header logo image


Page 10«..9101112..2030..»

Archive for the ‘Genetic Engineering’ Category

‘Moondust’ carnation uses genetic engineering to achieve its blue color – Batesville Daily Guard

Wednesday, September 2nd, 2020

The search for flowers with shades and hues different from what nature provides has been ongoing since humankind discovered the intricacies of plant breeding. But there are limits to the color range that can be achieved using traditional breeding techniques. For example, blue pigments are lacking from many plants.

But science now allows breeders to extend the natural range of colors, using genetic engineering. Moondust carnation, first grown commercially in 1997, is a mini-carnation with purple-mauve flowers that gets its blue color from petunia genes grafted into the DNA of the carnation.

Twelve scientists at an Australian company called Florigene labored for a decade to isolate the gene responsible for blue color in petunia and then transfer it into the carnation. To date, they have released five carnations with the "Moon" prefix, all with varying shades of mauve, blue, violet or purple.

Flower color expression is caused by the subtle blending of pigments contained in the vacuoles (think of vacuoles as storage closets in the cell) and plastid bodies (think of these as like chlorophyll, but with a color other than green) suspended in the cell sap. Just as the man at the paint store blends different pigments to a neutral base to color paint, flower color is caused by the subtle blends of several pigments.

But roses, carnations, lilies and orchids all lack a class of blue pigments called delphinidins, named after the violet-blue we see in delphinium. The gene for delphinidin production is what the Floragene scientists removed from petunia and transferred to the carnation.

The development of the blue carnation was not the primary goal of the research team; no, they wanted to make a blue rose. But, transplanting genes is easy to say but hard to do in the lab, so they honed their techniques on carnations a much easier species to manipulate than roses. The team has not given up on the idea of a blue rose, but it is now exploring the possibility of inserting genes from sea anemones into the rose to create the blue shades. The petunia gene didnt work in roses.

You may be thinking by now, "Ive seen blue carnations for years. Whats new about this?" True, there have been blue carnations available since the 1970s, but their blue was due to food color, not natural pigments.

Scott Admire with Little Rocks United Wholesale Florists said they used to dye white carnations shipped in from cut flower growers in Central and South America. The carnations would have to be shipped in as a "dry pack," exposed to neither water nor the floral preservative silver thiosulphate. The carnations would then suck in the pigment-laden water with a good deal of it ending up in the petals, turning the flowers the shade of blue you see atop a decorated birthday cake.

Carnation flowers sometime get "sleepy" and curl up. This is caused by the production of a plant growth hormone called ethylene, which is a part of the natural aging process in flower development. Cut flower growers combat this by using the silver-containing floral preservative that stops the production of ethylene. Floragene scientists are currently seeking clearance to introduce a line of plants that do not produce ethylene, thus eliminating the need for the silver thiosulphate treatment.

This ethylene-blocking technology is not new, and in fact, an Arkansas boy Dr. Randy Woodson from Fordyce and now a dean at Purdue University responsible for the agricultural research program patented a technique in the early 1990s using the "anti-sense" procedure. Using this technique, the gene for ethylene production is switched end-for-end in the DNA strand, rendering it inoperative. It's not clear if the Floragene technique uses this same "anti-sense" technology.

Are the scientists involved in creating genetically modified plants playing God? What about environmental concerns? For the former question, my theological credentials are suspect, but to the latter question I feel confident there is no significant environmental risk in growing blue carnations. The plants are essentially pollen sterile and carnations are harvested in the tight-bud stage, so the likelihood of out-crossing with wild carnations is remote.

See the original post:
'Moondust' carnation uses genetic engineering to achieve its blue color - Batesville Daily Guard

Read More...

The promise and perils of synthetic biology take center stage in a fast-paced new Netflix series – Science Magazine

Wednesday, September 2nd, 2020

Christian DitterNetflix6 episodes

The first season of the Netflix series Biohackers, consisting of six episodes released on the streaming platform on 20 August, tells a fictional tale centered around the sociotechnological movement known as do-it-yourself (DIY) biology, in which amateurs, professionals, anarchists, and civic-minded citizens push the boundaries of mainstream biology. The shows main characters include a wealthy biopharmaceutical executive, a group of medical students, a number of stereotypical biohackers making animals glow and plants play music, and a community of transhumanists intent on modifying their bodies for seemingly impractical endeavors.

Whereas biological experimentation was once the sole domain of trained professionals in well-stocked and well-funded institutional labs, the field has been democratized by the emergence of the open-source movement, plummeting sequencing costs, greater access to reagents and devices, the proliferation of online resources, and the emergence of tools and methodologies that enable nonexperts to genetically engineer organisms without years of professional training. [Valid concerns regarding some of the activities associated with the DIY bio community have been voiced by the Presidential Commission for the Study of Bioethical Issues (1).]

Medical student Mia Akerlund (right) meets biohackers pushing the boundaries of mainstream biology.

The show follows Mia Akerlund (played by Luna Wedler), a first-year medical student vying for a position at a prestigious biopharmaceutical firm headed by celebrated professor Tanja Lorenz (Jessica Schwarz). Akerlund and Lorenz clearly have some shared history, as well as their own secrets, although viewers are not privy to the details of either at the start of the series. For much of the first episodes, the relationship between these two enigmatic characters is revealed slowly through both flash-forwards and flashbacks. But we know that a big reveal is coming; the programs official description teases a secret so big it could change the fate of humanity.

Throughout the seasons six fast-paced episodes, the viewer is exposed to technologies and techniques that would be familiar to many professional scientists. And while the time frames of the various experiments conducted are often compressed for dramatic effect, Christian Ditterthe shows creator, writer, director, and showrunnergoes out of his way to present complex science as accurately as possible. In one montage, for example, we watch various biohackers, some with better aseptic technique than others, add reagents to microcentrifuge tubes, load polymerase chain reaction machines, and examine gels to assess whether they have accurately created a desired genomic sequence. In another scene, a student suffering from a degenerative disease seeks to develop his own cure in a secret lab, where he can work without burdensome oversight. The student injects himself with an unknown liquid, his purported cure. Here, the shows dialogue surrounding the cure and its antidote (to be administered if things go wrong) offers insight into how RNA interference therapies work.

But the show also serves as a pedagogical vehicle to raise many timely and interesting ethical, legal, and social concerns. From bioluminescent mammals to the collection of genetic material for clinical trials, the series storyline highlights how cavalierly we sometimes approach genomic data and genetic engineering. Later episodes depict even more egregious examples of biohacking, including organisms modified to transmit viruses as efficiently as possible. At one point, a character suggests that the ends of her research justify the experimental means, even when her methods demonstrate a gross disregard for test subjects who may suffer as a result.

The show also offers insight into some of the motivations that drive DIY biology efforts. For example, in one scene, a confidant of Akerlund expresses dismay that Lorenz is willing to sell a cheaply acquired drug to desperate patients for inflated prices. Such frustrations are what drive many citizens operating outside traditional institutions to develop their own pharmaceutical solutions.

It is ironic that Biohackers is set in Germany, one of the few places where genetic engineering experimentation outside of licensed facilities is illegal and can result in a fine or even imprisonment (2). Yet, given all that transpires in the show, one is left with the sense that such measures maybe justified.

References and Notes:1. Presidential Commission for the Study of Bioethical Issues, New directions: The ethics of synthetic biology and emerging technologies (2010).2. Sections 8 and 39 of the German Genetic Engineering Act [Gentechnikgesetz (GenTG)].

The reviewer is at Zvi Meitar Institute for Legal Implications of Emerging Technologies, Herzliya, Israel, and the Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA.

Read the original post:
The promise and perils of synthetic biology take center stage in a fast-paced new Netflix series - Science Magazine

Read More...

How Groups of Cells Cooperate to Build Organs and Organisms – The Scientist

Wednesday, September 2nd, 2020

Efforts to use regenerative medicinewhich seeks to address ailments as diverse as birth defects, traumatic injury, aging, degenerative disease, and the disorganized growth of cancerwould be greatly aided by solving one fundamental puzzle: How do cellular collectives orchestrate the building of complex, three-dimensional structures?

While genomes predictably encode the proteins present in cells, a simple molecular parts list does not tell us enough about the anatomical layout or regenerative potential of the body that the cells will work to construct. Genomes are not a blueprint for anatomy, and genome editing is fundamentally limited by the fact that its very hard to infer which genes to tweak, and how, to achieve desired complex anatomical outcomes. Similarly, stem cells generate the building blocks of organs, but the ability to organize specific cell types into a working human hand or eye has been and will be beyond the grasp of direct manipulation for a very long time.

But researchers working in the fields of synthetic morphology and regenerative biophysics are beginning to understand the rules governing the plasticity of organ growth and repair. Rather than micromanaging tasks that are too complex to implement directly at the cellular or molecular level, what if we solved the mystery of how groups of cells cooperate to construct specific multicellular bodies during embryogenesis and regeneration? Perhaps then we could figure out how to motivate cell collectives to build whatever anatomical features we want.

New approaches now allow us to target the processes that implement anatomical decision-making without genetic engineering. In January, using such tools, crafted in my lab at Tufts Universitys Allen Discovery Center and by computer scientists in Josh Bongards lab at the University of Vermont, we were able to create novel living machines, artificial bodies with morphologies and behaviors completely different from the default anatomy of the frog species (Xenopus laevis) whose cells we used. These cells rebooted their multicellularity into a new form, without genomic changes. This represents an extremely exciting sandbox in which bioengineers can play, with the aim of decoding the logic of anatomical and behavioral control, as well as understanding the plasticity of cells and the relationship of genomes to anatomies.

Deciphering how an organism puts itself together is truly an interdisciplinary undertaking.

Deciphering how an organism puts itself together is truly an interdisciplinary undertaking. Resolving the whole picture will involve understanding not only the mechanisms by which cells operate, but also elucidating the computations that cells and groups of cells carry out to orchestrate tissue and organ construction on a whole-body scale. The next generation of advances in this area of research will emerge from the flow of ideas between computer scientists and biologists. Unlocking the full potential of regenerative medicine will require biology to take the journey computer science has already taken, from focusing on the hardwarethe proteins and biochemical pathways that carry out cellular operationsto the physiological software that enables networks of cells to acquire, store, and act on information about organ and indeed whole-body geometry.

In the computer world, this transition from rewiring hardware to reprogramming the information flow by changing the inputs gave rise to the information technology revolution. This shift of perspective could transform biology, allowing scientists to achieve the still-futuristic visions of regenerative medicine. An understanding of how independent, competent agents such as cells cooperate and compete toward robust outcomes, despite noise and changing environmental conditions, would also inform engineering. Swarm robotics, Internet of Things, and even the development of general artificial intelligence will all be enriched by the ability to read out and set the anatomical states toward which cell collectives build, because they share a fundamental underlying problem: how to control the emergent outcomes of systems composed of many interacting units or individuals.

Many types of embryos can regenerate entirely if cut in half, and some species are proficient regenerators as adults. Axolotls (Ambystoma mexicanum) regenerate their limbs, eyes, spinal cords, jaws, and portions of the brain throughout life. Planarian flatworms (class Turbellaria), meanwhile, can regrow absolutely any part of their body; when the animal is cut into pieces, each piece knows exactly whats missing and regenerates to be a perfect, tiny worm.

The remarkable thing is not simply that growth begins after wounding and that various cell types are generated, but that these bodies will grow and remodel until a correct anatomy is complete, and then they stop. How does the system identify the correct target morphology, orchestrate individual cell behaviors to get there, and determine when the job is done? How does it communicate this information to control underlying cell activities?

Several years ago, my lab found that Xenopus tadpoles with their facial organs experimentally mixed up into incorrect positions still have largely normal faces once theyve matured, as the organs move and remodel through unnatural paths. Last year, a colleague at Tufts came to a similar conclusion: the Xenopus genome does not encode a hardwired set of instructions for the movements of different organs during metamorphosis from tadpole to frog, but rather encodes molecular hardware that executes a kind of error minimization loop, comparing the current anatomy to the target frog morphology and working to progressively reduce the difference between them. Once a rough spatial specification of the layout is achieved, that triggers the cessation of further remodeling.

The deep puzzle of how competent agents such as cells work together to pursue goals such as building, remodeling, or repairing a complex organ to a predetermined spec is well illustrated by planaria. Despite having a mechanistic understanding of stem cell specification pathways and axial chemical gradients, scientists really dont know what determines the intricate shape and structure of the flatworms head. It is also unknown how planaria perfectly regenerate the same anatomy, even as their genomes have accrued mutations over eons of somatic inheritance. Because some species of planaria reproduce by fission and regeneration, any mutation that doesnt kill the neoblastthe adult stem cell that gives rise to cells that regenerate new tissueis propagated to the next generation. The worms incredibly messy genome shows evidence of this process, and cells in an individual planarian can have different numbers of chromosomes. Still, fragmented planaria regenerate their body shape with nearly 100 percent anatomical fidelity.

Permanent editingof the encoded target morphology without genomic editing reveals a new kind of epigenetics.

So how do cell groups encode the patterns they build, and how do they know to stop once a target anatomy is achieved? What would happen, for example, if neoblasts from a planarian species with a flat head were transplanted into a worm of a species with a round or triangular head that had the head amputated? Which shape would result from this heterogeneous mixture? To date, none of the high-resolution molecular genetic studies of planaria give any prediction for the results of this experiment, because so far they have all focused on the cellular hardware, not on the logic of the softwareimplemented by chemical, mechanical, and electrical signaling among cellsthat controls large-scale outcomes and enables remodeling to stop when a specific morphology has been achieved.

Understanding how cells and tissues make real-time anatomical decisions is central not only to achieving regenerative outcomes too complex for us to manage directly, but also to solving problems such as cancer. While the view of cancer as a genetic disorder still largely drives clinical approaches, recent literature supports a view of cancer as cells simply not being able to receive the physiological signals that maintain the normally tight controls of anatomical homeostasis. Cut off from these patterning cues, individual cells revert to their ancient unicellular lifestyle and treat the rest of the body as external environment, often to ruinous effect. If we understand the mechanisms that scale single-cell homeostatic setpoints into tissue- and organ-level anatomical goal states and the conditions under which the anatomical error reduction control loop breaks down, we may be able to provide stimuli to gain control of rogue cancer cells without either gene therapy or chemotherapy.

During morphogenesis, cells cooperate to reliably build anatomical structures. Many living systems remodel and regenerate tissues or organs despite considerable damagethat is, they progressively reduce deviations from specific target morphologies, and halt growth and remodeling when those morphologies are achieved. Evolution exploits three modalities to achieve such anatomical homeostasis: biochemical gradients, bioelectric circuits, and biophysical forces. These interact to enable the same large-scale form to arise despite significant perturbations.

N.R. FULLER, SAYO-ART, LLC

BIOCHEMICAL GRADIENTS

The best-known modality concerns diffusible intracellular and extracellular signaling molecules. Gene-regulatory circuits and gradients of biochemicals control cell proliferation, differentiation, and migration.

BIOELECTRIC CIRCUITS

The movement of ions across cell membranes, especially via voltage-gated ion channels and gap junctions, can establish bioelectric circuits that control large-scale resting potential patterns within and among groups of cells. These bioelectric patterns implement long-range coordination, feedback, and memory dynamics across cell fields. They underlie modular morphogenetic decision-making about organ shape and spatial layout by regulating the dynamic redistribution of morphogens and the expression of genes.

BIOMECHANICAL FORCES

Cytoskeletal, adhesion, and motor proteins inside and between cells generate physical forces that in turn control cell behavior. These forces result in large-scale strain fields, which enable cell sheets to move and deform as a coherent unit, and thus execute the folds and bends that shape complex organs.

The software of life, which exploits the laws of physics and computation, is enabled by chemical, mechanical, and electrical signaling across cellular networks. While the chemical and mechanical mechanisms of morphogenesis have long been appreciated by molecular and cell biologists, the role of electrical signaling has largely been overlooked. But the same reprogrammability of neural circuits in the brain that supports learning, memory, and behavioral plasticity applies to all cells, not just neurons. Indeed, bacterial colonies can communicate via ionic currents, with recent research revealing brain-like dynamics in which information is propagated across and stored in a kind of proto-body formed by bacterial biofilms. So it should really come as no surprise that bioelectric signaling is a highly tractable component of morphological outcomes in multicellular organisms.

