cel. The other is follow-up for an ongoing clinical trial.

The Center for International Blood and Marrow Transplant Research, which has for years collected data on patients receiving cell therapies, will also assess the long-term safety of exa-cel, said Christopher Simard, Vertex's vice president of global patient safety. All planned U.S. exa-cel treatment centers will provide data to the center, he said. Gwendolyn Wu

Visualizing exa-cels benefit

William Hobbs, Vertexs head of hematology clinical development, outlined the clinical trial data showing exa-cels effectiveness. The results are clear: Treatment eliminates, for at least multiple years, the debilitating pain crises that people with sickle cell can experience and keeps them out of the hospital. Ned Pagliarulo

The need for treatment

There are a few drugs currently available to help manage sickle cell symptoms. But they dont fix the condition and dont work for everyone.

A bone marrow transplant of donor stem cells can cure the disease, but is available for less than 20% of people with sickle cell, said Alexis Thompson, division chief of hematology at Childrens Hospital of Philadelphia, who spoke at Tuesdays meeting on behalf of Vertex.

An estimated 100,000 people live with sickle cell in the U.S., and about one-fifth have a severe form of the disease, with recurring symptoms such as severe pain flare-ups, a lung condition known as acute chest syndrome, priapism and splenic sequestration. The resulting organ damage raises the risk of death.

Sickle cell also occurs at "disproportionately high rates" among people of color, particularly people of African ancestry, Thompson said.

People with sickle cell often live in low income areas and communities with high unmet medical need, further adding to substantial healthcare disparities, she said. Gwendolyn Wu

How much testing is enough?

The vastness of the human genome presents a thorny problem for assessing the risk of off-target CRISPR edits. Scientists trying to vet the risk have to focus their analysis, or risk being swamped by a deluge of data that might not actually help suss out potential problems.

Urnov, answering a question from the panelist and National Institutes of Health branch chief John Tisdale, said theres a limit to how much reassurance preclinical editing analyses can provide.

The technology is in fact ready for prime time, said Urnov. We're kind of reaching asymptotic places in terms of how we can de-risk it non-clinically. I don't know what else to do at this point in terms of understanding the benefit-risk.

One advantage to studying blood diseases, Bauer noted, is that samples can be easily obtained, tested and tracked over time.

Taby Ahsan, the committees chair, said Urnovs and Bauers answers set the stage for the rest of the days discussion: When is enough theoretical data sufficient to support a patient specific risk assessment? And where are we in that curve of risk mitigation? Ned Pagliarulo

Risk uncertainty

After deeply technical presentations by Urnov and Daniel Bauer about the risks of off-target edits, committee member Lisa Lee, a bioethics expert from Virginia Tech University, brought it back to the patient level by asking a simple question: If you were talking to a family about this kind of treatment, how would you characterize the consequences of off target edits?

Bauer acknowledged the uncertainty of the risk by noting how most of whats in the human genome doesnt code for any specific function, meaning an off-target edit might have no effect on patients.

The only way to know that is through careful follow up, he said. My guess is it's a relatively small risk in the scheme of the risk-benefit. And that's one of the goals, I would say, of doing this under very careful circumstances is to try to learn what that risk is so that we can continually improve those therapies. Jonathan Gardner

"A whole new world"

Fyodor Urnov is a leading expert on CRISPR gene editing and director of technology and translation at UC Berkeleys Innovative Genomics Institute. His task today: Condense the history of CRISPR gene editing research to a 20-minute presentation.

He starts by noting that, while CRISPR is still relatively new, it builds on decades of research into gene editing by other means.

I need to frame the state of our field of gene editing today by stepping 20 years back, Urnov said. So at the time, the sole method for targeted genetic engineering in human cells was an approach called gene targeting. It was inefficient and toxic, he added, ruling out therapeutic applications.

The discovery of CRISPR has brought about an exponential scale-up in gene editing research and its potential applications a shift Urnov documented with the stylized chart below.

Here we are in 2023, and we are proverbially in a whole new world. There are 27,000 references to the word cas9 in PubMed. Genome editing with cas9 and other tools has been shown to work in every basic and applied research setting where it can be tried, as well as in clinical trials of blood stem cells, T cells, the liver and the eye. Ned Pagliarulo

FDA lays out meeting purpose

OTP director Verdun kicked off the meeting by emphasizing the FDA convened the meeting to specifically evaluate the risk of off-target edits. Her comments again signaled that agency scientists are comfortable exa-cel helps patients and that its safety profile is otherwise acceptable.

Verdun also added a personal note about her experience with sickle-cell patients.

I've had the pleasure of taking care of several sickle cell patients and admire the courageous and resilient patient community, she said. I'm also reminded of the sickle cell disease patient-focused drug development program at FDA in which we heard directly from patients and their caregivers which highlighted the significant unmet need. Jonathan Gardner

Today's schedule

After some housekeeping, advisers will hear first from Nicole Verdun, the FDAs new head of the Office of Therapeutics Products, which oversees gene therapies like exa-cel.

The meeting includes two guest speaker presentations, from gene editing experts Fyodor Urnov, of the University of California, Berkeley, and Daniel Bauer, of Boston Childrens Hospital.

After a short break, Vertex and CRISPR Therapeutics will then walk through their data, followed by an open public hearing. The FDA is up in the afternoon and committee discussion is scheduled from 3:00 p.m. to 4:50 p.m Ned Pagliarulo

Why is this meeting important?

The advisory committee meeting is one of the final steps in
the FDAs review of exa-cel, which the agency expects to complete by Dec. 8. These meetings offer a rare window into the agencys thinking midway through an approval review, as well as a chance to publicly vet companies data.

In this case, the meeting will also serve as a forum for discussing CRISPR gene editing, which has become an important biomedical tool used by a growing number of biotechnology companies. The days agenda shows that advisers will hear from other experts about CRISPRs merits and risks, making Tuesdays meeting a mini-summit on the technology.

A positive review by the panel would up the chances that the regulator grants the therapy a historic approval, as well as boost other gene editing companies like Intellia Therapeutics. Ned Pagliarulo

How does exa-cel work?

Essentially, exa-cel combines CRISPR gene editing with bone marrow transplantation.

To construct the treatment, a patients stem cells are collected from their blood and sent to a manufacturing facility where they are edited using CRISPR/cas9. The DNA snip is made to a specific part of a gene called BCL11A that controls production of a protein known as fetal hemoglobin, or HbF. The edited cells are frozen and shipped back to the treating hospital, where, after a preparatory chemotherapy regimen, they are infused into the patient.

Churning out fetal hemoglobin, the new cells effectively mute the most prominent symptoms of sickle cell, which is caused by damaged adult hemoglobin warping red blood cells into a crescent.

Vertex and CRISPR Therapeutics are seeking approval of exa-cel in people aged 12 years and older who have sickle cell and experience the diseases characteristic pain crises. Ned Pagliarulo

What is the FDA thinking?

Documents published last week showed FDA staff to be laser focused on the risk of off-target edits, or when CRISPR makes unintended cuts to DNA other than what its been programmed to snip. Wayward edits could disrupt cell functions or cause damage that later leads to health problems.

Since unintended edits can disrupt gene expression if present in the coding or regulatory DNA sequences, it is critical that the specificity of [exa-cels targeting component] be thoroughly screened to ensure off-target genome editing is minimized, FDA staff wrote.

Vertex and CRISPR have done several analyzes to document and predict this risk with exa-cel specifically, and claim their work shows no detectable off-target editing.

But the FDA appears concerned that their work might have missed something, and wants its advisers to discuss whether any further study is needed. In particular, they raise questions about how well Vertex and CRISPRs analyses capture the risk of off-target edits in a broad population of people with sickle cell.

On the efficacy side of the equation, however, FDA scientists seemed supportive of exa-cels potential, describing Vertex and CRISPRs data as strongly positive at one point in the documents. Ned Pagliarulo

Visit link:
A play-by-play of the FDA's meeting on Vertex, CRISPR's sickle cell ... - BioPharma Dive

Glycoengineered keratinocyte library reveals essential functions of … – Nature.com

Generation of a glycogene knock out library in HaCaT keratinocytes

HaCaT is a human keratinocyte cell line capable of forming a stratified squamous epithelium, and thus allows evaluating the infection of the skin tropic HSV-1 in both cell and organotypic tissue culture. In order to address the role of specific glycan structures in the HSV-1 infectious cycle, we used precise gene editing to target glycosyltransferases involved in the early steps of core structure synthesis, and in major elongation and branching steps of the main glycosylation pathways, including N-linked glycosylation, mucin type O-linked glycosylation, as well as GSL and GAG synthesis (Fig.1a, Supplementary Table1).

For N-linked glycans we generated MGAT1, MGAT4A, MGAT4B, MGAT5, and MGAT5+4B knock outs (KO). MGAT1 adds the first N-acetylglucosamine to the C-2 of core 3-linked mannose, and lack of this enzyme results in elimination of all N-glycan maturation steps, yielding high-mannose type N-glycans as confirmed by MS-glycoprofiling (Fig.1a, Supplementary Fig.1). MGAT4A, MGAT4B and MGAT5 are responsible for N-glycan branching, where MGAT4A or MGAT4B initiate a 4-linked antenna on the 3-linked mannose, and MGAT5 performs 6-linked branching from the core 6-linked mannose. Lack of MGAT5 results in loss of tetra-antennary N-glycans, and loss of MGAT4 isoforms also strongly diminishes the content of tetra-antennary N-glycans (Supplementary Fig.1). In addition, KO of each of the three branching enzymes resulted in increased relative abundance of hybrid type N-glycans, whereas double KO of MGAT5 and MGAT4B increased the relative abundance of biantennary glycans (Supplementary Fig.1).

For mucin type O-linked glycans, we knocked out core 1 synthase (C1GALT1), its obligate chaperone COSMC (C1GALT1C1), core 2 synthase (GCNT1), as well as the major core 1-capping glycosyltransferase ST3GAL1. Loss of C1GALT1 or COSMC eliminates the 3-linked galactose (core 1 structure), results in truncation of O-linked glycans to the initiating -GalNAc, and prevents assembly on secreted -benzyl GalNAc precursor used in CORA O-glycoprofiling (Fig.1a, Supplementary Fig.2). GCNT1 is the predominant enzyme creating the branched core 2 structure by addition of 6-linked GlcNAc to the GalNAc. Loss of GCNT1 nearly abolished all the disialylated core 2 structures, though some structures matching the composition of monosialylated core 2 could still be detected. Such structures cannot be discriminated from isobaric core 1 structures, and a minor contribution from other GCNTs to core 2 synthesis cannot be excluded either. Finally, loss of ST3GAL1 significantly reduces the 3-linked sialic acid content and results in predominantly non-capped core 1 structures (Supplementary Fig.2). We also targeted the synthesis of GSLs and GAGs. Through KO of B4GALT5 or ST3GAL5, we generated cells with the truncated GSLs, glucosylceramide (GlcCer) and lactosylceramide (LacCer), respectively (Fig.1a). Furthermore, we knocked out B4GALT7, which adds a 4-linked galactose to the initiating xylose in GAG biosynthesis, effectively truncating all classes of GAGs on membrane proteoglycans (Fig.1a). The generated keratinocyte library represents a screening platform for roles of defined cell-surface presented glycan structures in HSV-1 biology in the context of natural infection.

To define the capacity of HSV-1 to complete the infectious cycle in glycoengineered keratinocytes, we infected confluent monolayers of the KO cell lines with HSV-1, and quantified HSV-1 DNA and infectious particles in the growth media at 17h post infection (hpi) by qPCR and plaque titration, respectively. As a measure for viral replication fitness, we calculated the ratio of genome copies/infectious particles for each KO. When infecting cells with truncated O-glycans (C1GALT1C1 KO) a decrease in viral titers was detected (Fig.1b, e). In contrast, the same cells generated close to normal levels of viral DNA (Fig.1c, f), suggesting decreased fitness of virions lacking elongated O-glycans (Fig.1d, g). This feature was unique to complete truncation, and not seen when eliminating branching or sialylation of O-glycans. In cells lacking N-glycan maturation (MGAT1 KO) we also found a lower number of infectious particles (Fig.1b, e) with an apparent decreased fitness as indicated by an increase in the ratio of DNA/infectious particles (Fig.1d, g). This apparent decrease in fitness was not detected in cells with loss of N-glycan branching, and in MGAT4A KO cells we even observed an overall increased viral output (Fig.1b, c). When analysing cells with GSL synthesis defects, we found that lack of LacCer sialylation (ST3GAL5 KO) accelerated virus production (Fig.1b, c, e, f), but without any change in viral fitness (Fig.1d, 1g). Finally, loss of cellular GAGs increased the production of viral particles (Fig.1c). In conclusion, most of the tested glycogene disruptions permitted HSV-1 replication, and only disruption of N- or O-glycan maturation impaired viral fitness. We next evaluated the impact of defined glycan classes to distinct stages of the HSV-1 infectious cycle, including binding and entry, viral assembly and properties of progeny virus, and cell-to-cell spread.

HSV-1 binds and enters human keratinocytes very rapidly, with around 30% of virions bound after 20min on ice, and 80% after 2h28. Most of the bound virions enter keratinocytes within 5min after warming28. Perturbations in each of the investigated glycosylation pathways modulated early virus-host interactions (Fig.2ae). Diminished core 2 O-glycan branching resulted in increased binding also reflected in subsequent entry experiments (Fig.2b, c). Lack of complex N-glycans and reduced 4-antenna branching (MGAT1 KO and MGAT4B KO) showed reduced binding, again also reflected in the entry experiments (Fig.2ac). Interestingly, deletion of MGAT4A, another isoform catalyzing the 4-antenna synthesis on N-glycans, likely on another subset of proteins or sites in proteins29,30, selectively affected viral entry (Fig.2b, c, e). Cells displaying truncated glycolipids showed a reduction in binding to around 50% of that of WT (Fig.2a, b). A similar effect was observed in both B4GALT5 and ST3GAL5 KOs, controlling the consecutive steps in the biosynthesis of the GSL GM3. In addition, an incremental reduction in entry was observed for B4GALT5 KO cells, suggesting involvement of glycolipids in both viral binding and entry to host cells (Fig.2c, e).

HSV-1 binding (20min (a) or 120min (b) on ice) and entry (5min at 37C after 120min on ice (c)) to KO cell lines. Data is shown as WT-normalized mean+SEM of 3 independent experiments for each KO cell line. Two-way ANOVA followed by Dunnetts multiple comparison test was used on raw data to evaluate differences from WT (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001). Proportion of virus bound at 20min compared to 120min (d) or proportion of virus entered at 5min compared to virus bound at 120min (e) is shown as mean+SEM of 3 independent experiments for each KO cell line from a total of 15 experiments. One-way ANOVA followed by Dunnetts multiple comparison test was used to evaluate differences from WT (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001). f HaCaT WT and B4GALT7 KO cells were probed for HSV-1 gC binding. Enzymatic treatments were included to evaluate contributions of HS and CS GAG chains. Representative of 2 independent experiments. g A simplified overview of GAG synthe sis. h HSV-1 gC binding to a panel of CHO KO cell lines. Data is shown as WT-normalized geometrical means of 3 independent experiments for each KO cell line, and the bar heights indicate mean+SEM. One sample t test was used to evaluate differences from 1. FDR at 5% was controlled by two-stage step-up method of Benjamini, Krieger and Yekutieli (*q<0.05, **q<0.01, ***q<0.001, ****q<0.0001). i Nectin 1 and HVEM surface expression. Data points show background subtracted median fluorescence intensity (MFI) from two independent experiments, and the bar heights indicate the mean. j Percentages of total quantified CS disaccharides in HaCaT WT (Supplementary Table2). k Percentages of total quantified HS disaccharides in HaCaT WT (Supplementary Table3). Source data are provided as a Source Data file for all graphs.

Then, we analysed cells impaired in GAG biosynthesis and found an almost complete loss of binding to cells presenting only the initiating xylose on proteoglycans (B4GALT7 KO). Although we still lack a clear understanding of finer structural requirements of GAGs presented on their core proteins in the context of total cellular glycome, this fits well with the known importance of heparan sulfate (HS) in the initial attachment of HSV-1 (Fig.2a, b)26. To further dissect the importance of GAG binding determinants we investigated the binding of recombinant HSV-1 gC to our HaCaT KO cells. The use of recombinant HSV-1 gC limited the interactions to a single viral protein known to bind to synthetic GAGs in vitro, similarly to intact HSV-127,31,32. As expected, no binding was detected on B4GALT7 KO cells (Fig.2f), and to further confirm the selectivity for HS we treated HaCaT WT cells with heparinases 1, 2, and 3. Loss of HS completely abolished gC binding suggesting minimal interaction with chondroitin sulfate (CS) or dermatan sulfate (DS) presented on the cell surface (Fig.2f). Minimal changes in cell staining for bound HSV-1 gC after chondroitinase ABC treatment further supported this interpretation. Next, we analyzed a library of glycoengineered CHO cells delineating the GAG biosynthesis pathways (Fig.2g) and quantified gC binding by flow cytometry (Fig.2h)33. This library included selective elimination of HS or CS (Extl2+Extl3 KO and Csgalnact1 KO+Csgalnact2 KO+Chsy1 KO, respectively), reduction in chain polymerization of HS or CS (Ext1 KO+Ext2 KO and Chpf KO, respectively), elimination of HS N-sulfation, also effectively diminishing follow-up O-sulfation (Ndst1 KO+Ndst2 KO), as well as elimination of 4-O sulfation of CS and DS units of CS chains (Chst11 KO+Chst12 KO+Chst13 KO+Chst14 KO). In addition, we used B4galt7 KO and B3gat3 KO cells to truncate all GAGs to the initiating xylose and a short linker trisaccharide, respectively (Fig.2g). In agreement with the HaCaT cell staining data, manipulation of HS synthesis and chain length substantially decreased gC binding showing that the interaction was entirely dependent on HS sulfation and not compensated by the presence of CS (Fig.2h). Accordingly, manipulation of CS synthesis only slightly decreased gC binding independent of the predominant 4-O sulfation (Fig.2h). As expected, truncation to the linker also eliminated gC binding (Fig.2h). To our surprise, some binding was retained upon complete GAG truncation, possibly representing unspecific binding due to gross changes in the glycocalyx. In conclusion, by using cell surface presented GAGs, we were able to identify sulfated HS as the major contributor to HSV-1 gC binding and show that CS sulfation is not necessary for interaction with CS, at least in the presence of HS. More generally, the binding and entry assays show that perturbations in the cellular glycome landscape have immediate effects to early virus-cell interactions, which can be further dissected as demonstrated for the interaction between gC and HS.

