Category Archives: Embryonic Stem Cells

Cell Freezing Media for Cell Therapy Market with Covid-19 Pandemic Analysis, Growth Rate, New Trend Analysis & Forecast To 2026 | Key Players:…

Global Cell Freezing Media for Cell Therapy Market Research Report 2020 to 2026 is segmented by product type, applications and enlists important features such as recent trends, Cell Freezing Media for Cell Therapy statistics, and growth factors to assist the users in planning the business strategies for setting up their business with huge market returns.

The study is also compiled on the basis of the latest and upcoming innovations, opportunities, and trends. In addition to SWOT analysis, the report also documents a detailed market analysis outlining every major player in the process. Based on the study, Global Acquire Market Research estimates that the market is likely to exhibit a steady CAGR growth.

Covid-19 pandemic affects most industries in the globe. Here at acquire market research we offer you comprehensive data of related industry which will help and support your business in all possible ways.

Expanding Operations In The Future? To Get The Perfect Launch Ask For A Free Custom Sample Report: https://www.acquiremarketresearch.com/sample-request/379423/

Major Market Players Covered In This Report: BioLife Solutions, Akron Biotechnology, GE Healthcare, Thermo Fisher Scientific, Biological Industries, Merck, WAK-Chemie Medical, Zenoaq

The key product type of Cell Freezing Media for Cell Therapy market are: With FBS, Without FBS

Cell Freezing Media for Cell Therapy Market Outlook by Applications: Human Embryonic Stem Cells, CAR-T Cell Therapy, Neural Stem Cell Therapy, Mesenchymal Stem Cell Therapy, Hematopoietic Stem Cell Transplantation, Other

Cell Freezing Media for Cell Therapy Market The ever increasing demand for the Cell Freezing Media for Cell Therapy and various business opportunities have boosted the growth of the Cell Freezing Media for Cell Therapy market According to the global Cell Freezing Media for Cell Therapy report, it is expected to strengthen its position in the near future. The report compiles several potential propositions related to Cell Freezing Media for Cell Therapys such as contribution, active and new entrants focusing on the Cell Freezing Media for Cell Therapy product, its specifications, and classification. Furthermore, the report represents sales margins and the competitive landscape of the industry.

Get Reasonable Discount on this Premium Report Here: https://www.acquiremarketresearch.com/discount-request/379423/

Based on regions, the market is classified into North America, Europe, Asia- Pacific, Middle East & Africa, and Latin America. The study is expected to provide detailed qualitative and quantitative information on the above-mentioned segments for every region and country covered under the scope of the study.

The Middle East and Africa (GCC Countries and Egypt) North America (the United States, Mexico, and Canada) South America (Brazil etc.) Europe (Turkey, Germany, Russia UK, Italy, France, etc.) Asia-Pacific (Vietnam, China, Malaysia, Japan, Philippines, Korea, Thailand, India, Indonesia, and Australia)

As a part of our Corporate Social Responsibility, we would like to announce that we would be contributing 15 % of our profits to USA, UK, Italy, Spain and India relief fund.

Pointers Covered Through This Global Cell Freezing Media for Cell Therapy Market Research Report: 1] Analysis of drivers, restraints, and opportunities 2] Discussion on sales patterns and methodologies 3] Profiling of leading key players across the globe 4] Detailed analysis of demand-supply chaining 5] Well explained SWOT and Porters Five technique 6] Analysis of key regions 7] Elaboration on the global competitive landscape

From the Cell Freezing Media for Cell Therapy market research reports, the following points are available with detailed study at every point:

Production Analysis Initiation of this Cell Freezing Media for Cell Therapy is analyzed based on top countries, types, and applications. Here, the report is expected to cover the price analysis of varied Cell Freezing Media for Cell Therapy market key players.

Profit and Sales Evaluation Both earnings and sales are verified for various components of this international Cell Freezing Media for Cell Therapy market. The report focuses on the price that plays a vital role in sales development in several regions.

Segments and Advantages In continuation of using earnings, this report studies the design and ingestion of its Cell Freezing Media for Cell Therapy market. This report also highlights the difference between usage and supply, export, and import data.

Competition In this section, many global Cell Freezing Media for Cell Therapy industry-top players have been enlisted based on their company profile, product portfolio, ability, price, cost, and revenue.

Other Analysis In addition to the aforementioned information, demand, and supply scrutiny to the Cell Freezing Media for Cell Therapy economy, contact information from leading producers, suppliers, and major consumers can also be sourced from the report.

Directly Purchase this research study here: https://www.acquiremarketresearch.com/buy-now/379423/?price=su

Why Buy This Report? The research report provides a complete analysis of the global Cell Freezing Media for Cell Therapy market to help players create powerful growth strategies and consolidate their position in the industry. The report presents a complete mapping of the market participants and the competitive landscape. Information on important sustainability strategies adopted by key companies along with their impact market growth and competition has been furnished in this report. All players can use the report to prepare themselves to face impending market challenges and compete in the global market.

About us:

Acquire Market Research is a market research-based company empowering companies with data-driven insights. We provide Market Research Reports with accurate and well-informed data, Real-Time with Real Application. A good research methodology proves to be powerful and simplified information that applied right from day-to-day lives to complex decisions helps us navigate through with vision, purpose, and well-armed strategies. At Acquire Market Research, we constantly strive for innovation in the techniques and the quality of analysis that goes into our reports.

Contact Us:

Sally Mach 555 Madison Avenue, 5th Floor, Manhattan, New York, 10022 USA Phone No: +1 (800) 663-5579 Email ID: [emailprotected]

More:
Cell Freezing Media for Cell Therapy Market with Covid-19 Pandemic Analysis, Growth Rate, New Trend Analysis & Forecast To 2026 | Key Players:...

Radiation Response of Murine Embryonic Stem Cells

To understand the mechanisms of disturbed differentiation and development by radiation, murine CGR8 embryonic stem cells (mESCs) were exposed to ionizing radiation and differentiated by forming embryoid bodies (EBs). The colony forming ability test was applied for survival and the MTT test for viability determination after X-irradiation. Cell cycle progression was determined by flow cytometry of propidium iodide-stained cells, and DNA double strand break (DSB) induction and repair by H2AX immunofluorescence. The radiosensitivity of mESCs was slightly higher compared to the murine osteoblast cell line OCT-1. The viability 72 h after X-irradiation decreased dose-dependently and was higher in the presence of leukemia inhibitory factor (LIF). Cells exposed to 2 or 7 Gy underwent a transient G2 arrest. X-irradiation induced H2AX foci and they disappeared within 72 h. After 72 h of X-ray exposure, RNA was isolated and analyzed using genome-wide microarrays. The gene expression analysis revealed amongst others a regulation of developmental genes (Ada, Baz1a, Calcoco2, Htra1, Nefh, S100a6 and Rassf6), downregulation of genes involved in glycolysis and pyruvate metabolism whereas upregulation of genes related to the p53 signaling pathway. X-irradiated mESCs formed EBs and differentiated toward cardiomyocytes but their beating frequencies were lower compared to EBs from unirradiated cells. These results suggest that X-irradiation of mESCs deregulate genes related to the developmental process. The most significant biological processes found to be altered by X-irradiation in mESCs were the development of cardiovascular, nervous, circulatory and renal system. These results may explain the X-irradiation induced-embryonic lethality and malformations observed in animal studies.

See the rest here:
Radiation Response of Murine Embryonic Stem Cells

Human Embryonic Stem Cells (HESC) Market Growth By Manufacturers, Type And Application, Forecast To 2026 – 3rd Watch News

Others

To get Incredible Discounts on this Premium Report, Click Here @ https://www.marketresearchintellect.com/ask-for-discount/?rid=218035&utm_source=3WN&utm_medium=888

The Human Embryonic Stem Cells (HESC) market report provides successfully marked contemplated policy changes, favorable circumstances, industry news, developments, and trends. This information can help readers fortify their market position. It packs various parts of information gathered from secondary sources, including press releases, web, magazines, and journals as numbers, tables, pie-charts, and graphs. The information is verified and validated through primary interviews and questionnaires. The data on growth and trends focuses on new technologies, market capacities, raw materials, CAPEX cycle, and the dynamic structure of the Human Embryonic Stem Cells (HESC) market.

This study analyzes the growth of Human Embryonic Stem Cells (HESC) based on the present, past and futuristic data and will render complete information about the Human Embryonic Stem Cells (HESC) industry to the market-leading industry players that will guide the direction of the Human Embryonic Stem Cells (HESC) market through the forecast period. All of these players are analyzed in detail so as to get details concerning their recent announcements and partnerships, product/services, and investment strategies, among others.

Sales Forecast:

The report contains historical revenue and volume that backing information about the market capacity, and it helps to evaluate conjecture numbers for key areas in the Human Embryonic Stem Cells (HESC) market. Additionally, it includes a share of each segment of the Human Embryonic Stem Cells (HESC) market, giving methodical information about types and applications of the market.

Reasons for Buying Human Embryonic Stem Cells (HESC) Market Report

This report gives a forward-looking prospect of various factors driving or restraining market growth.

It renders an in-depth analysis for changing competitive dynamics.

It presents a detailed analysis of changing competition dynamics and puts you ahead of competitors.

It gives a six-year forecast evaluated on the basis of how the market is predicted to grow.

It assists in making informed business decisions by performing a pin-point analysis of market segments and by having complete insights of the Human Embryonic Stem Cells (HESC) market.

This report helps the readers understand key product segments and their future.

Have Any Query? Ask Our Expert @ https://www.marketresearchintellect.com/need-customization/?rid=218035&utm_source=3WN&utm_medium=888

In the end, the Human Embryonic Stem Cells (HESC) market is analyzed for revenue, sales, price, and gross margin. These points are examined for companies, types, applications, and regions.

To summarize, the global Human Embryonic Stem Cells (HESC) market report studies the contemporary market to forecast the growth prospects, challenges, opportunities, risks, threats, and the trends observed in the market that can either propel or curtail the growth rate of the industry. The market factors impacting the global sector also include provincial trade policies, international trade disputes, entry barriers, and other regulatory restrictions.

About Us:

Market Research Intellect provides syndicated and customized research reports to clients from various industries and organizations with the aim of delivering functional expertise. We provide reports for all industries including Energy, Technology, Manufacturing and Construction, Chemicals and Materials, Food and Beverage, and more. These reports deliver an in-depth study of the market with industry analysis, the market value for regions and countries, and trends that are pertinent to the industry.

Contact Us:

Mr. Steven Fernandes

Market Research Intellect

New Jersey ( USA )

Tel: +1-650-781-4080

Our Trending Reports

Intelligent Network Market Size, Growth Analysis, Opportunities, Business Outlook and Forecast to 2026

Medical Device Connectors Market Size, Growth Analysis, Opportunities, Business Outlook and Forecast to 2026

Plant Based Protein Market Size, Growth Analysis, Opportunities, Business Outlook and Forecast to 2026

Property Management Market Size, Growth Analysis, Opportunities, Business Outlook and Forecast to 2026

Road Compactor Market Size, Growth Analysis, Opportunities, Business Outlook and Forecast to 2026

Visit link:
Human Embryonic Stem Cells (HESC) Market Growth By Manufacturers, Type And Application, Forecast To 2026 - 3rd Watch News

Cell Therapy Market Analysis Of Global Trends, Demand And Competition 2020-2028 – Jewish Life News

Trusted Business Insights answers what are the scenarios for growth and recovery and whether there will be any lasting structural impact from the unfolding crisis for the Cell Therapy market.

Trusted Business Insights presents an updated and Latest Study on Cell Therapy Market 2019-2026. The report contains market predictions related to market size, revenue, production, CAGR, Consumption, gross margin, price, and other substantial factors. While emphasizing the key driving and restraining forces for this market, the report also offers a complete study of the future trends and developments of the market.The report further elaborates on the micro and macroeconomic aspects including the socio-political landscape that is anticipated to shape the demand of the Cell Therapy market during the forecast period (2019-2029). It also examines the role of the leading market players involved in the industry including their corporate overview, financial summary, and SWOT analysis.