A few years ago, we studied the electrical dynamics that normally set the size and borders of the nascent Xenopus brain, and built a computer model of this process to shed light on how a range of various brain defects arise from disruptions to this bioelectric signaling. Our model suggested that specific modifications with mRNA or small molecules could restore the endogenous bioelectric patterns back to their correct layout. By using our computational platform to select drugs to open existing ion channels in nascent neural tissue or even a remote body tissue, we were able to prevent and even reverse brain defects caused not only by chemical teratogenscompounds that disrupt embryonic developmentbut by mutations in key neurogenesis genes.

Similarly, we used optogenetics to stimulate electrical activity in various somatic cell types totrigger regeneration of an entire tadpole tailan appendage with spinal cord, muscle, and peripheral innervationand to normalize the behavior of cancer cells in tadpoles strongly expressing human oncogenes such as KRAS mutations. We used a similar approach to trigger posterior regions, such as the gut, to build an entire frog eye. In both the eye and tail cases, the information on how exactly to build these complex structures, and where all the cells should go, did not have to be specified by the experimenter; rather, they arose from the cells themselves. Such findings reveal how ion channel mutations result in numerous human developmental channelopathies, and provide a roadmap for how they may be treated by altering the bioelectric map that tells cells what to build.

We also recently found a striking example of such reprogrammable bioelectrical software in control of regeneration in planaria. In 2011, we discovered that an endogenous electric circuit establishes a pattern of depolarization and hyperpolarization in planarian fragments that regulate the orientation of the anterior-posterior axis to be rebuilt. Last year, we discovered that this circuit controls the gene expressionneeded to build a head or tail within six hours of amputation, and by using molecules that make cell membranes permeable to certain ions to depolarize or hyperpolarize cells, we induced fragments of such worms to give rise to a symmetrical two-headed form, despite their wildtype genomes. Even more shockingly, the worms continued to generate two-headed progeny in additional rounds of cutting with no further manipulation. In further experiments, we demonstrated that briefly reducing gap junction-mediated connectivity between adjacent cells in the bioelectric network that guides regeneration led worms to regenerate head and brain shapes appropriate to other worm species whose lineages split more than 100 million years ago.

My group has developed the use of voltage-sensitive dyes to visualize the bioelectric pattern memory that guides gene expression and cell behavior toward morphogenetic outcomes. Meanwhile, my Allen Center colleagues are using synthetic artificial electric tissues made of human cells and computer models of ion channel activity to understand how electrical dynamics across groups of non-neural cells can set up the voltage patterns that control downstream gene expression, distribution of morphogen molecules, and cell behaviors to orchestrate morphogenesis.

The emerging picture in this field is that anatomical software is highly modulara key property that computer scientists exploit as subroutines and that most likely contributes in large part to biological evolvability and evolutionary plasticity. A simple bioelectric state, whether produced endogenously during development or induced by an experimenter, triggers very complex redistributions of morphogens and gene expression cascades that are needed to build various anatomies. The information stored in the bodys bioelectric circuitscan be permanently rewritten once we understand the dynamics of the biophysical circuits that make the critical morphological decisions. This permanent editing of the encoded target morphology without genomic editing reveals a new kind of epigenetics, information that is stored in a medium other than DNA sequences and chromatin.

Recent work from our group and others has demonstrated that anatomical pattern memories can be rewritten by physiological stimuli and maintained indefinitely without genomic editing. For example, the bioelectric circuit that normally determines head number and location in regenerating planaria can be triggered by brief alterations of ion channel or gap junction activity to alter the animals body plan. Due to the circuits pattern memory, the animals remain in this altered state indefinitely without further stimulation, despite their wildtype genomes. In other words, the pattern to which the cells build after damage can be changed, leading to a target morphology distinct from the genetic default.

N.R. FULLER, SAYO-ART, LLC

First, we soaked a planarian in voltage-sensitive fluorescent dye to observe the bioelectrical pattern across the entire tissue. We then cut the animal to see how this pattern changes in each fragment as it begins to regenerate.

We then applied drugs or used RNA interference to target ion channels or gap junctions in individual cells and thus change the pattern of depolarization/hyperpolarization and cellular connectivity across the whole fragment.

As a result of the disruption of the bodys bioelectric circuits, the planarian regrows with two heads instead of one, or none at all.

When we re-cut the two-headed planarian in plain water, long after the initial drug has left the tissue, the new anatomy persists in subsequent rounds of regeneration.

Cells can clearly build structures that are different from their genomic-default anatomical outcomes. But are cells universal constructors? Could they make anything if only we knew how to motivate them to do it?

The most recent advances in the new field at the intersection of developmental biology and computer science are driven by synthetic living machines known as biobots. Built from multiple interacting cell populations, these engineered machines have applications in disease modeling and drug development, and as sensors that detect and respond to biological signals. We recently tested the plasticity of cells by evolving in silico designs with specific movement and behavior capabilities and used this information to sculpt self-organized growth of aggregated Xenopus skin and muscle cells. In a novel environmentin vitro, as opposed to inside a frog embryoswarms of genetically normal cells were able to reimagine their multicellular form. With minimal sculpting post self-assembly, these cells form Xenobots with structures, movements, and other behaviors quite different from what might be expected if one simply sequenced their genome and identified them as wildtype X. laevis.

These living creations are a powerful platform to assess and model the computations that these cell swarms use to determine what to build. Such insights will help us to understand evolvability of body forms, robustness, and the true relationship between genomes and anatomy, greatly potentiating the impact of genome editing tools and making genomics more predictive for large-scale phenotypes. Moreover, testing regimes of biochemical, biomechanical, and bioelectrical stimuli in these biobots will enable the discovery of optimal stimuli for use in regenerative therapies and bioengineered organ construction. Finally, learning to program highly competent individual builders (cells) toward group-level, goal-driven behaviors (complex anatomies) will significantly advance swarm robotics and help avoid catastrophes of unintended consequences during the inevitable deployment of large numbers of artificial agents with complex behaviors.

Understanding how cells and tissues make real-time anatomical decisions is central to achieving regenerative outcomes too complex for us to manage directly.

The emerging field ofsynthetic morphology emphasizes a conceptual point that has been embraced by computer scientists but thus far resisted by biologists: the hardware-software distinction. In the 1940s, to change a computers behavior, the operator had to literally move wires aroundin other words, she had to directly alter the hardware. The information technology revolution resulted from the realization that certain kinds of hardware are reprogrammable: drastic changes in function could be made at the software level, by changing inputs, not the hardware itself.

In molecular biomedicine, we are still focused largely on manipulating the cellular hardwarethe proteins that each cell can exploit. But evolution has ensured that cellular collectives use this versatile machinery to process information flexibly and implement a wide range of large-scale body shape outcomes. This is biologys software: the memory, plasticity, and reprogrammability of morphogenetic control networks.

The coming decades will be an extremely exciting time for multidisciplinary efforts in developmental physiology, robotics, and basal cognition to understand how individual cells merge together into a collective with global goals not belonging to any individual cell. This will drive the creation of new artificial intelligence platforms based not on copying brain architectures, but on the multiscale problem-solving capacities of cells and tissues. Conversely, the insights of cognitive neurobiology and computer science will give us a completely new window on the information processing and decision-making dynamics in cellular collectives that can very effectively be targeted for transformative regenerative therapies of complex organs.

Michael Levinis the director of the Allen Discovery Center at Tufts University and Associate Faculty at Harvard Universitys Wyss Institute. Email him atmichael.levin@tufts.edu. M.L. thanks Allen Center Deputy DirectorJoshua Finkelsteinfor suggestions on the drafts of this story.

Originally posted here:
How Groups of Cells Cooperate to Build Organs and Organisms - The Scientist

Read More...

Viral Vector Manufacturing Market Forecast to Reach $1.47 Billion by 2025 – ResearchAndMarkets.com – Business Wire

Wednesday, September 2nd, 2020

DUBLIN--(BUSINESS WIRE)--The "Viral Vector Manufacturing Market - Forecasts from 2020 to 2025" report has been added to ResearchAndMarkets.com's offering.

The global viral vector manufacturing market is projected to grow at a CAGR of 22.09% to reach a market size of US$1,469.144 million in 2025 from US$443.592 million in 2019.

The viral vector market is primarily being driven by the growing adoption of adenoviral vectors, lentiviral vectors, as well as retroviral vectors. The growing adoption stems from the need for effectively transferring therapeutic gene into the target cells that are an integral part of the process that involves the insertion of a functional copy of a gene into a defective cell one of the preferred treatment options for most chronic diseases, which is known as Gene therapy.

Furthermore, the growing number of clinical trials, the increasing number of gene therapy, and the expanding cognizance of effective mode of disease treatment are further expected to drive the growth of the viral vector manufacturing market during the forecast period. Since vector designing, production, packaging, and release testing is subject to limited availability and faced with challenges due to the complex nature of technologies and platform and thus many players in this space often endeavor in striking strategic collaboration and acquisitions that cover many aspects like the delivery of clinical grade product under its ambit, to facilitate the successful collaboration development of viral agent-based products.

Moreover, the efficient ability to express the therapeutic genes and non-pathogenic nature is another factor that is responsible for driving the growth of this market. The other key factors that are expected to drive the growth of the market are the increasing investment in the biopharmaceutical production coupled with the growing aging population, healthcare expenditure, technological advancement, especially in the genetic engineering segment.

Moreover, the increasing accessibility of healthcare facilities, the growing demand for treatment of disease due to the increasing global burden of diseases are a few of the other factors that are poised to drive the growth of this market during the forecast period. Nevertheless, despite the transitioning of this niche industry to high manufacturing is one such factor that may restrain the growth of the market to a certain extent.

Therefore, with such growing recognition of the importance of viral vectors, various developments are taking place in the viral vector manufacturing market. For instance, in June 2020, it was announced by Emergent BioSolutions Inc. (NYSE: EBS) which is a global life sciences company that it is going to invest $75 Million in Canton Site and expand viral vector and gene therapy capability facilitating the reinforcement of its contract development and manufacturing (CDMO) capabilities.

Again, in June 2020, Oxford Biomedica (LSE: OXB) which is a major gene and cell therapy group, announced that it has signed an agreement of collaboration with the Vaccines Manufacturing and Innovation Centre (VMIC), a not-for-profit organization that has been established to provide the first strategic vaccine development and progressive manufacturing capability in the UK. Under this 5-year agreement, the organization will work towards enabling the manufacture of vaccines that are based on viral vector, to contribute towards a swift growth in the domestic capacity for this specialized field of vaccine manufacturing.

In April 2020, Merck KGaA (FWB: MRK) a leading science and technology company announced that a 100 million facility, second in Carlsbad, California USA that is intended to boost its BioReliance viral and gene therapy service offering to help their customers to aid their customers to commercialize the gene therapies that are brought about by viral vectors concomitantly helping innovators scale up their production that is in tandem with the quantum that allows them to reach out to more patients.

Earlier, in January 2020, the launch of ZYNTEGLO (autologous CD34+ cells encoding A-T87Q-globin gene) in Germany was announced by bluebird bio, Inc. (Nasdaq: BLUE). ZYNTEGLO is a one-time gene therapy that has been specifically developed for patients aged 12 years and older with transfusion-dependent -thalassemia (TDT) who do not possess 0/0 genotype.

In December 2019, it was announced that a leading supplier of services and technologies for the life sciences industry called Novasep launched oXYgene which is a fully integrated offering for the construction of facilities dedicated towards customers to aid them in their viral vector production.

In October 2019, it was reported that GE Healthcare Life Sciences which has now rebranded itself as Cytiva, was about to launch the KUBio box which is an adaptable, flexible and fully integrated environment for biomanufacturing to accelerate the production gene therapies based on of viral vector. These latest additions were intended to bring gene therapies swiftly to the market thereby contributing to the increased capacity in the viral vector area.

In March 2018, it was reported that Sartorius Stedim Biotech SA (SSB), which is a major international technology partner supplier of products and services biopharmaceutical industry has been selected by ABL Europe as its chief supplier of single-use systems whereby the new viral vector manufacturing capacity has been started in Strasbourg at its European facility. ABL Europe, a subsidiary of ABL Inc. provides dedicated viral vector GMP manufacturing services for oncolytic, vaccine and gene therapy projects in all stages of clinical development through to commercial launch.

Key Topics Covered

1. Introduction

1.1. Market Definition

1.2. Market Segmentation

2. Research Methodology

2.1. Research Data

2.2. Assumptions

3. Executive Summary

3.1. Research Highlights

4. Market Dynamics

4.1. Market Drivers

4.2. Market Restraints

4.3. Porters Five Forces Analysis

4.4. Industry Value Chain Analysis

5. Viral Vector Manufacturing Market Analysis, By Type

5.1. Introduction

5.2. Retroviral vectors

5.3. Lentiviral Vectors

5.4. Adenoviral Vectors

5.5. Others

6. Viral Vector Manufacturing Market Analysis, By Application

6.1. Introduction

6.2. Vaccinology

6.3. Gene Therapy

7. Viral Vector Manufacturing Market Analysis, By End-User

7.1. Introduction

7.2. Pharmaceutical & Biotechnology Companies

7.3. Research Institutes

7.4. Contract Research Organizations

8. Viral Vector Manufacturing Market Analysis, by Geography

8.1. Introduction

8.2. North America

8.3. South America

8.4. Europe

8.5. The Middle East & Africa

8.6. Asia-Pacific

9. Competitive Environment and Analysis

9.1. Major Players and Strategy Analysis

9.2. Emerging Players and Market Lucrativeness

9.3. Mergers, Acquisitions, Agreements, and Collaborations

9.4. Vendor Competitiveness Matrix

10. Company Profiles

10.1. Sirion-Biotech GmbH

10.2. Vigene Biosciences

10.3. Batavia Biosciences B.V.

10.4. Virovek

10.5. Lonza

10.6. Vector Biolabs

10.7. Cobra Biologics

10.8. MaxCyte, Inc.

10.9. Genelux

10.10. BioNTech SE

For more information about this report visit https://www.researchandmarkets.com/r/c3nfai

View original post here:
Viral Vector Manufacturing Market Forecast to Reach $1.47 Billion by 2025 - ResearchAndMarkets.com - Business Wire

Read More...

CAR T-Cell Optimization Starts in Production, Extends to Therapy – Genetic Engineering & Biotechnology News

Wednesday, September 2nd, 2020

Just as heat-seeking missiles race toward the infrared signatures of their targets, chimeric antigen receptor (CAR) T cells home in on cancer-associated or -specific antigens. Once the antigens are engaged, CAR T cells let fly with cytotoxic flak, granules containing perforin and granzymes, while activating supplementary tumor-killing mechanisms such as stromal sensitization and macrophage polarization. It is to be hoped that by the time the cytotoxic smoke clears, the cancer will have been destroyed.

The development of CAR T cells has revolutionized adoptive cellular therapies against cancer. CARs are genetically engineered to combine antigen- or tumor-specific-binding with T-cell activating domains. T cells, obtained from the patient (autologous cells) or from a healthy donor (allogeneic cells), are typically transduced with an engineered vector, expanded, and infused back into the patient for tumor eradication.

In the 10 years since its inception, the CAR T-cell field has progressed rapidly. Two CAR T-cell products have been approved for clinical use, and many more products are undergoing clinical trials or are in development. Although the field initially focused on B-cell malignancies, it is now advancing on solid tumors.

Despite its initial success, the CAR T-cell field must find ways to generate products that are potent, affordable, and available. To achieve enduring success, the CAR T-cell field is undertaking a range of initiatives. These include the engineering of bridging proteins for multiantigen targeting; the creation of nonviral allogeneic off-the-shelf products; the organization of vein-to-vein networks; and the development of precisely tuned therapies, that is, precisely timed and dosed therapies.

Cellular therapy is a living drug, declares Steve Shamah, PhD, senior vice president, Obsidian Therapeutics. As with any drug, damage can occur if the therapy is not carefully regulated. Our company focuses on creating controllable cell therapies by engineering CAR T cells or tumor-infiltrating lymphocytes to produce regulatable cytokines and proteins that can enhance functional activity, especially against solid tumors.

For example, the company is developing a platform that armors CAR T cells with immunomodulatory factors such as interleukin-15 (IL-15) or CD40 ligand. Shamah explains, These factors can enhance functional activity by driving T-cell expansion, conferring resistance to immunosuppression, improving antigen presentation, and inducing antigen spread. However, both factors can also produce systemic toxicity. Our technology modulates the level and timing of their activity in a fully controlled, dose-dependent manner using an FDA-approved small-molecule drug.