To follow up on our binding and entry data, we aimed to investigate the cellular landscape of HSV-1 entry receptors and other surface molecules that may have an impact on the early virus-cell interactions in the different knock out cells. We first quantified the surface expression levels of Nectin 1 and HVEM in WT HaCaT cells and found very low levels of the latter (Fig.2i, Supplementary Fig.3a, b). MGAT1 KO and B4GALT7 KO cells expressed significantly lower levels of Nectin 1 on the cell surface, whereas MGAT4 KO and GCNT1 KO expressed higher levels (Fig.2i). These results correlate well with the virus binding data, and may help explain the altered proportion of virus bound to cells with alterations in N-glycosylation and O-glycosylation pathways. Importantly, the selective effect on entry to MGAT4 KO was not correlated to availability of the receptor.

For B4GALT7 KO, Nectin 1 presentation decreased by approximately 60%, but this does not explain the complete loss of HSV-1 binding, which is likely a combination of a decrease in GAG and protein receptors. While gC mediates early virus-GAG interactions, facilitating subsequent interactions between gD and its cognate protein entry receptors, 3-O-sulfated HS has also been identified as an independent entry receptor for gD34,35. In order to evaluate the potential contribution of 3-O-sulfated HS to HSV-1 entry in skin cells, we performed disaccharide analysis of HaCaT WT and B4GALT7 KO cells, using our recently developed method, which allows detection of 3-O-sulfated HS36 (Fig.2 j, k, Supplementary Fig.4, Supplementary Table2 and 3). Except for hyaluronan, which is synthesized by a distinct family of enzymes, we did not detect any CS or HS disaccharides in B4GALT7 KO cells (Supplementary Fig.4). HaCaT WT cells expressed high levels of 4-O-sulfated or 6-O-sulfated CS, hyaluronan, as well as N-sulfated, N-/2-O-sulfated, N-/2-O/6-O-sulfated, and non-sulfated HS. We detected very low levels of 3-O-sulfated HS disaccharides, demonstrating that usage of these receptors for HSV-1 entry in human keratinocytes is limited. We therefore suggest that Nectin 1 is the most widely available HSV-1 entry receptor for gD in HaCaT keratinocytes.

The disaccharide expression profiles in skin cells provided additional insight into the gC binding data on the CHO cell library. Namely, N-sulfated GAG motifs required for gC binding to CHO cells were abundantly found on human keratinocytes, and likely play a significant role in vivo. On the contrary, 4-O-sulfated CS, abundantly found on skin cells, is unlikely to be a critical receptor for gC, as seen from CHO data.

We next looked into GSLs expressed in skin cells (Fig.3). We saw comparable levels of Nectin 1 on the surface of WT, B4GALT5 KO and ST3GAL5 KO cells (Fig.2i), and yet HSV-1 binding and entry to these cells was markedly decreased. We thus hypothesized that elongated GSLs may help deliver the viral entry receptors to membrane compartments accessible to incoming virus. We used antibodies and toxins recognizing various (glyco)lipid structures to illuminate their distribution in keratinocytes (Fig.3a). Ceramide and glucosylceramide, representing initial steps of GSL synthesis, were predominantly located intracellularly in WT cells, while some ceramide accumulation could be seen in B4GALT5 KO, devoid of elaborate GSLs (Fig.3b). Interestingly, expression of more complex GSLs was heterogeneous, and different cells appeared committed to a specific GSL subtype. Specifically, we detected Gb3 structures, synthesized from lactosylceramide precursor, in both WT, and ST3GAL5 KO cells with clear surface presentation, but not B4GALT
5 KO (Fig.3b, e). In contrast, GM3, the product of ST3GAL5, was only detected in WT cells (Fig.3b). GM3 partially co-localized with intracellular glucosylceramide-positive structures but were primarily expressed on the cell membrane (Fig.3c). Importantly, GM3 was abundantly found on apical cell surfaces accessible to the extracellular environment (Fig.3d). Gb3 and GM3 were expressed in mostly distinct subsets of cells, and a substantial proportion of skin cells remained unlabeled, presumably expressing more elaborate structures (Fig.3e). In conclusion, we show heterogeneous yet regulated expression of different GSLs in distinct cells and within different cellular compartments, which may be relevant for interaction with extracellular virus.

a The cartoon depicts a simplified human glycosphingolipid biosynthetic pathway. Glycolipid structures highlighted in magenta were probed by antibodies or fluorescently labeled toxins. be Cells grown on cover slips were fixed with 4% PFA and stained for different GSL structures. b Confocal micrographs show distribution of different GSLs in HaCaT WT, B4GALT5 KO and ST3GAL5 KO monolayers. Representative of two independent experiments, scale bars are indicated for each set of micrographs. c z-stack maximal intensity projection of HaCaT WT cells labeled with anti-GlcCer and anti-GM3 antibodies. Representative of 2 independent experiments, scale bar is indicated. d HaCaT WT cells labeled with anti-GM3 antibody. An individual z-slice within a stack is shown, with orthogonal cross sections of the z-volume included, and indicate apical expression of GM3. Nuclei are labeled with DAPI (blue). Representative of 2 independent experiments, scale bar is indicated. e The confocal micrograph shows spatially distinct distribution of Gb3 and GM3 GSLs in HaCaT WT, probed by FITC-labeled Shiga toxin B (StxB-FITC), and anti-GM3 antibody, respectively. Representative of 2 independent experiments, scale bar is indicated.

We next investigated late stages of viral replication in KO cells with changes in protein glycosylation capacity and altered viral propagation dynamics. We probed the expression of gD and gB that promote virion envelopment. In WT most of gD signal was confined to the cell surface, partially overlapping with E-cadherin (Fig.4a), while gB primarily localized to the perinuclear compartment and secondary envelopment sites with some surface presentation, consistent with the literature (Fig.4b)37. In contrast, C1GALT1C1 KO, C1GALT1 KO, and MGAT1 KO cells exhibited a weaker and more dispersed gD immunostaining pattern with partial cytoplasmic accumulation suggesting issues with envelope glycoprotein trafficking (Fig.4a). In addition, gB exhibited poorer surface and perinuclear localization and presented in large clusters within the cells (Fig.4b). ST3GAL1 KO cells, which did not exhibit defects in viral propagation dynamics, displayed similar gB staining as WT (Fig.4b), while exhibiting stronger gD signal (Fig.4a). Overall, the results suggest that lack of core 1 O-glycans or mature N-glycans causes defects in viral particle formation due to suboptimal incorporation of viral proteins, which would fit with the observed diminished titers or loss of fitness. In addition, using an HSV-1 strain with GFP-labeled capsid protein VP26 allowed us to observe differences in the localization of viral capsids. The capsids were found in nuclear assembly compartments, outer nuclear rim, and transitioning through the cytosol in WT and ST3GAL1 KO cells. We observed lower numbers of capsid assembly sites in the nucleus and rare association with the outer nuclear rim in C1GALT1C1 KO, C1GALT1 KO and MGAT1 KO cells, with the most pronounced effect in C1GALT1C1 KO (Fig.4a, b).

a HaCaT cells grown on cover slips were infected with MOI10 of HSV-1 K26-GFP and fixed and permeabilized at 14hpi followed by co-staining for HSV-1 gD (magenta) and E-cadherin (cyan). Histograms on the left indicate intensities of gD and E-cadherin signals across the confocal images (marked with black arrowheads). Pixel overlap from the two channels is shown in white. GFP labeled capsid proteins (VP26) are seen in green. Nuclei were stained with DAPI (blue). Stainings of mock-infected cells are included. Scale bar: 10m. Images are representative of two independent experiments. b HaCaT cells grown on cover slips were infected with MOI10 of HSV-1 K26-GFP and fixed and permeabilized at 14hpi followed by staining for HSV-1 gB (magenta). GFP labeled capsid proteins (VP26) are seen in green. Nuclei were stained with DAPI (blue). Scale bar: 10m. Magnified regions of merged images are indicated with dashed white boxes. Images are representative of 2 independent experiments. c HaCaT WT and C1GALT1C1 KO cells were infected with MOI3 or MOI10 of HSV-1 K26-GFP and viral capsids imaged by live microscopy at 14 and 20hpi. Fluorescent images overlaid with bright field images are also shown. Scale bar: 10m. Images are representative of two independent experiments.

We further explored the viral replication dynamics in WT and C1GALT1C1 KO cells by live imaging of GFP-labeled HSV-1. Features seen in thin optical sections (Fig.4a, b) were also reflected in widefield images (Fig.4c). In WT cells at 14 hpi, multiple capsid assembly sites could be seen in the nucleus and capsids were also associating with the nuclear envelope in most cells irrespective of the viral load (Fig.4c). In C1GALT1C1 KO cells less and smaller assembly sites could be seen, and capsids were less frequently associating with nuclear envelope. This association slightly improved later in infection (20hpi), but the capsid production did not intensify, suggesting HSV-1 infection is generally less robust in C1GALT1C1 KO (Fig.4c).

To evaluate the contribution of viral glycans to fitness of progeny virus for early interactions with wild type host cells, we added equal numbers of infectious particles, produced in propagation experiments, to WT keratinocyte monolayers following the previously outlined strategy. No defects in binding or entry were found with virions lacking different glycan structures (Fig.5ae). In fact, virions lacking O-glycan elongation were capable of accelerated binding, despite low viral titers of HSV-1 produced in C1GALT1C1 KO or C1GALT1 KO (Fig. 5). This suggests the observed propagation defects are related to host and viral factors influencing the formation of infectious virions and not their efficiency in establishing a new infection.

Binding (20min (a) or 120min (b) on ice) and entry (5min at 37C after 120min on ice (c)) of HSV-1 produced in different KO cell lines to HaCaT WT. Data is shown as WT-normalized mean+SEM of three independent experiments for eachglycoengineered virus species. Two-way ANOVA followed by Dunnetts multiple comparison test was used on raw data to evaluate differences from WT (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001). Proportion of virus bound at 20min compared to 120min (d) or proportion of virus entered at 5min compared to virus bound at 120min (e) is shown as mean+SEM of three independent experiments for each glycoengineered virus species from a total of 14 experiments. One-way ANOVA followed by Dunnetts multiple comparison test was used to evaluate differences from WT (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001). Source data are provided as a Source Data file for all graphs.

The effect of O-glycosylation on HSV-1 glycoprotein localization, and prior knowledge of O-glycosite modifications compelled us to investigate specific O-glycosites. Eliminating site-specific O-glycosylation may have a more profound effect on protein function than truncation of the O-glycan structure5, 38,39. Therefore, although O-glycan truncation had no deleterious effects on properties of infectious virions, it should not be excluded that individual O-glycosylation sites could play a functional role.

We have previously identified more than 70 O-glycosites on eight out of the 12 HSV-1 surface proteins, including the indispensable fusion machinery comprised of gB, gD, gH, and gL15. Based on available structural data and defined molecular mechanisms, we mutated five out of the identified 21gB O-glycosites and three out of five gD O-glycosites most likely to affect fusion and receptor binding, respectively (Figs.6a, 7a)15. We generated Ser/Thr to Ala substitutions alone or in combination to test cell-cell fusion efficiency using a split luciferase reporter assay as a proxy for viral entry (Supplementary Table4, Supplementary Fig.5). The assay quantifies fusion between two cell types, one (effector) lacking HSV-1 entry receptors and transfected with plasmids encoding the conserved fusion machinery, and the other (target) presenting HSV-1 entry receptors (Fig.6b)40. Each cell type is also transfected with plasmids encoding half of a split luciferase reporter, which upon cell fusion can form a functional enzyme generating luminescence. In addition, we quantified gB and gD surface expression by CELISA40. We used CHO cells, refractory to HSV-1 entry, as effector, and HEK293, an HSV-1 permissive epithelial cell line, as target. We quantified low levels of Nectin 1 and HVEM on HEK293 cells (Supplementary Fig.3c), suggesting other types of receptors and co-receptors may also be involved.

a HSV-1 gB structure (PDB: 2GUM) with select mutated O-glycan acceptor sites indicated within the dashed box. Respective previously identified O-glycans were drawn manually as yellow squares. Domains are numbered in roman numericals according to Heldwein et al., Science 2006. b The cartoon illustrates the principle of split luciferase assay. c Cell surface expression of gB O-glycosite Thr to Ala mutants evaluated by CELISA using mouse anti-gB antibodies. Data is shown as mean absorbance at 450nm+SD of three technical replicates and is representative of three independent experiments. One-way ANOVA followed by Dunnetts multiple comparison test was used to evaluate differences from WT (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001). d, e Cell-cell fusion activity over 240min using gB O-glycosite Thr to Ala mutants. Data from two independent experiments is shown, where mean normalized luminescence of three technical replicates at each time point is indicated by a dot. Mean values of the two independent experiments are shown as thin lines. Data is normalized to maximum luminescence reading at final time point using WT gB for each experiment. d Data related to gB domain I mutations. e Data related to gB domain V mutations. f Cell-cell fusion activity of gB mutants at t=120min. Data is shown as mean normalized luminescence from two independent experiments. Two-way ANOVA followed by Dunnetts multiple comparison test was used to evaluate differences from WT (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001). g Average percentages of cell surface expression and fusion efficiency at t=120min from two independent experiments are shown in side-by-side columns. Source data are provided as a Source Data file for all graphs.

a HSV-1 gD structure (PDB: 2C36) with select mutated O-glycan acceptor sites indicated. Positions after removal of signal peptide, often encountered in the literature, are indicated in brackets. Respective previously identified O-glycans were drawn manually as yellow squares. N-terminal region omitted in the crystal structure is drawn as a dashed line. b Cell surface expression of gD O-glycosite mutants evaluated by CELISA. Data is shown as mean absorbance at 450nm +SD of three technical replicates and is representative of three independent experiments. One-way ANOVA followed by Dunnetts multiple comparison test was used to evaluate differences from WT. c Cell-cell fusion activity over 240min using gD O-glycosite mutants. Data from two independent experiments is shown, where mean normalized luminescence of three technical replicates at each time point is indicated by a dot. Mean values of the two independent experiments are shown as thin lines. Data is normalized to maximum luminescence reading at final time point using WT gD for each experiment. d Cell-cell fusion activity of gD mutants at t=120min. Data is shown as mean normalized luminescence from two independent experiments. Two-way ANOVA followed by Dunnetts multiple comparison test was used to evaluate differences from WT. CHO cells stably expressing Nectin 1 (e, f) or HVEM (g, h) were used as target cells to evaluate cell-cell fusion activity using gD O-glycosite mutants. Cell-cell fusion activity over 180min using CHO-Nectin 1 (e) or CHO-HVEM (g) as target. Parental CHO cell line without entry receptors was use for background subtraction. Data is presented as in (c). Cell-cell fusion activity of gD mutants at t=120min using CHO-Nectin 1 (f) or CHO-HVEM (h) as target. Data is shown as mean normalized luminescence from two independent experiments. Two-way ANOVA followed by Dunnetts multiple comparison test was used to evaluate differences from WT (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001). Source data are provided as a Source Data file for all graphs.

For gB single site O-glycan mutants, we focused on the domain directly involved in fusion (I), where we identified three sites on antiparallel beta strands (T169, T267, T268), as well as the arm domain (V) comprised of two alpha helices that undergo structural rearrangements upon fusion, where we found one O-glycosite on each (T690 and T703) (Fig.6a)15, 41,42. All mutations except for T169A and T268A affected gB cell surface expression; T267A and T703A showed moderate reduction, whereas T690A showed increased expression (Fig.6c). T268A exhibited reduced fusion activity, as did T267A. Double or triple mutations in domain I severely decreased surface presentation and fusion activity (Fig.6c, d, f, g). The activity of domain V single mutants did not correlate with changes in surface expression, where T690A exhibited very low fusion activity despite increased surface presentation (Fig.6c, e, f, g). Interestingly, concomitant mutation of T703 (T690A T703A) could partially compensate for the strongly decreased activity of the T690A mutant (Fig.6g).

Though gD does not directly execute fusion, it initiates entry by binding to several different host receptors and compromised interaction with gD would translate to reduced fusion efficiency. For gD, one O-glycan site on the N-terminal tail of the protein (S33 (8)), involved in interaction with both Nectin 1 and HVEM, and two O-glycan sites on an alpha helix undergoing structural changes upon interaction with HVEM (T255 (230) and S260 (235)), were mutated (Fig.7a)15,43,44,45. All mutants maintained close to normal levels of cell surface expression of gD and fusion activity (Fig.7bd). To inspect possible contributions of gD mutations to interactions with distinct HSV-1 entry receptors, we utilized CHO cells overexpressing Nectin 1 or HVEM as target (Fig.7eh, Supplementary Fig.3a, b). Here we saw a modest reduction in Nectin 1-initiated cell-cell fusion, when T255 and S260 were collectively mutated (Fig.7e, f). A more pronounced reduction in cell-cell fusion efficiency was seen in HVEM-mediated entry upon introduction of these mutations (Fig.7g, h).