Get Sample Copy of this Report @ Cell Therapy Market Size, Share, Market Research and Industry Forecast Report, 2020-2027 (Includes Business Impact of COVID-19)

Industry Insights, Market Size, CAGR, High-Level Analysis: Cell Therapy Market

The global cell therapy market size was valued at USD 5.8 billion in 2019 and is projected to witness a CAGR of 5.4% during the forecast period. The development of precision medicine and advancements in Advanced Therapies Medicinal Products (ATMPS) in context to their efficiency and manufacturing are expected to be the major drivers for the market. In addition, automation in adult stem cell and cord blood processing and storage are the key technological advancements that have supported the growth of the market for cell therapy. The investment in technological advancements for decentralizing manufacturing of this therapy is anticipated to significantly benefit the market. Miltenyi Biotec is one of the companies that has contributed to the decentralization in manufacturing through its CliniMACS Prodigy device. The device is an all-in-one automated manufacturing system that exhibits the capability of manufacturing various cell types.

An increase in financing and investments in the space to support the launch of new companies is expected to boost the organic revenue growth in the market for cell therapy. For instance, in July 2019, Bayer invested USD 215 million for the launch of Century Therapeutics, a U.S.-based biotechnology startup that aimed at developing therapies for solid tumors and blood cancer. Funding was further increased to USD 250 billion by a USD 35 million contribution from Versant Ventures and Fujifilm Cellular Dynamics. The biomanufacturing companies are working in collaboration with customers and other stakeholders to enhance the clinical development and commercial manufacturing of these therapies. Biomanufacturers and OEMs such as GE healthcare are providing end-to-end flexible technology solutions to accelerate the rapid launch of therapies in the market for cell therapy. The expanding stem cells arena has also triggered the entry of new players in the market for cell therapy. Celularity, Century Therapeutics, Rubius Therapeutics, ViaCyte, Fate Therapeutics, ReNeuron, Magenta Therapeutics, Frequency Therapeutics, Promethera Biosciences, and Cellular Dynamics are some startups that have begun their business in this arena lately. Use-type Insights The clinical-use segment is expected to grow lucratively during the forecast period owing to the expanding pipeline for therapies. The number of cancer cellular therapies in the pipeline rose from 753 in 2018 to 1,011 in 2019, as per Cancer Research Institute (CRI). The major application of stem cell treatment is hematopoietic stem cell transplantation for the treatment of the immune system and blood disorders for cancer patients. In Europe, blood stem cells are used for the treatment of more than 26,000 patients each year. These factors have driven the revenue for malignancies and autoimmune disorders segment. Currently, most of the stem cells used are derived from bone marrow, blood, and umbilical cord resulting in the larger revenue share in this segment. On the other hand, cell lines, such as Induced Pluripotent Stem Cells (iPSC) and human Embryonic Stem Cells (hESC) are recognized to possess high growth potential. As a result, a several research entities and companies are making significant investments in R&D pertaining to iPSC- and hESC-derived products. Therapy Type Insights of Cell Therapy Market

An inclination of physicians towards therapeutic use of autologous and allogeneic cord blood coupled with rising awareness about the use of cord cells and tissues across various therapeutic areas is driving revenue generation. Currently, the allogeneic therapies segment accounted for the largest share in 2019 in the cell therapy market. The presence of a substantial number of approved products for clinical use has led to the large revenue share of this segment.

Furthermore, the practice of autologous tissue transplantation is restricted by the limited availability of healthy tissue in the patient. Moreover, this type of tissue transplantation is not recommended for young patients wherein tissues are in the growth and development phase. Allogeneic tissue transplantation has effectively addressed the above-mentioned challenges associated with the use of autologous transplantation. However, autologous therapies are growing at the fastest growth rate owing to various advantages over allogeneic therapies, which are expected to boost adoption in this segment. Various advantages include easy availability, no need for HLA-matched donor identification, lower risk of life-threatening complications, a rare occurrence of graft failure, and low mortality rate.

Regional Insights of Cell Therapy Market

The presence of leading universities such as the Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, and Yale Stem Cell Center that support research activities in U.S. is one of the key factor driving the market for cell therapy in North America. Moreover, strong regulatory and financing support from the federal bodies for expansion of this arena in U.S. as well as Canada is driving the market. In Asia Pacific, the market is anticipated to emerge as a lucrative source of revenue owing to the availability of therapies at lower prices coupled with growing awareness among the healthcare entities and patients pertaining the potential of these therapies in chronic disease management. Japan is leading the Asian market for cell therapy, which can be attributed to its fast growth as a hub for research on regenerative medicine. Moreover, the Japan government has recognized regenerative medicine and cell therapy as a key contributor to the countrys economic growth. This has positively influenced the attention of global players towards the Asian market, thereby driving marketing operations in the region.

Market Share Insights of Cell Therapy Market

Some key companies operating in this market for cell therapy are Fibrocell Science, Inc.; JCR Pharmaceuticals Co. Ltd.; Kolon TissueGene, Inc.; PHARMICELL Co., Ltd.; Osiris Therapeutics, Inc.; MEDIPOST; Cells for Cells; NuVasive, Inc.; Stemedica Cell Technologies, Inc.; Vericel Corporation; and ANTEROGEN.CO.,LTD. These companies are collaborating with the blood centers and plasma collection centers in order to obtain cells for use in therapeutics development. Several companies have marked their presence in the market by acquiring small and emerging therapy developers. For instance, in August 2019, Bayer acquired BlueRock Therapeutics to establish its position in the market for cell therapy. BlueRock Therapeutics is a U.S. company that relies on a proprietary induced pluripotent stem cell (iPSC) platform for cell therapy development. Several companies are making an entry in the space through the Contract Development and Manufacturing Organization (CDMO) business model. For example, in April 2019, Hitachi Chemical Co. Ltd. acquired apceth Biopharma GmbH to expand its global footprint in the CDMO market for cell and gene therapy manufacturing.

Segmentations, Sub Segmentations, CAGR, & High-Level Analysis overview of Cell Therapy Market Research Report This report forecasts revenue growth at global, regional, and country levels and provides an analysis of the latest industry trends in each of the sub-segments from 2019 to 2030. For the purpose of this study, this market research report has segmented the global cell therapy market on the basis of use-type, therapy-type, and region:

Use-Type Outlook (Revenue, USD Million, 2019 2030)

Clinical-use

By Therapeutic Area

Malignancies

Musculoskeletal Disorders

Autoimmune Disorders

Dermatology

Others

By Cell Type

Stem Cell Therapies

BM, Blood, & Umbilical Cord-derived Stem Cells

Adipose derived cells

Others

Non-stem Cell Therapies

Research-use

Therapy Type Outlook (Revenue, USD Million, 2019 2030)

Allogeneic Therapies

Autologous Therapies

Quick Read Table of Contents of this Report @ Cell Therapy Market Size, Share, Market Research and Industry Forecast Report, 2020-2027 (Includes Business Impact of COVID-19)

Trusted Business Insights Shelly Arnold Media & Marketing Executive Email Me For Any Clarifications Connect on LinkedIn Click to follow Trusted Business Insights LinkedIn for Market Data and Updates. US: +1 646 568 9797 UK: +44 330 808 0580

Read the original:
Cell Therapy Market Analysis Of Global Trends, Demand And Competition 2020-2028 - Jewish Life News

Why we still haven’t cloned humans it’s not just ethics – Business Insider – Business Insider

Two crossed lines that form an 'X'. It indicates a way to close an interaction, or dismiss a notification.

Two crossed lines that form an 'X'. It indicates a way to close an interaction, or dismiss a notification.

Following is a transcript of the video.

Narrator: We've been able to clone human embryos for about seven years. But as far as we know, no one's actually cloned a whole person. Turns out, ethics aren't the only thing holding scientists back. Cloning isn't the sci-fi marvel we think it is. It can be dangerous, often ineffective, and, most of all, we just haven't thought of a good enough reason to do it. So, here's why you'll probably never have to fight your evil clone.

This is Dolly. Just kidding, that's a regular sheep. This is Dolly, the first mammal cloned successfully from an adult cell. She was born in 1996 after scientists figured out how to remove the DNA from the egg cell of a Scottish Blackface sheep and basically replace it with the DNA of a mammary cell from a Finn Dorset sheep. They gave it a little electric shock to fuse the cell and get it replicating, placed the cells in the uterus of another sheep, and boom, clone. This method, called reproductive cloning, could theoretically be used on humans. But this is a best-case scenario. It took 277 tries for the scientists to get one Dolly. Nowadays, cloning mammals generally has a success rate of about 10% to 20%. Better than one in 277, but still a majorly inefficient process.

Jose Cibelli: Technically, it's not difficult to produce a clone embryo, but human cloning has other hurdles that need to be considered.

Narrator: To even research human cloning, scientists would need to ethically collect a large amount of donated eggs and find enough surrogates to carry them. But even if they made it through that logistical nightmare, the biggest issue is this:

Cibelli: They're gonna hurt the baby, or they're gonna hurt the person carrying the cloned fetus.

Narrator: Across the board, scientists have found that some embryos expire before they're implanted. Others result in miscarriages. And those that make it to term often die soon after birth or end up with severe abnormalities. Simply, these are risks that are easier to take when it comes to experimenting with sheep than with people. But arguably the biggest reason we haven't cloned a human being? There's not a good enough reason to.

In pop culture, cloning is used to bring people back from the dead. But that's not how it works. Cloning someone would only create a twin, not a replica, since identical twins have the same genetics, but not necessarily personalities. And a "Never Let Me Go" scenario, where organs are harvested from clones to save the rich, is not only unethical, but unnecessary. Why clone an entire person when you can just make the part you need? Something, theoretically, therapeutic cloning can solve.

Therapeutic cloning is almost identical to reproductive, except the cloned embryo is never implanted in a uterus. Instead, the embryo is cloned for the sole purpose of extracting stem cells. Stem cells have the incredible ability to turn into any other cell in the human body, which means they're great for developing new treatments for disease and have the potential to repair or regenerate tissues and organs.

But, no surprise, there are a lot of downsides with therapeutic cloning. The thing about stem cells is that they're a pretty limited resource. The most substantial source for embryonic stem cells? Three- to five-day-old embryos, cloned or otherwise. And when someone else's stem cells are transplanted into a patient, the body will sometimes fight them off like a disease. Some researchers believe that cloned stem cells, since they share the patient's DNA, would be less likely to be rejected. But this use case is still in the research stage.

And, finally, therapeutic cloning is an individualized treatment in a world where drug companies are more interested in standardized ones. And there are easier ways to create multipurpose cells nowadays, like the method for creating induced pluripotent stem cells. They're basically adult cells that have been reprogrammed to be a different type of cell.

Cibelli: The problem with therapeutic cloning, of course, is that you need a lab personnel that is qualified to do it, specific equipment to do it. Whereas the other technique, you can just buy a kit and one person can do it in a lab that has some expertise in tissue culture.

Narrator: Cloned cells still have an advantage when it comes to healthier mitochondria and the ability to grow into entire animals, whereas iPSCs often peter out. But since iPSCs safely and reliably do most everything but create entire living animals, why fund the harder, ethically ambiguous thing? So, cloning might actually have a bigger place in movies than it does in real life, because the money just isn't there. And just because we can do something doesn't mean we need to.

Ian: Your scientists were so preoccupied with whether or not they could, they didn't stop to think if they should.