The Obsidian platform, cytoDRiVE, adds a drug-responsive domain (DRD) onto a therapeutic protein of interest. DRD tags are misfolded or inherently unstable in the cell. However, they can be reversibly stabilized by the binding of approved small-molecule drugs. When the drug is absent, the DRD-tagged protein is turned off. When the drug is present, the DRD-tagged protein is turned on. When DRD tags are in place, the concentration of the small-molecule drug serves as a biological rheostat for controlling the dosing of the therapeutic protein.

Preclinical in vivo mouse studies assessed anti-CD19 CAR T cells that were engineered to express an IL-15-DRD that responded to the FDA-approved drug acetazolamide. In these studies, tumor regression was demonstrated.

Controlling the precise timing and expression level of these immunomodulatory factors in CAR T cells could significantly enhance safety and therapeutic efficacy, concludes Shamah.

Obsidian is currently focusing on the oncology space, but the company is also exploring other areas such as autoimmunity and even the regulation of transcription factors to enable controllable in vivoCRISPR-Cas9 gene editing.

Despite the remarkable success of CAR T-cell therapies, relapses can occur within six months for up to 50% of patients treated with anti-CD19 CAR T-cell therapy.Failures can occur due to loss of CD19 expression or to continued tumor proliferation. Aleta Biotherapeutics has developed a novel technology to reactivate CAR T cells in relapsed patients.

Our approach utilizes antigen-bridging proteins to coat tumors with CD19, says Paul Rennert, PhD, Aletas president and CSO. [The tumors are then] recognized by the patients anti-CD19 CAR T cells, essentially creating a cytotoxic synapse that results in tumor cell death.

To thwart anti-CD19 CAR T-cell therapy relapses, the company developed a bridging protein using the extracellular domain of CD19 and an anti-CD20 antibody domain. CD20 is an antigen present on the majority of B-cell malignancies. Rennert explains that these injected bridging proteins will coat the patients tumor cells with CD19, creating a target to activate or reactivate a patients anti-CD19 CAR T cells.

To show proof-of-principle, the company performed in vivo studies using a half-life-extended form of the bridging protein injected into mice carrying CD20-positive tumor cells and anti-CD19 CAR T cells. Rennert emphasizes, Our studies demonstrated this strategy can be used to reactivate CD19 CAR T cells to prevent and reverse relapses.

Other programs in development include a bridging protein for injection to improve outcomes in multiple myeloma patients treated with CAR T cells, and bridging protein programs for HER2-positive breast cancer patients with central nervous system metastases. The company is preparing investigational new drug applications for its technology and plans to start Phase I trials in 2021.

Assessing whether engineered CAR T-cell and T-cell receptor (TCR) therapies have successfully attacked and penetrated solid tumors (and not normal cells) can be like finding the proverbial needle in the haystack. Traditional methods using immunohistochemistry are useful for immune profiling, but they cannot differentiate engineered versus endogenous cells, points out Christopher Bunker, PhD, senior director of business development, Advanced Cell Diagnostics, a Bio-Techne brand. We developed a means to easily detect and track engineered therapeutic cells and delineate their pharmacokinetics within the tumor microenvironment of intact tumor biopsies, as well as their on-target/off-tumor activity.

Enter RNAscope, an RNA in situ hybridization technology that can enable single-cell spatial transcriptomics. RNAscope, Bunker asserts, is the only off-the-shelf method that can specifically detect engineered CAR T cells and TCR T cells in solid tumor patient biopsies.

Most cell therapies employ lentivirus transduction. Because CAR or TCR transgenes have unique sequences in the viral untranslated regions, these can be used as tags for identification of engineered cell therapies with RNAscope probes. The technology utilizes pools of paired oligos that can be thought of as a ZZ pair, where the paired 3 ends hybridize to ~50 bases of target mRNA, and where the paired 5 ends hybridize to a signal amplification module, which is built through sequential hybridization steps. The signal amplification of paired oligos results in an assay able to detect individual transcripts that appear as visible and quantifiable dots.

Its a little like planting and lighting Christmas trees, quips Bunker. The ZZ pairs plant trees along the mRNA with branches that are decorated either with fluorophores or chromogens. Although the primary technology currently features four colors, the company has developed a HiPlex (12-plex) assay and foresees even higher-plex assays with different detection methods.

We envision assays based on our core technologies that enable spatial analysis of perhaps a hundred transcripts in combination with tens of proteins, Bunker projects. In the context of cell therapy development, these will enable a more comprehensive understanding of tumor biology and immune cell profiles to determine the most effective treatment strategy for a patient, as well as for monitoring efficacy of solid tumor cellular therapies.

Companies developing CAR T-cell products are also eyeing a future involving GMP production. Thus, a critical early question is how to choose the best T-cell medium for manufacturing processes. To test the suitability of a CAR T-cell growing medium, companies must assess factors such as cell viability, cell expansion, cytokine profiles, and cell purity. A medium suitable for a CAR T-cell manufacturing process also needs to support rapid activation and CAR transduction. Additionally, the selected medium needs to be compatible with a variety of donors.

There are many available choices for T-cell culture media ranging from do-it-yourself recipes to commercially available one-size-fits-all complete formulations. CellGenix has developed a novel T-cell medium that avoids the use of human serum. Sebastian Warth, PhD, a senior scientist at CellGenix, explains, To achieve consistent results, human serum requires extensive testing prior to its use for production of cellular products due to lot-to-lot inconsistencies. Since human serum is a limited resource and might not be available in large quantities, it is unfavorable for commercial-scale manufacturing. Furthermore, the human origin of serum poses a certain risk of containing adventitious agents and is, therefore, a risk to the safety of the T-cell therapy product.

The companys TCM GMP-Prototype medium provides a serum-free and xeno-free product for early-onset T-cell expansion. According to Warth, key advantages include promotion of sustained viability, support for expansion of CD4+ and CD8+ T cells, promotion of a central memory and early differentiated memory T-cell phenotype, and maintenance of a high proportion of cytokine-producing cells including polyfunctional cells. Further, it was optimized for and verified with CAR T cells.

While autologous CAR T-cell therapies have proven highly successful, they also require a long and expensive manufacturing process. The dream of being able to utilize off-the-shelf allogeneic T cells is on the horizon.

Devon J. Shedlock, PhD, senior vice president, research and development,Poseida Therapeutics, reports, With our technology, we are able to genetically modify cells to create a fully allogenic, or off-the-shelf, product that does not require additional immunosuppression treatment like earlier generation approaches. We also have developed technology to allow us to make hundreds of doses from a single manufacturing run from healthy donors, thereby dropping the cost substantially.

According to Shedlock, the technology consists of three key aspects: 1) the piggyback DNA Modification System, 2) the Cas-CLOVER site-specific gene editing system, and 3) the Booster Molecule.

The PiggyBac DNA Modification System is a nonviral technology for stably integrating genes into DNA. One key feature is that piggyBac preferentially inserts into less mature T cells, enabling the production of therapies that have a high proportion of stem cell memory T cells, or Tscm cells.

Viral technologies are virtually excluded from Tscm cells, Shedlock states. However, Tscm cells are the ideal cell type for cell-based therapies because they have the ability to engraft and potentially last a lifetime, can produce wave after wave of more differentiated cells to attack the tumor, and are much more tolerable with low levels of adverse events compared to other CAR T-cell products.

The companys Cas-CLOVER gene editing technology is a hybrid gene editing technology used to edit the T cells to make allogeneic products. Cas-CLOVER works well in resting T cells, which is important in preserving Tscm cells in a fully allogeneic CAR T-cell product, Shedlock elaborates. It also is a very precise and clean system. This is a particularly important safety issue for allogeneic products that may be given to many patients.

The Booster Molecule is added during manufacture and is temporarily expressed on the cell surface to allow cell stimulation. Normally when allogeneic CAR T-cell products are created, the T-cell receptor must be eliminated to avoid the graft-versus-host reaction, which is a major safety issue. Importantly, this booster stimulation occurs while preserving the Tscm phenotype.

Poseida Therapeutics expects to launch a clinical trial for its multiple myeloma allogeneic product late this year or early next year. The company will also begin clinical trials later in 2021 on its pan-solid tumor allogeneic program.

Creation of partnerships can help drive development of CAR T-cell therapeutics from concept through clinical trials. Advanced therapies require advanced supply chain and data management, advises Minh Hong, PhD, head of autologous cell therapy, Lonza Pharma & Biotech. Prior biopharmaceutical models of mass production and distributionand the systems that support themare not effective for personalized therapeutics. As manufacturing demand increases for autologous cell therapies, there is an overarching need to both industrialize and simplify the entire supply chain ecosystem.

Hong says the overall project needs to be considered from a more comprehensive perspective: Due to the criticality of the starting material, everything from cell sourcing, patient coordination and scheduling for collection/infusion, transportation logistics, and manufacturing logistics needs to be coordinated, ensuring the highest standards, regulatory compliance, and safety throughout the process.

To meet these needs, Lonza is building a network of partners to develop a fully integrated vein-to-vein solution, that is, a system that includes all touch points involved in patient scheduling and sample collection, through material shipping logistics, manufacturing, and eventually the infusion of the cell therapy back into the patient. The partner network, Hong indicates, will help participants define smart workflows and execute an integration strategy. Hong sums up the networks therapeutic implications as follows: We believe these partnerships will decrease time to clinical program setup.

Lonza has more than a 20-year history of providing clinical and commercial manufacturing. Hong asserts, Our company brings to the table our process development and manufacturing experience along with proprietary solutions including a manufacturing execution system solution, MODA-ESTM, for electronic batch records and manufacturing traceability. In addition, we have announced partnerships with Vineti for a supply chain orchestration system and Cryoport to aid in shipping and logistics.

Lonza is also looking beyond CAR T-cell therapies. We would not limit our solutions and partnerships to autologous cell therapies, Hong declares. We can envision solutions for our in vivo viral vector manufacturing clients as well as our traditional allogeneic cell therapy clients.

View post:
CAR T-Cell Optimization Starts in Production, Extends to Therapy - Genetic Engineering & Biotechnology News

Read More...

Genetic Engineering Market 2020 Share Growing Rapidly with Recent Trends, Revenue, Top Players and Forecast to 2027 – Scientect

Wednesday, September 2nd, 2020

Fort Collins, Colorado Reports Globe recently added the Genetic Engineering Market research report which has an in-depth scenario analysis of the market size, share, demand, growth, trends, and the forecast from 2020-2027. The report deals with the impact analysis of the COVID-19 pandemic. The COVID-19 pandemic has affected export-import, demand and industry trends and is expected to have economic effects on the market. The report provides a comprehensive analysis of the impact of the pandemic across the industry and an overview of a post-COVID-19 market scenario.

The report mainly mentions definitions, classifications, applications, and market reviews of the Genetic Engineering industry. It also includes product portfolios, manufacturing processes, cost analyzes, structures and the gross margin of the industry. It also offers a comprehensive analysis of key competitors, their regional breakdown and market size.

Global Genetic Engineering Market to reach USD XX billion by 2025.Global Genetic Engineering Market valued approximately USD XX billion in 2017 is anticipated to grow with a healthy growth rate of more than XX% over the forecast period 2019-2026.

The report covers extensive analysis of the key market players in the market, along with their business overview, expansion plans, and strategies. The key players studied in the report include:

The report provides a comprehensive analysis in an organized manner in the form of tables, graphs, charts, figures, and diagrams. The organized data paves the way for thorough examination and research of the current and future outlook of the market.

The examination of the Genetic Engineering industry provides an in-depth analysis of the key market drivers, opportunities, challenges, and their impact on the working of the market. The technological advancements and product developments, driving the demands of the market are also covered in the report.

The report provides comprehensive data on the Genetic Engineering market and its trends to assist the reader in formulating decisions to accelerate the business. The report provides a complete overview of the economic scenario of the market, along with benefits and limitations.

Genetic Engineering market report contains industrial chain analysis and value chain analysis to provide a comprehensive view of the Genetic Engineering market. The study is composed of market analysis along with a detailed analysis of the application segments, product types, market size, growth rate, and current and emerging trends in the industry.

The report further studies the segmentation of the market based on product types offered in the market and their end-use/applications.

By Devices:

By Techniques:

By End-User:

By Application:

Geographically, the market is spread across several key geographical regions, and the report covers the regional analysis as well as the production, consumption, revenue, and market share in those regions for the forecast period of 2020-2027. The regions includeNorth America, Latin America, Europe, Asia-Pacific, and the Middle East and Africa.

Radical Coverage of the Genetic Engineering Market:

Thank you for reading our report. For further queries on the report and customization, please connect with us. Our team will ensure you get the report best suited for your needs.

How Reports Globe is different than other Market Research Providers

The inception of Reports Globe has been backed by providing clients with a holistic view of market conditions and future possibilities/opportunities to reap maximum profits out of their businesses and assist in decision making. Our team of in-house analysts and consultants works tirelessly to understand your needs and suggest the best possible solutions to fulfill your research requirements.

Our team at Reports Globe follows a rigorous process of data validation, which allows us to publish reports from publishers with minimum or no deviations. Reports Globe collects, segregates, and publishes more than 500 reports annually that cater to products and services across numerous domains.

Contact us:

Mr. Mark Willams

Account Manager

US: +1-970-672-0390

Email:[emailprotected]

Web:reportsglobe.com

View original post here:
Genetic Engineering Market 2020 Share Growing Rapidly with Recent Trends, Revenue, Top Players and Forecast to 2027 - Scientect

Read More...

From Cotton to Brinjal: Fraudulent GMO Project in India Sustained by Deception – CounterPunch

Wednesday, September 2nd, 2020

Insecticidal Bt (Bacillus thuringiensis) cotton is the first and only GM (genetically modified) crop that has been approved in India. It has been cultivated in the country for more than 20 years. In a formal statement to the Supreme Court of India, the Indian government has asserted that hybrid Bt cotton is an outstanding success. It therefore argues that Bt cotton is a templatefor the introduction of GM food crops.

However, over the last week, two important webinars took place that challenged the governments stance. The first was on Bt cotton and involved a panel of internationally renowned scientists who conclusively debunked the myth of Bt cotton success in India. The webinar, organised by the Centre for Sustainable Agriculture and Jatan, focused on an evidence-based evaluation of 18 years of approved Bt cotton cultivation in India.

The second webinar discussed the case of Bt brinjal, which the countrys apex regulatory body, the Genetic Engineering Appraisal Committee (GEAC), has brought to the brink of commercialisation. The webinar highlighted deep-seated problems with regulatory processes in India and outlined how the GEAC is dogged by secrecy, conflicts of interest and (scientific) fraud: participants outlined how the GEAC has been colluding with crop developers and seed companies to drive GM crops into agriculture.

Bt cotton failure

The panel for the Bt cotton webinar (YouTube: Bt Cotton in India: Myths & Realities An Evidence-Based Evaluation) on 24 August included Dr Andrew Paul Gutierrez, senior emeritus professor in the College of Natural Resources at the University of California at Berkeley; Dr Keshav Kranthi, former director of Central Institute for Cotton Research in India; Dr Peter Kenmore, former FAO representative in India, and Dr Hans Herren, World Food Prize Laureate.

Dr Herren said that the failure of Bt cotton is a classic representation of what an unsound science of plant protection and faulty direction of agricultural development can lead to.

He explained:

Bt hybrid technology in India represents an error-driven policy that has led to the denial and non-implementation of the real solutions for the revival of cotton in India, which lie in HDSS (high density short season) planting of non-Bt/GMO cotton in pure line varieties of native desi species and American cotton species.

He argued that a transformation of agriculture and the food system is required; one that entails a shift to agroecology, which includes regenerative, organic, biodynamic, permaculture and natural farming practices.

Dr Kenmore said that Bt cotton is an aging pest control technology:

It follows the same path worn down by generations of insecticide molecules from arsenic to DDT to BHC to endosulfan to monocrotophos to carbaryl to imidacloprid. In-house research aims for each molecule to be packaged biochemically, legally and commercially before it is released and promoted. Corporate and public policy actors then claim yield increases but deliver no more than temporary pest suppression, secondary pest release and pest resistance.

Recurrent cycles of crises have sparked public action and ecological field research which creates locally adapted agroecological strategies.

He added that this agroecology:

now gathers global support from citizens groups, governments and UN-FAO. Their robust local solutions in Indian cotton do not require any new molecules, including endo-toxins like in Bt cotton.

Prof Gutierrez presented the ecological reasons as to why hybrid Bt cotton failed in India: long season Bt cotton introduced in India was incorporated into hybrids that trapped farmers into biotech and insecticide treadmills that benefited GMO seed manufacturers.

He noted:

The cultivation of long-season hybrid Bt cotton in rainfed areas is unique to India. It is a value capture mechanism that does not contribute to yield, is a major contributor to low yield stagnation and contributes to increasing production costs.