In conclusion, we identified functionally relevant O-glycan acceptor amino acids on gB, directly executing fusion, but no effects were observed for the initial engager gD in the presence of multiple host entry receptors in HEK293 cells. However, compound mutations in gD affected isolated receptor-mediated entry.

Lastly, we investigated the roles of the specific classes of glycans in direct cell-to-cell spread mediated in part by gE/gI via cell contacts of 2D grown keratinocytes, and unrestricted spread in 3D skin culture, facilitated by tissue destruction and release of free virions (Fig.8a).

a The cartoon illustrates different modes of HSV-1 cell-to-cell spread in 2D glycoengineered HaCaT cell monolayers in the context of a plaque assay, and spread in 3D organotypic skin models. b Plaque diameter on cell monolayers infected with HSV-1 Syn17+ at 48hpi. Data is presented as violin diagrams that include measurements from 3 independent experiments for each KO cell line, with 50 plaques measured for each experiment. Paired WT data includes measurements from 15 independent experiments. The dashed lines within the plots indicate median diameter, whereas the dotted lines indicate the interquartile range. One-way ANOVA followed by Games-Howells multiple comparison test was used to evaluate differences from WT (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001). Source data are provided as a Source Data file. c Cell monolayers grown on cover slips were infected with 200 PFU (MOI<0.0005) of HSV-1 K26-GFP and overlaid with semi-solid media for 48h followed by fixation and staining for HSV-1 gE (magenta). GFP labeled capsid proteins (VP26) are seen in green. Confocal images at two different magnifications were taken to illustrate overviews of plaques (4 combined tiles at 10x, upper panels) as well as gE expression at higher resolution (63x, lower panels). Scale bars for the different magnifications are indicated.

We first performed plaque assays with 2D grown cells, where dissociation of progeny virions is impeded by the dense overlay media, making direct cell-to-cell spread as the predominant mode of spread. Perturbations in core 1 O-glycan biosynthesis resulted in increased plaque size, most notably in C1GALT1 KO and ST3GAL1 KO cells (Fig.8b). Upon plaque immunostaining, WT cells and KO cells exhibiting increased plaque size showed strong gE expression on the cell surface (Fig.8c). In cells lacking N-linked glycan maturation (MGAT1 KO) and those lacking MGAT4B (MGAT4B KO; MGAT5+MGAT4B KO), resulting in reduced 4-antenna branching, we found a markedly reduced cell-to-cell spread (Fig.8b). MGAT1 KO cells showed less pronounced and more punctate gE expression, which may be linked to N-glycosylation sites on gE and help explain the limited spread capacity. Surprisingly, accelerated spread was observed in MGAT4A KO cells, which also contributes to 4-antenna branching, and a similar tendency was observed for MGAT5 KO, devoid in 6-linked antenna branching.

To assess viral spread in tissue, we infected fully developed 3D epidermises built with the glycoengineered cells (Fig.9a). Different spread characteristics were observed, when viral spread was not limited to cell-to-cell contacts mediated by gE/gI complex. In wild type HaCaT skin equivalents trans epidermal lesions were observed at 36 hpi (Fig.9b, c). To avoid selection bias, we generated ten subsequent tissue sections separated by 30microns and scanned whole sections, which allowed to visualize and compare the extent of the viral lesions (Fig.9a, b, Supplementary Fig.6). We identified lesions spanning several sections and measured the cross-section areas corresponding to the central regions of those lesions (Fig.9d). Large lesions were seen in MGAT1 KO tissues, contrasting the small plaques observed in 2D (Fig.9c, d). Most N-glycan branching KO tissues, especially MGAT4A KO and MGAT5+4B KO, permitted only limited spread in the top layers of the epidermis. MGAT4B KO allowed formation of bigger lesions, but the tissue penetrance was limited, which was also the case for tissues with reduced core 1 sialylation (ST3GAL1 KO) (Fig.9c, d). No significant spread defects were noted for tissues with disruptions in GSL and GAG synthesis.

ad Fully differentiated 3D skin models built with glycoengineered cells were infected with HSV-1 Syn17+ for 36h followed by fixation in formalin and embedding in paraffin. a The cartoon illustrates the procedure for evaluating HSV-1 spread in organotypic skin tissues. FFPE tissues were sectioned every 30m for 10 consecutive slices containing two sections each, spanning from the center of the tissue outwards in two directions. b Consecutive sections were stained with a polyclonal FITC-labeled anti-HSV-1 antibody to visualize virus lesions and whole sections imaged with a microscope slide scanner. An example section series is shown with HSV-1 lesions outlined in purple. c Representative lesions were selected from the scanned section series for each KO tissue. Nuclei were labeled with DAPI. d Three lesions spanning several sections were identified for each KO tissue and lesion area measured at the centermost section. Data is shown as mean+SEM with individual measurement values indicated as black dots. One-way ANOVA followed by Dunnetts multiple comparison test was used to evaluate differences from WT (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001). Source data are provided as a Source Data file.

See the original post:
Glycoengineered keratinocyte library reveals essential functions of ... - Nature.com

Fact Sheet: Biotechnology – Center for Arms Control and Non-Proliferation

What is Biotechnology?

Biotechnology is the application of biological systems and materials to create new technologies, products, and services that offer qualitative improvements to human and environmental health. Biotechnology encompasses many disciplines including genetics, molecular biology, bioengineering, pharmaceuticals, agriculture, bioinformatics and more. Among the most widely recognized applications of biotechnology is genomic manipulation, or gene editing. Through gene editing, scientists can make highly targeted alterations to the DNA sequences of a living organisms genome. This enables better understanding of genetic and hereditary diseases and can be used to make DNA more resilient to certain viruses or bacteria. The field also enables the creation of biosynthetics, or novel materials created through organic chemistry, that exhibit superior characteristics and greater environmental sustainability than traditional petroleum-based compounds.

There will be increased risk that biotech advancements make the weaponization of biological and chemical agents more likely. There is no universal standard governing the proliferation of biotech, and export controls fail to keep pace with the rapid development of this sector. The spread of biosynthetic tools will enable more research labs around the world to explore pathogen research and the engineering of novel viruses. Moreover, existing safeguards and governance regimes may not be sufficient to prevent the accidental or nefarious spread of dangerous new compounds, toxins and infectious diseases. The risk of non-state access to various biotechnologies also means that bioterrorism will present as a threat, potentially in the form of intentional sabotage of agricultural systems via release of genetically modified organisms. The multiplying effect of AI-enabled research and development will also contribute to the proliferation of biotechnology inways that have yet to be understood.

View original post here:
Fact Sheet: Biotechnology - Center for Arms Control and Non-Proliferation

"My Film is For the Pigs": Heather Dewey-Hagborg on Hybrid: an … – Filmmaker Magazine

Heather Dewey-Hagborg is on a mission to confront the uncomfortable future, especially when it comes to emerging tech. Stranger Visions features portrait sculptures crafted from analyses of genetic material the transdisciplinary artist, educator and filmmaker literally picked up in public places (one persons discarded cigarette butt is anothers way into a strangers DNA). T3511, a collaboration with cinematographer Toshiaki Ozawa (Laurie Andersons Heart of a Dog), sees an anonymous saliva sample become fodder for the alchemizing of the perfect romantic partner.

Now theres Hybrid: an Interspecies Opera, perhaps Dewey-Hagborgs most ambitious work to date. Opening at NYCs Fridman Gallery on November 1, the multimedia project includes a short documentary/personal narrative set to an original score alongside a set of (robotically-constructed and clay-fired) memorial pig sculptures, which allude to the xenotransplantation topic at hand as well as the question of whether genetically engineering bovine for the sole purpose of harvesting hearts for human transplantation is the ethical easy call Big Tech would like us to make (and believe).

Just prior to the artworks New York debut, Filmmaker reached out to Dewey-Hagborg to learn more about enmeshing the scientific and the personal to shape a career in biopolitical art.

Filmmaker: What initially led you to explore the biomedical realm?

Dewey-Hagborg: This started more than 10 years ago, when I became entranced with emerging possibilities of genomics in my project Stranger Visions. The first community biohacker lab had just opened in Brooklyn (Genspace), and I became a member and learned all about DNA. What I realized at that time was that so much was happening so fast in biotech, but it wasnt getting the same critical, artistic attention as digital technology was. Well, this is still true, and I am committed to changing that.

Filmmaker: How did Hybrid: an Interspecies Opera originate? Do you see it as an extension of that previous work Stranger Visions and (your collaboration with Toshiaki Ozawa) T3511?

Dewey-Hagborg: Yes, but also it is a pretty different approach for me in a number of ways. The musical collaboration with Bethany Barrett was something very new for me, and now working on transforming it into a live opera performance (which will premiere next year at the Exploratorium on March 7 and 8) is a really exciting but also very challenging new direction. The film itself has some similarities to T3511 in that both are unusual forms of documentary and exist as records of my practice, but also hopefully transcend this to stand as emotionally relatable media that draws the viewer into contemplating those topics of DNA privacy and xenotransplantation, respectively, more deeply.

Filmmaker: How did this idea of turning this piece into an opera come about? What was the actual process of developing the score and working with the various musicians?

Dewey-Hagborg: I was invited to work on a new piece about gene editing by the MIT Museum and guest curator William Myers. I had been intrigued by xenotransplantation for quite some time because it was the place where the most simultaneous gene edits had occurredin order to make pigs essentially more human. Usually I like to work hands-on in the lab, but with this piece getting access to the kinds of labs that do this work was really prohibitively difficult, because of the controversy surrounding it and the fragile nature of this very experimental new technique. Additionally, it was during the height of COVID.

So, I started the project with a lot of research. I began interviewing scientists that study pigs and xenotransplantation, as well as archaeologists who study the evolution of the pig. I really wanted to get at this question of whether gene editing was something radically new or a continuation of 10 millennia of domestication and selective breeding (as molecular biologists often posit). I began having these Zoom sessions and recording them, then I started working with the wordstranscribing them, editing themand was struck by the beauty, poetry, humor and drama I was hearing from my interlocutors. I just started pulling sentences and arranging them into small poems, and suddenly I heard them in my head in opera voice. I thought, Maybe that is the form this should take. Maybe music should convey the emotional layers of this emerging technology.

I wrote the libretto and went through several iterations and experiments until finally a friend recommended composer Bethany Barrett, who is based in Berlin. She wrote the music and sent me the names of singers she wanted to work with, and we just continued to pass ideas and recordings back and forth.

Now, in working on the live production, I have a music director, Sam Faustine; an associate director, Becca Wolff; and a local crew of singers in San Francisco. Its really an incredible team. (Also, the staff at the Exploratorium has been wonderful.) We rehearse together because my (speaking) parts are intertwined with the singing. It is such an amazing feeling standing onstage and hearing these powerful voices sing the words I wrote live.

Filmmaker: Why do you bring personal narrative into your art?

Dewey-Hagborg: When I was an undergraduate art student I was taught not to: I was told to keep my work conceptual, impersonal, abstract. And while I love work like that too, ultimately it was not my voice. The personal for me is authentic. I want to put my subjectivity forward. I really enjoy enmeshing the scientific and the personal, the messy and the clean. I call it writing through. I like to write my experience through the scientific and technological critique. It feels real to me and more honest than a standard documentary would. And I hope it brings an emotional layer that people can relate to. But every project is different, and I try to listen to the material and orient my approach in a way to best serve its dimensions.

Filmmaker: Youve spoken in the past about your discomfort with both corporations and governments having such easy access to our genetic material be it through seemingly benign ancestry tracing sites or even COVID testingand you also seem similarly uneasy with xenotransplantation and genetically engineering pigs for human hearts (i.e., for humanitys greater good). So, what sorts of change do you ultimately hope to accomplish through your biopolitical art?

Dewey-Hagborg: Some issues are very straightforward, but most are complex and contain layers of tradeoffs. Xenotransplantation is clearly a morally complicated issue. The goal with my work generally is to question the status quo, to advocate for critical attention and debate to topics that are under-discussed. With all the reports in the last year of the remarkable progress in xenotransplantation, there is little to no discussion of the animals whose lives are taken. This is not to say I advocate for a ban on the practice, but I dont think it makes sense to completely skip over discussing the moral dilemma, when we are setting structures into place now that will frame how the future unfolds. When I started the project, I tried to get access to the leading xenotransplantation company in the US to shoot and they told me straight up, We dont want people thinking about pigs. So, my film is for the pigs.

View post:
"My Film is For the Pigs": Heather Dewey-Hagborg on Hybrid: an ... - Filmmaker Magazine

China warns a ‘certain country’ is making ethnic bioweapons – Interesting Engineering

China has blown the whistle on the potential dangers of what it calls "genetic weapons" that could prove to be an incredibly potent weapon of mass destruction, the Global Times reports. On Monday, October 20, 2023, the Chinese Ministry of State Security released a statement on WeChat warning that a "certain" foreign non-governmental organization (NGO) had recruited Chinese "volunteers" to collect biodiversity distribution data under the guise of biological species research to steal China's species data.

The ministry did not name the countries in question, nor did it offer evidence to support the claim. While not a new claim, the existence of such weapons has long been dismissed by the mainstream scientific community as a "conspiracy theory." In a February 2020 report, the South China Morning Post (SCMP) reports, the Council on Strategic Risks stated that bioweapons as a deterrent were "irrelevant" because no country was safe from the effects of a pandemic.

Continued here:
China warns a 'certain country' is making ethnic bioweapons - Interesting Engineering

Navigating the horizon: The future of frontier tech – EU Reporter

In the rapidly evolving landscape of technology, innovation continues to shape the world we live in. Frontier tech, also known as cutting-edge or emerging technology, is at the forefront of this transformation. These emerging technologies are poised to redefine industries, disrupt traditional business models, and push the boundaries of what's possible. In this article, we will explore the future of frontier tech, examining key trends and their potential impacts on society and the economy, writes Colin Stevens.

Artificial Intelligence (AI)

Artificial Intelligence, often referred to as AI, has already made significant strides, but its future holds even greater promise. The integration of AI into various aspects of our lives, from autonomous vehicles and healthcare diagnostics to customer service chatbots, is set to become more profound. AI will continue to evolve, with more advanced algorithms, natural language understanding, and improved machine learning models. This progress will lead to better decision-making, increased efficiency, and the emergence of new applications we haven't even imagined yet. Quantum Computing

Quantum computing is a game-changer in the world of computing. Unlike classical computers, quantum computers harness the power of quantum bits (qubits) to perform complex calculations exponentially faster. As quantum technology matures, it will revolutionize fields like cryptography, materials science, drug discovery, and optimization problems. The ongoing research in quantum algorithms and hardware will pave the way for practical quantum computers that can tackle some of the world's most challenging problems.

5G and Beyond

The rollout of 5G networks is just the beginning of a new era in wireless communication. Beyond 5G, we will witness the development of 6G technology, which promises even higher data speeds, lower latency, and greater connectivity. These advancements will fuel the growth of the Internet of Things (IoT), enabling more interconnected devices and immersive experiences. 6G may also enable entirely new applications, such as holographic communication and remote surgery.

Blockchain and Cryptocurrency

Blockchain technology, known for its role in enabling cryptocurrencies like Bitcoin, has applications that extend beyond digital currency. Decentralized finance (DeFi), non-fungible tokens (NFTs), and smart contracts are just a few examples of how blockchain is disrupting traditional finance, art, and legal processes. In the future, we can expect to see more widespread adoption of blockchain in various sectors, including supply chain management, voting systems, and identity verification. Biotechnology and Genetic Engineering

Advances in biotechnology and genetic engineering are poised to revolutionize healthcare, agriculture, and even our understanding of life itself. Gene editing techniques like CRISPR-Cas9 offer the potential to cure genetic diseases, create more resilient crops, and address environmental challenges. As our understanding of the human genome deepens, we may also see breakthroughs in personalized medicine and enhanced human capabilities.

Augmented and Virtual Reality

Augmented Reality (AR) and Virtual Reality (VR) technologies are making their way into various industries, including gaming, healthcare, education, and manufacturing. In the future, AR glasses and VR headsets could become more compact, affordable, and versatile, enabling immersive experiences for everyday tasks. The blending of physical and digital worlds through AR will lead to a wide range of applications, from interactive navigation to enhanced training and remote collaboration.

Space Exploration and Commercialization

Space exploration is no longer the exclusive domain of governments. Private companies like SpaceX, Blue Origin, and Virgin Galactic are rapidly advancing the possibilities of commercial space travel and colonization. These developments have the potential to unlock new economic opportunities in space mining, satellite services, and interplanetary tourism.

Challenges and Considerations

While frontier tech holds immense promise, it also raises important ethical, regulatory, and security concerns. As technology continues to evolve, society must grapple with issues like data privacy, cybersecurity, AI bias, and the ethical implications of genetic engineering. Striking a balance between innovation and responsibility will be a key challenge in the future.

The future of frontier tech is a journey into uncharted territory, where the boundaries of what's possible are constantly expanding. As AI, quantum computing, 5G, blockchain, biotechnology, AR/VR, and space exploration continue to advance, they will create new opportunities and challenges for society. Staying informed and engaged with these emerging technologies will be crucial in shaping a future that harnesses their potential for the benefit of all. The path ahead is full of possibilities, and it's up to us to navigate the horizon of frontier tech responsibly and wisely.

About the author: Colin Stevens founded EU Reporter in 2008. He has more than 30 years of experience as a TV producer, journalist and news editor. He is a past president of the Press Club Brussels (2020-2022) and was awarded an Honorary Doctor of Letters at Zerah Business School (Malta and Luxembourg) for leadership in European journalism.