Abby Tang: So, in the research for this video, I did come across one very interesting tidbit, and that is the announcement of cloned human baby Eve, who was born on December 26, 2002. And the source of this announcement is a company called Clonaid, which was formed in 1997 by the Raelian cult. And they're a cult that believes that humans were cloned from aliens and the only way for us to reach immortality is to clone ourselves. It's been 18 years, and we haven't gotten any proof that baby Eve exists or has ever existed, but the company is still alive and well. So if any proof does come through, we will update you.

Read the original here:
Why we still haven't cloned humans it's not just ethics - Business Insider - Business Insider

COVID-19 Impact and Recovery Analysis on Cell Freezing Media for Cell Therapy Market 2020-2026 by BioLife Solutions, Akron Biotechnology, GE…

Heres recently issued report on the Global Cell Freezing Media for Cell Therapy Market that allows you to offer a brief analysis of the market size, demand, supply chain, distribution channels, futuristic trends, market growth elements and so on. It offers a comprehensive analysis of assorted business aspects like global Cell Freezing Media for Cell Therapy market trends, recent technological advancements, market shares, size and new innovations. What is more, this analytical knowledge has been compiled through knowledge searching techniques like primary and secondary analysis for Cell Freezing Media for Cell Therapy industry. Moreover, an professional team of researchers throws lightweight on numerous static additionally as dynamic aspects of the worldwide Cell Freezing Media for Cell Therapy market.

NOTE: Our reports include the analysis of the impact of COVID-19 on this industry. Our new sample is updated which correspond in new report showing impact of Covid-19 on Industry trends. Also we are offering 20% discount

Obtain sample copy of Cell Freezing Media for Cell Therapy market report: https://calibreresearch.com/report/global-cell-freezing-media-therapy-market-5285#request-sample

Geographically, the worldwide Cell Freezing Media for Cell Therapy market has been analyzed in numerous regions like North America, geographical region, geographical region, Asia-Pacific, Africa, Europe and Asian nation. The worldwide Cell Freezing Media for Cell Therapy market region is dominating this market among the forthcoming future. Worldwide Cell Freezing Media for Cell Therapy market is that the skilled and correct study of assorted business views like key players, key geographies, divers, restraints, opportunities and challenges. This global analysis report has been mass on the concept of assorted market segments and sub-segments associated with the worldwide market.

Global Cell Freezing Media for Cell Therapy market competition by prime manufacturers, with Cell Freezing Media for Cell Therapy sales volume, value (USD/Unit), revenue (Million USD) and market share for each manufacturer. The report provides an in-depth analysis of the worldwide Cell Freezing Media for Cell Therapy Market progressing to cut back time to plug for products and services, cut back operational value, improve accuracy and operational performance. Besides this, the report elaborates crucial aspects such as fresh product establishment, greater expenditure in R&D and increasing demand in the manufacturing differentiable growth opportunities in the Cell Freezing Media for Cell Therapy Market globally.

Checkout Inquiry For Buying or Customization of Cell Freezing Media for Cell Therapy Market Report: https://calibreresearch.com/report/global-cell-freezing-media-therapy-market-5285#inquiry-for-buying

The complete profile of the manufacturers are mentioned. And so the capability, production, price, revenue, cost, gross, margin of profit, sales volume, sales revenue, consumption, rate of growth, import and export, future strategies and therefore the technological developments that they are creating are also enclosed among the report. The historical knowledge and forecast knowledge from 2020 to 2026.

Leading companies reviewed in the Cell Freezing Media for Cell Therapy report are:

BioLife Solutions Akron Biotechnology GE Healthcare Thermo Fisher Scientific Biological Industries Merck WAK-Chemie Medical Zenoaq

The Cell Freezing Media for Cell Therapy Market report is segmented into following categories:

The Cell Freezing Media for Cell Therapy market report is segmented into Type by following categories; With FBS Without FBS

The Cell Freezing Media for Cell Therapy market report is segmented into Application by following categories; Human Embryonic Stem Cells CAR-T Cell Therapy Neural Stem Cell Therapy Mesenchymal Stem Cell Therapy Hematopoietic Stem Cell Transplantation Other

Checkout FREE Report Sample of Cell Freezing Media for Cell Therapy Market Report for Better Understanding: https://calibreresearch.com/report/global-cell-freezing-media-therapy-market-5285#request-sample

The report provides substantial knowledge relating to the market share that every one in every of these firms presently garner across this business, in tandem with the market share that they are expected to amass by the highest of the forecast period. Also, the report elaborates on details regarding the products manufactured by every of these companies, that may facilitate new entrants and outstanding stakeholders work on their competition and strategy portfolios. To not mention, their decision-making method is vulnerable to get easier on account of the actual fact that the Cell Freezing Media for Cell Therapy System market report additionally enumerates a gist of the products value trends and so the profit margins of each firm among the industry.

This will enable the readers to focus on Cell Freezing Media for Cell Therapy market product specifications, current competitive manufacturers in Cell Freezing Media for Cell Therapy market and also the market revenue with gain. Worldwide Industry Analyze Cell Freezing Media for Cell Therapy Market by competitive manufacturers, regions and applications of Cell Freezing Media for Cell Therapy market, forecast up to 2026. This report analyses the scope of Cell Freezing Media for Cell Therapy market. This will be achieved by Cell Freezing Media for Cell Therapy previous historical information, analysing qualitative insights info, demonstrable projections regarding global Cell Freezing Media for Cell Therapy market size.

View post:
COVID-19 Impact and Recovery Analysis on Cell Freezing Media for Cell Therapy Market 2020-2026 by BioLife Solutions, Akron Biotechnology, GE...

Cellular Analysis Market Promising Growth Opportunities and Forecast 2017-2025 – Owned

The compositional, genetic, or functional heterogeneousness between diseased and healthy tissues is one of the key challenges faced by the field of drug development and discovery. The lack of knowledge in this field challenges the development of accurate disease models that can be used for drug development, can lead to misinterpretation of the levels of biomarkers, and responses of patients to specific therapies.

Get Exclusive PDF Sample Copy Of This Report:https://www.tmrresearch.com/sample/sample?flag=B&rep_id=788

The complex nature of heterogeneous tissues has encouraged the development of several tools for the analysis of single cell. A number of such tools, for applications such as transcriptomic analysis, cell genomic analysis, and multiplex proteomic analysis have surfaced in the global market in the past few years and continue to be used at an enormous pace across a number of applications, especially in the field of drug development and discovery.

There has been significant advancements in the functionality and level of accuracy of cell analysis tools and companies are increasingly investing in research and development activities to introduce more accurate tools. The competitive landscape and the variety of products that the market has to offer are changing at a rapid pace and a number of new technologies capable of comprehensively analyzing cells at a molecular-level are expected to hit the market in the next few years. Even at present, several cellular analysis technologies exist that can assay for a large number of cell surface markets and secreted proteins. The cellular analysis market is expected to expand at a promising pace in the next few years as well, thanks to the rising investments from across the globe.

Cellular analysis technology has wide applications in cell identification, target identification and validation, and cell counting. The technology is widely used in analysis of cell interaction, cell proliferation, cell structure, cell viability, and cell signaling/cell transduction. Prominent applications of cellular analysis include its uses in various phases of the drug discovery process, oncology, bloodcell analysis, and toxicity testing. Increased applications of cellular analysis in understanding the development processes of various cells types occupies a seminal role in the discovery of personalized medicine and in finding potential treatment of different diseases, which has offered momentum to the cellular analysis market growth.

The cellular analysis market is anticipated to witness significant growth opportunities along the forecast period. General rise in healthcare spending, coupled with increasing corporate funding for cell-based researches, has boosted the growth of the market across various regions. Cell identification is one of the key applications of cellular analysis and helps in the identification of blood cells based on their size and morphology; this helps in tumor detection. Along with other target identification technologies, the high-content screening in cellular analysis technology helps in getting accurate information on drug toxicity and hence facilitates drug discovery.

The report cellular analysis market provides the market share and size of key application segments in different countries across key regions. The research analysis provides in-depth analysis into major drivers and market challenges, current and emerging trends, and technological advances in life sciences, new product launches, and the factors shaping the competitive landscape of the cellular analysis market. The insights offered in the report help new and established players devise impactful strategies to strengthen their market presence.

Buy This Report @https://www.tmrresearch.com/checkout?rep_id=788<ype=S

The cellular analysis market is primarily driven by the rising incidence of chronic and infectious diseases, increased corporate funding for cell-based research activities, and the increasing number of patients suffering from cancer. Furthermore, technological advancements in cellular analysis instruments, combined with potential developments in life-sciences industry related to drug discovery, are the crucial factors boosting the cellular analysis market.

However, the high costs of installing cell analysis systems, coupled with substantial investments required for cell-based researches for oncology and drug discovery, are likely to hinder the growth of the cellular analysis market. In addition, the ethical concerns related to embryonic stem cell research along with stringent regulations for the approval of new cellular analysis systems are major challenges faced by the market players. On the other hand, automation in oncology and the emergence of contract research organizations offering high-content screening services, combined with a large untapped market in the Asia Pacific region, are expected to create promising growth opportunities for the cellular analysis market players.

Based on geography, North America, Europe, and Asia occupy a major share in the cellular analysis market. North America is a prominent region for growth, led by technological advancements in cellular analysis systems and considerable funding by the different governments and private players. Asia Pacific is fast emerging as a promising region to create new revenue streams for market players. A large part of untapped markets in countries such as India, Japan, and China, combined with the increasing government support for cancer and stem cell research, are the crucial factors fuelling the growth of the cellular analysis market in the region.

The research study provides an extensive analysis of the key offerings of the market players and the strategies adopted by them to strengthen their market share. Companies in the cellular analysis market actively launch new products, enter into partnerships and agreements, and make acquisitions to consolidate their market presence. Prominent players operating in the cellular analysis market include Thermo Fisher Scientific, Inc, Beckman Coulter Inc. Bio-Rad Laboratories, Inc., Merck KGaA, Agilent Technologies, Inc, GE Healthcare, Olympus Corporation, Tecan Group Ltd., Nikon Corporation, and Becton, Dickinson and Company.

To know more about the table of contents, you can click @https://www.tmrresearch.com/sample/sample?flag=T&rep_id=788

About TMR Research

TMR Research is a premier provider of customized market research and consulting services to busi-ness entities keen on succeeding in todays supercharged economic climate. Armed with an experi-enced, dedicated, and dynamic team of analysts, we are redefining the way our clients conduct business by providing them with authoritative and trusted research studies in tune with the latest methodologies and market trends.

Go here to read the rest:
Cellular Analysis Market Promising Growth Opportunities and Forecast 2017-2025 - Owned

Why are scientists trying to manufacture organs in space? – The Conversation US

This Bioculture System will let biologists learn about how space impacts human health by studying cells grown in the microgravity environment of the International Space Station.

Gravity can be a real downer when you are trying to grow organs.

Thats why experiments in space are so valuable. They have revealed a new perspective into biological sciences, including insights into making human tissues.

Gravity influences cellular behavior by impacting how protein and genes interact inside the cells, creating tissue that is polarized, a fundamental step for natural organ development. Unfortunately, gravity is against us when we try to reproduce complex three dimensional tissues in the lab for medical transplantation. This is difficult because of the intrinsic limitations of bio-reactors used on Earth.

I am a stem cell biologist and interested on brain health and evolution. My lab studies how the human brain is formed inside the womb and how alterations in this process might have lifelong consequences to human behavior, such as in autism or schizophrenia. Part of that work includes growing brain cells in space.

To build organized tissues in the lab, scientists use scaffolds to provide a surface for cells to attach based on a predetermined rigid shape. For example, an artificial kidney needs a structure, or scaffold, of a certain shape for kidney cells to grow on. Indeed, this strategy helps the tissue to organize in the early stages but creates problems in the long run, such as eventual immune reactions to these synthetic scaffolds or inaccurate structures.

[Deep knowledge, daily. Sign up for The Conversations newsletter.]