Prof Gutierrez asserted that increases in cotton farmer suicides are related to the resulting economic distress.

He argued:

A viable solution to the current GM hybrid system is adoption of improved non-GM high-density short-season fertile cotton varieties.

Presenting data on yields, insecticide usage, irrigation, fertiliser usage and pest incidence and resistance, Dr Keshav Kranthi said that a critical analysis of official statistics (eands.dacnet.nic.inandcotcorp.gov.in) shows that Bt hybrid technology has not been providing any tangible benefits in India either in yield or insecticide usage.

He said that cotton yields are the lowest in the world in Maharashtra, despite being saturated with Bt hybrids and the highest use of fertilisers. Yields in Maharashtra are less than in rainfed Africa where there is hardly any usage of technologies such as Bt, hybrids, fertilisers, pesticides or irrigation.

It is revealing that Indian cotton yields rank 36th in the world and have been stagnant in the past 15 years and insecticide usage has been constantly increasing after 2005, despite an increase in area under Bt cotton.

Dr Kranthi argued that research also shows that the Bt hybrid technology has failed the test of sustainability with resistance in pink bollworm to Bt cotton, increasing sucking pest infestation, increasing trends in insecticide and fertiliser usage, increasing costs and negative net returns in 2014 and 2015.

Dr Herren said that GMOs exemplify the case of a technology searching for an application:

It is essentially about treating symptoms, rather than taking a systems approach to create resilient, productive and bio-diverse food systems in the widest sense and to provide sustainable and affordable solutions in its social, environmental and economic dimensions.

He went on to argue that the failure of Bt cotton is a classic representation of what an unsound science of plant protection and a faulty direction of agricultural development can lead to:

We need to push aside the vested interests blocking the transformation with the baseless arguments of the world needs more food and design and implement policies that are forward looking We have all the needed scientific and practical evidence that the agroecological approaches to food and nutrition security work successfully.

Bt brinjal the danger is back

The governments attempt to use a failed technology as a template for driving GMOs into agriculture has been exposed. Nevertheless, the GEAC has been moving forward with late-stage trials of Bt brinjal, while ignoring the issues and arguments against its commercialisation that were forwarded a decade ago.

In February 2010, the Indian government placed an indefinite moratorium on the release of Bt brinjal after numerous independent scientific experts from India and abroad had pointed out safety concerns based on data and reports in the biosafety dossier that Mahyco, the crop developer, had submitted to the regulators.

The then Minister of the Ministry of Environment and Forests Jairam Ramesh had instituted a unique four-month scientific enquiry and public hearings. His decision to reject the commercialisation of Bt brinjal was supported by advice from the renowned scientists.Their collective appraisals demonstrated serious environmental and biosafety concerns.

Jairam Ramesh pronounced a moratorium on Bt brinjal in February 2010 by stating:

it is my duty to adopt a cautious, precautionary principle-based approach and impose a moratorium on the release of Bt brinjal, till such time independent scientific studies establish, to the satisfaction of both the public and professionals, the safety of the product from the point of view of its long-term impact on human health and environment, including the rich genetic wealth existing in brinjal in our country.

The moratorium has not been lifted and the conditions he set out have still not been met. Moreover, five high-level reportshave advised against the adoption of GM crops in India. Appointed by the Supreme Court, the Technical Expert Committee (TEC) Final Report (2013) was scathing about the prevailing regulatory system and highlighted its inadequacies. The TEC went a step further by recommending a 10-year moratorium on the commercial release of all GM crops.

The regulatory process was shown to lack competency, possessed endemic conflicts of interest and demonstrated a lack of expertise in GMO risk assessment protocols, including food safety assessment and the assessment of environmental impacts.

Ten years on and regulators have done nothing to address this woeful state of affairs. As we have seen with the relentless push to get GM mustard commercialised, the problems persist. Through numerous submissions to the Supreme Court, Aruna Rodrigues has described how GM mustard is being forced through withflawed tests (or no tests) and a lack of public scrutiny.Regulators are seriously conflicted: they promote GMOs openly, fund them and then regulate them.

And this is precisely what the webinar Bt brinjal the danger is back (watch onYouTube) discussed on 27 August. Organised by the Coalition for a GM-Free India, the webinar was arranged because the regulators have again brought to the brink of commercialisation a new Bt brinjal event a different Bt brinjal than the 2010 version. Also included in the webinar were the experiences of Bt brinjal introduction in Bangladesh.

Dr Ramanjaneyulu (Centre for Sustainable Agriculture) highlighted how need has never been established for Bt brinjal of which India is a recognised centre of diversity. The argument for Bt brinjal in the run-up to Jairam Rameshs moratorium was that pesticide use is a problem in containing the brinjal fruit and shoot borer. He noted that Bt brinjal was promoted by Monsanto, USAID and Cornell University, but serious protocol violations, environmental contamination concerns and potential adverse health impacts were discovered.

He outlined simple non-pesticidal, agroecological management practises that can and are being used to deal with the brinjal fruit and shoot borer.

Farida Akhter of UBINIG (Policy Research for Development Alternative) outlined how the introduction of Bt brinjal in Bangladesh was not needed but imposed on the country, which has 248 varieties of brinjal. Where pesticide use is problematic, she argued that it concerns hybrid varieties rather than traditional cultivars of which 24 varieties are resistant to fruit and shoot borer.

Akhter said that poor quality brinjal and financial losses for farmers have been major issues. Many have abandoned Bt brinjal, but farmers have received incentives to cultivate and where they have done so, fertiliser use has increased and there have been many pest attacks, with 35 different types of pesticides applied.

The Bill Gates-funded Cornell Alliance for Science, a public relations entity that promotes GM agriculture, and USAID, which serves the interests of the GMO biotech sector, tried to sell Bt brinjal on the basis it would save people from the overuse of pesticides and related illnesses. But Akhter argued that Bangladesh was targeted because the Philippines and India had rejected Bt brinjal. Again, protocol violations occurred leading to its introduction and Akhter concluded that there was no scientific basis for Bt brinjal: its introduction was political.

As for India, event EE1, the initial Bt brinjal, has now been replaced by event 142, a different Bt brinjal. Kavitha Kuruganti (Alliance for Sustainable and Holistic Agriculture) explained this in the webinar and notes that the GEAC, immediately after the 2010 moratorium was announced, went straight ahead and sanctioned new trials for this Bt brinjal. The GEAC basically stated that the moratorium did not apply to this version, while ignoring all the criticisms about lack of competence, conflicts of interest, non-transparency and protocol violations. It was effectively business as usual!

With event EE1, Kuruganti implied that the GEAC acted more like a servant for Mayco and its Monsanto master. Nothing has changed. She noted the ongoing revolving door between crop developers (even patent holders) and regulators. As before, developers-cum-lobbyists were actually sitting on regulatory bodies as event 142 was proceeding.

Under public-private-partnership arrangements, event 142 has been licensed to private companies for biosafety testing/commercialisation. Despite major concerns, the GEAC has pressed ahead with various trials. In May 2020, under lockdown, Kuruganti notes that the GEAC held a virtual meeting and sanctioned what were effectively final trials prior to commercialisation. She explains that important information and vital data is not in the public domain.

According to Kuruganti, the regulator sits with the crop developer and the companies and grant biosafety clearance, claiming all tests (soil, pollen flow, toxicity, etc) are complete. What is also disturbing is that these licensed companies have closed and opened under new names (with the same people in charge), thereby making accountability and liability fixing very difficult if something were to go wrong further down the line.

She concludes that the story of event 142 is even worse than event EE1:

Once again, they are certainly hiding things that they dont want conscientious scientists and aware citizens to see and know.

Taken together, the two webinars highlighted how hybrid Bt cotton is being deceptively used as a template for rolling out GM food crops: a fraud being used to promote another fraud in order to force unnecessary GMOs into Indian agriculture.

See the original post:
From Cotton to Brinjal: Fraudulent GMO Project in India Sustained by Deception - CounterPunch

Read More...

Nucleic Acid Isolation and Purification Market : The Rescuer in COVID-19 Crisis, Says TMR – Owned

Wednesday, September 2nd, 2020

Nucleic acid isolation and purification is an initial step in molecular biology studies and recombinant DNA techniques. The process of isolation includes mechanical and chemical disruption, enzymatic digestion, while the purification involves combination of extraction/precipitation, chromatography, centrifugation, electrophoresis, and affinity separation.

View Report : https://www.transparencymarketresearch.com/nucleic-acid-isolation-purification-market.html

This technique has wide applications in the field of genetic engineering, life science research, forensics and molecular diagnostics. Nucleic acid isolation helps in processing of more sample in less time, minimizes nucleic acid loss ad degradation and increases laboratory efficiency and effectiveness. The purified product can thus be used in recombinant technology methods, and be used in targeted purposes in research.

The rising demand of pure nucleic acids in pharmaceutical and biotechnological industries, and growing use of nucleic acid-based tests in diagnosis have propelled the growth of this market across the globe. Moreover, increasing applications of these techniques in various fields such as genetic engineering, life science research, forensics and molecular diagnostics and government funding in R&Ds and the recent technological innovations are expected to fuel the growth of this market.

Get PDF Sample Copy of Report: (Including TOC, List of Tables & Figures, Chart) : https://www.transparencymarketresearch.com/sample/sample.php?flag=B&rep_id=9521

In addition, emergence of new technologies in molecular diagnostics, expression analysis and genotyping would create an opportunity for the suppliers in future. However, low market penetration of automated nucleic acid isolation and purification procedures in developing countries, is a key factor restraining the growth of global nucleic acid isolation and purification market. Similarly, higher prices of the instruments associated with these procedures is one of the major challenges for this market. New product development, mergers and acquisition and partnership are some of the key trends in nucleic acid isolation and purification market.

The market is segmented into technology, application, product, end user, and geography. Based on various type of technology, the market can be segmented as: columnbased, magnetic bead-based, reagent-based, and other (anion exchange-based, glass fiber-based) for DNA and RNA isolation and purification. Column-based technology for DNA isolation and purification commands the largest share of the global market. Magnetic bead-based technologyis poised to grow at fastest rate.

Request For Covid19 Impact Analysis https://www.transparencymarketresearch.com/sample/sample.php?flag=covid19&rep_id=9521

Application segments of this market includes genomic DNA isolation and purification, micro RNA isolation and purification, blood DNA isolation and purification, mRNA isolation and purification, plasmid DNA isolation and purification, total RNA isolation and purification, and PCR clean up. The plasmid DNA isolation and purification is the leading market segment by application. The end-users of the market are academic research, hospitals and diagnostic centers, pharmaceutical and biotechnology companies, contract research organizations, and other end users. Nucleic acid isolation and purification market have its major share in academic research organizations.

Read our Case study at :https://www.transparencymarketresearch.com/casestudies/innovative-medical-device-manufacturing-start-up

North America holds the leading position in the Nucleic Acid Isolation and Purification market followed by Europe. Asia Pacific is the most promising market for the growth of market due to various emerging economies. The market in the region is easy to penetrate and it is expected to have a huge future scope in the region, especially India and China, so the players are looking invest more in Asia-Pacific region. Some of the key players in global Nucleic Acid Isolation and Purification Market are Agilent Technologies Inc. (U.S.), Bio-Rad Laboratories Inc. (U.S.), Danaher Corporation (U.S.), Illumina Inc. (U.S.), Life Technologies (U.S.), Promega Corporation (U.S.)., among others.

More Trending Reports by Transparency Market Research 1.https://www.prnewswire.com/news-releases/global-radiation-therapy-market-to-reach-us-8-6-bn-by-2026product-approvals-to-drive-growth-transparency-market-research-300998453.html

2.https://www.biospace.com/article/dna-test-kits-market-increase-in-demand-for-ancestry-testing-to-drive-market/

See more here:
Nucleic Acid Isolation and Purification Market : The Rescuer in COVID-19 Crisis, Says TMR - Owned

Read More...

Bacterial Superglue Allows Adhesion to the Gut – Genetic Engineering & Biotechnology News

Wednesday, September 2nd, 2020

Before bacteria colonize a tissue in the human body, such as the intestine, they have to attach. Not only that, they have to achieve firm adhesion under hydrodynamic flow. New research reports a molecular mechanism behind an ultrastable protein complex responsible for resisting shear forces and adhering bacteria to cellulose fibers in the human gut. The results explain how gut microbes regulate cell adhesion strength at high shear stress through intricate molecular mechanisms including dual-binding modes, mechanical allostery, and catch bonds.

The researchers used single-molecule force spectroscopy (SMFS), single-molecule FRET (smFRET), and molecular dynamics (MD) simulations to uncover that two different binding modes allow bacteria to withstand the shear forces in the body. The findings are published in Nature Communications in the paper titled, High force catch bond mechanism of bacterial adhesion in the human gut.

Cellulose is a major building block of plant cell walls, consisting of molecules linked together into solid fibers. For humans, cellulose is indigestible, and the majority of gut bacteria lack the enzymes required to break down cellulose.

However, recently genetic material from the cellulose-degrading bacterium R. champanellensis was detected in human gut samples. Bacterial colonization of the intestine is essential for human physiology, and understanding how gut bacteria adhere to cellulose broadens our knowledge of the microbiome and its relationship to human health.

The bacterium under investigation uses an intricate network of scaffold proteins and enzymes on the outer cell wall, referred to as a cellulosome network, to attach to and degrade cellulose fibers. These cellulosome networks are held together by families of interacting proteins.

Of particular interest is the cohesin-dockerin interaction responsible for anchoring the cellulosome network to the cell wall. This interaction needs to withstand shear forces in the body to adhere to fiber. This vital feature motivated the researchers to investigate in more detail how the anchoring complex responds to mechanical forces.

By using a combination of single-molecule atomic force microscopy, single-molecule fluorescence, and molecular dynamics simulations, Michael Nash, PhD, assistant professor with joint appointments at the University of Basel, department of chemistry, and at ETH Zurich, department of biosystems science & engineering, along with collaborators from LMU Munich and Auburn University, studied how the complex resists external force.

Two binding modes allow bacteria to stick to surfaces under shear flow

They were able to show that the complex exhibits a rare behavior called dual binding mode, where the proteins form a complex in two distinct ways. The researchers found that the two binding modes have very different mechanical properties, with one breaking at low forces of around 200 piconewtons and the other exhibiting a much higher stability breaking only at 600 piconewtons of force.

Further analysis showed that the protein complex displays a behavior called a catch bond, meaning that the protein interaction becomes stronger as force is ramped up. The dynamics of this interaction are believed to allow the bacteria to adhere to cellulose under shear stress and release the complex in response to new substrates or to explore new environments.

We clearly observe the dual binding modes, but can only speculate on their biological significance. We think the bacteria might control the binding mode preference by modifying the proteins. This would allow switching from a low to high adhesion state depending on the environment, Nash explained.

By shedding light on this natural adhesion mechanism, these findings set the stage for the development of artificial molecular mechanisms that exhibit similar behavior but bind to disease targets. Such materials could have applications in bio-based medical superglues or shear-enhanced binding of therapeutic nanoparticles inside the body. For now, we are excited to return to the laboratory and see what sticks, said Nash.

Read the original here:
Bacterial Superglue Allows Adhesion to the Gut - Genetic Engineering & Biotechnology News

Read More...

Viewpoint: Is there a scientific basis to ban gene drive technology that can rid us of virus-carrying rodents and mosquitoes? – Genetic Literacy…

Tuesday, August 18th, 2020

Gene drives may be invaluable tools to control the spread of parasites, invasive species, and disease carriers. But the technology has faced strong opposition from activist groups and some mainstream scientists based on environmental and food safety. Are these concerns valid?

On June 30, some 80 environmental organizations, led by Greenpeace EU, Friends of the Earth Europe and Save Our Seeds, signed an open letter to the European Commission asking for support for a global moratorium on gene drive technology. The advocacy groups claimed that the release of gene drives poses serious and novel threats to biodiversity and the environment at an unprecedented scale and depth.

Citing a report by the European Network of Scientists for Social and Environmental Responsibility (ENSSER), the coalition wrote:

in light of the unpredictabilities, the lack of knowledge and the potentially severe negative impacts on biodiversity and ecosystems, any releases (including experimental) of Gene Drive Organisms into the environment be placed on hold to allow proper investigation until there is sufficient knowledge and understanding.

The environmental claims were unsupported by any documents other than the report by ENSSER, a controversial group of anti-biotechnology activist scientists co-founded by Gilles-ricSralini, best known for his retracted and discredited 2012 paper linking GMOs to cancer in rats.