Share this article:

Continued here:
Navigating the horizon: The future of frontier tech - EU Reporter

Stanisaw Lem’s Prescient Vision of Artificial Life – The MIT Press Reader

As with the best science fiction, Lems novel The Invincible has as much to teach us about our present situations as any futures we may face.

In the grand tradition of H. G. Wells and Jules Verne, Stanisaw Lems The Invincible tells the story of a space cruiser sent to an obscure planet to determine the fate of a sister spaceship whose communication with Earth has abruptly ceased. Landing on the planet Regis III, navigator Rohan and his crew discover a form of life that has apparently evolved from autonomous, self-replicating machines perhaps the survivors of a robot war. Rohan and his men are forced to confront the classic quandary: What course of action can humanity take once it has reached the limits of its knowledge? In The Invincible, Lem has his characters confront the inexplicable and the bizarre: the problem that lies just beyond analytical reach.

The following is literary critic and theorist N. Katherine Hayles foreword to the 2020 edition of Lems classic novel, originally published in Polish in 1964.

Science fiction has famously predicted many of the important technologies of the 20th century: space travel, satellites, the atomic bomb, television, the internet, and virtual reality, to name a few. In The Invincible, Stanisaw Lem predicts another: artificial life. Although speculations about self-reproducing artificial systems date from the 1940s, the scientific field received its name from Christopher Langton only in 1986, more than two decades after the original publication of The Invincible (1964). One of the central controversies in artificial life is whether evolutionary programs and devices are actually alive (the strong version), or whether they merely simulate life (the weak version). Researchers who follow the strong version argue that the processes embedded in software programs such as genetic algorithms are as natural as life itself; what is artificial is the medium in which these processes take place.

The issue prompted Robert Rosen, among others, to speculate about the essential characteristics of life itself, not only as it evolved on Earth in carbon-based life forms but also about the possibility of life-as-it-could-be in exoplanetary systems, arguing that silicon-based artificial life forms may provide insight into these theoretical speculations.

The Invincible presents a fascinating hybridization of these different views. Dr. Laudas hypothesis proposes that a space ship from the Lycran system landed on Regis III millions of years ago; while the biological visitors perished, the automata did not. There then followed an evolutionary struggle between the automata and the planets indigenous life forms, on the one hand, and between the different kinds of automata, on the other. Such a scenario requires that the survive and reproduce mandate that governs life on Earth could also operate on this planet. Lem minimally fulfills the requirement by postulating that the automata could manufacture themselves with modifications dictated by evolutionary processes. Clearly his interest is not in filling out how this might take place (John von Neumann, encountering a similar problem, imagined metal parts floating on a lake that could self-assemble). Rather, Lems focus is on envisioning an artificial life form that won the evolutionary competition on Regis III for profoundly different reasons than did Homo sapiens on Earth.

The effect is achieved by introducing a significant factor that has a monumental impact on evolutionary trajectories: rather than fulfilling their energy needs through ingesting food, the automata on Regis III evolve to use solar power. The smaller the artificial organism, the less energy it needs. Hence the evolutionary driver is toward smaller forms, which overcome not through superior intellect but through swarm intelligence. Lem added to this the ability of the swarm of flies to generate immensely powerful electromagnetic fields, which meant that the tiny automata are not only the evolutionary winners on their planet but a powerful force against the invading humans. Their tiny size notwithstanding, their awesome potential illuminates the profound ambiguity of the works title, which can be taken to refer either to the spaceships proud name or to the swarms of alien automata that threaten it.

From a broader cosmic perspective, the best of human science, engineering, and weaponry may reveal humans to be completely out of our depth, mere kindergarteners bidding for a place in the universes adult civilizations.

Contemporary research in artificial life has validated Lems insight that swarms of artificial beings require only a few simple rules to manifest complex behaviors and hence each member needs to carry only a little cognitive power onboard. Computer simulations that have accurately depicted swarm behaviors in fish, birds, bees, and other biota demonstrate that each individual responds only to the four or five closest to it, with rule sets that take up only a few lines of code. For example, a school of fish swimming to evade a predator is guided by the fish closest to the predator. The direction this most imperiled individual follows determines how the entire school will run as it flashes back and forth, a simple strategy that makes excellent sense, since the fish that has the most to lose will try hardest to escape. Although each fishs behaviors are simple, the collective result nevertheless generates swarm intelligence of considerable complexity.

Decades before these ideas became disseminated within the scientific community, Lem intuited that different environmental constraints might lead to radically different evolutionary results in automata compared to biological life forms. Although on Earth the most intelligent species (i.e., humans) has tended to fare the best, their superior intelligence comes with considerable costs: a long period of maturation; a lot of resources invested in each individual; socialization patterns that emphasize pair bonding and community support; and a premium on individual achievement. But these are not cosmic universals, and different planetary histories might result in the triumph of very different kinds of qualities.

The contrasts between humans and the automata swarm are brought out most poignantly in the scene between Captain Horpach and First Officer Rohan, in which the captain delegates to Rohan the decision whether to put another crew member in grave danger to determine if the missing four men have indeed perished, as seems all but certain, or whether one or more might still be alive. The assumptions that make this gamble even remotely worth taking are revealing: human life is precious; human solidarity depends on the crews belief that everything possible will be done to save them if they are in peril; and every human is unique and therefore uniquely valuable. None of these, of course, holds true for the swarm, whose individual members are virtually identical to one another, with each tiny automaton easily replaced and therefore disposable. Consequently, none is valuable in itself; only the swarm has evolutionary survival value. The contest, then, is not only between different life forms but also between the different values that have resulted from the divergent evolutionary pathways of humans on Earth and the flies on this strange planet. As with Solaris, Lem suggests that assumptions born and bred of Earth may appear hopelessly provincial in light of human encounters with radically different life forms. From a broader cosmic perspective, the best of human science, engineering, and weaponry may reveal humans to be completely out of our de
pth, mere kindergarteners bidding for a place in the universes adult civilizations. The reduction of crew members to infancy when attacked by the flies may be a metaphor for this realization.

Of all the human characters, Rohan has the strongest claim to have encountered the planet on its own terms. He has traversed its terrain with his own feet; he has mixed his sweat with its crevices, valleys, and hills; he has breathed its native atmosphere into his lungs. The insight he gains from his heroic trek therefore commands our respect. When he concludes that not everything everywhere is for us [humans], we are right to hear in this pronouncement Lems own challenge to the anthropocentric assumptions that continue to dominate human ethical frameworks as well as human exploitations of planet Earth. As with the best science fiction, The Invincible has as much to teach us about our present situations as any futures we may face.

N. Katherine Hayles is Distinguished Research Professor of English at the University of California, Los Angeles.

Read the original:
Stanisaw Lem's Prescient Vision of Artificial Life - The MIT Press Reader

Screen and treat essential to beat cervical cancer – University of Cape Town News

In 2020 the World Health Organization (WHO) publicised a global strategy to accelerate the elimination of cervical cancer as a public health problem. To achieve this goal, the organisation recommends that 90% of girls be fully vaccinated with the human papilloma virus(HPV) vaccine by the age of 15; and that 70% of women be screened with a high-performance test by the age of 35.

For more than four decades, the University of Cape Towns (UCT) Professor Lynette Denny one of the countrys and continents leading cervical cancer researchers has been at the forefront of this fight. During a recent conference, arranged to shine a spotlight on the advances in prostate and cervical cancer research organised by the International Centre for Genetic Engineering and Biotechnology (ICGEB) Professor Denny highlighted the importance of adopting effective screen-and-treat strategies to adequately address the burden of this disease on women.

We started looking at alternative strategies to the pap smear because of our failure to make a dent on [the burden caused by] cervical cancer.

Denny told the audience that her team at the Khayelitsha Cervical Cancer Screening Project (KCCSP), have for years been testing and evaluating alternative, effective methods to the pap smear a well-known procedure used to test for cervical cancer. The KCCSP fast tracks research into cervical cancer prevention and treatment options and provides vital HPV screening and treatment. The clinic is located on the Khayelitsha Community Health Clinic precinct in Site B.

Evaluating alternative strategies

We started looking at alternative strategies to the pap smear because of our failure to make a dent on [the burden caused by] cervical cancer, Denny said.

Cervical cancer is the fourth most common cancer globally, and in 2020 claimed the lives of approximately 350000 women. More than 80% of cases occur in low- to middle-income countries in sub-Saharan Africa, Melanesia, Asia and Southeast Asia. In South Africa, thousands of cases of cervical cancer are diagnosed annually, and the prognosis is seldom good.

Over the past couple of years, Denny and her team have explored several alternative techniques to the pap smear. One, she explained, included evaluating the effectiveness of visual inspection with acetic acid (VIA), both with and without magnification a simple and inexpensive test used to detect cervical pre-cancerous lesions. The next alternative was exploring the use of visual automated evaluation using artificial intelligence and machine learning. This technique shows promise, but, said Denny, further investigation into its effectiveness is still required. The third option was assessing the feasibility of molecular testing using nucleic acid amplification(NAATs). NAATS is a unique HPV DNA test that checks the presence of specific cancer-causing strains, and this technique revealed some pleasantly surprising results.

Using HPV NAATs as the primary screening test prevents cancer and saves more lives than [the use of] VIA cytology as the primary screening test, Denny said. [Therefore] the WHO now encourages the use of HPV NAATs once testing infrastructure is operational and affordable.

Randomised control trial

While on a quest to locate effective, safe and affordable methods to prevent cervical cancer, Denny said the team designed a randomised clinical control trial to investigate the efficacy and feasibility of two specific screen-and-treat strategies. More than 6000 non-pregnant women, who were previously unscreened for HPV, were recruited from clinics in Khayelitsha and participated in the study.

She said a group of women were randomly selected for an HPV [NAATs] screen-and-treat round, in which HPV-positive women received prior therapy; while the second group of participants followed a VIA screen-and-treat approach and HPV-positive women received prior therapy as well.

Performance characteristics of HPV and VIA as the primary screening tests you can see that [with] HPV testing, we [achieved] a 90% sensitivity [to detecting pre-cancerous lesions], 85% specificity, and a 99% negative predictive value. This is very important for national screening programmes.

However, she pointed out that the VIA screen-and-treat results revealed a below 50% sensitivity to detecting pre-cancerous lesions, an 80% specificity and 97% negative predictive value.

If we compare these two screen-and-treat strategies, to reduce the accumulative prevalence of CIN2+ [cervical cancer] by 36 months, we see that we needed 23 patients screened [with HPV NAATS] to prevent one case of CIN2, compared to 50 cases of VIA. This gives you a graphic description, Denny said.

The way forward

However, she added, the right screen-and-treat strategy depends on the environment and the clinics location.

The impact of cervical cancer and mortality must be measured and demonstrated. Without knowing the impact, the process of secondary prevention will fail.

And as scientists and clinicians work towards winning the war on cervical cancer, Denny said there is an urgent need to meticulously evaluate screen-and-treat strategies, while keeping various contexts in the country, the continent and the world top of mind. Further, she said, performing situational analyses to assess these contexts prior to introducing the preferred strategy is essential to ensure that it will benefit the patient and will not break the system.

We need to create a menu of options. What exactly is needed for successful [screen-and-treat] implementation? The ultimate goal is the elimination of cervical cancer as a public health problem, Denny said. The impact of cervical cancer incidence and mortality must be measured and demonstrated. Without knowing the impact, the process of secondary prevention will fail.

Excerpt from:
Screen and treat essential to beat cervical cancer - University of Cape Town News

The potential of CO2-based production cycles in biotechnology to … – Nature.com

Paraschiv, S. & Paraschiv, L. S. Trends of carbon dioxide (CO2) emissions from fossil fuels combustion (coal, gas and oil) in the EU member states from 1960 to 2018. Energy Rep. 6, 237242 (2020).

Article Google Scholar

International Energy Agency (IEA). CO2 Emissions in 2022. CO2 Emiss. 2022 (2023). https://doi.org/10.1787/12ad1e1a-en.

Vom Berg, C., Carus, M., Stratmann, M. & Dammer, L. Renewable Carbon as a Guiding Principle for Sustainable Carbon Cycles. Renew. Carbon Initiat. (2022).

Wang, H., Peng, X., Zhang, H., Yang, S. & Li, H. Microorganisms-promoted biodiesel production from biomass: A review. Energy Convers. Manag. X 12, 100137 (2021).

CAS Google Scholar

Shears, J. Is there a role for synthetic biology in addressing the transition to a new lowcarbon energy system? Microb. Biotechnol. 12, 824827 (2019).

Article PubMed PubMed Central Google Scholar

Srisawat, P., Higuchi-Takeuchi, M. & Numata, K. Microbial autotrophic biorefineries: Perspectives for biopolymer production. Polym. J. 54, 11391151 (2022).

Article CAS Google Scholar

Lee, R. A. & Lavoie, J.-M. From first- to third-generation biofuels: Challenges of producing a commodity from a biomass of increasing complexity. Anim. Front 3, 611 (2013).

Article Google Scholar

Caltzontzin-Rabell, V. et al. Raw materials for a biomass-based industry. in Biofuels and Biorefining 2552 (Elsevier, 2022). https://doi.org/10.1016/B978-0-12-824116-5.00010-6.

Yang, F., Hanna, M. A. & Sun, R. Value-added uses for crude glycerola byproduct of biodiesel production. Biotechnol. Biofuels 5, 13 (2012).

Article CAS PubMed PubMed Central Google Scholar

Food and Agriculture Organization of the United Nations (FAO). Sustainable Food and Agriculture. online at https://www.fao.org/sustainability/news/detail/en/c/1274219/ (2020).

Gitz, V., Meybeck, A., Lipper, L., Young, C. & Braatz, S. Climate change and food security: Risks and responses. Food and Agriculture Organization of the United Nations (2016).

Cotton, C. A., Claassens, N. J., Benito-Vaquerizo, S. & Bar-Even, A. Renewable methanol and formate as microbial feedstocks. Curr. Opin. Biotechnol. 62, 168180 (2020).

Article CAS PubMed Google Scholar

Jiang, W. et al. Metabolic engineering strategies to enable microbial utilization of C1 feedstocks. Nat. Chem. Biol. 17, 845855 (2021).

Article CAS PubMed Google Scholar

Ewis, D. et al. Electrochemical reduction of CO2 into formate/formic acid: A review of cell design and operation. Sep. Purif. Technol. 316, 123811 (2023).

Article CAS Google Scholar

Li, P., Gong, S., Li, C. & Liu, Z. Analysis of routes for electrochemical conversion of CO2 to methanol. Clean. Energy 6, 967975 (2022).

Article Google Scholar

Lee, M. Y. et al. Current achievements and the future direction of electrochemical CO2 reduction: A short review. Crit. Rev. Environ. Sci. Technol. 50, 769815 (2020).

Article CAS Google Scholar

Izadi, P. & Harnisch, F. Microbial | electrochemical CO2 reduction: To integrate or not to integrate? Joule 6, 935940 (2022).

Article Google Scholar

Nitopi, S. et al. Progress and Perspectives of Electrochemical CO2 Reduction on Copper in Aqueous Electrolyte. Chem. Rev. 119, 76107672 (2019).

Article CAS PubMed Google Scholar

Santos Correa, S., Schultz, J., Lauersen, K. J. & Soares Rosado, A. Natural carbon fixation and advances in synthetic engineering for redesigning and creating new fixation pathways. J. Adv. Res. 47, 7592 (2023).

Article CAS PubMed Google Scholar

Bar-Even, A., Noor, E. & Milo, R. A survey of carbon fixation pathways through a quantitative lens. J. Exp. Bot. 63, 23252342 (2012).

Article CAS PubMed Google Scholar

Claassens, N. J. Reductive Glycine Pathway: A Versatile Route for One-Carbon Biotech. Trends Biotechnol. 39, 327329 (2021).

Article CAS PubMed Google Scholar

Stephens, S., Mahadevan, R. & Allen, D. G. Engineering Photosynthetic Bioprocesses for Sustainable Chemical Production: A Review. Front. Bioeng. Biotechnol. 8, 610723 (2021).

Article PubMed PubMed Central Google Scholar

Zhang, S. et al. Main components of free organic carbon generated by obligate chemoautotrophic bacteria that inhibit their CO2 fixation. iScience 25, 105553 (2022).

Article ADS CAS PubMed PubMed Central Google Scholar

Sarma, S. et al. Valorization of microalgae biomass into bioproducts promoting circular bioeconomy: a holistic approach of bioremediation and biorefinery. 3 Biotech 11, 378 (2021).

Article PubMed PubMed Central Google Scholar

Veaudor, T. et al. Recent Advances in the Photoautotrophic Metabolism of Cyanobacteria: Biotechnological Implications. Life 10, 71 (2020).

Article ADS CAS PubMed PubMed Central Google Scholar

Yoon, J. & Oh, M.-K. Strategies for Biosynthesis of C1 Gas-derived Polyhydroxyalkanoates: A review. Bioresour. Technol. 344, 126307 (2022).

Article CAS PubMed Google Scholar

Bengelsdorf, F. R. et al. Industrial Acetogenic Biocatalysts: A Comparative Metabolic and Genomic Analysis. Front. Microbiol. 7, 115 (2016).

Article Google Scholar

Bourgade, B., Minton, N. P. & Islam, M. A. Genetic and metabolic engineering challenges of C1-gas fermenting acetogenic chassis organisms. FEMS Microbiol. Rev. 45, 120 (2021).

Article Google Scholar

Liew, F. E. et al. Carbon-negative production of acetone and isopropanol by gas fermentation at industrial pilot scale. Nat. Biotechnol. 40, 335344 (2022).

Article CAS PubMed Google Scholar

Yurimoto, H., Shiraishi, K. & Sakai, Y. Physiology of Methylotrophs Living in the Phyllosphere. Microorganisms 9, 809 (2021).