By contrast, in weightless conditions, cells can freely self-organize into their correct three-dimensional structure without the need for a scaffold substrate. By removing gravity from the equation, we researchers might learn new ways of building human tissues, such as cartilage and blood vessels that are scaffold-free, mimicking their natural cellular arrangement in an artificial setting. While this is not exactly what happens in the womb (after all the womb is also subject to gravity), weightless conditions does give us an advantage.

And this is precisely what is happening at the International Space Station.

These experiments help researchers optimize tissue growth for use in basic science, personalized medicine and organ transplantation.

But there are other reasons why we should manufacture organs in space. Long-term space missions create a series of physiological alterations in the body of astronauts. While some of these alterations are reversible with time, others are not, compromising future human spaceflights.

Studying astronauts bodies before and after their mission can reveal what goes wrong on their organs, but provides little insights on the mechanisms responsible for the observed alterations. Thus, growing human tissues in space can complement this type of investigation and reveal ways to counteract it.

Finally, all forms of life that we know about have evolved in the presence of microgravity. Without gravity, our brains might have evolved in a different trajectory, or our livers might not filter liquids as it does on Earth.

By recreating embryonic organ formation in space, we can anticipate how the human body in the womb would develop. There are several research initiatives going on in my lab with human brain organoids at ISS, designed to learn the impact of zero gravity on the developing human brain. These projects will have profound implications for future human colonization (can humans successfully reproduce in space?). These studies will also improve the generation of artificial organs that are used for testing drugs and treatments on Earth. Will better treatments for neurodevelopmental and neurodegenerative conditions that affects millions of people come from research in space?

See more here:
Why are scientists trying to manufacture organs in space? - The Conversation US

Stem Cell Characterization and Analysis Tool Market By Covid-19 Impact Analysis Volume and Value, Revenue, Business Opportunity, Future Projections,…

The research study Stem Cell Characterization and Analysis Tool market 2020 launched by ABRReports.com provides the detailed analysis of the current market status, investment plans, production and consumption, price trends, and analysis by the market player, by region, by type, by application and etc, and custom research can be added according to specific requirements

Get Sample Copy of the report at https://www.abrreports.com/industry-insights/covid-19-impact-on-2020-2026-global-and-regional-stem-cell-characterization-and-analysis-tool-industry-production-sales-and-consumption-status-and-prospects-professional-market-research-report-standard-version?form=request-report-sample

The prime objective of this report is to help the user understand the market in terms of its definition, segmentation, market potential, influential trends, and the challenges that the market is facing with 10 major regions and 50 major countries. Deep researches and analysis were done during the preparation of the report. The readers will find this report very helpful in understanding the market in depth.

The key players covered in the Stem Cell Characterization and Analysis Tool Market research report are: By Market Players: Osiris Therapeutics, Inc. Cytori Therapeutics, Inc. Astellas Pharma Inc. Caladrius Biosciences, Inc. Cellular Engineering Technologies Inc. U.S. Stem Cell, Inc. BioTime Inc. TEMCELL Technologies Inc. BrainStorm Cell Therapeutics Inc.

By Type Services Software Instruments Accessories Consumables Reagent and Assay Kits

By Application Neurological Disorders Orthopedic Treatments Oncology Disorders Diabetes Other Therapeutic Applications Drug Development and Discovery Embryonic Stem Cells Research

Click to access full report and Table of Content at https://www.abrreports.com/industry-insights/covid-19-impact-on-2020-2026-global-and-regional-stem-cell-characterization-and-analysis-tool-industry-production-sales-and-consumption-status-and-prospects-professional-market-research-report-standard-version

The data and the information regarding the market are taken from reliable sources such as websites, annual reports of the companies, journals, and others and were checked and validated by the industry experts. The facts and data are represented in the report using diagrams, graphs, pie charts, and other pictorial representations. This enhances the visual representation and also helps in understanding the facts much better.

Key pointers of the Table of Contents:

Chapter 1 Industry Overview Chapter 2 Global Stem Cell Characterization and Analysis Tool Competition by Types, Applications, and Top Regions and Countries Chapter 3 Production Market Analysis Chapter 4 Global Stem Cell Characterization and Analysis Tool Sales, Consumption, Export, Import by Regions (2015-2020) Chapter 5 North America Stem Cell Characterization and Analysis Tool Market Analysis Chapter 6 East Asia Stem Cell Characterization and Analysis Tool Market Analysis Chapter 7 Europe Stem Cell Characterization and Analysis Tool Market Analysis Chapter 8 South Asia Stem Cell Characterization and Analysis Tool Market Analysis Chapter 9 Southeast Asia Stem Cell Characterization and Analysis Tool Market Analysis Chapter 10 Middle East Stem Cell Characterization and Analysis Tool Market Analysis Chapter 11 Africa Stem Cell Characterization and Analysis Tool Market Analysis Chapter 12 Oceania Stem Cell Characterization and Analysis Tool Market Analysis Chapter 13 South America Stem Cell Characterization and Analysis Tool Market Analysis Chapter 14 Company Profiles and Key Figures in Stem Cell Characterization and Analysis Tool Business Chapter 15 Global Stem Cell Characterization and Analysis Tool Market Forecast (2021-2026) Chapter 16 Conclusions

Purchase the research study @

https://www.abrreports.com/industry-insights/covid-19-impact-on-2020-2026-global-and-regional-stem-cell-characterization-and-analysis-tool-industry-production-sales-and-consumption-status-and-prospects-professional-market-research-report-standard-version/checkout?option=one

About Us:

ABR Reports (Advanced Business Research Reports) is the premium market research reselling company that offers market research reports to individuals, organizations, and industries to enhance and strengthen the decision making process. With an associate thoroughgoing list of market research Publishers, we tend to cut across all the business verticals covering 5000+ micro markets and offer market size and share analysis, industry trends, information on products, regional market, and keen business insights to our clients.

Contact Us:

Scott Harris Sales Manager Email ID: [emailprotected] Phone No.: +1-561-448-7424

Excerpt from:
Stem Cell Characterization and Analysis Tool Market By Covid-19 Impact Analysis Volume and Value, Revenue, Business Opportunity, Future Projections,...

A novel system to map protein interactions reveals evolutionarily conserved immune evasion pathways on transmissible cancers – Science Advances

Abstract

Around 40% of humans and Tasmanian devils (Sarcophilus harrisii) develop cancer in their lifetime, compared to less than 10% for most species. In addition, devils are affected by two of the three known transmissible cancers in mammals. Immune checkpoint immunotherapy has transformed human medicine, but a lack of species-specific reagents has limited checkpoint immunology in most species. We developed a cut-and-paste reagent development system and used the fluorescent fusion protein system to show that immune checkpoint interactions are conserved across 160,000,000 years of evolution, CD200 is highly expressed on transmissible tumor cells, and coexpression of CD200R1 can block CD200 surface expression. The systems versatility across species was demonstrated by fusing a fluorescent reporter to a camelid-derived nanobody that binds human programmed death ligand 1. The evolutionarily conserved pathways suggest that naturally occurring cancers in devils and other species can be used to advance our understanding of cancer and immunological tolerance.

Metastatic cancer affects most mammals, but the cancer incidence can vary widely across phylogenetic groups and species (Fig. 1 and table S1) (13). In humans, the lifetime risk of developing cancer is around 40% (4). This figure is in stark contrast to a general cancer incidence of 3% for mammals, 2% for birds, and 2% for reptiles reported by the San Diego Zoo (N = 10,317) (2, 5). A more recent study at the Taipei Zoo reported cancer incidence of 8, 4, and 1% for mammals, birds, and reptiles, respectively (N = 2657) (6). Cancer incidence in domestic animals is generally less than 10% (N = 202,277) (3). However, two studies performed 40 years apart reported that greater than 40% of Tasmanian devils develop spontaneous, often severe neoplasia in their lifetime (5, 7). Devils are also unique because they are affected by two of the three known naturally occurring transmissible cancers in vertebrate species (8, 9). Transmissible cancers are a distinct form of cancer in which the tumor cells function as an infectious pathogen and an allograft. Dogs (Canis lupus familiaris) are the only other vertebrate species affected by a transmissible cancer (10), and interestingly, some breeds of dogs also have high cancer incidence (3, 11).

Metastatic cancer has been reported in nearly all mammalian orders, and MHCs have been the most intensely studied molecules in most orders. In the past decade, studies of immune checkpoint molecules (PD1, PDL1, and CTLA4) have become a primary focus in humans and rodents. However, immune checkpoint studies in other species are limited, particularly at the protein level, because of the lack of species-specific reagents. This creates a vast gap in our understanding of the evolution of the mammalian immune system. The numbers in the columns represent the number studies matching Web of Science search results between 2009 and 2019. See table S1 for search terms.

The devil facial tumor (DFT) disease was first detected in Northwest Tasmania and has been a primary driver of an 80% decline in the wild Tasmanian devil population (8, 12). The clonal DFT (DFT1) cells have been continually transmitted among devils and are estimated to have killed at least 10,000 individuals since at least 1996. In 2014, a second independent transmissible Tasmanian DFT (DFT2) was found in wild devils (9), and 23 cases have been reported to date (13). Genetic mismatches, particularly in the major histocompatibility complex (MHC) genes, should lead to rejection of these transmissible tumors. Consequently, the role of devil MHC has been a focus of numerous studies (Fig. 1 and table S1) to understand the lack of rejection of the transmissible tumors. These studies have revealed that the DFT1 cells down-regulate MHC class I (MHC-I) expression (14), a phenomenon observed in many human cancers. In contrast to DFT1 cells, the DFT2 cells do express MHC-I (15). DFT1 and DFT2 cells have 2884 and 3591 single-nucleotide variants, respectively, that are not present in 46 normal devil genomes (16). The continual transmission of DFT1 and DFT2, despite MHC-I expression by DFT2 cells and genetic mismatches between host and tumor, suggests that additional pathways are likely involved in immune evasion.

Human cancer treatment has been transformed in the past decade by manipulating interactions among immune checkpoint molecules. These have proven broadly effective in part because they function across many different MHC types and tumor mutational patterns. However, these pathways have received little attention in transmissible cancers and other naturally occurring cancers in nonmodel species (Fig. 1 and table S1) (1719). We have previously shown that the inhibitory immune checkpoint molecule programmed death ligand 1 (PDL1) is expressed in the DFT microenvironment and is up-regulated by interferon- (IFN-) in vitro (17). This finding led us to question which other immune checkpoint molecules play a role in immune evasion by the transmissible cancers and the devils high spontaneous cancer incidence. Understanding immune evasion in a natural environment will support DFT vaccine development to help protect this endangered species (20) and has the potential to identify protein interactions that are conserved across divergent species to improve translational success of animal models (19). Unfortunately, a persistent limitation for immunology in nontraditional study species is a lack of species-specific reagents. Wildlife biologists and veterinarians are at the front lines of emerging infectious disease outbreaks, but they often lack species-specific reagents to fulfill the World Health Organizations call for cross-cutting R&D preparedness and perform mechanistic immunological investigations.

To solve the paucity of reagents available for Tasmanian devils and address ongoing limitations for nontraditional study species, we developed a Fluorescent Adaptable Simple Theranostic (FAST) protein system that builds on the diverse uses of fluorescent proteins previously reported (2123). This simple system can be used for rapid development of diagnostic and therapeutic (i.e., theranostic) immunological toolkits for any animal species (Fig. 2). We demonstrate the impact of the FAST system by using it to confirm seven receptor-ligand interactions among 12 checkpoint proteins in devils. We demonstrate the versatility of the system across species by fusing a fluorescent reporter to a well-characterized camelid-derived nanobody that binds human PDL1 (24).