The European parliament has already supported such a moratorium, an act that echoes EUs precautionary approach to genetic engineering, transgenic organisms and gene editing. The EU stated reasons include:

Recent advances in genetics and synthetic biology, particularly the development of CRISPR gene editing tools, have given scientists a powerful way to address problems created by pests, from mosquitoes to rodents, that vector disease to humans. In classical genetics, genes that offer adaptation benefits to individuals tend to increase their occurrence in the population while genes that reduce fitness tend to disappear.

Gene drives are genetic sequences designed to spread strongly and become present in every individual of a targeted species after a few generations. The genes may offer benefits, be neutral for adaptation purposes, or hinder their carriers survival and reproduction potential.Generation after generation, it would relentlessly copy and paste the gene it carried, until the gene and the desired trait was present in every descendant.Because the spread of a trait happens over generations, a gene drive works best in species that reproduce quickly, like insects and rodents

Gene drives are the first genetic constructs that can theoretically affect a population in its entirety, and quickly. It could even lead to the extinction of entire species, as gene drive critics allege. Species distinction has been part of life and evolution for all of Earths history. Although the data are fuzzy and contested, the UN Convention on Biological Diversity concluded that 150-200 plant, insect bird, and mammal species go extinct every day.

The likelihood that a gene drive will destroy a species in part or in whole, such as the infectedAedes aegyptimosquito species that carries the Zika, dengue and chingunya viruses and offers no known environmental benefits, is nonetheless daunting to some. On the one hand, gene drives could be used to eradicate disease such as malaria and yellow fever by controlling the mosquitoes that transmit them. On the other hand, critics fear that the technology will open a Pandoras Box; removing a species that theoretically could resultin what is popularly and controversially known as the butterfly effect.

As imagined by MIT meteorologist Edward Lorenz 60 years ago, a tiny environmental changesay an extinction of a pestcould dramatically and unpredictably result in unpredictable or even catastrophic consequences (Lorenz imagined abutterflyflapping its wings and causing a typhoon).

In the last few years, various groups have called for a global moratorium on gene drives. Such attempts were resisted at the 2016 and 2018 United Nations Conventions on Biological Diversity, mainly due to the strong opposition of many scientists and sub-Saharan African nations hardest hit by disease-vectored pests. Nevertheless, gene drive opponents have gained traction and gene drive research and applications face significant regulatory obstacles across the world (see Genetic Literacys Global Gene Editing Regulation tracker for a country-by-country analysis).

What does the scientific evidence say about gene drives and their environmental consequences?

There are over 3,000 mosquito species, likely a fraction of the number of species that have existed over some 100 million years. A handful of these (Aedes, Anopheles, and Culex species) are disease vectors and transmit infections such as malaria, yellow fever, the West Nile virus, Zika, and dengue fever. Mosquito-borne disease account for more than 17% of all infectious diseases and cause more than 700,000 deaths every year. These mosquitoes are mostly invasive in their ecological distributions.

Ultimately, there seem to be few things that mosquitoes do that other organisms cant do just as wellexcept perhaps for one, reported Nature magazine ina 2010 article A World Without Mosquitoes.

They are lethally efficient at sucking blood from one individual and mainlining it into another, providing an ideal route for the spread of pathogenic microbes. The Nature article concluded that wiping out mosquitoes wouldnt be a badthing. In fact, they could restore rather than harm the ecosystem. The same can be inferred for most parasitic insects, which are specialized to a particular host and normally dont have an extended ecological interactions network.

Invasive species also cause significant environmental hazards. Cane toads, having no natural predators, are slowly taking over the Australian continent from the northeast. Invasive fish from the red sea are wrecking havoc in the Mediterranean marine ecosystems. Rodents have spread in every conceivable corner of the earth, displacing vulnerable local fauna.

Gene drives might be one of the only ways to contain their spread, protecting biodiversity. They can be a powerful conservation tool that targets only the organism of interest, unlike contemporary pest management techniques such as the use of insecticides that attack all insects indiscriminately, or introduction of natural predators from other ecosystems (that by default disturb the food chains and interactions network).

It is possible for a DNA sequence to jump from one species to the other through a process called horizontal gene transfer. This theoretically could happen between insects, which appears to lend support to the argument that there is at least a small chance for a gene drive to move from species to species with unforeseen consequences.

The truth is that gene drives can be designed to target a very specific area of the genome, unique for a species. The modern gene drives use the precise CRISPR base editing technologies to spread to the population. In the off chance that the DNA encoding the gene drive will enter the reproductive cells of an individual from the other species, the editing system will have no template to act upon and the gene will be lost. One may argue that CRISPR has a chance for off-target activity, but a gene drive needs maximum efficiency to act as a gene drive. If the CRISPR doesnt work at 100%, the DNA sequence will be subject to the typical laws of inheritance and will disappear from the genetic pool

The ability to introduce genetic information to a wild population, which will spread to every individual, is unfortunately a dual use technology. The technology can theoretically be exploited to make biological weapons, though theres no indication that such a weapon is or has been developed. As gene drives can work well across many generations and require a large amount of offspring, they are unable to directly harm humans, crops, and farm animals. But a gene drive could be used to enhance the fitness of a crop-eating insect or a disease-carrying rodent.

The solution to this potential hazard is more research (and definitely not a research moratorium). Anyone with the means (which are considerable, so no lone bioterrorists or rogue scientists) and intent to cause harm can already research into such applications and will ignore aUN-imposed technology ban. The research community needs to develop the means to detect and monitor any malicious gene drive release and counter any offensive use.

The question on who and how should approve gene drive projects isnt easy to answer. A gene drive isnt contained by country borders, and the outdated GMO regulation framework existing in most countries is scientifically outdated and practically inadequate to handle such applications.

Moreover, the technology cannot be monopolized by a few countries or private companies. Each project is different. The approval should be a result of consensus among numerous stakeholders. There should also be a defined way to monitor how the gene drive spreads and how to handle liability claims if there are negative effects.

With populism growing and fewer people willing to trust the judgment of regulators and scientists, the rhetoric around complex innovations has become increasingly polarized, with both sides stuck fighting a high-stakes battle for public opinion. The issue is complex, and any decisions cannot be left to scientists, state organizations, and companies alone. But it also cannot be left solely in the hands of environmental organizations with little or no understanding of the science and with an ideological agenda that doesnt necessarily serve the public.

Environmental groups have often resorted to hyperbole as the debate over gene drives has unfolded. At the UN Convention on Biological Diversity in Sharm el Sheikh, Egypt, in 2018, a coalition of activists compared gene drives to the atomic bomb and accused researchers of using malaria as a Trojan horse to cover up the development of agricultural gene drives for corporate profit.A handful of small NGOs in the US, collectively known as SynBioWatch, have taken to describing gene-drive researchers as a cabal. The Canadian anti-biotechnology organization ETC Group claims aggressively spreads misinformation on social media, including claims that gene-drive honeybees could supposedly be controlled with a beam of light.

Meanwhile, Florida Keys is experiencing the largest dengue fever outbreak in a decade, with close to 40 cases already documented. The outbreak has led the Florida Keys Mosquito Control District to enter a partnership with UK-based, US-owned Oxitec that could lead to the Keys becoming the first U.S. trial site for genetically modified Aedes aegypti mosquitoes.

With a technology that can prevent hundreds of thousands of deaths per year, it is unethical to peremptorily ban it because it doesnt fit a few peoples worldview of what is natural. One may argue that governments and regulators should have no say whether one species should go extinct or not. But one can also question why activist groups in North America or Europe should be able to insert themselves in life and death decisions, preventing initiatives across the globe that could save millions of lives and protect our populations health and crops, and promote biological diversity.

Kostas Vavitsas, PhD, is a Senior Research Associate at the University of Athens, Greece. He is also a steering committee member of EUSynBioS. Follow him on Twitter@konvavitsas

Excerpt from:
Viewpoint: Is there a scientific basis to ban gene drive technology that can rid us of virus-carrying rodents and mosquitoes? - Genetic Literacy...

Read More...

Biotechnology Could Change the Cattle Industry. Will It Succeed? – Singularity Hub

Tuesday, August 18th, 2020

When Ralph Fisher, a Texas cattle rancher, set eyes on one of the worlds first cloned calves in August 1999, he didnt care what the scientists said: He knew it was his old Brahman bull, Chance, born again. About a year earlier, veterinarians at Texas A&M extracted DNA from one of Chances moles and used the sample to create a genetic double. Chance didnt live to meet his second self, but when the calf was born, Fisher christened him Second Chance, convinced he was the same animal.

Scientists cautioned Fisher that clones are more like twins than carbon copies: The two may act or even look different from one another. But as far as Fisher was concerned, Second Chance was Chance. Not only did they look identical from a certain distance, they behaved the same way as well. They ate with the same odd mannerisms; laid in the same spot in the yard. But in 2003, Second Chance attacked Fisher and tried to gore him with his horns. About 18 months later, the bull tossed Fisher into the air like an inconvenience and rammed him into the fence. Despite 80 stitches and a torn scrotum, Fisher resisted the idea that Second Chance was unlike his tame namesake, telling the radio program This American Life that I forgive him, you know?

In the two decades since Second Chance marked a genetic engineering milestone, cattle have secured a place on the front lines of biotechnology research. Today, scientists around the world are using cutting-edge technologies, from subcutaneous biosensors to specialized food supplements, in an effort to improve safety and efficiency within the $385 billion global cattle meat industry. Beyond boosting profits, their efforts are driven by an imminent climate crisis, in which cattle play a significant role, and growing concern for livestock welfare among consumers.

Gene editing stands out as the most revolutionary of these technologies. Although gene-edited cattle have yet to be granted approval for human consumption, researchers say tools like Crispr-Cas9 could let them improve on conventional breeding practices and create cows that are healthier, meatier, and less detrimental to the environment. Cows are also being given genes from the human immune system to create antibodies in the fight against Covid-19. (The genes of non-bovine livestock such as pigs and goats, meanwhile, have been hacked to grow transplantable human organs and produce cancer drugs in their milk.)

But some experts worry biotech cattle may never make it out of the barn. For one thing, theres the optics issue: Gene editing tends to grab headlines for its role in controversial research and biotech blunders. Crispr-Cas9 is often celebrated for its potential to alter the blueprint of life, but that enormous promise can become a liability in the hands of rogue and unscrupulous researchers, tempting regulatory agencies to toughen restrictions on the technologys use. And its unclear how eager the public will be to buy beef from gene-edited animals. So the question isnt just if the technology will work in developing supercharged cattle, but whether consumers and regulators will support it.

Cattle are catalysts for climate change. Livestock account for an estimated 14.5 percent of greenhouse gas emissions from human activities, of which cattle are responsible for about two thirds, according to the United Nations Food and Agriculture Organization (FAO). One simple way to address the issue is to eat less meat. But meat consumption is expected to increase along with global population and average income. A 2012 report by the FAO projected that meat production will increase by 76 percent by 2050, as beef consumption increases by 1.2 percent annually. And the United States is projected to set a record for beef production in 2021, according to the Department of Agriculture.

For Alison Van Eenennaam, an animal geneticist at the University of California, Davis, part of the answer is creating more efficient cattle that rely on fewer resources. According to Van Eenennaam, the number of dairy cows in the United States decreased from around 25 million in the 1940s to around 9 million in 2007, while milk production has increased by nearly 60 percent. Van Eenennaam credits this boost in productivity to conventional selective breeding.

You dont need to be a rocket scientist or even a mathematician to figure out that the environmental footprint or the greenhouse gases associated with a glass of milk today is about one-third of that associated with a glass of milk in the 1940s, she says. Anything you can do to accelerate the rate of conventional breeding is going to reduce the environmental footprint of a glass of milk or a pound of meat.

Modern gene-editing tools may fuel that acceleration. By making precise cuts to DNA, geneticists insert or remove naturally occurring genes associated with specific traits. Some experts insist that gene editing has the potential to spark a new food revolution.

Jon Oatley, a reproductive biologist at Washington State University, wants to use Crispr-Cas9 to fine tune the genetic code of rugged, disease-resistant, and heat-tolerant bulls that have been bred to thrive on the open range. By disabling a gene called NANOS2, he says he aims to eliminate the capacity for a bull to make his own sperm, turning the recipient into a surrogate for sperm-producing stem cells from more productive prized stock. These surrogate sires, equipped with sperm from prize bulls, would then be released into range herds that are often genetically isolated and difficult to access, and the premium genes would then be transmitted to their offspring.

Furthermore, surrogate sires would enable ranchers to introduce desired traits without having to wrangle their herd into one place for artificial insemination, says Oatley. He envisions the gene-edited bulls serving herds in tropical regions like Brazil, the worlds largest beef exporter and home to around 200 million of the approximately 1.5 billion head of cattle on Earth.

Brazils herds are dominated by Nelore, a hardy breed that lacks the carcass and meat quality of breeds like Angus but can withstand high heat and humidity. Put an Angus bull on a tropical pasture and hes probably going to last maybe a month before he succumbs to the environment, says Oatley, while a Nelore bull carrying Angus sperm would have no problem with the climate.

The goal, according to Oatley, is to introduce genes from beefier bulls into these less efficient herds, increasing their productivity and decreasing their overall impact on the environment. We have shrinking resources, he says, and need new, innovative strategies for making those limited resources last.

Oatley has demonstrated his technique in mice but faces challenges with livestock. For starters, disabling NANOS2 does not definitively prevent the surrogate bull from producing some of its own sperm. And while Oatley has shown he can transplant sperm-producing cells into surrogate livestock, researchers have not yet published evidence showing that the surrogates produce enough quality sperm to support natural fertilization. How many cells will you need to make this bull actually fertile? asks Ina Dobrinski, a reproductive biologist at the University of Calgary who helped pioneer germ cell transplantation in large animals.

But Oatleys greatest challenge may be one shared with others in the bioengineered cattle industry: overcoming regulatory restrictions and societal suspicion. Surrogate sires would be classified as gene-edited animals by the Food and Drug Administration, meaning theyd face a rigorous approval process before their offspring could be sold for human consumption. But Oatley maintains that if his method is successful, the sperm itself would not be gene-edited, nor would the resulting offspring. The only gene-edited specimens would be the surrogate sires, which act like vessels in which the elite sperm travel.

Even so, says Dobrinski, Thats a very detailed difference and Im not sure how that will work with regulatory and consumer acceptance.

In fact, American attitudes towards gene editing have been generally positive when the modification is in the interest of animal welfare. Many dairy farmers prefer hornless cowshorns can inflict damage when wielded by 1,500-pound animalsso they often burn them off in a painful process using corrosive chemicals and scalding irons. In a study published last year in the journal PLOS One, researchers found that most Americans are willing to consume food products from cows genetically modified to be hornless.

Still, experts say several high-profile gene-editing failures in livestock and humans in recent years may lead consumers to consider new biotechnologies to be dangerous and unwieldy.

In 2014, a Minnesota startup called Recombinetics, a company with which Van Eenennaams lab has collaborated, created a pair of cross-bred Holstein bulls using the gene-editing tool TALENs, a precursor to Crispr-Cas9, making cuts to the bovine DNA and altering the genes to prevent the bulls from growing horns. Holstein cattle, which almost always carry horned genes, are highly productive dairy cows, so using conventional breeding to introduce hornless genes from less productive breeds can compromise the Holsteins productivity. Gene editing offered a chance to introduce only the genes Recombinetics wanted. Their hope was to use this experiment to prove that milk from the bulls female progeny was nutritionally equivalent to milk from non-edited stock. Such results could inform future efforts to make Holsteins hornless but no less productive.

The experiment seemed to work. In 2015, Buri and Spotigy were born. Over the next few years, the breakthrough received widespread media coverage, and when Buris hornless descendant graced the cover of Wired magazine in April 2019, it did so as the ostensible face of the livestock industrys future.

But early last year, a bioinformatician at the FDA ran a test on Buris genome and discovered an unexpected sliver of genetic code that didnt belong. Traces of bacterial DNA called a plasmid, which Recombinetics used to edit the bulls genome, had stayed behind in the editing process, carrying genes linked to antibiotic resistance in bacteria. After the agency published its findings, the media reaction was swift and fierce: FDA finds a surprise in gene-edited cattle: antibiotic-resistant, non-bovine DNA, read one headline. Part cow, part bacterium? read another.

Recombinetics has since insisted that the leftover plasmid DNA was likely harmless and stressed that this sort of genetic slipup is not uncommon.