Article CAS PubMed PubMed Central Google Scholar

Pea, D. A., Gasser, B., Zanghellini, J., Steiger, M. G. & Mattanovich, D. Metabolic engineering of Pichia pastoris. Metab. Eng. 50, 215 (2018).

Article PubMed Google Scholar

Zhang, W. et al. Current advance in bioconversion of methanol to chemicals. Biotechnol. Biofuels 11, 111 (2018).

Article Google Scholar

Nattermann, M. et al. Engineering a new-to-nature cascad
e for phosphate-dependent formate to formaldehyde conversion in vitro and in vivo. Nat. Commun. 14, 2682 (2023).

Article ADS CAS PubMed PubMed Central Google Scholar

Collas, F. et al. Engineering the biological conversion of formate into crotonate in Cupriavidus necator. bioRxiv (2023). https://doi.org/10.1101/2023.03.14.532570.

Gregory, G. J., Bennett, R. K. & Papoutsakis, E. T. Recent advances toward the bioconversion of methane and methanol in synthetic methylotrophs. Metab. Eng. 71, 99116 (2022).

Article CAS PubMed Google Scholar

Guerrero-Cruz, S. et al. Methanotrophs: Discoveries, Environmental Relevance, and a Perspective on Current and Future Applications. Front. Microbiol. 12, 128 (2021).

Article Google Scholar

Fei, Q. et al. Bioconversion of natural gas to liquid fuel: Opportunities and challenges. Biotechnol. Adv. 32, 596614 (2014).

Article CAS PubMed Google Scholar

Kalyuzhnaya, M. G. et al. Highly efficient methane biocatalysis revealed in a methanotrophic bacterium. Nat. Commun. 4, 2785 (2013).

Article ADS CAS PubMed Google Scholar

Kwon, M., Ho, A. & Yoon, S. Novel approaches and reasons to isolate methanotrophic bacteria with biotechnological potentials: recent achievements and perspectives. Appl. Microbiol. Biotechnol. 103, 18 (2019).

Article CAS PubMed Google Scholar

Bar-Even, A., Noor, E., Lewis, N. E. & Milo, R. Design and analysis of synthetic carbon fixation pathways. Proc. Natl Acad. Sci. USA. 107, 88898894 (2010).

Article ADS CAS PubMed PubMed Central Google Scholar

Liang, B., Zhao, Y. & Yang, J. Recent Advances in Developing Artificial Autotrophic Microorganism for Reinforcing CO2 Fixation. Front. Microbiol. 11, 592631 (2020).

Article PubMed PubMed Central Google Scholar

Klein, V. J., Irla, M., Gil Lpez, M., Brautaset, T. & Fernandes Brito, L. Unravelling Formaldehyde Metabolism in Bacteria: Road towards Synthetic Methylotrophy. Microorganisms 10, 220 (2022).

Article CAS PubMed PubMed Central Google Scholar

Keller, P. et al. Generation of an Escherichia coli strain growing on methanol via the ribulose monophosphate cycle. Nat. Commun. 13, 113 (2022).

Article Google Scholar

Zhan, C. et al. Reprogramming methanol utilization pathways to convert Saccharomyces cerevisiae to a synthetic methylotroph. Nat. Catal. 6, 435450 (2023).

Article ADS CAS Google Scholar

Tuyishime, P. et al. Engineering Corynebacterium glutamicum for methanol-dependent growth and glutamate production. Metab. Eng. 49, 220231 (2018).

Article CAS PubMed Google Scholar

Chen, F. Y. H., Jung, H. W., Tsuei, C. Y. & Liao, J. C. Converting Escherichia coli to a Synthetic Methylotroph Growing Solely on Methanol. Cell 182, 933946.e14 (2020).

Article CAS PubMed Google Scholar

Gassler, T. et al. The industrial yeast Pichia pastoris is converted from a heterotroph into an autotroph capable of growth on CO2. Nat. Biotechnol. 38, 210216 (2020).

Article CAS PubMed Google Scholar

Gassler, T., Baumschabl, M., Sallaberger, J., Egermeier, M. & Mattanovich, D. Adaptive laboratory evolution and reverse engineering enhances autotrophic growth in Pichia pastoris. Metab. Eng. 69, 112121 (2022).

Article CAS PubMed Google Scholar

Gleizer, S. et al. Conversion of Escherichia coli to Generate All Biomass Carbon from CO2. Cell 179, 12551263.e12 (2019).

Article CAS PubMed PubMed Central Google Scholar

Baumschabl, M. et al. Conversion of CO2 into organic acids by engineered autotrophic yeast. Proc. Natl Acad. Sci. 119, 110 (2022).

Article Google Scholar

Noor, E., Flamholz, A., Liebermeister, W., Bar-Even, A. & Milo, R. A note on the kinetics of enzyme action: A decomposition that highlights thermodynamic effects. FEBS Lett. 587, 27722777 (2013).

Article CAS PubMed Google Scholar

Flamholz, A., Noor, E., Bar-Even, A. & Milo, R. EQuilibrator - The biochemical thermodynamics calculator. Nucl. Acids Res. 40, 770775 (2012).

Article Google Scholar

Noor, E. et al. Pathway Thermodynamics Highlights Kinetic Obstacles in Central Metabolism. PLoS Comput. Biol. 10, e1003483 (2014).

Article PubMed PubMed Central Google Scholar

Read the original post:
The potential of CO2-based production cycles in biotechnology to ... - Nature.com

The Race To Destroy PFAS, the Forever Chemicals – Slashdot

An anonymous reader shares a report: PFAS stands for "per- and polyfluoroalkyl substances," a family of upwards of 15,000 or more human-made and incredibly durable chemical compounds that have been used in countless industrial and consumer applications for decades. Firefighting foams, waterproof hiking boots, raincoats, nonstick frying pans, dental floss, lipstick, and even the ink used to label packaging -- all can contain PFAS. The compounds are ubiquitous in drinking water and soil, even migrating to Arctic sea ice. PFAS are called forever chemicals because once present in the environment, they do not degrade or break down. They accumulate, are transferred throughout the watershed, and ultimately persist. The quest to reduce the amount of PFAS in the environment is what led me to an industrial park in a southern suburb of Grand Rapids, Michigan. The jar of PFAS concentrate in my hand is part of a demonstration arranged by my hosts, Revive Environmental, during a tour of the company's PFAS destruction site, one of the first in the country to operate commercially and at scale. A few yards in front of me sits the company's PFAS "Annihilator" in a white shipping container.

The Annihilator represents just one of several technologies now vying to break down and destroy PFAS. These span the gamut from established processes like electrochemical oxidation and supercritical water oxidation to emerging techniques relying on ultraviolet light, plasma, ultrasound, or catalyst-driven thermal processes. Some are deployed in field tests. Other companies are actively running pilot programs, many with various divisions of the US Department of Defense and other government agencies. And many other technologies are still undergoing laboratory research. There's good reason for this. Not only are PFAS everywhere around us; they're also in us. Humans can't break down PFAS, and our bodies struggle to clear them from our systems. Studies suggest they're in my blood and yours -- the majority of Americans,' in fact -- and they have been linked to increased risks of kidney and testicular cancer, decreased infant birthweights, and high blood pressure. And that's only what we know about now: researchers continue to grapple with the full impacts of PFAS on human and environmental health.

Read more here:
The Race To Destroy PFAS, the Forever Chemicals - Slashdot

18 Human Genetic Engineering – Clemson University

Melissa Nolan

By the end of this chapter, students should be able to:

Those beautiful blue eyes you inherited from your mother are actually a result of a complex science known as Genetics. The scientific field of genetics studies genes in our DNA. Genes are units of heredity transferred from a parent to offspring and determine some characteristic of offspring. Your genes are responsible for coding all of your traits- including hair color, eye color, and so on. In recent years, scientists began exploring the concept of gene editing, which is the deliberate manipulation of genetic material to achieve desired results. Gene editing can potentially alter any given trait in an organism- from height to hair texture to susceptibility for certain diseases.

Gene editing applied to humans is referred to as Human Genetic Engineering, or HGE. There is extensive debate in and out of the scientific community regarding the ethics of HGE. Much of this debate stems from how this technology will affect society, and vice versa. Individuals may harbor concerns about the rise of designer babies or scientists playing God by determining the traits of an individual. On the contrary, HGE presents potential cures to diseases caused by genetic mutations. Human Genetic Engineering (HGE) is a novel technology which presents various ethical concerns and potential consequences. HGE should be approached cautiously and with extensive governmental regulation given its history, its current state, and the potential it has to change the world in the future.

Genetic Encoding of Proteins by MIT OpenCourseWare is licensed under CC BY-NC-SA 2.0

HGE utilizes CRISPR/Cas9 gene editing tools to cut out specific genes and replace them with a newly designed gene.

HGE encompasses a variety of methods which all work to produce a deliberate change in the human genome. The most common and prevalent way to edit the human genome is via CRISPR/Cas9. CRISPR stands for clustered regularly interspaced short palindromic repeats, and Cas9 is a protein that functions as scissors to cut DNA/genes. The CRISPR/Cas9 system originally developed as a part of a bacterias immune system, which can recognize repeats in DNA of invading viruses, then cut them out. Since then, scientists have harnessed the CRISPR/Cas9 system to cut DNA sequences of their choice and then insert new DNA sequences in their place.

The CRISPR/Cas9 system allows for designer genomes, and rapid engineering of any cells programming. With the use of CRISPR/Cas9, scientists can cut out certain traits from an individuals cells and insert new traits into those same cells.

CRISPR Cas9 System by Marius Walter is licensed under CC-BY-SA-4.0

Gene therapy is a recently-developed technology which can be applied to both somatic and germline genome editing.

Gene therapy concepts were initially introduced in the 1960s, utilizing outdated methods, such as recombinant DNA technology and viral vectors, to edit microorganisms genomes. Recombinant DNA consists of genetic material from multiple sources. The first experiments involved transferring a genome from one bacteria to another via a viral vector. Soon after was the first successful transformation of human cells with foreign DNA. The success of the experiment prompted public concern over the ethics of gene therapy, and led to political regulation. In the gene therapy report of the Presidents Commission in the United States, germline genome editing was deemed problematic over somatic genome editing. Also, non-medical genome editing was deemed problematic over medical genome editing. Germline genome editing occurs when scientists alter the genome of an embryo, so that the entire organism has altered genes and the traits can be passed to offspring. Somatic genome editing involves editing only a few cells in the entire organism so that traits can not be passed down to offspring. In response to the report, the rDNA Advisory Committee of the National Institutes of Health was formed and proposed the first guidelines for the gene therapy clinical trials. This is an example of technological determinism, in which technology determines the development of its social structure and cultural values or regulations.

In the past few decades, gene editing has advanced exponentially, introducing state-of-the-art technologies such as the CRISPR/Cas9 system, which was developed to induce gene modifications at very specific target sites. Thus, gene editing became a major focus for medical research (Tamura, 2020). Gene editing has led to the potential for development of treatment strategies for a variety of diseases and cancers. So far, somatic genome editing has shown promise in treating leukemia, melanoma, and a variety of other diseases. In this way, HGE may be demonstrative of cultural determinism, in which the culture we are raised presents certain issues which necessitate the development of a specific technology.

DNA CRISPR Scissors by Max Pixel is licensed under CC0 1.0

CRISPR/Cas9 is the primary technology proposed for use in HGE. HGE presents a variety of pros and cons to society.

Somatic genome editing in HGE via the CRISPR/Cas9 system has proven to be effective at editing specific genome sites. Since 2015, genome editing technologies have been used in over 30 human clinical trials and have shown positive patient outcomes. The treatment of disease may be a positive benefit of HGE, but there are also various potential risks. Various forms of deliberative democracies formed in recent years to address scientific and ethical concerns in HGE. Deliberative democracies afrm the need to justify technological decisions made by citizens and their representatives with experts in the field via deliberation. Overall, the consensus remains that the pros and cons of HGE are not equivalent enough to justify widespread use of the technology.

Current human clinical trials show successful transformation of human immune cells to HIV-resistant cells. This implies that HGE may be the cure for HIV(Hu, 2019). Other successful somatic genome editing trials treated myeloma, leukemia, sickle cell disease, various forms of epithelial cancers, and hemophilia. Thus, gene editing has provided novel treatment options for congenital diseases and cancers (Tamaura, 2020). Congenital diseases are those present from birth, and typically have a genetic cause. For these reasons, scientific summits concluded HGE is ethical for research regarding somatic genome editing in congenital diseases and cancers.

There are many safety concerns regarding CRISPR applications, mainly in germline genome editing. As a result of technological determinism, a leading group of CRISPR/Cas9 scientists and ethicists met for the international Summit on Human Gene Editing. The summit determined that heritable genome research trials may be permitted only following extensive research on risks and benefits of HGE. However, the summit concluded that federal funding cannot be used to support research involving human embryos with germline editing techniques. These decisions were made to avoid potential risks such as the following.

The major concerns regarding germline genome editing in HGE include: serious injury or disability, a blurry line between therapeutic applications of HGE and medical applications, misapplications, potential for eugenics ( the study of how to arrange reproduction within a human population to increase the occurrence of heritable characteristics regarded as desirable), and inequitable access to the technology.

HGE is a complex technology which presents a variety of risk factors for the coming decades. Deliberative democracy is necessary to keep this technology in check, ethically.

The future of HGE is uncertain and requires immense forethought. The American Society of Human Genetics workgroup developed a position statement on human germline engineering. The statement argues that it is inappropriate to perform germline gene editing that culminates in human pregnancy; and that
in vitro(outside of an organism) germline editing should be permitted with appropriate oversight. It also states future clinical human germline editing requires ethical justification, compelling medical rationale, and evidence that supports its clinical usage. Many of these decisions were made based on the potential concerts over the future possibilities of the technology.

At the societal level, there may be concerns related to eugenics, social justice, and accessibility to technology. Eugenics could potentially reinforce prejudice and enforce exclusivity in certain physical traits. Traits can be preselected for, thus labeling some as good and others as unfavorable. This may perpetuate existing racist ideals, for example.

Moreover, germline genome editing may also increase the amount of inequality in a society. Human germline editing is likely to be very expensive and access may be limited to certain geographic regions, health systems, or socioeconomic statuses. Even if human genetic engineering is only used for medical purposes, genetic disease could become an artifact of class, location, or ethnic group. Therefore, preclinical trials are necessary to establish validity, safety, and efficacy before any wide scale studies are initiated.

Others argue that HGE may lessen genetic diversity in a human population, creating a biological monoculture that could lead to disease susceptibility and eventual extinction. Analyses have predicted that there will be negligible effect on diversity and will more likely ensure the health and longevity of humans (Russel, 2010). Legacy thinking may be responsible for the hesitations towards continuing forward with HGE, as there are also many potential pros for genetic engineering. Legacy thinking is using outdated thinking strategies and actions which may not be useful anymore.

In an alternative modernity, we can imagine HGE as an end-all for most congenital diseases and cancers. Moreover, it may be used in germline gene editing to prevent certain birth defects or heritable diseases. So, although HGE has a variety of potential risk factors, there is also great promise for novel medical therapies in the coming decades. The continued use of this technology should be approached cautiously and with extensive governmental regulation, allowing for research regarding its medical applications only.

In 2016, germline gene editing was proven feasible and effective in chickens by leading researchers in genetic engineering, Dimitrov and colleagues. In this study, scientists used CRISPR/Cas9 to target the gene for an antibody/ immunoglobulin commonly produced in chickens. Antibodies are proteins produced in immune response. In the resulting population, the chickens grew normally and healthily with modified antibodies which conferred drug resistance. This study was the first to prove that germline editing is both feasible and effective.

HGE is a rapidly expanding field of research which presents novel possibilities for the coming decades. HGE utilizes CRISPR/Cas9 gene editing tools to cut out specific genes and replace them with a newly designed gene. As important as this technology is, it is also important to recognize how new it is. Gene therapy research began in the 1960s, with somatic cell editing only commencing in the past two decades. This has presented many advantages for the potential treatment of congenital diseases, but also presents various risks. Those risks stem from germline gene editing and include eugenics and inequitable access to the technology creating large socio economic divides. In the future, more regulation should be placed on the advancement of HGE research before larger-scale studies take place.