(A) Schematic diagram of FAST protein therapeutic and diagnostic (i.e., theranostic) features. POI, protein of interest. (B) Graphic overview of FAST protein system including key steps: (i) characterize gene of interest (GOI) in silico; (ii) design expression vectors; (iii) digest FAST base vectors and insert alternative genes of interest or colors; (iv) transfect FAST vectors into mammalian cells and monitor using fluorescent microscopy or flow cytometry; (iv) purify the protein using 6xHis tag, visualize fluorescent color to show that protein is in frame and correctly folded. Image of microfuge tubes shows 100 l of mCitrine, mOrange, and mCherry FAST proteins (1 mg/ml) excited with blue light with an amber filter. Full protocols for vector construction and protein testing are available in the Supplementary Materials. (C) Results of flow cytometry binding assay with devil 41BB FAST proteins. The colored lines in the histograms show binding of devil 41BB fused to mTagBFP, mCerulean3, mAzurite, mCitrine, mOrange, mCherry, or mNeptune2 to CHO cells transfected with devil 41BBL, and the black lines show binding to untransfected CHO cells. FSC, forward scatter; SSC, side scatter.

In humans, checkpoint proteins have been targets of immunotherapy in clinical trials, but the functional role and binding patterns of these proteins are unknown for most other species. We have used the FAST system to show that the inhibitory checkpoint protein CD200 is highly expressed on DFT cells, opening the door to single-cell phenotyping of circulating tumor cells (CTCs) in devil blood. Furthermore, we are the first to report that coexpression of CD200R1 can block surface expression of CD200 in any species. Understanding how clonal tumor cells graft onto new hosts, evade immune defenses and metastasize within a host will identify evolutionarily conserved immunological mechanisms to help improve cancer, infectious disease, and transplant outcomes for human and veterinary medicine.

Initially, we developed FAST proteins to determine whether monomeric fluorescent proteins could be fused to devil proteins and secreted from mammalian cells (Fig. 2A and table S2). We used 41BB (TNFRSF9) for proof-of-concept studies by fusing the extracellular domain of devil 41BB checkpoint molecule to monomeric fluorescent proteins (Fig. 2, A and B, and fig. S1). We used wild-type Chinese hamster ovary (CHO) cells and CHO cells transfected with 41BBL (TNFSF9) to confirm specificity of the 41BB FAST proteins and demonstrate that the fluorescent proteins [mTag-blue fluorescent protein (BFP), mCerulean3, mAzurite, mCitrine, mOrange, mCherry, and mNeptune2] remained fluorescent when secreted from mammalian cells (Fig. 2C).

We chose mCherry, mCitrine, mOrange, and mBFP for ongoing FAST protein development. Initial batches of FAST proteins were purified using the 6xHis tag and eluted with imidazole. Following purification of FAST protein the color can be immediately observed with blue light and an amber filter unit, allowing confirmation that the fluorescent protein DNA coding sequences were in frame and the proteins were properly folded. After combining, concentrating, and sterile filtering the eluted fractions, 100 l at 1 mg/ml was aliquoted and visualized again using blue light to confirm fluorescent signal (Fig. 2B). A full step-by-step protocol and set of experimental templates for creating and testing FAST proteins for any species are available online in the Supplementary Materials.

We chose candidate immune checkpoint molecules for FAST protein development (Fig. 3A and table S2) based on targets of human clinical trials and then selected devil genes for which a reliable sequence was available either in the published devil genome or transcriptomes (19, 25, 26). We transfected the FAST protein expression vectors (table S3) into CHO cells and tested the supernatant against CHO cell lines expressing full-length receptors. 41BB FAST proteins in supernatant exhibited strong binding to 41BBL cell lines, but the fluorescent signals from most other FAST proteins were too weak to confirm binding to the expected receptors (fig. S2). As FAST proteins do not require secondary reagents, we next incubated target cells with purified FAST proteins and added chloroquine to block the lysosomal protein degradation pathway. This allowed us to take advantage of receptor-mediated endocytosis, which can allow accumulation of captured fluorescent signals inside the target cells (27). This protocol adjustment allowed confirmation that CD47-mCherry, CD200-mBFP, CD200-mOrange, CD200R1-mBFP, and CD200R1-mOrange, and PD1-mCitrine bound to their expected receptors (Fig. 3B). We also demonstrated the flexibility of the FAST proteins by showing that alternative fusion conformations (fig. S1, C and D), such as type II proteins (e.g., mCherry-41BBL) and a devil Fc tag (e.g., CD80-Fc-mCherry) bound to their expected ligands (Fig. 3B). The stability of the fusion proteins was demonstrated using supernatants that were stored at 4C for 2 months before use in a 1-hour live-culture assay with chloroquine (fig. S3).

(A) Diagram of soluble FAST proteins and full-length proteins used for testing of FAST proteins. 41BBL is a type II transmembrane protein; all other proteins are type I. CD80 and CTLA4 soluble FAST proteins included a devil immunoglobulin G (IgG) Fc tag. Arrows indicate interactions confirmed in this study. TNF, tumor necrosis factor. (B) Histograms showing binding of FAST proteins to CHO cells expressing full-length devil proteins. Target CHO cells were cultured with chloroquine to block lysosomal degradation of FAST proteins and maintain fluorescent signal during live-culture binding assays with purified FAST proteins (2 g per well) for 30 min or 18 hours to assess receptor-ligand binding (N = 1 per time point).

To further streamline the reagent development process, we next took advantage of the single-step nature of FAST proteins (i.e., no secondary antibodies or labels needed) in live-cell coculture assays (Fig. 4A). Cell lines secreting 41BB-mCherry, 41BBL-mCherry, or CD80-Fc-mCherry FAST proteins were mixed with cell lines expressing full-length 41BB, 41BBL, or CTLA4-mCitrine and cocultured at a 1:1 ratio overnight with chloroquine. Singlet cells were gated (Fig. 4B) and binding of mCherry FAST proteins to carboxyfluorescein diacetate succinimidyl ester (CFSE) or mCitrine-labeled target cells was analyzed (Fig. 4C). The strongest fluorescent signal from 41BB-mCherry, 41BBL-mCherr, and CD80-Fc-mCherry was detected when cocultured with their predicted receptors, 41BBL, 41BB, and CTLA4, respectively.

(A) Schematic of coculture assays to assess checkpoint molecule interactions (absent, weak, and strong). Cells were mixed and cultured overnight with chloroquine. Protein binding and/or transfer were assessed using flow cytometry. (B) Gating strategy for coculture assays. (C) CHO cells that secrete 41BBL-mCherry, 41BB-mCherry, or CD80-Fc-mCherry were cocultured overnight with target CHO cells that express full-length 41BB, 41BBL, or CTLA4. 41BB and 41BB-L were labeled with CFSE, whereas full-length CTLA4 was directly fused to mCitrine. Cells that secrete mCherry FAST proteins appear in the upper left quadrant. Cells expressing full-length proteins and labeled with CFSE or mCitrine appear in the lower right quadrant. Cells in the upper right quadrant represent binding of mCherry FAST proteins to full-length proteins on carboxyfluorescein diacetate succinimidyl ester (CFSE) or mCitrine-labeled cells. Results shown are representative of n = 3 per treatment. (D) CTLA4-Fc-mCherry FAST protein binding to DFT cells. DFT1 C5065 cells transfected with control vector (black), 41BB (gray), CD80 (red), or CD86 (blue) were stained with CTLA4-Fc-mCherry supernatant with chloroquine. Results are representative of N = 2 replicates per treatment.

The fluorescent binding signal of CD80-Fc-mCherry was lower than expected, so we next reexamined our Fc tag construct. In humans and all other mammals examined to date, the immunoglobulin G (IgG) heavy chain has glycine-lysine (Gly-Lys) residues at the C terminus; the initial devil IgG constant region sequence available to us had an incomplete C terminus, and thus, our initial CD80-Fc-mCherry vector did not have the C-terminal Gly-Lys. We subsequently made a new FAST-Fc construct with CTLA4-Fc-mCherry, which exhibited strong binding to both CD80 and CD86 transfected DFT cells (Fig. 4D).

Analysis of previously published devil and DFT cell transcriptomes suggested that CD200 mRNA is highly expressed in DFT2 cells and peripheral nerves, moderately expressed in DFT1 cells, and lower in other healthy devil tissues (Fig. 5A) (25, 26, 28). As CD200 is an inhibitory molecule expressed on most human neuroendocrine neoplasms (29), and both DFT1 and DFT2 originated from Schwann cells (26, 30), we sought to investigate CD200 expression on DFT cells at the protein level. Staining of wild-type DFT1 and DFT2 cells with CD200R1-mOrange FAST protein showed minimal fluorescent signal (Fig. 5B). However, overexpression of CD200 using a human EF1 promoter yielded a detectable signal with CD200R1-mOrange binding to CD200 on DFT1 cells. A weak signal from CD200-mOrange was detected on DFT1 cells overexpressing CD200R1 (Fig. 5B). To confirm naturally expressed CD200 on DFT cells, we digested CD200 and 41BB FAST proteins using tobacco etch virus (TEV) protease to remove the linker and fluorescent reporter. The digested proteins were then used to immunize mice for polyclonal serum production. We stained target CHO cell lines with preimmune or immune mouse sera collected after three-times immunizations. Only the immune sera showed strong binding to the respective CD200 and 41BB target cell lines (Fig. 5C). After the final immunization (four times), we collected another batch of sera and tested it on DFT1 and DFT2 cells (Fig. 5D). In agreement with the transcriptomic data for DFT cells (25), the polyclonal sera revealed high levels of CD200 on DFT cells, but low levels of 41BB.

(A) GOIs for this study are plotted as a log2-transformed transcripts per million (TPM) heat map with dark blue indicating the most highly expressed genes. Technical replicates (N = 2) from separate flasks were used for the cell lines (C5065, RV) and biological replicates (N = 2) were used for primary tissues, except peripheral nerve (PN) (N = 1). (B) Wild-type DFT1.C5065, DFT2.JV, DFT2.SN, and DFT1.C5065 transfected to overexpress CD200 or CD200R1 were stained with either CD200R1-mOrange or CD200-mOrange FAST protein. Histograms filled with blue or red highlight expected strong binding interactions. The percentage of events that falls within the marker is shown. Results are representative of N = 2 replicates per treatment. (C) Mice were immunized with 41BB or CD200 FAST proteins. Black, preimmune; gray, immune sera from a mouse immunized with 41BB; red, preimmune; blue, immune sera from a mouse immunized with CD200. CHO cells transfected with either full-length 41BB or CD200 were stained with sera and then anti-mouse AF647. Results are representative of N = 2 per treatment. (D) Sera were used to screen two strains each of DFT1 and DFT2 cells for 41BB and CD200 expression. Results are representative of N = 3 per treatment. (E) DFT1 C5065 transfected with either vector control, CD200, or CD200R1 was stained with purified polyclonal anti-CD200 and anti-mouse IgG AF647 (black, no antibodies; red, secondary antibody only; blue, primary and secondary antibody). Results are representative of N = 2 per treatment.

In humans, overexpression of some checkpoint proteins can block surface expression of heterophilic binding partners in cis (e.g., CD80 and PDL1) (31). As a potential route for disrupting the inhibitory effects of CD200 on antitumor immunity, we tested whether overexpression of CD200R1 on DFT cells could reduce CD200 surface expression. We stained a DFT1 strain, C5065, and DFT1 C5065 cells transfected to overexpress CD200 or CD200R1 with polyclonal anti-CD200 sera and secondary anti-mouse IgG Alexa Fluor 647 (AF647). We detected no surface protein expression of CD200 DFT1 cells overexpressing CD200R1 (Fig. 5E).