Is there any risk with the plasmid? I would say theres none, says Tad Sonstegard, president and CEO of Acceligen, a Recombinetics subsidiary. We eat plasmids all the time, and were filled with microorganisms in our body that have plasmids. In hindsight, Sonstegard says his teams only mistake was not properly screening for the plasmid to begin with.

While the presence of antibiotic-resistant plasmid genes in beef probably does not pose a direct threat to consumers, according to Jennifer Kuzma, a professor of science and technology policy and co-director of the Genetic Engineering and Society Center at North Carolina State University, it does raise the possible risk of introducing antibiotic-resistant genes into the microflora of peoples digestive systems. Although unlikely, organisms in the gut could integrate those genes into their own DNA and, as a result, proliferate antibiotic resistance, making it more difficult to fight off bacterial diseases.

The lesson that I think is learned there is that science is never 100 percent certain, and that when youre doing a risk assessment, having some humility in your technology product is important, because you never know what youre going to discover further down the road, she says. In the case of Recombinetics. I dont think there was any ill intent on the part of the researchers, but sometimes being very optimistic about your technology and enthusiastic about it causes you to have blinders on when it comes to risk assessment.

The FDA eventually clarified its results, insisting that the study was meant only to publicize the presence of the plasmid, not to suggest the bacterial DNA was necessarily dangerous. Nonetheless, the damage was done. As a result of the blunder,a plan was quashed forRecombinetics to raise an experimental herd in Brazil.

Backlash to the FDA study exposed a fundamental disagreement between the agency and livestock biotechnologists. Scientists like Van Eenennaam, who in 2017 received a $500,000 grant from the Department of Agriculture to study Buris progeny, disagree with the FDAs strict regulatory approach to gene-edited animals. Typical GMOs are transgenic, meaning they have genes from multiple different species, but modern gene-editing techniques allow scientists to stay roughly within the confines of conventional breeding, adding and removing traits that naturally occur within the species. That said, gene editing is not yet free from errors and sometimes intended changes result in unintended alterations, notes Heather Lombardi, division director of animal bioengineering and cellular therapies at the FDAs Center for Veterinary Medicine. For that reason, the FDA remains cautious.

Theres a lot out there that I think is still unknown in terms of unintended consequences associated with using genome-editing technology, says Lombardi. Were just trying to get an understanding of what the potential impact is, if any, on safety.

Bhanu Telugu, an animal scientist at the University of Maryland and president and chief science officer at the agriculture technology startup RenOVAte Biosciences, worries that biotech companies will migrate their experiments to countries with looser regulatory environments. Perhaps more pressingly, he says strict regulation requiring long and expensive approval processes may incentivize these companies to work only on traits that are most profitable, rather than those that may have the greatest benefit for livestock and society, such as animal well-being and the environment.

What company would be willing to spend $20 million on potentially alleviating heat stress at this point? he asks.

On a windy winter afternoon, Raluca Mateescu leaned against a fence post at the University of Floridas Beef Teaching Unit while a Brahman heifer sniffed inquisitively at the air and reached out its tongue in search of unseen food. Since 2017, Mateescu, an animal geneticist at the university, has been part of a team studying heat and humidity tolerance in breeds like Brahman and Brangus (a mix between Brahman and Angus cattle). Her aim is to identify the genetic markers that contribute to a breeds climate resilience, markers that might lead to more precise breeding and gene-editing practices.

In the South, Mateescu says, heat and humidity are a major problem. That poses a stress to the animals because theyre selected for intense productionto produce milk or grow fast and produce a lot of muscle and fat.

Like Nelore cattle in South America, Brahman are well-suited for tropical and subtropical climates, but their high tolerance for heat and humidity comes at the cost of lower meat quality than other breeds. Mateescu and her team have examined skin biopsies and found that relatively large sweat glands allow Brahman to better regulate their internal body temperature. With funding from the USDAs National Institute of Food and Agriculture, the researchers now plan to identify specific genetic markers that correlate with tolerance to tropical conditions.

If were selecting for animals that produce more without having a way to cool off, were going to run into trouble, she says.

There are other avenues in biotechnology beyond gene editing that may help reduce the cattle industrys footprint. Although still early in their development, lab-cultured meats may someday undermine todays beef producers by offering consumers an affordable alternative to the conventionally grown product, without the animal welfare and environmental concerns that arise from eating beef harvested from a carcass.

Other biotech techniques hope to improve the beef industry without displacing it. In Switzerland, scientists at a startup called Mootral are experimenting with a garlic-based food supplement designed to alter the bovine digestive makeup to reduce the amount of methane they emit. Studies have shown the product to reduce methane emissions by about 20 percent in meat cattle, according to the New York Times.

In order to adhere to the Paris climate agreement, Mootrals owner, Thomas Hafner, believes demand will grow as governments require methane reductions from their livestock producers. We are working from the assumption that down the line every cow will be regulated to be on a methane reducer, he told the New York Times.

Meanwhile, a farm science research institute in New Zealand, AgResearch, hopes to target methane production at its source by eliminating methanogens, the microbes thought to be responsible for producing the greenhouse gas in ruminants. The AgResearch team is attempting to develop a vaccine to alter the cattle guts microbial composition, according to the BBC.

Genomic testing may also allow cattle producers to see what genes calves carry before theyre born, according to Mateescu, enabling producers to make smarter breeding decisions and select for the most desirable traits, whether it be heat tolerance, disease resistance, or carcass weight.

Despite all these efforts, questions remain as to whether biotech can ever dramatically reduce the industrys emissions or afford humane treatment to captive animals in resource-intensive operations. To many of the industrys critics, including environmental and animal rights activists, the very nature of the practice of rearing livestock for human consumption erodes the noble goal of sustainable food production. Rather than revamp the industry, these critics suggest alternatives such as meat-free diets to fulfill our need for protein. Indeed, data suggests many young consumers are already incorporating plant-based meats into their meals.

Ultimately, though, climate change may be the most pressing issue facing the cattle industry, according to Telugu of the University of Maryland, which received a grant from the Bill and Melinda Gates Foundation to improve productivity and adaptability in African cattle. We cannot breed our way out of this, he says.

This article was originally published on Undark. Read the original article.

Image Credit: RitaE from Pixabay

View original post here:
Biotechnology Could Change the Cattle Industry. Will It Succeed? - Singularity Hub

Read More...

Boundless Bio Announces Publication in Nature Genetics Detailing the Association Between Extrachromosomal DNA-Based Oncogene Amplification and Poor…

Tuesday, August 18th, 2020

SAN DIEGO--(BUSINESS WIRE)--Boundless Bio, a company developing innovative new therapies directed to extrachromosomal DNA (ecDNA) in aggressive cancers, today announced research published in the journal Nature Genetics that demonstrates that ecDNA-based oncogene amplification drives poor outcomes for patients across many cancer types.

The manuscript, Frequent extrachromosomal oncogene amplification drives aggressive tumors, was co-authored by Boundless Bio scientists Nam-phuong Nguyen, Ph.D., and Kristen Turner, Ph.D., and scientific founders Paul Mischel, M.D., Distinguished Professor at the University of California San Diego (UC San Diego) School of Medicine and a member of the Ludwig Institute for Cancer Research; Vineet Bafna, Ph.D., Professor of Computer Science & Engineering, UC San Diego; Howard Chang, M.D., Ph.D., Virginia and D.K. Ludwig Professor of Cancer Genomics and Genetics, Stanford University; and Roel Verhaak, Ph.D., Professor and Associate Director of Computational Biology, The Jackson Laboratory.

The researchers used intensive computational analysis of whole-genome sequencing data from more than 3200 tumor samples in The Cancer Genome Atlas (TCGA) and the Pan-Cancer Analysis of Whole Genomes (PCAWG), totaling over 400 TB of raw sequencing data, to observe the impact of ecDNA amplification on patient outcomes. The researchers observed that ecDNA amplification occurs in many types of cancers, but not in normal tissue or in whole blood, and that the most common recurrent oncogene amplifications frequently arise on ecDNA. Notably, ecDNA-based circular amplicons were found in 25 of 29 cancer types analyzed, and at high frequency in many cancers that are considered to be amongst the most aggressive histological types, such as glioblastoma, sarcoma, and esophageal carcinoma. In addition, patients whose cancers carried ecDNA had significantly shorter survival, even when controlled for tissue type, than patients whose cancers were not driven by ecDNA-based oncogene amplification.

The findings demonstrate that ecDNA play a critical role in cancer, providing a mechanism for achieving and maintaining high copy number oncogene amplification and genetic heterogeneity while driving enhanced chromatin accessibility and elevating oncogene transcription. ecDNA amplifications are associated with aggressive cancer behavior, potentially by providing tumors with additional routes to circumvent current treatments and other evolutionary bottlenecks. The shorter overall survival, even when stratified by tumor type, raises the possibility that cancer patients whose tumors are driven by ecDNA may not be as responsive to current therapies and may be in need of new forms of treatment.

This important study builds on our rapidly expanding knowledge about ecDNA, showing, for the first time, that ecDNA amplifications are present in a broad range of cancer tumor types, said Jason Christiansen, Ph.D., Chief Technology Officer of Boundless Bio. These results point to the urgent need for therapies that can target ecDNA and interfere with their ability to drive aggressive cancer growth, resistance, and recurrence.

By detecting and characterizing the role that ecDNA play in driving hard-to-treat cancers, we are drawing a more accurate map of the cancer genome, said Dr. Mischel. It is our goal to take these findings and apply them to the development of powerful anti-cancer therapies for individuals with ecDNA-driven cancers.

About ecDNA

Extrachromosomal DNA, or ecDNA, are distinct circular units of DNA containing functional genes that are located outside cells chromosomes and can make many copies of themselves. ecDNA rapidly replicate within cancer cells, causing high numbers of oncogene copies, a trait that can be passed to daughter cells in asymmetric ways during cell division. Cancer cells have the ability to upregulate or downregulate oncogenes located on ecDNA to ensure survival under selective pressures, including chemotherapy, targeted therapy, immunotherapy, or radiation, making ecDNA one of cancer cells primary mechanisms of recurrence and treatment resistance. ecDNA are rarely seen in healthy cells but are found in many solid tumor cancers. They are a key driver of the most aggressive and difficult-to-treat cancers, specifically those characterized by high copy number amplification of oncogenes.

About Boundless Bio

Boundless Bio is a next-generation precision oncology company interrogating a novel area of cancer biology, extrachromosomal DNA (ecDNA), to deliver transformative therapies to patients with previously intractable cancers.

For more information, visit http://www.boundlessbio.com.

Follow us on LinkedIn and Twitter.

About Boundless Bios Spyglass Platform

Boundless Bios Spyglass platform is a comprehensive suite of proprietary ecDNA-driven and pair-matched tumor models along with proprietary imaging and molecular analytical tools that enables Boundlesss researchers to interrogate ecDNA biology to identify a pipeline of novel oncotargets essential to the function of cancer cells that are enabled by ecDNA. The Spyglass platform facilitates Boundless innovation in the development of precision therapeutics specifically targeting ecDNA-driven tumors, thereby enabling selective treatments for patients whose tumor genetic profiles make them most likely to benefit from our novel therapeutic candidates.

See the article here:
Boundless Bio Announces Publication in Nature Genetics Detailing the Association Between Extrachromosomal DNA-Based Oncogene Amplification and Poor...

Read More...

Unless true origin of coronavirus is identified, another Chinese pandemic is in the offing – WION

Tuesday, August 18th, 2020

To date, no one has stated the urgent universal need to aggressively investigate the true origin of SARS-CoV-2, the coronavirus responsible for COVID-19, better than Karl and Dan Sirotkin in their August 12, 2020 article Might SARSCoV2 Have Arisen via Serial Passage through an Animal Host or Cell Culture?

Despite claims from prominent scientists that SARSCoV2 indubitably emerged naturally, the etiology of this novel coronavirus remains a pressing and open question: Without knowing the true nature of a disease, it is impossible for clinicians to appropriately shape their care, for policymakers to correctly gauge the nature and extent of the threat, and for the public to appropriately modify their behaviour.

As the authors correctly note, serial passage, that is, the repeated re-infection within an animal or human population allows a virus to specifically adapt to the infected species.

That process occurs naturally in the wild, but it can be greatly accelerated in the laboratory by deliberate serial passaging of viruses in cell culture systems or animals, potentially leaving few or no traces as to whether the adapted viruses are naturally-occurring or laboratory-manipulated.

That type of "gain of function" experimentation can become particularly dangerous if viruses are adapted for human infection by serial passaging them through cell cultures and animal models that have been genetically-modified to express human receptors.

There are numerous scientific publications describing serial passaging of coronaviruses through humanised cell cultures and animal models, thus potentially creating a new coronavirus pre-adapted for human infection.

At present, the scientific consensus is that SARS-CoV-2 came from bats, but how it evolved to infect humans remains unknown.

China has claimed that a bat coronavirus named RaTG13 is the closest relative to SARS-CoV-2, but RaTG13 is not actually a virus because no biological samples exist. It is only a genomic sequence of a virus for which there are now serious questions about its accuracy.

In contrast, Dr Li-Meng Yan, a Chinese virologist and whistleblower, has implied that RaTG13 may have been used to divert the worlds attention away from the true source of the COVID-19 pandemic, a novel coronavirus that originated in military laboratories overseen by China's Peoples Liberation Army and created by the manipulation of Zhoushan coronaviruses ZC45 and/or ZXC21.

SARS-CoV-2 has signs of serial passaging and the direct genetic insertion of novel amino acids sequences for which no natural evolutionary pathway has been identified.

Although SARS-CoV-2 appears to have the backbone of bat coronaviruses, its spike protein, which is responsible for binding to the human cell and its membrane fusion-driven entry, has sections that do not appear in any closely-related bat coronaviruses.

SARS-CoV-2s receptor binding domain, the specific element that binds to the human cell, has a ten times greater binding affinity than the first SARS virus that caused the 2002-2003 pandemic.

Furthermore, SARS-CoV-2 appears to be pre-adapted for human infection and has not undergone a similar natural mutation process within the human population that was observed during the 2002-2003 SARS outbreak.

Those observations plus the inexplicable genetic distance between SARS-CoV-2 and any of its potential bat predecessors suggest an accelerated evolutionary process obtained by laboratory-based serial passaging through genetically-engineered mouse models containing humanised receptors previously developed by China.

The other unique feature of SARS-CoV-2 is a furin polybasic cleavage site that facilitates membrane fusion between the virus and the human cell and widely known for its ability to enhance pathogenicity and transmissibility, but also is not present in any closely related bat coronaviruses.

There are no readily-available animal models to produce a unique furin polybasic cleavage site by serial passaging, but techniques for the artificial insertion of such furin polybasic cleavage sites by genetic engineering have been used for over ten years.

To paraphrase Karl and Dan Sirotkin, unless the zoonotic hosts necessary for completing a natural jump from animals to humans are identified, the dualuse gainoffunction research practice of viral serial passage and the artificial insertion of unique viral features should be considered viable routes by which SARS-CoV-2 arose and the COVID-19 pandemic was initiated.

Lawrence Sellin, PhD is a retired US Army Reserve colonel. He has previously worked at the US Army Medical Research Institute of Infectious Diseases and conducted basic and clinical research in the pharmaceutical industry. His email address is lawrence.sellin@gmail.com.

(Disclaimer: The opinions expressed above are the personal views of the author and do not reflect the views of ZMCL.)

Go here to read the rest:
Unless true origin of coronavirus is identified, another Chinese pandemic is in the offing - WION

Read More...

SinoPharm’s Inactivated Coronavirus Vaccine | In the Pipeline – Science Magazine

Tuesday, August 18th, 2020

So now we have some clinical data on yet another category of vaccine: SinoPharms inactivated coronavirus candidate. This is one of the classic vaccine techniques, where an infectious virus is altered by some sort of protein-denaturing treatment (heating or reactive chemistry) to make it noninfectious. But such particles can retain enough of their protein surfaces to set off a useful immune response the tricky part is inactivating the virus enough so that it cant infect cells and replicate, but not so much that it presents totally different proteins to the immune system and raises a response that wont help against the real virus.

In SinoPharms case, they inactivated the coronavirus with beta-propiolactone, which is a classic protein-alkylating compound. BPL is a strained four-membered ring that is ready to be attacked and opened by pretty much any sort of nucleophile, including protein side chains from amino acids such as Cys or Lys. The compound is used for chemical disinfection (surgical instruments and the like), but thats not a casual application, because its carcinogenic by itself. It works out for such applications, though, because its very volatile (and thus easy to remove by vacuum or heating), much like another small reactive and toxi) strained ring compound, ethylene oxide. So theres no danger in using BPL to inactivate a virus the question, as mentioned, is going to be whether youve inactivated it too much.