1. What is the primary technology proposed for use in HGE?

A. Recombinant DNA technology

B. CRISPR/Cas9

C. Bacterial Transformation

D. Immunoglobulin

2. When was gene therapy concepts first introduced?

A. 1920s

B. 1940s

C. 1960s

D. 1980s

3. What is a major ethical concern regarding HGE addressed in this chapter?

A. Potential for ageism

B. Gene editing is only 50% effective

C. HGE can only be used in Caucasians

D. Potential for eugenics

Answers:

Baltimore, D. et. al.(2015). A prudent path forward for genomic engineering and germline gene modification. Science. https://doi.org/10.1126/science.aab1028

Brokowski, C., & Adli, M. (2019). CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool. Journal of Molecular Biology. https://doi.org/10.1016/j.jmb.2018.05.044

Cong, L., Ran, F., & Zhang, F. (2013). Multiplex Genome Engineering Using CRISPR/Cas9 Systems. Science. https://doi.org/10.1126/science.1231143

Dimitrov, L., et. al. (2016). Germline Gene Editing in Chickens by Efficient CRISPR-Mediated Homologous Recombination in Primordial Germ Cells. Plos One. https://doi.org/10.1371/journal.pone.0154303

Hu, C. (2019). Safety of Transplantation of CRISPR CCR5 Modified CD34+ Cells in HIV-Infected Subjects with Hematological Malignancies. U.S National Library of Medicine. https://clinicaltrials.gov/ct2/show/NCT03164135

Ormond, K., et. al.(2017). Human Germline Genome Editing. AJHG. https://doi.org/10.1016/j.ajhg.2017.06.012

Russell P.(2010) The Evolutionary Biological Implications of Human Genetic Engineering, The Journal of Medicine and Philosophy: A Forum for Bioethics and Philosophy of Medicine. https://doi.org/10.1093/jmp/jhq004

Tamura, R., & Toda, M. (2020). Historic Overview of Genetic Engineering Technologies for Human Gene Therapy. Neurologia medico-chirurgica. https://doi.org/10.2176/nmc.ra.2020-0049

Thomas, C. (2020). CRISPR-Edited Allogeneic Anti-CD19 CAR-T Cell Therapy for Relapsed/Refractory B Cell Non-Hodgkin Lymphoma. ClinicalTrials. https://clinicaltrials.gov/show/NCT04637763

Go here to see the original:
18 Human Genetic Engineering - Clemson University

Genetic Engineering: A Serious Threat to Human Society

By Zachary Rom|Considering Another Side Essays

Scientists have been trying to create synthetic life, life created in lab, for many years. The first breakthrough in this process happened about thirty years ago when genetic engineers began to genetically modify organisms (Savulescu). These engineers physically move genes across species in order to improve an organism or to cause an organism to function differently. Even though this process sounds as if it happens only in fantasy games, genetically modified organisms are common. For example, genetically modified crops are used every day in the worlds food supply and genetically modified bacteria have been used in medicine, chemical manufacturing, and bio warfare (Pickrell). Slowly, genetic engineering has become a powerful tool in many different fields. Recently, genetic engineerings potential power increased when Craig Venter, a famous geneticist and entrepreneurs, recreated a living organism out of synthetic chemicals. His success proved to genetic engineers that functioning genomes can be made purely of synthetic chemicals. This power would allow genetic engineers to build new artificial genomes instead of having to modify naturally existing genomes. Genetic engineers now have the chance to broaden their fields applications. However, genetic engineering is unpredictable and dangerous, and broadening the application of genetic engineering only furthers the risks. Genetically engineered organisms pose lethal and economic risks to human society.

The availability of genomic information and genetic engineering technology creates a lethal threat to humanity because terrorists can use both the information and technology to recreate deadly pathogens, such as the poliovirus. The naturally occurring poliovirus killed and paralyzed millions of people for many years. In 1988, a worldwide vaccination campaign against the virus nearly exterminated it from the environment, and this solved the poliovirus epidemic. However, in 2002, well intentioned scientists decided to recreate the poliovirus for research means. Using the genomic sequence of the poliovirus found on a public database and commercially available machines, these scientists synthesized fragments of viral genomes into a functional poliovirus (Avise 7). These scientists proved that deadly pathogens can be recreated from genetic engineering techniques. Also, the information and technology used in genetic engineering is readily available and relativity cheap (Kuzma and Tanji 3). Mixing the power to recreate a deadly pathogen with the public availability of genetic engineering information and technology creates a lethal risk to humanity when terrorist exist in society. Terrorist could use genetic engineering to reinstate the poliovirus into the environment, and the virus would kill and paralyze more people. Luckily, these scientists were filled with good intent; however, there is nothing to prevent terrorists from harming innocent lives. Recreating deadly pathogens makes genetic engineering dangerous enough; however, genetic engineers also have the potential to improve the effectiveness of deadly pathogens, such as Y. pestis.

Genetic engineers can make deadly pathogens, such as Y. pestis, resistant to modern antibiotics, and these pathogens could kill innocent people if used as a weapon. Y. pestis, also known as the black plague, wreaked havoc on humanity during the Middle Ages by killing millions of people. In response to a Y. pestis threat during the 20th century, scientists developed an effective vaccine for the pathogen. However, genetic engineers at Biopreparat, a Russian biological warfare agency, engineered a new Y. pestis strain with genetic resistance to modern antibiotics and natural human immunity (Avise 6). The genetically engineered Y. pestis was more deadly and effective than the natural Y. pestis that killed millions of people during the Middle Ages. Biopreparats research proved that deadly pathogens can be genetically engineered into superior forms that are resistant to modern medicine. If this strain of Y. pestis was released, a black plague would devastate current human society. Militaries could use the same genetic engineering techniques that Biopreparat used to create deadly biological weapons. With this ability to make deadly pathogens resistant to modern medicine, genetically engineered organisms become lethal weapons that cannot be stopped. Other than lethal weapons, genetically engineered organisms can produce lethal chemical compounds when they are used as a manufacturing tool in the chemical industry.

Showa Denkos genetically modified bacteria produced a lethal L-tryptophan amino acid that killed and disabled people who took the companys food supplements. In 1989, an epidemic of eosinophilia myalgia syndrome, a syndrome that is characterized by a high eosinophil count and severe muscle pain, struck the United States (Genetic Engineering: Too Good to Go Wrong 9). This epidemic killed a hundred people and physically disabled ten thousand patients, some of which were paralyzed. Doctors eventually discovered that L-tryptophan, an amino acid used as a food supplement, was causing the epidemic. In 1990, the Journal of the American Medical Association reported that only people who took the L-tryptophan supplement made by Showa Denko, a Japanese biotech company, came down with EMS. Showa Denkos genetically engineered organisms produced corrupted forms of L-tryptophan that were dangerous to human health (Smith 4).

Many chemical companies want to use genetically engineered organisms to produce chemicals because it is cheaper than normal manufacturing methods. If chemical companies begin to rely on genetically engineered organisms to produce food and medical chemicals, the public could be at risk for another dangerous outbreak of lethal chemicals. Using genetically engineered organisms to cutting down manufacturing costs seems as if it will help the economy; however, genetically engineered organisms, specifically anti-material organisms, can hurt economies more than help them.

Genetic engineers possess the ability to create anti-material organisms that can degrade infrastructure and man-made materials, and malicious people can use these organisms to tear down societys infrastructures and economies. In nature, there are many organisms with the ability to degrade infrastructure and man-made materials. These microbes cost governments and industries millions of dollars in biodeterioration and biodegradation damages. For instance, bacteria are the leading cause of road and runway deterioration. In Houston, Texas, microbes have been known to degrade the concrete in the citys sewage systems, and the city has spent millions of dollars trying to contain the problem. High-tech companies, such as airlines and fuel companies, constantly have their facilities and machinery being degraded away by anti-material organisms. These natural organisms cause enough damage to infrastructure, and fixing the damage is expensive and time consuming (Sunshine Project 2). Similarly to the artificially made poliovirus, genetic engineers have the potential to recreate or improve these naturally occurring anti-material organisms. In theory, malicious people could unleash genetically engineered anti-material organisms on infrastructures worldwide, and this would create an expensive cleanup project for governments and companies. With these expensive damages, genetically engineered organisms can destroy economies. The same economic and environmental dangers of anti-material organisms can also be seen in genetically modified crops.

Genetically modified crops will negatively impact the economy and environment because engineered genetic resistance is ineffective at stopping natural parasites in the long term. Farmers use genetically modified crops because these crops contain a genetic resistance to parasites, such as insect pests and microbes. In evolution, two organisms that are in a parasitic relationship evolve in a balance with each other. When genetically modified plan
ts are placed into a natural environment, parasites will evolve in a direction that allows them to bypass the genetic resistance engineered into the crops. Since the majority of crop parasites go through successive generations at a fast pace, these parasites will quickly evolve into a population that can surpass the genetic resistance. This evolutionary process makes the benefits of genetically modified crops short lived. Farmers, who pay more for genetically modified seed than natural seed, then have to pay for harmful and expensive pesticides to protect their crops. In the end, farmers will lose money due to the increased costs of buying genetically modified crops and dangerous pesticides. Also, dangerous chemicals, such as DDT, will be reintroduced into the environment (Avise 73). The ineffectiveness of genetically modified crops creates an economic and environmental risk to human society in the long run since farmers will be losing more money and introducing dangerous chemicals into the environment.

Genetically engineered organisms pose an enormous risk to human society on a lethal and economic front. Natural lethal pathogens, such as the poliovirus and Y. pestis, can be recreated or improved, and malicious people could use these genetically engineered pathogens to kill millions of people. Chemicals manufactured by genetically modified bacteria have proven to be harmful to human health, which was the case during the EMS epidemic in the United States. On an economic front, genetically engineered organisms increase costs instead of minimizing them, and they harm the environment. Anti-material organisms can be created to deteriorate infrastructures, and this would cost governments and industries millions of dollars in repair costs. Also, genetically modified crops in the long term will cost farmers more money than they save because the advantages of the genetically modified crops will be nullified by evolving parasites. Genetically engineered organisms have a huge potential to harm society. However, researching new methods and applications of genetic engineering will not stop because scientists believe in the vast opportunities of the field. In order to keep human society safe, scientists must exhaust all options before turning to the power of genetic engineering. It is an unwise idea to rely on genetic engineering since it is unpredictable and imprecise form of engineering.

Visit link:
Genetic Engineering: A Serious Threat to Human Society

Genetic Engineering and Ethics: Are We Ready? | The Voice

Written by Audrey Eaves

Advancements in science and technology have enabled the possibility of human genetic cloning and engineering. In contemporary society, these biological technologies are controversial. Many governmental, scientific, and religious organizations are fervently opposing genetic engineering due to controversy in the context of safety and moral outcomes. Nevertheless, advocates and supporters argue that these technologies are fundamental to providing remedies via regenerative medicine through genetically identical human cells, organs, or tissues. Other health areas such as cosmetic and reconstructive surgeries, infertility, burn treatments, heart disease, cancer, and diabetes can benefit from the new technologies available through gene therapies. Gene therapy can help millions suffering from disease and disorders. Biomedical researchers are working on effective solutions regarding some major genetic disorders such as sickle-cell and hemophilia, but there are always risks.

Genetic engineering certainly has its dilemmas, but it also has a moral and ethical value in contemporary society, therefore, a new branch of ethics is born: bioethics. Bioethics refers to the application of medical and biological sciences in appropriate, humane, and responsible ways. Supporters see genetic engineering and cloning as a viable way to duplicate organs and tissues for patients who otherwise would not be able to find transplants and could escape lifetimes of medications with undesirable side effects. Yet, some are concerned that if done incorrectly, genetic engineering could actually introduce new disorders that would subsequently circulate in the population and thus become a permanent aspect of the worlds population.

The majority of biomedical researchers view genetic engineering as a crucial tool for medicine, especially in the provision of solutions for diverse terminal health issues. Consider these daunting statistics from Kidney.org: the average wait time for a needed kidney is three to five years, and some patients cannot wait that long. According to another source, Donate Life America, 8,000 people die every year waiting for an organ, 80% of which are kidneys. However, in a world where slavery, human organ harvesting, and black markets continue to be a problem, genetic engineering and cloning could provide even darker opportunities for these human rights crimes. A realistic approach in the context of humanitys place in the world and a code of ethics to form the foundation of human genetic engineering practices is needed.

Religious factions are by no means the only moral compass of society, but they tend to be the loudest sounding alarms of anything that is morally questionable. While their objections sometimes (but not always) deviate from science and can frustrate progressive efforts, they provide a necessary role in a symbiotic system of checks and balances within the scientific communities they oppose. It is constructively beneficial that science should always be questioned and forced to prove itself before diving headfirst into the deep waters of the latest and greatest technological discoveries.

Embryonic engineering and cloning in particular draws criticism from people of various faiths who argue that the creation of embryos for the purposes of research does not respect life. A number of religious faiths assert that embryos should be assigned personhood. This particular characterization disarms objectification practices that are currently in place regarding human embryos. During the process of embryonic research, excess embryos are created and destined for destruction, which is another challenge for bioethics. However, this is nothing new, as the process of IVF does similarly for couples who struggle with infertility. Matters of human wastefulness always arise in these waters. Even with natural pregnancies, research shows that half of the embryos fail to implant or are lost. While embryonic loss does occur in natural pregnancies, most people do not equate laboratory embryonic loss with infant mortality, which implies they have a different moral value to most of society. Does regarding human embryos as mere objects that can be used in any desirable way make them lack the nascent aspect of human life and significance? Whatever side one falls on the argument, it is vital to encourage the cultivation of a society that views life as having great intrinsic value. This understanding and respect for life creates the difference between barbarism and civilization.

There are yet other faiths who place great spiritual importance on what goes inside their bodies. This can apply both to what is in their food as well as medical treatments. For these groups, there could be a moral dilemma posed by significant genetic modification of food and medicine. For example, various genes are being injected into peppers and tomatoes to make them grow faster and more hearty. Animal and human cells are also used in the production of some vaccines. This raises the question of how many human and animal genes can be present in vegetables or medicine without it being considered unsuitable for vegans or the millions of religious adherents who abstain from certain animal and human by-products, such as with Islam, Jehovahs Witnesses, and Judaism. While these unique groups of people are ultimately responsible for their own decisions, sensitivity to diverse belief systems must be a consideration of the scientific community as well.

Despite all the current ethical concerns regarding genetic engineering and human cloning, the practice still has tremendous potential in light of more conclusive scientific research studies on this particular subject. However, the challenges experienced in past genetic experiments should be a major factor in discouraging a rushed start of biogenetics. More research should be developed to review the ethical and moral considerations in genetic engineering practices. A full understanding of what we are doing and its consequences needs some time to catch up with the technology. Most important is the conviction and cultivation of a society that protects and enhances life in all of its scientific endeavors.

The rest is here:
Genetic Engineering and Ethics: Are We Ready? | The Voice

Fully Human Ovarian Organoid That Supports Egg Cell Maturation Created …

Scientists at the Wyss Institute for Biologically Inspired Engineering at Harvard University, Harvard Medical School (HMS), and Duke University, in collaboration with Gameto, report that they have created a living, fully human ovarian organoid that supports egg cell maturation, develops follicles, and secretes sex hormones. This ovaroid model enables the study of human ovarian biology without the need to take tissue from patients and could enable the development of new treatments for conditions like infertility, ovarian cancer, and more, according to the researchers.

Through an agreement with Harvards Office of Technology Development (OTD), the technology has been licensed to Gameto, which is using it to develop therapeutics for diseases of the female reproductive system. The ovaroids are described in detail Directed differentiation of human iPSCs to functional ovarian granulosa-like cells via transcription factor overexpression in eLife.

An in vitro model of human ovarian follicles would greatly benefit the study of female reproduction. Ovarian development requires the combination of germ cells and several types of somatic cells. Among these, granulosa cells play a key role in follicle formation and support for oogenesis. Whereas efficient protocols exist for generating human primordial germ cell-like cells (hPGCLCs) from human induced pluripotent stem cells (hiPSCs), a method of generating granulosa cells has been elusive, write the investigators.

Here, we report that simultaneous overexpression of two transcription factors (TFs) can direct the differentiation of hiPSCs to granulosa-like cells. We elucidate the regulatory effects of several granulosa-related TFs and establish that overexpression of NR5A1 and either RUNX1 or RUNX2 is sufficient to generate granulosa-like cells. Our granulosa-like cells have transcriptomes similar to human fetal ovarian cells and recapitulate key ovarian phenotypes including follicle formation and steroidogenesis.

When aggregated with hPGCLCs, our cells form ovary-like organoids (ovaroids) and support hPGCLC development from the premigratory to the gonadal stage as measured by induction of DAZL expression. This model system will provide unique opportunities for studying human ovarian biology and may enable the development of therapies for female reproductive health.

Our new method of fully human ovaroid production is several times faster than existing human/mouse hybrid methods, and replicates many of the critical functions of these organs, marking a significant step forward in our ability to study female reproductive health in the lab. In the future, similar technology could also treat infertility by growing egg cells from peoplewhose own eggs arent viable, said co-first author Merrick Pierson Smela, a graduate student in the lab of George Church, PhD, at the Wyss Institute and HMS.

Creating the granulosa cells on their own was a significant accomplishment, but making an ovaroid out of only granulosa cells wouldnt tell us anything about their ability to support the maturation of germ cells, which was what we wanted to be able to studyin vitro, said co-first author Christian Kramme, PhD, the vice president of cell engineering at Gameto and a former graduate student in Churchs group at the Wyss Institute and HMS. This process had been replicated previously using hPGCLCs and mouse somatic cells, but with this new technology, we now have the ability to do it with a fully human model.

The Wyss team is continuing to develop its human ovaroid model and plans to integrate additional ovarian cell types, including hormone-producing theca cells, to more fully replicate the complex functions of the human ovary. They also hope to improve their culture system to allow their germ cells to fully develop into egg cells, and determine the optimal dosage of the different TFs. Gameto, meanwhile, has conducted preclinical studies of a derived co-culture system for egg maturation in humans with leading national fertility clinics.

View post:
Fully Human Ovarian Organoid That Supports Egg Cell Maturation Created ...

Genetic Engineering with Dr. Nagase – by Daniel Nagase MD

Here we will look at how the mRNA sequences for pfizer and modernas COVID'-19 injection can cause problems with the human gene Line-1.

Primer on Line-1 Current till 2022

First lets look at the code of Line-1:

Line-1 Accession: L19088.1

(Sequence taken from the national library of medicine - where you can find multiple versions and fragments of LINE-1. I picked the longest version, as thats least likely to have missing parts that were cropped during isolation of the gene. https://www.ncbi.nlm.nih.gov/nuccore/?term=Human+LINE1)

Id paste the whole thing but the part were interested is at the end.

6001 isnt the year yet, its the base pair number. (Each group is 10 base pairs, except the last batch in this cut and paste which is 9.) The start of the gene is base pair 1, and it goes all the way to base pair 6059 for the gene Line-1. (A group of 3 base pairs forms a codon which can code for an amino acid. Chain a bunch of amino acids together and voila! A protein!)

So whats so particular about the end of Line-1?

We have 37 a s in a row. Why is that important? Because the moderna and pfizer mRNA injections for COVID have something very similar.