In addition to high expression of CD200 on neuroendocrine neoplasms (29), CD200 is used as a diagnostic marker for several human blood cancers (32). DFT cells metastasize in the majority of cases (33), and our transcriptome results (Fig. 5A) suggest that CD200 mRNA is more highly expressed in DFT cells than in peripheral blood mononuclear cells (PBMCs) (25, 26). As a result, we tested whether CD200 could be used to identify DFT cells in blood. We stained PBMCs and DFT2 cells separately with polyclonal anti-CD200 sera and anti-mouse AF647 and then analyzed CD200 expression by flow cytometry (fig. S4A). We then mixed the stained PBMCs and DFT2 cells at ratios of 1:10 (fig. S4A) and 1:5 (fig. S4B) and analyzed the mixed populations. PBMCs showed minimal CD200 expression and background staining (fig. S4), whereas CD200 was highly expressed on DFT2 cells. CD200+ DFT2 cells were readily distinguishable from PBMCs.

As our RNA sequencing (RNA-seq) results only included mononuclear cells, we next performed a pilot test to determine whether DFT cells could be spiked into whole devil blood and identified via flow cytometry using CD200 staining. DFT1 and DFT2 cells were labeled with CellTrace violet (CTV), and 10,000 cells were diluted directly into 100 l of whole blood from a healthy devil (N = 1 per treatment; n = 1 devil). The cells were then stained with purified polyclonal anti-CD200 with and without secondary anti-mouse IgG AF647 before red blood cell (RBC) lysis. Initial results showed that DFT2 cells expressed CD200 above the leukocyte background but that DFT1 cells could not be distinguished from leukocytes (fig. S5). To eliminate the secondary antibody step from the whole blood staining protocol, we next labeled the polyclonal anti-CD200 and normal mouse serum (NMS) with a no-wash Zenon mouse IgG AF647 labeling reagent (n = 1 per treatment; n = 2 devils). This system again showed that CD200 expression could be used to identify DFT2 cells in blood (Fig. 6, A to E), suggesting that CD200 is a candidate marker for identification of metastasizing DFT2 cells.

Color dot plots showing DFT cells in green (CFSE), PBMCs in black, DFT Alexa Fluor 647+ (AF647+) cells in red, and PBMC AF647+ in blue. Forward- and side-scatter plot of DFT2.JV cells alone (A) and DFT2.JV cells mixed with PBMCs (B). (C) Color dot plot showing dead cells stained with 4,6-diamidino-2-phenylindole (DAPI) (right quadrants) and CFSE-labeled DFT cells (upper quadrants). (D) The top row shows unmixed PBMCs. The middle row and bottom row show PBMCs mixed with DFT1.C5065 (middle) and DFT2.JV (bottom) cells. Cell mixtures were either untreated or incubated with Zenon AF647labeled NMS or Zenon AF647labeled -CD200 serum. AF647+ DFT (red) and PBMC (blue) are in the right quadrants. (E) Histogram overlays to highlight AF647+ (right quadrants) from DFT1-PBMC and DFT2-PBMC mixtures. Cells were analyzed on the Beckman Coulter MoFlo Astrios. (F) FAST nanobody proof of concept was accomplished using supernatant from untransfected ExpiCHO cells or ExpiCHO cells secreting human antiPDL1-mCitrine nanobody. Nanobody supernatant was used undiluted or at 1:10 or 1:100 dilutions in media and used to stain CHO cells that express either human PDL1 or human CD80. Results are representative of N = 2 per treatment.

Last, to test whether the FAST system could be applied to other species (e.g., camelid-derived nanobody) and applications (FAST nanobody), we reverse-translated the protein sequence for an anti-human PDL1 nanobody (24) and inserted the codon-optimized DNA sequence into a FAST mCitrine vector. The assembled plasmid was transfected into ExpiCHO cells, and the supernatant was tested for binding to CHO cells stably transfected with either full-length human PDL1 or human CD80; the human proteins were fused to miRFP670 (Addgene no. 79987) in a FAST vector. The nanobody supernatant was used undiluted or at 1:10 or 1:100 dilutions. The nanobody showed strong binding to PDL1-expressing cells, but not CD80-expressing cells (Fig. 6F).

Naturally occurring cancers provide a unique opportunity to study immune evasion and the metastatic process across diverse hosts and environments. The exceptionally high cancer rate in Tasmanian devils coupled with the two transmissible tumors currently circulating in the wild warrants a thorough investigation of the devil immune system. However, taking advantage of these natural disease models has been out of reach for most species because of a lack of reagents. The FAST protein system that we developed here is well suited to discovering additional DFT markers and, more generally, to filling the reagent gap for nontraditional species. For proteins like 41BB that have high affinity for 41BBL, FAST proteins can be used as detection reagents directly from supernatant. For other molecules with lower receptor-ligand affinity, the FAST proteins can be purified, digested with a protease to remove the nontarget proteins, and used for production of higher-affinity binding proteins (e.g., antibodies, aptamers, and nanobodies).

The versatility of the FAST system was demonstrated by fusing a validated human anti-PDL1 nanobody derived from a camel (Camelus bactrianus) heavy-chain variable region to mCitrine. The nanobody-reporter fusion allowed direct testing of the nanobody from supernatant without the need for purification or secondary labeling and provided a 1:1 ratio of nanobody and reporter to allow quantification of target proteins. In addition to fusing nanobodies to fluorescent proteins, fluorescently labeled target proteins could be used with nanobody display libraries to pull down or sort nanobodies that bind the target protein.

The simple cut-and-paste methods for vector assembly lend the FAST protein system to entry-level immunology and molecular biology skill sets. In addition, the ability of FAST proteins to be used in live coculture assays and with elimination of secondary reagents will increase efficiency and reduce experimental error for advanced human and mouse cancer immunology studies. For example, previous high-throughput studies have used a two-step staining process (i.e., recombinant protein and secondary antibody) to screen more than 2000 protein interactions (34); this type of assay can be streamlined using FAST proteins to eliminate the need for secondary antibodies. Fc tags or other homodimerization domains can be incorporated into FAST proteins to increase binding for low-affinity interactions and to assess potential Fc receptormediated functions.

Production of recombinant proteins in cell lines that closely resemble the physiological conditions of the native cell type (i.e., mammalian proteins produced in mammalian cell lines) is more likely to yield correct protein folding, glycosylation, and function than proteins produced using evolutionarily distant cell lines. The fluorescent fusion proteins developed here take advantage of natural receptor expression and cycling processes (e.g., CTLA4 transendocytosis) in eukaryotic target cells; bacterial protein production methods are not amenable to coculture with eukaryotic target cells in immunological assays. Our demonstration of the FAST protein system in CHO cells suggest that this method can be efficiently integrated into existing research and development pipelines for humans and other vertebrate species.

A primary question in transmissible tumor research is why genetically mismatched cells are not rejected by the host. Successful infection of devils with DFT cells relies on the ability of the tumor allograft to evade and manipulate host defenses. The missing-self hypothesis suggests that the lack of constitutive MHC-I expression on DFT1 cells should lead to natural killer (NK) cellmediated killing of the allograft tumor cells. Here, we used the FAST protein system to develop a tool set to address this question and show that DFT1 and DFT2 cells express CD200 at higher levels than most other devil tissues examined to date. CD200 has been shown to directly inhibit NK cells in other species (35), so overexpression of CD200 is a potential mechanism of immune evasion of NK responses by DFT cells.

We hypothesize that CD200 could be particularly important in DFT transmission as the CD200-CD200R pathway is critical to the initial stages of establishing transplant and allograft tolerance in other species (36). In line with this hypothesis, a recent study reported that overexpressing several checkpoint molecules, including CD200, PDL1, and CD47, in mouse embryonic stem cells could be used to generate teratomas that could establish long-term allograft tolerance in fully immunocompetent hosts (37). We have previously reported that PDL1 mRNA and protein are up-regulated on DFT2 cells in response to IFN- (17), and our transcriptome results show that CD47 is expressed at moderate to high levels in DFT cells. Here, we show that overexpression of CD200R1 on DFT1 eliminates binding of our polyclonal anti-CD200 antibodies, suggesting that DFT cells overexpressing CD200R1 could be used to test the role of CD200 in allograft tolerance. Alternatively, genetic ablation of CD200 in DFT cells could be used as a complementary approach to examine the role of immune checkpoint molecules in DFT allograft tolerance. Low MHC-I expression is a primary means of immune evasion by DFT1 cells, and disrupting the CD200-CD200R1 pathway could facilitate improved recognition of DFT1 cells by CD8 T cells by enhancing IFN-mediated MHC-I up-regulation. Recent work in mice has identified immunosuppressive natural regulatory plasma cells that express CD200, LAG3, PDL1, and PDL2; we have previously identified PDL1+ cells with plasma cell morphology near or within the DFT microenvironment (17).

Previous DFT vaccine efforts have used killed DFT cells with adjuvants (38, 39). A similar approach to treat gliomas in dogs reported that tumor lysate with CD200 peptides nearly doubled progression-free survival compared to tumor lysate alone (40). Like devils, several breeds of dog are prone to cancer, and these genetically outbred large animal models provide a fertile ground for testing cancer therapies. The CD200 peptides are reported to provide agonistic function through CD200-like activation receptors (CD200R4) rather than by blocking CD200R1 (40). The functional role of CD200-CD200R pathway in devils remains to be elucidated, but the CD200R1NPLY inhibitory motif and key tyrosine residues are conserved in devil CD200R (19, 41, 42), demonstrating that this motif is conserved over 160 million years of evolutionary history (43). In addition to agonistic peptides, several other options for countering CD200-CD200R immune inhibition are possible. Human chronic lymphocytic leukemia cells often express high levels of CD200, which can be down-regulated in response to imiquimod (44). Likewise, we have previously shown that DFT1 cells down-regulate expression of CD200 mRNA in vitro in response to imiquimod treatment (25). In one of the longest running and most in-depth studies of host-pathogen coevolution, CD200R was shown to be under selection in rabbits in response to a myxoma virus biocontrol agent (45). As DFT1 and DFT2 have been circulating in devils for more than 20 and 5 years, respectively, it will be important to monitor CD200/R expression and the potential evolution of paired activating and inhibitory receptors in these natural disease models.

Immunophenotyping and single-cell RNA-seq of CTCs have a potential to identify key gene expression patterns associated with metastasis and tissue invasion. CD200 is a potential marker for the identification of CTCs from devil blood. As proof of concept, DFT2 cells could be identified in devil blood spiked with DFT2 cells. As CTCs are likely to be rare in the blood of most infected devils, CD200 alone would be insufficient for identifying DFT1 cells. Additional surface DFT markers would be required to purify CTCs for metastases and tissue invasion analyses. The FAST protein system provides a simple procedure to facilitate the production of a panel of DFT markers to help identify key proteins in the metastatic process.

In summary, the simple cut-and-paste production of the vectors and single-step testing pipeline of the FAST system provided multiple benefits. The FAST system allowed us to characterize receptor-ligand interactions and to identify evolutionarily conserved immune evasion pathways in naturally occurring transmissible cancers. Our initial implementation of the system confirmed numerous predicted protein interactions in a marsupial species and documented high expression of the inhibitory molecule CD200 on DFT cells. The high expression of CD200 in devil nervous tissues and neuroendocrine tumors, down-regulation of CD200 in response to imiquimod, and binding of CD200 to CD200R1 are consistent with results from human and mouse studies. Consequently, the CD200/R pathway provides a promising immunotherapy and vaccine target for DFTs (20). Beyond this study, FAST proteins meet the key attributes needed for reagent development, such as being straightforward to make, stable, versatile, renewable, cheap, and amenable to high-throughput testing. The direct fusion of the reporter protein to the protein of interest allows for immediate feedback during transfection, supernatant testing, and protein purification; proteins with frameshifts, introduced stop codons, or folded improperly will not fluoresce and can be discarded after a simple visualization, rather than only after extensive downstream testing. Efficient mapping of immune checkpoint interactions across species can identify evolutionarily conserved immune evasion pathways and appropriate large-animal models with naturally occurring cancer. This knowledge could inform veterinary and human medicine in the fields of immunological tolerance to tissue transplants, infectious disease, and cancer.