Patients in the Phase I trial got 2.5, 5, or 10 micrograms of this agent at Day 0, Day 28, and a third time at Day 56. There were 24 patients in each group, plus an equal-sized placebo group that just got alum adjuvant injections. In the Phase II trial, the 5 microgram dose was chosen, and there were two groups: injection at Day 0 and Day 14, or injection at Day 0 and Day 21, with 84 patients in each group and a 28-patient placebo group for each. Median ages were around the early 40s, slightly more men than women. Adverse reactions appear to have been nothing remarkable pain at the injection site mostly, with very little systemic stuff like fever or fatigue, which certainly appears to be the mildest profile of the vaccines that weve seen so far.

As for neutralizing antibodies, it looks like the three-dose Phase I trial had an odd dose-response. The medium dose was actually slightly worse than either the low or high one. Meanwhile, in the Phase II, which was done with that medium five-microgram dose, the antibody response (measured two weeks after the second dose) was not as strong as with the full three-dose schedule, but the 0/21 day dosing schedule led to a better response than the 0/14 one. It appears from the Phase II data that one of the 42 patients who were tested for antibody response in that group did not seroconvert at all. The geometric mean titer values for the neutralizing antibodies (247 for the 0/21 group) appear to be in the range of other Phase I data reported, although its not easy to make a head-to-head comparison with any certainty. There is no comparison in the study with a convalescent plasma group, but as weve been seeing, those samples tend to be pretty variable themselves. There are also no data on T-cell responses.

So this is a rather preliminary report (as the authors themselves note), but its the first one we have on an inactivated vaccine. Like all of the others so far except the J&J Ad26 one, this candidate will also need a booster shot. The small and mild adverse-event reactions here are really the main thing that stands out if youre a glass half full person, then you can be glad about that, but if youre a glass-half-empty one, you might wonder about the overall robustness of the immune response. Were going to need more data to make any calls about that, and (just as with every other vaccine under development!) the real numbers were waiting on for efficacy. How many people will this (or any) vaccine protect, and how well? Stay tuned.

Read more:
SinoPharm's Inactivated Coronavirus Vaccine | In the Pipeline - Science Magazine

Read More...

Engineered COVID-19-Infected Mouse Bites Researcher Amid ‘Explosion’ of Risky Coronavirus Research – CounterPunch

Tuesday, August 18th, 2020

Photograph by Nathaniel St. Clair

University researchers genetically engineer a human pandemic virus. They inject the new virus into a laboratory mouse. The infected mouse then bites a researcher..It is a plot worthy of a Hollywood blockbuster about risky coronavirus research.

But according to newly obtained minutes of the Institutional Biosafety Committee (IBC) of the University of North Carolina (UNC), Chapel Hill, these exact events need not be imagined. They occurred for real between April 1st and May 6th this year.

The identity of the bitten coronavirus researcher has not been revealed except that they were working in a high security BSL-3 virus lab when the accident happened.

According to Richard Ebright, an epidemiologist from Rutgers University, the UNC incident underscores an important development in virus research since the pandemic began:

There has been an explosion of research involving fully infectious SARS-CoV-2 over the last six months.Research with infectious SARS-CoV-2 now is occurring in every, or almost every, BSL-3 facility in the US and overseas.

This strong upsurge is affirmed by Edward Hammond of Prickly Research, Austin, TX, former Director of the Sunshine Project, an NGO that tracked the post 9/11 expansion of the US Biodefense program.

It is evident that swarms of academic researchers with little prior experience with coronaviruses have leapt into the field in recent months.

For Hammond, this explosion represents a hazard:

We need to be clear headed about the risk. The first SARS virus was a notorious source of laboratory-acquired infections and there is a very real risk that modified forms of SARS-CoV-2 could infect researchers, especially inexperienced researchers, with unpredictable and potentially quite dangerous results. The biggest risk is the creation and accidental release of a novel form of SARS-CoV-2 a variant whose altered characteristics might undermine global efforts to stop the pandemic by evading the approaches being taken to find COVID vaccines and treatments.

And, continues Hammond: Each additional lab that experiments with CoV-2 amplifies the risk.

Richard Ebright concurs, telling Independent Science News in an email that this research is:

in many cases being performedbyresearchers who have no prior experience in BSL-3 operations and pathogens research, and who therefore pose elevated risk of laboratory accidents withBSL-3 pathogens.

Ebright is also concerned that some influential experimenters are now calling for reduced oversight:

The UNC incident also underscores that calls by some, notably Columbia University virologist Vincent Racaniello (Podcast at 01:35mins onwards), to allow virus-culture and virus-production research with fully infectious SARS-CoV-2 at BSL-2 are egregiously irresponsible and absolutely unacceptable.

Other researchers are also calling for restraint. In a paper titled Prudently conduct the engineering and synthesis of the SARS-CoV-2 virus, researchers from China and the US critiqued the synthesis in February of a full length infectious clone (Gao et al., 2020; Thao et al., 2020). And, in concluding, these researchers asked a question that is even more pertinent now than then Once the risks [of a lab escape] become a reality, who or which organization should take responsibility for them?

The accident at the University of North Carolina (UNC) is now in the public domain but only thanks to a FOIA request submitted by Hammond (in line with NIH guidelines) and shared with Independent Science News.

Despite the FOIA request, apart from the fact that UNC classified it as an official Reportable Incident, i.e. that must be reported to National Institutes of Health (NIH) in Washington DC, scarcely any information about the accident is available.

In part this is because the minutes of the relevant IBC meeting (May 6th, 2020, p109) are extremely brief. They do not provide any details of the fate of the bitten researcher. Nor do they state, for example, whether the researcher developed an active infection, nor whether they developed symptoms, nor if they transmitted the recombinant virus to anyone else. Neither do they reveal what kind of recombinant virus was being used or the purpose of the experiment.

To try to learn more, Independent Science News emailed the lab of Ralph Baric at UNC, which, based on their research history is the most likely coronavirus research group involved (Roberts et al., 2007; Menachery et al., 2015), the University Biosafety Officer, and UNC Media relations.

Only the latter replied:

The April 2020 incident referred to in the University Institutional Biosafety Committee meeting minutes involved a mouse-adapted SARS-CoV-2 strain used in the development of a mouse model system.

Ralph Baric UNC Gillings School of Public Health-web.

The researcher did not develop any symptoms and noinfection occurredas a result of the incident.

Our questions in full and the full UNC reply are available here.

The second reason for this lack of information is that the UNC redacted the names of Principal Investigators (PIs) whose research required biosafety scrutiny, along with many of the experimental specifics.

Nevertheless, unredacted parts of minutes from IBC meetings held in 2020 contain descriptions of experiments that potentially encompass the accident. They include:

Application 75223:

(a full-length infectious clone refers to a viable DNA copy of the coronavirus, which is ordinarily an RNA virus)

and

Application 73790:

and

Application 74962:

In all, any one of eight sets of different experiments approved by the UNC Chapel Hill IBC in 2020 proposed infecting mice with live infectious and mutant SARS-CoV-2-like coronaviruses under BSL-3 conditions and therefore could have led to the accident.

According to Hammond the lack of transparency represented by the sparse minutes and especially the redactions represent a violation of sciences social contract:

At the dawn of recombinant DNA, at the request of the scientific community itself, following the fabled Asilomar conference, the United States government took the position of not regulating genetic engineering in the lab. The deal that big science struck with the government was that, in return for not being directly regulated, principal investigators would take personal responsibility for lab biosafety, involve the public in decision-making, and accept public accountability for their actions.

The NIH Guidelines and Institutional Biosafety Committee system of self-regulation by researchers is founded upon the principal of personal responsibility of PIs and the promise of transparency. The redaction of the researchers identities from IBC meeting minutes, in order to hide the activities of researchers and avoid accountability for accidents, fundamentally contradicts the core principles of the US oversight system and violates the commitments that science made.

Richard Ebright goes further:

There is no justification for UNCs redactionof the names of the laboratory heads andthe identities of pathogens. UNCs redactions violate conditions UNC agreed to in exchange for NIH funding of UNCs research and, if not corrected, should result in the termination of current NIH funding, and the loss of eligibility for future NIH funding, of UNCs research.

Are universities doing too many risky experiments on coronaviruses?

The second concern of researchers contacted by Independent Science News is that unnecessary and dangerous experiments will be conducted as a result of the COVID-19 pandemic. According to Richard Ebright:

The UNC incident shows that serious laboratory accidents with SARS-CoV-2can occur even in a lab having extremely extensive experience in BSL-3 operations and unmatched expertise in coronavirus research, and underscores the risks associated with uncontrolled proliferation of research on SARS-CoV-2, especially for labs lacking prior experience in BSL-3 operations and coronavirus research.

For this reason Ebright argues that:

It is essential that a national needs-assessment and biosafety assessment be performed for research involving fully infectious SARS-CoV-2. It also is essential that a risk-benefit review be performed before approving research projects involving fully infectious SARS-CoV-2something that currently does not occurto ensure that potential benefits to the public outweigh the real risks to laboratory workers and the public.

This concern over risks and benefits is shared by Edward Hammond. Using FOIA again he has further discovered that researchers at the University of Pittsburgh (whose identity is redacted) plan to make what Hammond calls Corona-thrax.

In short, according to its Institutional Biosafety Committee, Pittsburgh researchers intend put the spike protein of SARS-CoV-2 (which allows the virus to gain entry into human cells) into Bacillus anthracis which is the causative agent of anthrax.

The anthrax strain proposed to be used for this experiment is disarmed but, Hammond agrees with Gao et al., (2020) that the balance of risks and benefits appears not to be receiving adequate consideration.

This experiment was nevertheless approved by the Institutional Biosafety Committee of the University of Pittsburgh. But by redacting the name of the laboratory from the minutes and also every name of the members of the committee which approved it, the University has supplied a de facto response to the final question posed by Gao et al.: who will take responsibility for risky coronavirus research?

References

Gao, P., Ma, S., Lu, D., Mitcham, C., Jing, Y., & Wang, G. (2020). Prudently conduct the engineering and synthesis of the SARS-CoV-2 virus.Synthetic and systems biotechnology,5(2), 59-61.Menachery, V. D., Yount, B. L., Debbink, K., Agnihothram, S., Gralinski, L. E., Plante, J. A., & Randell, S. H. (2015). A SARS-like cluster of circulating bat coronaviruses shows potential for human emergence.Nature medicine,21(12), 1508-1513.Roberts, A., Deming, D., Paddock, C. D., Cheng, A., Yount, B., Vogel, L., & Zaki, S. R. (2007). A mouse-adapted SARS-coronavirus causes disease and mortality in BALB/c mice.PLoS Pathog,3(1), e5.Thao, T. T. N., Labroussaa, F., Ebert, N., Vkovski, P., Stalder, H., Portmann, J., & Gultom, M. (2020). Rapid reconstruction of SARS-CoV-2 using a synthetic genomics platform.BioRxiv.

This article first appeared in Independent Science News.

Read more here:
Engineered COVID-19-Infected Mouse Bites Researcher Amid 'Explosion' of Risky Coronavirus Research - CounterPunch

Read More...

Protein Expression Technology Market is anticipated to record a CAGR of around 10% over the forecast period ie 2019-2027 – Scientect

Tuesday, August 18th, 2020

A new report published by Research nester, titledProtein Expression Technology Market: Global Demand Analysis & Opportunity Outlook 2027delivers detailed overview of the global protein expression technology marketin terms of market segmentation by expression system, by application, by end user and by region.

Based on expression systems, the global protein expression technology market is segmented into prokaryotic, insect, mammalian, yeast and others; by application into drug discovery, protein purification, protein-protein interaction, disease diagnostics & monitoring and others; by end user into pharmaceutical companies, biotechnology companies, academic research and others.

The global protein expression technology market is anticipated to record a CAGR of around 10% over the forecast period i.e. 2019-2027.

Protein expression is among the most fundamental biological processes. It refers to processes which describe how living cells or organisms synthesize, modify and regulate proteins. Expression of a protein includes three processes, namely, transcription, translation and post-translational modification. These processes play a vital role in the expression of a gene and its regulation. Increasing investment for R & D development by pharmaceutical and biotechnology companies along with rising demand for therapeutic proteins for the treatment of various ailments is anticipated to propel the growth of the market over the forecast period. Additionally, one major advantage of protein and gene expression is the ability to detect both hereditary and environmental abnormalities of a given disease, if linked to disease presentation. This is also projected to positively impact the growth of the market.

Furthermore, application of soluble receptors, monoclonal antibodies, engineered proteins, peptides, and their conjugates as drugs is expected to drive the growth of the market.

The Final Report will cover the impact analysis of COVID-19 on this industry (Global and Regional Market).

Request for a sample of this research report @https://www.researchnester.com/sample-request-679

On the basis of region, the global protein expression technology market is segmented into five major regions including North America, Europe, Asia Pacific, Latin America and Middle East & Africa region. The market in North America is anticipated to witness a significant growth during the forecast period on the back of technological advancements for the manufacturing and research of personalized medicine with the help of proteomics. Moreover, funding provided by various governmental and non-governmental organizations for research in this region is also expected to foster the growth of market. The market in Asia Pacific is also estimated to show a considerable growth over the forecast period owing to the increased prevalence of chronic diseases due to sedentary lifestyle of people.

The Final Report will cover the impact analysis of COVID-19 on this industry (Global and Regional Market).

Download Sample Report Here:https://www.researchnester.com/sample-request-679

Application of Therapeutic Protein in Medicine

Most of the human diseases are in some way related to dysfunction of a specific protein. Therapeutic protein produced by the protein expression technology provides treatment for a variety of diseases, such as diabetes, cancer, hemophilia, infectious diseases, and anemia. These proteins formed by recombinant technology and genetic engineering have wide array of applications in medicine. A few common therapeutic proteins are FC fusion proteins, hormones, interleukins, enzymes, anticoagulants, and others. Thus, the application of these therapeutic protein in medicine for the remedy of the numerous diseases is anticipated to propel the growth of the market.

However, high cost involved with the research of expression proteins along with stringent regulation and policies for their approval as drugs are expected to operate as a key restraint to the growth of the global protein expression technology market over the forecast period.

Further, for the in-depth analysis, the report encompasses the industry growth drivers, restraints, supply and demand risk, market attractiveness, BPS analysis and Porters five force model.

This reportalso provides the existing competitive scenario of some of the key players of the global protein expression technology market which includes company profiling of Thermo Fisher Scientific Inc., Merck KGaA, Promega Corporation, Agilent Technologies, GenScript, Takara Holdings Inc., Bio-Rad Laboratories Inc., Qiagen, Lonza and other prominent players. The profiling enfolds key information of the companies which encompasses business overview, products and services, key financials and recent news and developments. On the whole, the report depicts detailed overview of the global protein expression technology marketthat will help industry consultants, equipment manufacturers, existing players searching for expansion opportunities, new players searching possibilities and other stakeholders to align their market centric strategies according to the ongoing and expected trends in the future.

The Final Report will cover the impact analysis of COVID-19 on this industry (Global and Regional Market).

Request Report [emailprotected]https://www.researchnester.com/sample-request-679

About Research Nester

Research Nester is a leading service provider for strategic market research and consulting. We aim to provide unbiased, unparalleled market insights and industry analysis to help industries, conglomerates and executives to take wise decisions for their future marketing strategy, expansion and investment etc. We believe every business can expand to its new horizon, provided a right guidance at a right time is available through strategic minds. Our out of box thinking helps our clients to take wise decision so as to avoid future uncertainties.

For More Update Follow-LinkedIn|Twitter|Xing

Contact for more Info:

AJ Daniel

Email:[emailprotected]

U.S. Phone: +1 646 586 9123

U.K. Phone: +44 203 608 5919

Read More Related Reports:

Metal Packaging MarketConveyor Ovens MarketCommercial Aircraft Wing MarketAutomotive Carbon Wheels MarketDefense Fuel Cell MarketGyro Compass MarketTranscatheter Aortic Valve Replacement System MarketExpanded Perlite MarketShoe Packaging MarketSolar Equipment MarketSmart Switches MarketMicroscope digital cameras MarketAutomotive Door Stabilizer MarketSport Cap And Closure MarketDemulsifier Market

Original post:
Protein Expression Technology Market is anticipated to record a CAGR of around 10% over the forecast period ie 2019-2027 - Scientect

Read More...