(a stands for adenine in DNA. t stands for thymine, g is guanine, and c is cytosine For a short explanation of DNA and RNA please check out Dr. Syed Haiders substack where one of my dear readers found the article that I needed to complete this one:)

Dr. Syed Haider

If I had finished this article earlier, I would have been missing this key piece, so thank you College of Physicians and Surgeons of British Columbia, for delaying my article but making it better in the process!

Fight with Medical College Lawyers

So back to the Human Gene Line-1, it makes up 17-20% of the Human Genome.

Now if we look at Modernas sequence here:

Moderna Covid-19 mRNA (Elasomeran)

And then Pfizers mRNA for COVID-19:

Pfizer Sequence - BNT-162B2

70 a s preceeded by gcauaugac. (u in the moderna and pfizer isnt true uracil - a nucleotide component that makes up RNA. It is methyl pseudo uracil, an artificial modified version made to prevent cells from destroying the spike protein mRNA.)

Well if either the pfizer or moderna versions of the spike protein mRNA are reverse transcribed, then that long chain of a s will turn into a long chain of t s that would base pair (attach) to any gene with a long tail of a s like Line-1.

AND theres many copies of Line-1 throughout the human genome.

So 17-20% of the human genome could be targeted because pfizer and moderna put a long tail of a s on the ends of their mRNA?

Thats exactly what I was thinking

Well, I wasnt quite sure. I had my suspicions, but no scientific article that could quite make those suspicions suspiciously suspect. Thats when the study found by one of my readers in Dr. Sayed Haiders substack baked the cake.

Line-1 and Poly-a

Now thanks to this fresh study by Rudolf Jaenisch and Liguo Zhang, I had evidence the proteins made by the Line-1 gene had an affinity for Poly-a that is the long chains of a s, coincidentally also found in Modernas and Pfizers COVID mRNA injections. These Poly-as are also in the Line-1 gene itself. When a Line-1 mRNA with a long Poly-a is in the cytoplasm (outside the nucleus) the L1ORF2p proteins made by Line-1 preferentially bind to the poly-A stretch at the end of the LINE1 mRNA, AND CARRY IT INTO THE NUCLEUS!

Because what happens if L1ORF2p proteins that bind to the Poly-a stick to the long stretch of a 's in the Pfizer and Moderna Spike Protein mRNA?

AND THEN CARRY THAT INTO THE NUCLEUS?!?

OMFG

The Pfizer and Moderna spike protein mRNAs already resist breakdown within the cytoplasm because of their engineered 5 Cap and their Methyl pseudo uracil nucleotides resist exonucleases. They already live longer than natural mRNAs.

Now theres a mechanism (Line-1 ORF1 and ORF2 proteins) to take them into the nucleus?

AND that mechanism has a reverse transcriptase AND an endonuclease to insert it into DNA?

Accidental Engineering?

Geoengineering?

or

Genetic Engineering.

Theres a Discrepancy! (in the study)

In the February 13th article about Line-1 and Poly-a, they find that the SARS-CoV-2 virus likes to make Poly-a tails as well. They found the virus had Poly-a tails on its Nucleocapsid mRNA and that it integrated into the DNA of cells infected with the SARS-CoV-2 virus.

Then they did another experiment where they took just the mRNA for the nucleocapsid and transfected it into cells. They didnt use the whole virus as would be the case in an infection. What they found was transfection did not result in DNA integration of viral genes. (Insertion of virus genes into the DNA)

Transfection is what happens when you take Pfizer or Modernas COVID-19 injection! Lipid nanoparticles transfect your cells with Spike protein mRNA. They dont infect your cells with SARS-CoV-2 like youd get from standing too close to someone without a maskright? (So this experiment showed that a transfection like getting an mRNA injection didnt alter DNA right?)

Not quite

NOT

This studys authors dont go into how long the Poly-as are in a virus infection, but the original Wuhan strain it looks like it has a 33 base pair Poly-a tail.

SARS-CoV-2 Genome (Original Wuhan)

They came to the conclusion that transfection didnt cause viral genes to get integrated into a cells DNA whereas an infection with SARS-CoV-2 did?

Are they trying to say the virus changes the DNA more than a transfection vaccine using mRNA?

But the Poly-a tail they used in their transfection experiment was 25% SHORTER than the Poly-a tail in the SARS-CoV-2 virus experiment!

Whats even worse is that the transfection Poly-a tail is 32% shorter than Line-1s natural Poly-a tail, and 75% shorter than the Pfizer Spike Protein Poly-a tail.

What is wrong with them?

Arent they comparing Apples to Bicycles?

Why do an OK experiment, when for the same amount of time and nucleotide you could do a TITANIC experiment?

Forget about nucleocapsid protein. Forget about someones donated pUC57-2019-ncov plasmid, a kind gift from Christine A. Roden from the Amy S. Gladfelter laboratory (University of North Carolina at Chapel Hill).

GO TO A VACCINE CLINIC AND BORROW SOME Pfizer and Moderna mRNA!

You know the ones with 100 base pair and 70 base pair Poly-a TAILS?

What are they afraid of?

Growing spike proteins in a dish?

They know how to wear gloves right?

They know how to work under a biohazard hood right?

It does not make sense to do a pancake mix experiment when for the same time and $ they could have done the Pompeii of all experiments.

Discrepancy Analysis (Heuristics)

The clue. The very suspicious clue is the name of one of the studys authors, Rudolf Jaenisch.

I dont know the guy. Ive never met him. Im thinking hes a great guy who knows a thing or two about cell biology.

So why the suspicion?

Do I think a serious cell biologist like him is afraid of spike proteins?

Not really. I think Rudolf Jaenisch might be afraid of a different kind of spike. The kind of lead spike thats attached to a brass casing with flammable powder.

Let me explain.

The only reason I know the name Rudolf Jaenisch is because Dr. Robert Malone trash talked him during an interview I did in November 2021.

Watch the video:

9:45 Dr. Malone: "I work closely with government."

10:43 Dr. Malone: "I was alerted by a CIA officer..."

40:30 Dr. Nagase: Backstory.

45:30 Dr. Nagase: Cancer and reverse transcriptase

47:03 Dr. Malone: drops off call

57.53 Dr. Malone: comes back cautioning against speculating about reverse transcriptase.

58:30 Dr. Malone: "We're under intense pressure... we have to be super careful about our messenging and what we're stating...not useful to specu
late about things like integration (of DNA from reverse transcribed RNA)

59:26 Malone: "I really think one does need to be a little cautious about interpreting some of these papers like the PNAS paper regarding reverse transcriptase by Rudy Jaenisch, WHO HAS A MULTI DECADE HISTORY OF OVER INTERPRETING RETRO VIROLOGY AND PUBLISHING IRREPRODUCIBLE FINDINGS. So that's my parting gentle comment is that we do have to be really careful not to provide opportunities for our haters to attack us."

Malone trashing Rudy Jaenisch

Why is the inventor of mRNA technology trashing another cell biologist?

Dr. Malone:

Works closely with government?

Was alerted by a CIA officer about Wuhan?

Trash Talks his Cell Biology buddy Rudolf Jaenisch?

Maybe Rudolf Jaenisch could have done the experiment with the COVID mRNA injections instead of donated nucleocapsid RNA. (Maybe he did do the same experiment with Pfizer and Moderna)

But to save his life, and not end up like JFK, he didnt publish it.

Post Script: If it indeed was the case that Spike protein mRNA was deliberately gene edited into people, theoretically it would be possible to do the reverse. That is gene edit it out of people. The question then would be what do we gene edit it out with?

My first idea was use Line-1 itself to Edit Out spike protein genes. But Line-1 itself isnt 100% benign, as it has been thought to have a role sometimes in cancers. However, an extra copy of natural Line-1 might be better than an unnatural copy of "spike protein.

Post Post Script: You can hear Dr. Malone in the back ground at 1:23 trying to talk over Dr. Weismann because hes getting into Uncomfortable territory. (fyi, Dr. Weismann is way smarter than me.)

Dr. Weismann vs Dr. Malone at 1:23

Read more here:
Genetic Engineering with Dr. Nagase - by Daniel Nagase MD

Human enhancement: Genetic engineering and evolution – OUP Academic

Abstract

Genetic engineering opens new possibilities for biomedical enhancement requiring ethical, societal and practical considerations to evaluate its implications for human biology, human evolution and our natural environment. In this Commentary, we consider human enhancement, and in particular, we explore genetic enhancement in an evolutionary context. In summarizing key open questions, we highlight the importance of acknowledging multiple effects (pleiotropy) and complex epigenetic interactions among genotype, phenotype and ecology, and the need to consider the unit of impact not only to the human body but also to human populations and their natural environment (systems biology). We also propose that a practicable distinction between therapy and enhancement may need to be drawn and effectively implemented in future regulations. Overall, we suggest that it is essential for ethical, philosophical and policy discussions on human enhancement to consider the empirical evidence provided by evolutionary biology, developmental biology and other disciplines.

Lay Summary: This Commentary explores genetic enhancement in an evolutionary context. We highlight the multiple effects associated with germline heritable genetic intervention, the need to consider the unit of impact to human populations and their natural environment, and propose that a practicable distinction between therapy and enhancement is needed.

There are countless examples where technology has contributed to ameliorate the lives of people by improving their inherent or acquired capabilities. For example, over time, there have been biomedical interventions attempting to restore functions that are deficient, such as vision, hearing or mobility. If we consider human vision, substantial advances started from the time spectacles were developed (possibly in the 13th century), continuing in the last few years, with researchers implanting artificial retinas to give blind patients partial sight [13]. Recently, scientists have also successfully linked the brain of a paralysed man to a computer chip, which helped restore partial movement of limbs previously non-responsive [4, 5]. In addition, synthetic blood substitutes have been created, which could be used in human patients in the future [68].

The progress being made by technology in a restorative and therapeutic context could in theory be applied in other contexts to treat non-pathological conditions. Many of the technologies and pharmaceutical products developed in a medical context to treat patients are already being used by humans to enhance some aspect of their bodies, for example drugs to boost brain power, nutritional supplements, brain stimulating technologies to control mood or growth hormones for children of short stature. Assistive technology for disabled people, reproductive medicine and pharmacology, beside their therapeutic and restorative use, have a greater potential for human enhancement than currently thought. There are also dual outcomes as some therapies can have effects that amount to an enhancement as for example, the artificial legs used by the South African sprinter Oscar Pistorius providing him with a competitive advantage.

This commentary will provide general ethical considerations on human enhancement, and within the several forms of so-called human biomedical enhancement, it will focus on genetic engineering, particularly on germline (heritable) genetic interventions and on the insights evolutionary biology can provide in rationalizing its likely impact. These insights are a subject often limited in discussions on genetic engineering and human enhancement in general, and its links to ethical, philosophical and policy discussions, in particular [9]. The rapid advances in genetic technology make this debate very topical. Moreover, genes are thought to play a very substantial role in biological evolution and development of the human species, thus making this a topic requiring due consideration. With this commentary, we explore how concepts based in evolutionary biology could contribute to better assess the implications of human germline modifications, assuming they were widely employed. We conclude our brief analysis by summarizing key issues requiring resolution and potential approaches to progress them. Overall, the aim is to contribute to the debate on human genetic enhancement by looking not only at the future, as it is so often done, but also at our evolutionary past.

The noun enhancement comes from the verb enhance, meaning to increase or improve. The verb enhance can be traced back to the vulgar Latin inaltiare and late Latin inaltare (raise, exalt), from altare (make high) and altus (high), literally grown tall. For centuries human enhancement has populated our imagination outlined by stories ranging from the myths of supernormal strengths and eternal life to the superpowers illustrated by the 20th century comic books superheroes. The desire of overcoming normal human capacities and the transformation to an almost perfect form has been part of the history of civilization, extending from arts and religion to philosophy. The goal of improving the human condition and health has always been a driver for innovation and biomedical developments.

In the broadest sense, the process of human enhancement can be considered as an improvement of the limitations of a natural version of the human species with respect to a specific reference in time, and to different environments, which can vary depending on factors such as, for example, climate change. The limitations of the human condition can be physical and/or mental/cognitive (e.g. vision, strength or memory). This poses relevant questions of what a real or perceived human limitation is in the environment and times in which we are living and how it can be shifted over time considering social norms and cultural values of modern societies. Besides, the impact that overcoming these limitations will have on us humans, and the environment, should also be considered. For example, if we boost the immune system of specific people, this may contribute to the development/evolution of more resistant viruses and bacteria or/and lead to new viruses and bacteria to emerge. In environmental terms, enhancing the longevity of humans could contribute to a massive increase in global population, creating additional pressures on ecosystems already under human pressure.

Two decades ago, the practices of human enhancement have been described as biomedical interventions that are used to improve human form or functioning beyond what is necessary to restore or sustain health [10]. The range of these practices has now increased with technological development, and they are any kind of genetic, biomedical, or pharmaceutical intervention aimed at improving human dispositions, capacities, or well-being, even if there is no pathology to be treated [11]. Practices of human enhancement could be visualized as upgrading a system, where interventions take place for a better performance of the original system. This is far from being a hypothetical situation. The rapid progress within the fields of nanotechnology, biotechnology, information technology and cognitive science has brought back discussions about the evolutionary trajectory of the human species by the promise of new applications which could provide abilities beyond current ones [12, 13]. If such a possibility was consciously embraced and actively pursued, technology could be expected to have a revolutionary interference with human life, not just helping humans in achieving general health and capabilities commensurate with our current ones but helping to overcome human limitations far beyond of what is currently possible for human beings. The emergence of new technologies has provided a broader range of potential human interventions and the possibility of transitioning from external changes to our bodies (e.g. external prosthesis) to internal ones, especially when considering genetic manipulation, whose changes can be permanent and transmissible.

The advocat
es of a far-reaching human enhancement have been referred to as transhumanists. In their vision, so far, humans have largely worked to control and shape their exterior environments (niche construction) but with new technologies (e.g. biotechnology, information technology and nanotechnology) they will soon be able to control and fundamentally change their own bodies. Supporters of these technologies agree with the possibility of a more radical interference in human life by using technology to overcome human limitations [1416], that could allow us to live longer, healthier and even happier lives [17]. On the other side, and against this position, are the so-called bioconservatives, arguing for the conservation and protection of some kind of human essence, with the argument that it exists something intrinsically valuable in human life that should be preserved [18, 19].

There is an ongoing debate between transhumanists [2022] and bioconservatives [18, 19, 23] on the ethical issues regarding the use of technologies in humans. The focus of this commentary is not centred on this debate, particularly because the discussion of these extreme, divergent positions is already very prominent in the public debate. In fact, it is interesting to notice that the moderate discourses around this topic are much less known. In a more moderate view, perhaps one of the crucial questions to consider, independently of the moral views on human enhancement, is whether human enhancement (especially if considering germline heritable genetic interventions) is a necessary development, and represents an appropriate use of time, funding and resources compared to other pressing societal issues. It is crucial to build space for these more moderate, and perhaps less polarized voices, allowing the consideration of other positions and visions beyond those being more strongly projected so far.

Ethical and societal discussions on what constitutes human enhancement will be fundamental to support the development of policy frameworks and regulations on new technological developments. When considering the ethical implications of human enhancement that technology will be available to offer now and in the future, it could be useful to group the different kinds of human enhancements in the phenotypic and genetic categories: (i) strictly phenotypic intervention (e.g. ranging from infrared vision spectacles to exoskeletons and bionic limbs); (ii) somatic, non-heritable genetic intervention (e.g. editing of muscle cells for stronger muscles) and (iii) germline, heritable genetic intervention (e.g. editing of the CC chemokine receptor type 5 (CCR5) gene in the Chinese baby twins, discussed later on). These categories of enhancement raise different considerations and concerns and currently present different levels of acceptance by our society. The degree of ethical, societal and environmental impacts is likely to be more limited for phenotypic interventions (i) but higher for genetic interventions (ii and iii), especially for the ones which are transmissible to future generations (iii).

The rapid advances in technology seen in the last decades, have raised the possibility of radical enhancement, defined by Nicholas Agar, as the improvement of human attributes and abilities to levels that greatly exceed what is currently possible for human beings [24]. Genetic engineering offers the possibility of such an enhancement by providing humans a profound control over their own biology. Among other technologies, genetic engineering comprises genome editing (also called gene editing), a group of technologies with the ability to directly modify an organisms DNA through a targeted intervention in the genome (e.g. insertion, deletion or replacement of specific genetic material) [25]. Genome editing is considered to achieve much greater precision than pre-existing forms of genetic engineering. It has been argued to be a revolutionary tool due to its efficiency, reducing cost and time. This technology is considered to have many applications for human health, in both preventing and tackling disease. Much of the ethical debate associated with this technology concerns the possible application of genome editing in the human germline, i.e. the genome that can be transmitted to following generations, be it from gametes, a fertilized egg or from first embryo divisions [2628]. There has been concern as well as enthusiasm on the potential of the technology to modify human germline genome to provide us with traits considered positive or useful (e.g. muscle strength, memory and intelligence) in the current and future environments.

To explore some of the possible implications of heritable interventions we will take as an example the editing (more specifically deletion using CRISPR genome editing technology) of several base pairs of the CCR5 gene. Such intervention was practised in 2018 in two non-identical twin girls born in China. Loss of function mutations of the CCR5 had been previously shown to provide resistance to HIV. Therefore, the gene deletion would be expected to protect the twin baby girls from risk of transmission of HIV which could have occurred from their father (HIV-positive). However, the father had the infection kept under control and the titre of HIV virus was undetectable, which means that risk of transmission of HIV infection to the babies was negligible [29].