The objectives of this study were to fill a major gap in our understanding of the mammalian immune system and to understand how genetically mismatched transmissible tumors evade host immunity. To achieve this goal, we developed a recombinant protein system that directly fuses proteins of interest to a fluorescent reporter protein. The first phase was to determine whether the fluorescent protein remained fluorescent after secretion from mammalian cells and to confirm that proteins bound to their predicted receptors (i.e., ligands). Initial testing was performed in CHO cells and follow-up assays used devil cells. To reduce the risk of false positives in binding assays, we tested each FAST protein against the expected target protein and additional nontarget proteins. To further demonstrate the functionality of this system for antibody development, mice were immunized with either 41BB or CD200 proteins. Pre- and postimmunization polyclonal sera were used to confirm that the proteins used for immunization induced antibodies that specifically bound to surface-expressed recombinant proteins and native proteins on DFT cells. Last, to demonstrate the flexibility of the system, we replicated a known anti-human PDL1 nanobody that we fused to mCitrine. This shows that the FAST system can be used to target human proteins, to produce recombinant proteins derived from other species (e.g., camelid-derived nanobody), and for functions other than receptor-ligand interactions.

Target gene DNA sequences for vector construction were retrieved from Genbank, Ensembl, or de novo transcriptome assemblies (table S2). Target DNA was amplified from a complementary DNA template or existing plasmids using primers and polymerase chain reaction (PCR) conditions shown in tables S2 and S4 using Q5 High-Fidelity 2X Master Mix (New England Biolabs no. M0494L). Primers were ordered with 5 base extensions that overlapped expression vectors on either side of the restriction sites. The amplified products were identified by gel electrophoresis and purified using the NucleoSpin PCR and Gel Clean Up Kit (Macherey-Nagel no. 740609.5). Alternatively, DNA sequences were purchased as double-stranded DNA gBlocks (table S5) (Integrated DNA Technologies) for direct assembly into expression vectors.

All new plasmids were assembled using the NEBuilder kit (New England Biolabs; NEB no. E5520S) following the manufacturers recommendations unless otherwise noted. DNA inserts, digested plasmids, and NEBuilder master mix were incubated for 60 min at 50C and then transformed into DH5 included with the NEBuilder kit. Plasmid digestions were performed following manufacturer recommendations and generally subjected to Antarctic phosphatase (New England Biolabs no. M0289S) treatment to prevent potential reannealing. Sleeping Beauty transposon vectors pSBbi-Hyg (Addgene no. 60524), pSBbi-BH (Addgene no. 60515), pSBtet-Hyg (Addgene no. 60508), and pSBtet-RH (Addgene no. 60500) were gifts to Addgene from E. Kowarz (46). The pCMV(CAT)T7-SB100 containing the cytomegalovirus (CMV) promoter and SB100X transposase was a gift to Addgene from Z. Izsvak (Addgene no. 34879) (47). We first constructed an all-in-one Sleeping Beauty vector by inserting a CMV promoter and SB100X transposase from pCMV(CAT)T7-SB100 (47) into pSBi-BH (46) (tables S3 and S4). This was accomplished by using pAF111-vec.FOR and pAF111.1.REV primers to amplify an overlap region from pSBbi-BH (insert 1) and pAF111-2.FOR and pAF111-2.REV to amplify the CMV-SB100X region from pCMV(CAT)T7-SB100 (insert 2). The purified amplicons were then used for NEBuilder assembly of pAF111. The final all-in-one vectors pAF112 (hygromycin resistance and luciferase) and pAF123 (hygromycin resistance) were assembled from the pAF111 components. pAF112 was assembled by amplifying the Luc2 luciferase gene (insert 1) from pSBtet-Hyg and the P2A-hygromycin resistance gene (insert 2) from pSBbi-BH and inserting into the pAF111 Bsu36 I digest using NEBuilder. pSBbi-Hyg was Bsu36 Idigested to obtain the hygromycin resistance gene, and this fragment was inserted into Bsu36 Idigested pAF111 using T4 ligase cloning to replace the BFP-P2A-hygromycin segment in pAF111.

All full-length gene coding sequences except CTLA4 were cloned into the pAF112 Sfi I digest (table S2). All full-length vectors also contain luciferase with T2A peptide linked to the hygromycin resistance protein; luciferase was included for use in downstream functional testing that was not part of this study. Tasmanian devil CTLA4 was cloned into a NotI-HF and Xma I digest of pAF100 that was used in a different study but is derived from vectors pAF112 and pAF138. In addition, we also used devil PDL1 (CHO.pAF48) and 41BBL (CHO.pAF56) cell lines developed using a vector system described previously (17).

Plasmids containing fluorescent protein coding sequences mCerulean3-N1 (Addgene no. 54730), mAzurite-N1 (Addgene no. 54617), mOrange-N1 (Addgene no. 54499), and mNeptune2-N1 (Addgene no. 54837) were gifts to Addgene from M. Davidson. mTag-BFP was amplified from pSBbi-BH, mCitrine was amplified from pAF71, and mCherry was amplified from pTRE-Dual2 (Clontech no. PT5038-5). pAF137 was constructed by amplifying the devil 41BB extracellular domain with primers pAF137-1.FOR and pAF137-1.REV and amplifying mCherry with pAF137-2a.FOR and pAF137-2.REV (tables S3 and S4). 5 extensions on pAF137-1.FOR and pAF137-2.REV were used to create overlaps for NEBuilder assembly of pAF137 from a pAF123 Sfi Idigested base vector. 3 extensions on pAF137-1.REV and pAF137-2a.FOR were used to create the linker that included an Xma I/Sma I restriction site, TEV cleavage tag, GSAGSAAGSGEF linker peptide, and 6xHis tag between the gene of interest and fluorescent reporter. The GSAGSAAGSGEF was chosen because of the low number of large hydrophobic residues and less repeated nucleic acids than are needed with other flexible linkers such as (GGGS)4. The pAF137 primer extensions also created 5 Not I and 3 Nhe I sites in the FAST vector to facilitate downstream swapping of functional genes and to create a Kozak sequence (GCCGCCACC) upstream of the FAST protein open-reading frame. Following confirmation of correct assembly via DNA sequencing, the FAST 41BB-mCherry (pAF137) was digested and used as the base vector (Fig. 2B and fig. S1, A and B) for development of FAST vectors with alternative fluorescent proteins. This was accomplished by digestion of pAF137 with Sal I and Nhe I and then inserting PCR-amplified coding sequences for other fluorescent proteins using NEBuilder (tables S3 and S4).

Type I FAST (extracellular N terminus and cytoplasmic C terminus) protein vectors were constructed by digestion of 41BB FAST vectors with Not I and either Xma I or Sma I (Fig. 2B and fig. S1, A and B) and then inserting genes of interest (tables S2 to S4). To create an Fc-tagged FAST protein, we fused the extracellular domain of devil CD80 to the Fc region of the devil IgG (fig. S1C). The Fc region was amplified from a devil IgG plasmid provided by L. Corcoran (Walter and Eliza Hall Institute of Medical Research). All secreted FAST proteins in this study used their native signal peptides, except for 41BBL. 41BBL is a type II transmembrane protein in which the signal peptide directly precedes the cytoplasmic and transmembrane domains of the protein (cytoplasmic N terminus and extracellular C terminus). As type I FAST vectors cannot accommodate this domain architecture, we developed an alternative base vector for type II transmembrane FAST proteins (fig. S1D). To increase the probability of efficient secretion of type II FAST proteins from CHO cells, we used the hamster interleukin-2 (IL-2) signal peptide (accession no. NM_001281629.1) at the N terminus of the protein, followed by a Sal I restriction site, mCherry, an Nhe I restriction site, 6xHis tag, GSAGSAAGSGEF linker, TEV cleavage site, Xma I/Sma I restriction site, the gene of interest, and a Pme I restriction site following the stop codon.

Following transformation of assembled plasmids, colony PCR was performed as an initial test of the candidate plasmids. Single colonies were inoculated directly into a OneTaq Hot Start Quick-Load 2X Master Mix (NEB no. M0488) with primers pSB_EF1a_seq.FOR (atcttggttcattctcaagcctcag) and pSB_bGH_seq.REV (aggcacagtcgaggctgat). PCR was performed with 60C annealing temperature for 25 to 35 cycles. Colonies yielding appropriate band sizes were used to inoculate Luria broth with ampicillin (100 g/ml) for bacterial outgrowth overnight at 37C and 200 rpm. The plasmids were purified using standard plasmid kits and prepared for Sanger sequencing using the BigDye Terminator v3.1 Cycle Sequencing Kit (Thermo Fisher Scientific no. 4337455) with pSB_EF1a_seq.FOR and pSB_bGH_seq.REV primers. The BigDye Terminator was removed using Agencourt CleanSEQ (Beckman Coulter no. A29151) before loading samples to a 3500xL Genetic Analyzer (Applied Biosystems) for sequencing by fluorescence-based capillary electrophoresis.

DFT1 cell line C5065 and DFT2 cell line JV were cultured at 35C with 5% CO2 in cRF10 [10% complete RPMI (Gibco no. 11875-093) with 2 mM l-glutamine, supplemented with 10% heat-inactivated fetal bovine serum, and 1% antibiotic-antimycotic (Thermo Fisher Scientific no. 15240062)]. RPMI without phenol red (Sigma-Aldrich no. R7509) was used to culture FAST protein cell lines when supernatants were collected for downstream flow cytometry assays. Devil peripheral blood cells were cultured in cRF10 at 35C with 5% CO2. CHO cells were cultured at 37C in cRF10 during transfections and drug selection but were otherwise cultured at 35C in cRF5 (5% complete RPMI). For production of purified recombinant proteins, stably transfected CHO cells were cultured in suspension in spinner flasks in chemically defined, serum-free CHO EX-CELL (Sigma-Aldrich no. 14361C) media supplemented with 8 mM l-glutamine, 10 mM Hepes, 50 M 2-ME, 1% (v/v) antibiotic-antimycotic, and 1 mM sodium pyruvate and without hygromycin.

Stable transfections of CHO and DFT cells were accomplished by adding 3 105 cells to each well in six-well plates in cRF10 and allowing the cells to adhere overnight. The next day, 2 g of plasmid DNA was added to 100 l of phosphate-buffered saline (PBS) in microfuge tubes. Polyethylenimine (PEI) (linear, molecular weight, 25,000; Polysciences no. 23966-2) was diluted to 60 g/ml in PBS and incubated for at least 2 min. The PEI solution (100 l) was added to the 100 l of plasmid DNA in each tube to achieve a 3:1 ratio of PEI:DNA. The solution was mixed by gentle pipetting and incubated at room temperature for 15 min. While the solution was incubating, the media on the CHO cells were replaced with fresh cRF10. All 200 l from each DNA:PEI mix was then added dropwise to the CHO cells and gently rocked side to side and front to back to evenly spread the solution throughout the well. The plates were then incubated overnight at 37C with 5% CO2. The next day, the plates were inspected for fluorescence, and then the media were removed and replaced with cRF10 containing hygromycin (1 mg/ml) (Sigma-Aldrich no. H0654). The media were replaced with fresh cRF10 hygromycin (1 mg/ml) every 2 to 3 days for the next 7 days until selection was complete. The cells were then maintained in hygromycin (0.2 mg/ml) in cRF5 at 35C with 5% CO2. Supernatant was collected 2 to 3 weeks after transfection and stored at 4C for 2 months to assess stability of secreted FAST proteins.