Make way for sustainable, healthy and delicious food! – Innovation Origins

Tuesday, August 18th, 2020

Vitamin B12 is only found in animal proteins, i.e. in meat and dairy. This is why vegans often take supplements in order to get the amount they need. A sustainable diet implies eating less animal protein, as the Netherlands Nutrition Centre (Stichting Voedingscentrum) also recommends. But how can we make sure that our sustainable food is both healthy and delicious?

According to Corn van Dooren, nutritionist and expert on sustainable eating at the Netherlands Nutrition Centre, this is the key question for the future. A liter of cola has less environmental impact than a liter of milk. Of course, that doesnt mean that you are better off drinking cola because it doesnt provide any nutrients. In Van Doorens opinion, you have to look at sustainable food in a variety of ways: You cant do without food, but you can do without flying. Get your food close to home and in season.

In 2018, Van Dooren earned his PhD on the topic of how sustainable (as in, environmental impact) and healthy (as in, nutritional quality) existing dietary patterns are. He devised the Sustainable Nutrient-Rich Food index, a mathematical model that offers nutritional advice. He divided products into four categories: red, white, brown, and green. Van Dooren: The green category comprising vegetables, mushrooms, legumes, soy products, and fruit, for example, scores the highest on both aspects sustainability and healthy nutritional value. The model also shows, in addition to the composition of a product, where it was grown and how its sustainability is assessed, Van Dooren explains.

The Wheel of Five is a guideline for healthy food in the Netherlands, but it does not yet include a sustainable index. The last update of the wheel was made in 2016. It stated: eat more vegetables (from 200 grams to 250 grams per day), less meat (maximum 500 grams per week), and eat more plant-based foods, such as a weekly portion of legumes and a handful of nuts every day. Those 500 grams could be discussed from a sustainability perspective, because in that case, perhaps meat once or twice per week would then be the most optimal option.

If you eat the maximum amount of 500 grams of protein per week, then the ratio of animal and vegetable protein is 50/50. If you eat almost no meat as part of the Wheel of Five, but you do eat dairy, egg, and fish, then you end up with a ratio of 40/60. On average, Dutch people now get 61% of their protein from animal sources and 39% from plant-based sources. According to the Dutch Council for the Environment and Infrastructure, RLi), this figure needs to be reversed by 2030. This means that the percentage of animal protein in our diet must then be reduced to 40% of our total protein consumption.

In that whole protein transition, as we like to call the offer of alternatives to meat, there was Finnish research into vitamin B12 that can, of course, help people take that step more easily. In any case, Van Dooren regards fermentation, which the Finnish researchers used, as an interesting and exciting innovative development: We should take a closer look into areas around small organisms, such as bacteria, fungi, yeasts, and algae, and see what can be gained from that.

Fermentation is by no means new. It is mainly known for ensuring that vegetables, fruit, fish, and dairy products have a longer shelf life. It is a natural process, also known as controlled fermentation, which can also provide new flavors and products that have plenty of nutritional value. For instance, microbiologist Professor. Dr. Eddy Smit at Wageningen University has cultured tempeh from fermented lupine beans. Apparently, there is as much vitamin B12 in that as there is in a piece of steak.

The greatest voyage of discovery to come lies in the combination of common nutrients with existing natural products, Van Dooren believes: What we know is that legumes and bacteria work together in removing nitrogen from the air. As a consequence, you would no longer need artificial fertilizer. It would be extremely interesting if you were able to combine those kinds of interactions with other plants.

That you can use that particular characteristic of legumes in other crops is also very exciting news for Van Dooren. You end up with a technology that is very far-reaching, albeit not universally accepted. It will have to be done using genetic engineering, which is something that a proportion of consumers will not accept.

Of course, the flavor is also important when it comes to gaining acceptance for meat and dairy substitutes, Van Dooren adds. Soy, for example, is the closest thing to dairy in terms of nutritional value, but it tastes completely different. The German company Made With Luve makes dairy substitutes using lupine. This is a crop that can grow very well in northwestern Europe, which is also a major advantage. Soy is difficult to grow here.

Step by step, sustainable and healthy food is coming ever-closer. Van Dooren considers that true change will still take some time: Now the main emphasis of current innovations is to ensure that meat substitutes are as similar in taste and texture to meat as possible. That they produce the same sensation as meat. The real innovation will be, of course, when a new generation comes along that spurs us on to forget about meat altogether. That is making products that are so special and which have a totally new flavor sensation that transcends the taste of meat.

The rest is here:
Make way for sustainable, healthy and delicious food! - Innovation Origins

Read More...

Novome Biotechnologies Expands Therapeutic Focus and Platform Capabilities with Acquisition of Preclinical Projects and Intellectual Property from…

Wednesday, August 12th, 2020

Transfer of Caribous novel microbial IP will enable new therapeutic opportunities for Novome

License to CRISPR-Cas9 intellectual property controlled by Caribou will accelerate the development of preclinical candidates built on Novomes proprietary synthetic biology platform

SOUTH SAN FRANCISCO, Calif., Aug. 12, 2020 (GLOBE NEWSWIRE) -- Novome Biotechnologies, Inc., a biotechnology company engineering first-in-class, living medicines for chronic diseases, today announced that it has taken assignment to certain microbial intellectual property, and has non-exclusively licensed foundational CRISPR-Cas9 intellectual property controlled by Caribou Biosciences, a leading CRISPR genome editing company, to expand its therapeutic pipeline and platform capabilities.

This is an important milestone for Novome that should unlock new therapeutic avenues while we accelerate the pace of preclinical development at the company. The ability to leverage the efficient and flexible CRISPR-Cas9 system will allow us to rapidly iterate on GEMM strain designs to generate the most promising therapeutic candidates, said Blake Wise, Chief Executive Officer of Novome. Additionally, we are excited to leverage the progress made by Caribous microbial group and advance this promising science.

Under the terms of an assignment agreement, Novome acquired ownership of certain intellectual property and preclinical projects related to undisclosed therapeutic areas. Additionally, pursuant to a license agreement, Novome received a non-exclusive license to foundational CRISPR-Cas9 intellectual property controlled by Caribou for genetic modification of bacterial species for administration as therapeutics in humans. Terms of the agreements have not been disclosed. Novome will have full control of development, manufacturing, and commercialization of any product candidates covered by either the assignment agreement or the license agreement.

Novome developed the first platform for controlled and robust colonization of the human gut with engineered therapeutic bacteria, its Genetically Engineered Microbial Medicines (GEMMs) platform. The Company is focused on advancing its lead hyperoxaluria program through Phase 1 clinical proof-of-concept work and expanding its platform and pipeline to address additional disease indications.

Novome was founded in 2016 by scientists from Stanford University and the University of California, Berkeley, based on research performed in the laboratory of Scientific Co-founder Dr. Justin Sonnenburg, Associate Professor, Stanford University. The founding team, Drs. Will DeLoache, Weston Whitaker, Zachary Russ, and Liz Shepherd, combines deep expertise in synthetic biology and the study of the gut microbiota. Their work has led to numerous peer-reviewed scientific publications, as well as the filing of a portfolio of patents, both developed at Novome and licensed exclusively from Stanford.

About Genetically Engineered Microbial MedicinesGenetically Engineered Microbial Medicines (GEMMs) are proprietary bacterial strains designed to colonize the gut at a controllable abundance and express therapeutic transgenes at clinically meaningful levels. Colonization is maintained using a daily dose of prebiotic polysaccharide that GEMMs are engineered to depend upon for their survival.

About NovomeNovome Biotechnologies, Inc. is a biotechnology company focused on engineering defined activities into the human gut microbiota to treat chronic diseases. The Company has developed the first-ever platform for controlled colonization of the gut with engineered bacteria, enabling first-in-class living therapeutics: Genetically Engineered Microbial Medicines (GEMMs). Novome is utilizing its proprietary GEMMs platform in its lead preclinical program in hyperoxaluria, which is focused on the development of a therapeutic strain of bacteria that degrades oxalate to prevent the formation of kidney stones. Efforts are also directed to the expansion of its proprietary synthetic biology platform into additional indications.

Source: Novome Biotechnologies, Inc.

Go here to see the original:
Novome Biotechnologies Expands Therapeutic Focus and Platform Capabilities with Acquisition of Preclinical Projects and Intellectual Property from...

Read More...

Scientists Gene-Hack Cotton Plants to Make Them Every Color of the Rainbow – Singularity Hub

Wednesday, August 12th, 2020

Imagine this: Youre on a drive through cotton country. The suns out, tops down. Its a beautiful, totally normal day. Only, what was once a sea of white puff balls has transformed into a multi-hued swirl. Lines of deep purple, bright yellow, midnight blue. All the colors in the rainbowand your t-shirt drawer, as it so happens.

Today, youd do well to check your water. But in the future, colorful cotton could be the normAustralian scientists are having early success genetically modifying the crop to make it multicolored. And although color is their latest project, theyre also working to make synthetic-like, stretchy cotton.

The team hopes their new cotton plants might eventually be grown widely and made into clothes, helping to displace the toxic dyes and synthetic materials used by the fashion industry.

And thats a worthy cause.

The numbers are hard to pin down exactly, but theres little doubt that how we make clothes could be more environmentally friendly. In textile manufacturing, which often takes place in developing countries, those harmful dyes can cause health problems for workers and do more damage as toxic runoff. Also, the life cycle of clothing isnt as long as it once was. While some items will get a second life by way of a thrift store, it all eventually makes its way to a landfill, where the synthetic materials in many clothes can take centuries to break down.

The holy grail then? Non-toxic, compostable clothing. And while its still early, the tools of synthetic biology and genetic engineering may well prove a big part of the solution.

To be totally clear, cotton doesnt only grow in one color. (This was news to me, so maybe it is to you too.) Some varieties, dating back millennia, are naturally dark chocolate, light brown, and even mauve. These were traditionally used in handwoven textiles, but with the Industrial Revolution, naturally pigmented cotton gave way to white cotton because it had longer, higher quality fibers, you could dye it any color on the cheap, and it didnt require specialized equipment or methods to harvest.

In the 80s and 90s, as people became more environmentally conscious, there was a revival of naturally pigmented cotton. You could make clothes from it without dye and suppliers were often small, organic farmers. There was even work to make it amenable to industrial looms. Sally Fox, for example, developed varieties with longer fibers in an array of colors.

Still, naturally colored cotton is generally more expensive to produce, the color range is limited, and the fiber quality is lower than white cotton.

Enter genetic engineering.

As far back as 1993, people were talking about adding color genes to cotton. Two biotech companies, Agracetus and Calgene, had plans to splice in genes from the indigo plant to make cotton for blue jeans. Of course it will work, Ken Barton, vice-president of research and development at Agracetus, said at the time. Give a scientist enough time and money and he can do anything. Of course, were still dying jeans 27 years laterbut maybe the time has come.

As with all things in the realm of biology, the devils in the details, but our tools for manipulating nature have advanced in the last few decades too.

An array of tiny, brilliantly colored buds of cotton tissue are sitting in a few dozen petri dishes in a Canberra greenhouse (check out images here). In one dish, the cotton is raspberry red; in the other its yellow like a mango. The tissue, which carries genes for color spliced in by scientists at Australias scientific research agency, CSIRO, is only the first step, but its a promising one. In the next few months, the team, led by senior research scientist, Colleen MacMillan, will coax the tissue into full-grown cotton plants.

If all goes to plan, the cotton fiber will be just as colorful as the petri dish tissue. The team points to splotches of color on leaves of tobacco plants carrying the cotton genes as likely evidence theyll take. If the leaves of the cotton plants are similarly colored, the cotton fiber will be too.

Weve seen some really beautiful bright yellows, sort of golden-orangey colors, through to some really deep purple, Filomena Pettolino, a scientist on MacMillans team, told Australias ABC News. The team is also working on black cotton, which would be a significant achievementblack dyes are notoriously the nastiest, most toxic of the lot. And the less dye the better.

Though theyre favored for speed and quality, synthetic dyes can include formaldehyde and heavy metals which stain the skin and cause cancer. That early-90s dream to make jeans with blue cotton? Its just as relevant today. In the Chinese province of Xintang, where 300 million pairs of jeans are dyed each year, the toxic runoff flows into rivers by the gallon.

In parallel to their work in multicolored cotton, the team has a longer-term project to make synthetic-like cotton. Synthetics like polyester and nylon make their way into the environment from washing machineswhich pull off and flush microfibers from the fabricand of course, they also line landfills. The team is screening thousands of plants, hunting for proteins with just the right properties: stretchy, wrinkle-free, and maybe even waterproof.

Were looking into the structure of cotton cell walls and harnessing the latest tools in synthetic biology to develop the next generation cotton fiber, CSIRO scientist Dr Madeline Mitchell said. Weve got a whole bunch of different cotton plants growing; some with really long thin fibers, others like the one we call Shaun the Sheep, with short, woolly fibers.

It remains to be seen whether this next-gen cotton can keep up with fashions insatiable demand for new huesthough black is never out of styleif it can yield as much as a standard cotton plant, and what it will cost farmers.

First, though, this team (or another) will need to prove they can grow the stuff and produce seeds at scale. But if it works, you or someone you know may one day rock a pair of fully compostable, bright purple yoga pants of gene-hacked cotton.

Image credit: Crystal de Passill-Chabot / Unsplash

See original here:
Scientists Gene-Hack Cotton Plants to Make Them Every Color of the Rainbow - Singularity Hub

Read More...

Baseceuticals Offers Oncolytic Virus Services to Accelerate the Development in Gene Therapy – Press Release – Digital Journal

Wednesday, August 12th, 2020

Baseceuticals recently announced its release of the oncolytic virus service aiming to accelerate the development of gene therapy.

New York, USA - August 12, 2020 - Baseceuticals, the major division of Creative Biogene, mainly targets on gene therapy to provide various services for researchers and institutes to develop new drugs, including genetic modification, gene delivery systems and preclinical trials, recently announced its release of the oncolytic virus service aiming to accelerate the development of gene therapy.

As a global leader in the field of gene therapy, Baseceuticals provides high-quality oncolytic virus services based on an excellent and mature platform. Relying on the most advanced technology and the most advanced equipment, the experienced technical team has successfully completed many oncolytic virus projects, including oncolytic virus construction, engineering and verification. After communicating and analyzing the specific situation, Baseceuticals can propose the best strategy for the project to meet specific needs.

Oncolytic viruses have been used for treatment in clinical trials, and an oncolytic virus product T-VEC has been approved by the FDA. Oncolytic virus therapy has been recognized as a promising and effective cancer treatment method. Compared with radiotherapy and chemotherapy, it is easier to destroy tumor cells.

Oncolytic viruses (OV) are a group of tumor-killing viruses with replication ability that can effectively multiply in cancer cells without damaging healthy cells. According to development, oncolytic viruses can be divided into two categories, namely natural viruses and genetically modified viruses. Among them, natural viruses include broad and natural variants of weak viruses. The main advantage of oncolytic viruses is that they can quickly produce virus particles and genetically engineer other genes to enhance anti-tumor immunity, increase tumor cell sensitivity and improve patient safety. Oncolytic virus services provided by Baseceuticals include oncolytic virus construction, oncolytic virus engineering, oncolytic virus verification, and development of disease-specific oncolytic virus therapy.

Highlights of Oncolytic Virus Service in Baseceuticals:

1. Years of rich experience in oncolytic virus services2. Leading equipment and first-class technology3. Fast turnaround time and reliable results4. A variety of oncolytic viruses are available5. Reasonable price and quality service6. Customize services to meet specific requirements through feasible suggestions

"Baseceuticals provides high-quality oncolytic virus services, our technical research team specializes in efficient systems and procedures in projects related to oncolytic viruses," said Marcia Brady, she also claimed, "Our oncolytic virus service starts with free communication and then feasible suggestions to meet your specific needs. We are confident to provide the best oncolytic virus service at an affordable price and reliable results."

About Baseceuticals

With years of experience and advanced technologies, Baseceuticals provides worldwide customers with innovative products and services to greatly enhance the clinical application and drug launches. As a division of Creative Biogene, Baseceuticals has become a well-recognized industry leader to support scientists from research institutes, government, pharmaceutical companies, diagnosis industries and testing laboratories.

Media ContactCompany Name: Creative BiogeneContact Person: Marcia BradyEmail: Send EmailPhone: 1-631-619-7922Country: United StatesWebsite: https://baseceuticals.creative-biogene.com

Continue reading here:
Baseceuticals Offers Oncolytic Virus Services to Accelerate the Development in Gene Therapy - Press Release - Digital Journal

Read More...

Page 10«..9101112..2030..»


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