From an ethical ground, based on current acceptable practices, this case has been widely criticized by the scientific community beside being considered by many a case of human enhancement intervention rather than therapy [29, 30]. One of the questions this example helps illustrate is that the ethical boundary between a therapy that corrects a disorder by restoring performance to a normal scope, and an intervention that enhances human ability outside the accepted normal scope, is not always easy to draw. For the sake of argument, it could be assumed that therapy involves attempts to restore a certain condition of health, normality or sanity of the natural condition of a specific individual. If we take this approach, the question is how health, normality and sanity, as well as natural per se, are defined, as the meaning of these concepts shift over time to accommodate social norms and cultural values of modern societies. It could be said that the difficulty of developing a conceptual distinction between therapy and enhancement has always been present. However, the potential significance of such distinction is only now, with the acceleration and impact of technological developments, becoming more evident.

Beyond ethical questions, a major problem of this intervention is that we do not (yet?) know exactly the totality of the effects that the artificial mutation of the CCR5 may have, at both the genetic and phenotypic levels. This is because we now know that, contrary to the idea of one gene-one trait accepted some decades ago, a geneor its absencecan affect numerous traits, many of them being apparently unrelated (a phenomenon also known as pleiotropy). That is, due to constrained developmental interactions, mechanisms and genetic networks, a change in a single gene can result in a cascade of multiple effects [31]. In the case of CCR5, we currently know that the mutation offers protection against HIV infection, and also seems to increase the risk of severe or fatal reactions to some infectious diseases, such as the influenza virus [32]. It has also been observed that among people with multiple sclerosis, the ones with CCR5 mutation are twice as likely to die early than are people without the mutation [33]. Some studies have also shown that defective CCR5 can have a positive effect in cognition to enhance learning and memory in mice [34]. However, its not clear if this effect would be translated into humans. The example serves to illustrate that, even if human enhancement with gene editing methods was considered ethically sound, assessing the totality of its implications on solid grounds may be difficult to achieve.

Beyond providing the opportunity of enhancing human capabilities in specific individua
ls, intervening in the germline is likely to have an impact on the evolutionary processes of the human species raising questions on the scale and type of impacts. In fact, the use of large-scale genetic engineering might exponentially increase the force of niche construction in human evolution, and therefore raise ethical and practical questions never faced by our species before. It has been argued that natural selection is a mechanism of lesser importance in the case of current human evolution, as compared to other organisms, because of advances in medicine and healthcare [35]. According to such a view, among many others advances, natural selection has been conditioned by our niche-construction ability to improve healthcare and access to clean water and food, thus changing the landscape of pressures that humans have been facing for survival. An underlying assumption or position of the current debate is that, within our human species, the force of natural selection became minimized and that we are somehow at the end-point of our evolution [36]. If this premise holds true, one could argue that evolution is no longer a force in human history and hence that any human enhancement would not be substituting itself to human evolution as a key driver for future changes.

However, it is useful to remember that, as defined by Darwin in his book On the Origin of the Species, natural selection is a process in which organisms that happen to be better adapted to a certain environment tend to have higher survival and/or reproductive rates than other organisms [37]. When comparing human evolution to human genetic enhancement, an acceptable position could be to consider ethically sound those interventions that could be replicated naturally by evolution, as in the case of the CCR5 gene. Even if this approach was taken, however, it is important to bear in mind that human evolution acts on human traits sometimes increasing and sometimes decreasing our biological fitness, in a constant evolutionary trade-off and in a contingent and/or neutralin the sense of not progressiveprocess. In other worlds, differently from genetic human enhancement, natural selection does not aim at improving human traits [38]. Human evolution and the so-called genetic human enhancement would seem therefore to involve different underlying processes, raising several questions regarding the implications and risks of the latter.

But using genetic engineering to treat humans has been proposed far beyond the therapeutic case or to introduce genetic modifications known to already occur in nature. In particular, when looking into the views expressed on the balance between human evolution and genetic engineering, some argue that it may be appropriate to use genetic interventions to go beyond what natural selection has contributed to our species when it comes to eradicate vulnerabilities [17]. Furthermore, when considering the environmental, ecological and social issues of contemporary times, some suggest that genetic technologies could be crucial tools to contribute to human survival and well-being [2022]. The possible need to engineer human traits to ensure our survival could include the ability to allow our species to adapt rapidly to the rate of environmental change caused by human activity, for which Darwinian evolution may be too slow [39]. Or, for instance, to support long-distance space travel by engineering resistance to radiation and osteoporosis, along with other conditions which would be highly advantageous in space [40].

When considering the ethical and societal merits of these propositions, it is useful to consider how proto-forms of enhancement has been approached by past human societies. In particular, it can be argued that humans have already employedas part of our domestication/selective breeding of other animalstechniques of indirect manipulation of genomes on a relatively large scale over many millennia, albeit not on humans. The large-scale selective breeding of plants and animals over prehistoric and historic periods could be claimed to have already shaped some of our natural environment. Selective breeding has been used to obtain specific characteristics considered useful at a given time in plants and animals. Therefore, their evolutionary processes have been altered with the aim to produce lineages with advantageous traits, which contributed to the evolution of different domesticated species. However, differently from genetic engineering, domestication possesses inherent limitations in its ability to produce major transformations in the created lineages, in contrast with the many open possibilities provided by genetic engineering.

When considering the impact of genetic engineering on human evolution, one of questions to be considered concerns the effects, if any, that genetic technology could have on the genetic pool of the human population and any implication on its resilience to unforeseen circumstances. This underlines a relevant question associated with the difference between health and biological fitness. For example, a certain group of animals can be more healthyas domesticated dogsbut be less biologically fit according to Darwins definition. Specifically, if such group of animals are less genetically diverse than their ancestors, they could be less adaptable to environmental changes. Assuming that, the human germline modification is undertaken at a global scale, this could be expected to have an effect, on the distribution of genetically heritable traits on the human population over time. Considering that gene and trait distributions have been changing under the processes of evolution for billions of years, the impact on evolution will need to be assessed by analysing which genetic alterations have been eventually associated with specific changes within the recent evolutionary history of humans. On this front, a key study has analysed the implications of genetic engineering on the evolutionary biology of human populations, including the possibility of reducing human genetic diversity, for instance creating a biological monoculture [41]. The study argued that genetic engineering will have an insignificant impact on human diversity, while it would likely safeguard the capacity of human populations to deal with disease and new environmental challenges and therefore, ensure the health and longevity of our species [41]. If the findings of this study were considered consistent with other knowledge and encompassing, the impact of human genetic enhancements on the human genetic pool and associated impacts could be considered secondary aspects. However, data available from studies on domestication strongly suggests that domestication of both animals and plans might lead to not only decreased genetic diversity per se, but even affect patterns of variation in gene expression throughout the genome and generally decreased gene expression diversity across species [4244]. Given that, according to recent studies within the field of biological anthropology recent human evolution has been in fact a process of self-domestication [45], one could argue that studies on domestication could contribute to understanding the impacts of genetic engineering.

Beyond such considerations, it is useful to reflect on the fact that human genetic enhancement could occur on different geographical scales, regardless of the specific environment and geological periods in which humans are living and much more rapidly than in the case of evolution, in which changes are very slow. If this was to occur routinely and on a large scale, the implications of the resulting radical and abrupt changes may be difficult to predict and its impacts difficult to manage. This is currently highlighted by results of epigenetics studies, and also of the microbiome and of the effects of pollutants in the environment and their cumulative effect on the development of human and non-human organisms alike. Increasingly new evidence indicates a greater interdependence between humans and their environments (including other microorganisms), indicating that modifying the environment can have direct an
d unpredictable consequences on humans as well. This highlight the need of a systems level approach. An approach in which the bounded body of the individual human as a basic unit of biological or social action would need to be questioned in favour of a more encompassing and holistic unit. In fact, within biology, there is a new field, Systems Biology, which stresses the need to understand the role that pleiotropy, and thus networks at multiple levelse.g. genetic, cellular, among individuals and among different taxaplay within biological systems and their evolution [46]. Currently, much still needs to be understood about gene function, its role in human biological systems and the interaction between genes and external factors such as environment, diet and so on. In the future if we do choose to genetically enhance human traits to levels unlikely to be achieved by human evolution, it would be crucial to consider if and how our understanding of human evolution enable us to better understand the implications of genetic interventions.

New forms of human enhancement are increasingly coming to play due to technological development. If phenotypic and somatic interventions for human enhancement pose already significant ethical and societal challenges, germline heritable genetic intervention, require much broader and complex considerations at the level of the individual, society and human species as a whole. Germline interventions associated with modern technologies are capable of much more rapid, large-scale impacts and seem capable of radically altering the balance of humans with the environment. We know now that beside the role genes play on biological evolution and development, genetic interventions can induce multiple effects (pleiotropy) and complex epigenetics interactions among genotype, phenotype and ecology of a certain environment. As a result of the rapidity and scale with which such impact could be realized, it is essential for ethical and societal debates, as well as underlying scientific studies, to consider the unit of impact not only to the human body but also to human populations and their natural environment (systems biology). An important practicable distinction between therapy and enhancement may need to be drawn and effectively implemented in future regulations, although a distinct line between the two may be difficult to draw.

In the future if we do choose to genetically enhance human traits to levels unlikely to be achieved by human evolution, it would be crucial to consider if and how our understanding of humans and other organisms, including domesticated ones, enable us to better understand the implications of genetic interventions. In particular, effective regulation of genetic engineering may need to be based on a deep knowledge of the exact links between phenotype and genotype, as well the interaction of the human species with the environment and vice versa.

For a broader and consistent debate, it will be essential for technological, philosophical, ethical and policy discussions on human enhancement to consider the empirical evidence provided by evolutionary biology, developmental biology and other disciplines.

This work was supported by Fundao para a Cincia e a Tecnologia (FCT) of Portugal [CFCUL/FIL/00678/2019 to M.A.].

Conflict of interest: None declared.

Pham

P

Roux

S

Matonti

F

Post-implantation impedance spectroscopy of subretinal micro-electrode arrays, OCT imaging and numerical simulation: towards a more precise neuroprosthesis monitoring tool

J Neural Eng

2013

10

046002

Maghami

MH

Sodagar

AM

Lashay

A

Visual prostheses: the enabling technology to give sight to the blind

J Ophthal Vis Res

2014

9

494

505

Weitz

AC

Nanduri

D

Behrend

MR

Improving the spatial resolution of epiretinal implants by increasing stimulus pulse duration

Sci Transl Med

2015

7

318ra203.

Bouton

CE

Shaikhouni

A

Annetta

NV

Restoring cortical control of functional movement in a human with quadriplegia

Nature

2016

533

247

50

Geddes

L.

First paralysed person to be reanimated offers neuroscience insights. Technique moves mans arm by decoding his thoughts and electrically stimulating his own muscles

Nat News

2016

533

Squires

JE.

Artificial blood

Science

2002

295

1002

5

Lowe

KC.

Blood substitutes: from chemistry to clinic

J Mater Chem

2006

16

See the original post:
Human enhancement: Genetic engineering and evolution - OUP Academic

genetic engineering – Process and techniques | Britannica

Most recombinant DNA technology involves the insertion of foreign genes into the plasmids of common laboratory strains of bacteria. Plasmids are small rings of DNA; they are not part of the bacteriums chromosome (the main repository of the organisms genetic information). Nonetheless, they are capable of directing protein synthesis, and, like chromosomal DNA, they are reproduced and passed on to the bacteriums progeny. Thus, by incorporating foreign DNA (for example, a mammalian gene) into a bacterium, researchers can obtain an almost limitless number of copies of the inserted gene. Furthermore, if the inserted gene is operative (i.e., if it directs protein synthesis), the modified bacterium will produce the protein specified by the foreign DNA.

A subsequent generation of genetic engineering techniques that emerged in the early 21st century centred on gene editing. Gene editing, based on a technology known as CRISPR-Cas9, allows researchers to customize a living organisms genetic sequence by making very specific changes to its DNA. Gene editing has a wide array of applications, being used for the genetic modification of crop plants and livestock and of laboratory model organisms (e.g., mice).

The correction of genetic errors associated with disease in animals suggests that gene editing has potential applications in gene therapy for humans. Gene therapy is the introduction of a normal gene into an individuals genome in order to repair a mutation that causes a genetic disease. When a normal gene is inserted into a mutant nucleus, it most likely will integrate into a chromosomal site different from the defective allele; although this may repair the mutation, a new mutation may result if the normal gene integrates into another functional gene. If the normal gene replaces the mutant allele, there is a chance that the transformed cells will proliferate and produce enough normal gene product for the entire body to be restored to the undiseased phenotype.

Genetic engineering has advanced the understanding of many theoretical and practical aspects of gene function and organization. Through recombinant DNA techniques, bacteria have been created that are capable of synthesizing human insulin, human growth hormone, alpha interferon, a hepatitis B vaccine, and other medically useful substances. Plants may be genetically adjusted to enable them to fix nitrogen, and genetic diseases can possibly be corrected by replacing dysfunctional genes with normally functioning genes.

Genes for toxins that kill insects have been introduced in several species of plants, including corn and cotton. Bacterial genes that confer resistance to herbicides also have been introduced into crop plants. Other attempts at the genetic engineering of plants have aimed at improving the nutritional value of the plant.

In 1980 the new microorganisms created by recombinant DNA research were deemed patentable, and in 1986 the U.S. Department of Agriculture approved the sale of the first living genetically altered organisma virus, used as a pseudorabies vaccine, from which a single gene had been cut. Since then several hundred patents have been awarded for genetically altered bacteria and plants. Patents on genetically engineered and genetically modified organisms, particularly crops and other foods, however, were a contentious issue, and they remained so into the first part of the 21st century.

Special concern has been focused on genetic engineering for fear that it might result in the introduction of unfavourable and possibly dangerous traits into microorganisms that were previously free of theme.g., resistance to antibiotics, production of toxins, or a tendency to cause disease. Indeed, possibilities for misuse of genetic engineering were vast. In particular, there was significant concern about genetically modified organisms, especially modified crops, and their impacts on human and environmental health. For example, genetic manipulation may potentially alter the allergenic properties of crops. In addition, whether some genetically modified crops, such as golden rice, deliver on the promise of improved health benefits was also unclear. The release of genetically modified mosquitoes and other modified organisms into the environment also raised concerns.

In the 21st century, significant progress in the development of gene-editing tools brought new urgency to long-standing discussions about the ethical and social implications surrounding the genetic engineering of humans. The application of gene editing in humans raised significant ethical concerns, particularly regarding its potential use to alter traits such as intelligence and beauty. More practically, some researchers attempted to use gene editing to alter genes in human sperm, which would enable the edited genes to be passed on to subsequent generations, while others sought to alter genes that increase the risk of certain types of cancer, with the aim of reducing cancer risk in offspring. The impacts of gene editing on human genetics, however, were unknown, and regulations to guide its use were largely lacking.

Visit link:
genetic engineering - Process and techniques | Britannica

Arthritis – Symptoms and causes – Mayo Clinic

Overview Osteoarthritis vs. rheumatoid arthritis Open pop-up dialog box

Close

Osteoarthritis, the most common form of arthritis, involves the wearing away of the cartilage that caps the bones in your joints. Rheumatoid arthritis is a disease in which the immune system attacks the joints, beginning with the lining of joints.

Arthritis is the swelling and tenderness of one or more joints. The main symptoms of arthritis are joint pain and stiffness, which typically worsen with age. The most common types of arthritis are osteoarthritis and rheumatoid arthritis.

Osteoarthritis causes cartilage the hard, slippery tissue that covers the ends of bones where they form a joint to break down. Rheumatoid arthritis is a disease in which the immune system attacks the joints, beginning with the lining of joints.

Uric acid crystals, which form when there's too much uric acid in your blood, can cause gout. Infections or underlying disease, such as psoriasis or lupus, can cause other types of arthritis.

Treatments vary depending on the type of arthritis. The main goals of arthritis treatments are to reduce symptoms and improve quality of life.

The most common signs and symptoms of arthritis involve the joints. Depending on the type of arthritis, signs and symptoms may include:

Sign up for free, and stay up to date on research advancements, health tips and current health topics, like COVID-19, plus expertise on managing health.

To provide you with the most relevant and helpful information, and understand which information is beneficial, we may combine your email and website usage information with other information we have about you. If you are a Mayo Clinic patient, this could include protected health information. If we combine this information with your protected health information, we will treat all of that information as protected health information and will only use or disclose that information as set forth in our notice of privacy practices. You may opt-out of email communications at any time by clicking on the unsubscribe link in the e-mail.

Subscribe!

You'll soon start receiving the latest Mayo Clinic health information you requested in your inbox.

Please, try again in a couple of minutes

Retry

The two main types of arthritis osteoarthritis and rheumatoid arthritis damage joints in different ways.

The most common type of arthritis, osteoarthritis involves wear-and-tear damage to a joint's cartilage the hard, slick coating on the ends of bones where they form a joint. Cartilage cushions the ends of the bones and allows nearly frictionless joint motion, but enough damage can result in bone grinding directly on bone, which causes pain and restricted movement. This wear and tear can occur over many years, or it can be hastened by a joint injury or infection.

Osteoarthritis also causes changes in the bones and deterioration of the connective tissues that attach muscle to bone and hold the joint together. If cartilage in a joint is severely damaged, the joint lining may become inflamed and swollen.

In rheumatoid arthritis, the body's immune system attacks the lining of the joint capsule, a tough membrane that encloses all the joint parts. This lining (synovial membrane) becomes inflamed and swollen. The disease process can eventually destroy cartilage and bone within the joint.

Risk factors for arthritis include:

Severe arthritis, particularly if it affects your hands or arms, can make it difficult for you to do daily tasks. Arthritis of weight-bearing joints can keep you from walking comfortably or sitting up straight. In some cases, joints may gradually lose their alignment and shape.

Sept. 15, 2021

Continued here:
Arthritis - Symptoms and causes - Mayo Clinic