Sixteen days after transfection, the first batch of FAST protein cell lines was adapted to a 1:1 mix of cRF5 and chemically defined, serum-free CHO EX-CELL media for 1 to 2 days to facilitate adaptation of the adherent CHO cells to suspension culture in serum-free media. At least 5 107 cells were then transferred to ProCulture spinner flasks (Sigma-Aldrich no. CLS45001L and no. CLS4500250) and stirred at 75 rpm at 35C in 5% CO2 on magnetic stirring platforms (Integra Bioscience no. 183001). Cells were maintained at a density ranging from 5 105 to 2 106 cells/ml for 8 to 14 days. Supernatant was collected every 2 to 3 days, centrifuged at 3200 relative centrifugal force (RCF) for 10 min, stored at 4C, and then purified using the KTA start protein purification system (GE Life Sciences no. 29022094). The supernatant was diluted 1:1 with 20 mM sodium phosphate (pH 7.4) and then purified using HisTrap Excel columns (GE Life Sciences no. 17-3712-05) according to the manufacturers instructions. Samples were passed through the columns using a flow rate of 2 ml/min at 4C; all wash and elution steps were done at 1 ml/min. Elution from HisTrap columns (GE Life Sciences no. 17-3712-05) was accomplished using 0.5 M imidazole and fractionated into 1-ml aliquots using the Frac30 fraction collector (GE Life Sciences no. 29023051). Fluorescence of FAST proteins was checked via brief excitation (Fig. 2) on a blue light transilluminator with an amber filter unit. In the case of mCherry, chromogenic color was visible without excitation. Fractions containing target proteins were combined and diluted to 15 ml with cold PBS, dialyzed (Sigma-Aldrich no. PURX60005) in PBS at 4C, 0.22-m sterile-filtered (Millipore no. SLGV033RS), and concentrated using Amicon Ultra centrifugal filter units (Sigma-Aldrich no. Z706345). The protein concentration was quantified using the 280-nm absorbance on a NanoDrop spectrophotometer. Extinction coefficients using for each protein were calculated using the ProtParam algorithm (48). The proteins were then aliquoted into microfuge tubes and frozen at 80C until further use. The CTLA4-Fc-mCherry protein was designed, assembled, and tested separately from the other FAST proteins and was tested directly in supernatant without purification.

CHO cells expressing full-length proteins were thawed in cRF10 and then maintained in cRF5 with hygromycin (0.2 mg/ml). The adherent CHO cells were washed with PBS and incubated with trypsin for 5 min at 37C to remove cells from the culture flask. Trypsin was diluted five times with cRF5 and centrifuged at 200 RCF for 5 min. Cells were resuspended in cRF5, counted (viability >95% in all cases), and resuspended and aliquoted for assays as described below.

Supernatants (cRF5) were collected from CHO cells expressing devil 41BB extracellular domain fused to either mCherry (pAF137), mCitrine (pAF138), mOrange (pAF164), mBFP (pAF139), mAzurite (pAF160), mCerulean3 (pAF161), or mNeptune2 (pAF163) (tables S2 to S4). The supernatant was spun for 10 min at 3200 RCF to remove cells and cellular debris and then stored at 4C until further use. CHO cells expressing devil 41BBL (CHO.pAF56) and untransfected CHO cells were prepared as described above. Flow cytometry tubes were loaded with 5 104 target CHO cells per well in cRF5, centrifuged 500 RCF for 3 min, and then resuspended in 200 l of supernatant from the 41BB FAST cell lines (N = 1 per treatment). The tubes were then incubated for 15 min at 4C, centrifuged at 500 RCF for 3 min, resuspended in 400 l of cold fluorescence-activated cell sorting (FACS) buffer, and stored on ice until the data were acquired on a Beckman Coulter Astrios flow cytometer (Fig. 2C). All flow cytometry data were analyzed using FCS Express 6 Flow Cytometry Software version 6 (De Novo Software).

U-bottom 96-well plates were loaded with 1 105 target CHO cells per well in cRF5, centrifuged 500 RCF for 3 min, and then resuspended in 175 l of cRF5 supernatant from FAST cell lines collected 11 days after transfection (N = 1 per treatment). The plates were then incubated for 30 min at room temperature, centrifuged at 500 RCF for 3 min, resuspended in 200 l of cold FACS buffer, centrifuged again, and fixed with FACS fix buffer [PBS, 0.02% NaN3, 0.4% formalin, glucose (10 g/liter)]. The cells were transferred to tubes, diluted with FACS buffer, and analyzed on a Beckman Coulter Astrios flow cytometer (fig. S2).

Purified FAST proteins were diluted to 20 g/ml in cRF5, aliquoted into V-bottom 96-well transfer plates, and then stored at 37C until target cells were ready for staining. Target cells were resuspended in cRF5 with 100 M chloroquine, and 100,000 cells per well were aliquoted into U-bottom 96-well plates. One hundred microliters of the diluted FAST proteins (N = 1 per treatment, two time points per treatment) was then transferred from the V-bottom plates into the U-bottom 96-well plates containing target cells. The final volumes and concentrations were 200 l per well in cRF5 with 50 M chloroquine and 2 g per well of FAST proteins. One set of plates was incubated at 37C for 30 min, and another set of plates was incubated at 37C overnight. The cells were then centrifuged 500 RCF for 3 min, the media decanted, and incubated for 5 min with 100 l of trypsin to dislodge adherent cells. The cells were then washed with 200 l of cold FACS buffer, fixed, resuspended in cold FACS buffer, and transferred to tubes for analysis on the Astrios flow cytometer (Fig. 3B).

The protocol for using FAST protein supernatants was the same above as the preceding experiment except for the modifications described here. Supernatants were collected 2 to 3 weeks after transfection, centrifuged at 3200 RCF for 10 min, and stored at 4C for 2 months. Before staining for flow cytometry, the supernatant was 0.22-m filtered. Supernatant was then loaded into V-bottom 96-well plates to facilitate rapid transfer to staining plates and stored at 37C until target cells were ready for staining. Target cells were prepared as described above except for being diluted in cRF5 with 100 M chloroquine. A total of 2 105 cells per well (100 l) were then loaded into U-bottom 96-well plates. One hundred microliters of FAST protein supernatant (N = 1 per treatment) was then transferred from the V-bottom plates to achieve 50 M chloroquine, and the cells were then incubated at 37C for 60 min. The plates were then washed, fixed, and analyzed on the Astrios flow cytometer (fig. S3). A similar procedure was used for staining stably transfected DFT cells with CTLA4-Fc-mCherry, except that the supernatant was used fresh (Fig. 4D).

CHO cells expressing full-length CTLA4 with a C-terminal mCitrine and CHO cells expressing full-length 41BB or 41BBL were labeled with 5 M CFSE; CFSE and mCitrine were analyzed using the same excitation laser (488 nm) and emission filters (513/26 nm). A total of 1 105 FAST proteinsecreting cells were mixed with 1 105 target cells in cRF5 with 50 M chloroquine and incubated overnight at 37C in 96-well U-bottom plates (Fig. 4A). The next day, the cells were rinsed with PBS, trypsinized, washed, fixed, and resuspended in FACS buffer before running flow cytometry. Cells were gated on forward and side scatter (FSC SSC) and for singlets (FSC-H FSC-A) (Fig. 4B). Data shown in Fig. 4C are representative of N = 3 technical replicates per treatment. Data were collected using a Beckman Coulter MoFlo Astrios and analyzed using FCS Express.

RNA-seq data were generated during previous experiments, aligned against the reference Tasmanian devil genome Devil_ref v7.0 (GCA_000189315.1), and summarized into normalized read counts as previously described (25, 26). Transcripts per millionnormalized read counts were calculated in R, and a heat map was produced from log2-converted values using the heatmap.2 function of gplots.

A total of 50,000 DFT cells per well were aliquoted into U-bottom 96-well plates, washed with 150 l of cRF10, and resuspended in 100 l of warm cRF10 containing 100 M chloroquine. Five micrograms of FAST protein per well was then added and mixed by pipetting. The plates were then incubated at 37C for 30 min. The cells were then transferred to microfuge tubes without washing, stored on ice, and analyzed on a Beckman Coulter MoFlo Astrios (N = 2 per treatment).

CD200 and 41BB FAST proteins were digested overnight with TEV protease (Sigma-Aldrich no. T4455) at 4C in PBS. The cleaved linker and 6xHis tag were then removed using a His SpinTrap kit (GE Healthcare no. 28-9321-71). Digested proteins in PBS were diluted 1:1 in Squalvax (OZ Biosciences no. SQ0010) to a final concentration of 0.1 g/l and were mixed using interlocked syringes to form an emulsion. Immunization of BALB/c mice for antibody production was approved by the University of Tasmania Animal Ethics Committee (no. A0014680). Preimmune sera were collected before subcutaneous immunization with at least 50 l of the emulsion. On day 14 after immunization, the mice were boosted using a similar procedure. On day 50, the mice received a booster with proteins in IFAVax (OZ Biosciences no. IFA0050); mice immunized with CD200 again received subcutaneous injections, whereas 41BB mice received subcutaneous and intraperitoneal injections. Preimmune and sera collected after three-times immunizations were then tested by flow cytometry against CHO cells expressing either 41BB or CD200. CHO cells were prepared as described above, and 2 105 cells were incubated with mouse serum diluted 1:200 in PBS for 30 min at 4C. The cells were then washed two times and stained with 50 l of anti-mouse IgG AF647 diluted 1:1000 in FACS buffer. The cells were then washed two times, stained with 4,6-diamidino-2-phenylindole (DAPI) to identify live cells, and analyzed on a CyAn ADP flow cytometer (Fig. 5C). CD200 and 41BB expression on DFT cells was tested using a procedure similar to the CHO cell staining, except that the sera used were collected after four-times immunizations and was diluted 1:500 and analyzed on the BD FACSCanto II (Fig. 5D).

Approximately, 200 l of NMS or anti-CD200 serum day 157 (after four-times immunizations) was purified using an Ab SpinTrap (GE Healthcare no. 28-4083-47) according to the manufacturers instructions. Serum was diluted 1:1 with 20 mM sodium phosphate and binding buffer (pH 7.0) and eluted with 0.1 M glycine-HCl (pH 2.7), and the pH was neutralized with 0.1 M glycine-HCl (pH 2.7). The eluted antibodies were then concentrated using an Amicon Ultra 0.5 centrifugal unit (Merck no. UFC500308) by centrifuging at 14,000 RCF for 30 min at 4C and then washing the antibodies with 400 l of PBS twice. The protein concentration was then quantified on a NanoDrop spectrophotometer at 280 nm using the extinction coefficients for IgG.

A total of 50,000 DFT cells per well were aliquoted into U-bottom 96-well plates and washed with 200 l of cold FACS buffer. Purified polyclonal anti-CD200 was diluted to 2.5 g/ml in cold FACS buffer, and the cells in appropriate wells were resuspend in 100 l per well (0.25 g per well) diluted antibody; wells that did not receive antibody were resuspended in 100 l of FACS buffer. The cells were incubated on ice for 20 min and then washed with 200 l of FACS buffer. While incubating, anti-mouse IgG AF647 was diluted to 1 g/ml in cold FACS buffer and then used to resuspend cells in the appropriate wells. The plates were incubated on ice for 20 min and then washed with 100 l of cold FACS buffer. The cells were then resuspended in 200 l of FACS fix and incubated on a rocking platform at room temperature for 15 min. The cells were then centrifuged 500 RCF for 3 min at 4C, resuspended in 200 l of FACS buffer, and stored at 4C until they were analyzed on a FACSCanto II (N = 2 per treatment) (Fig. 5E).

Blood collection from Tasmanian devils was approved by the University of Tasmania Animal Ethics Committee (permit no. A0014599) and the Tasmanian Department of Primary Industries, Parks, Water and Environment. Blood was collected from the jugular vein and stored in EDTA tubes for transport to the laboratory. Blood was processed within 3 hours by diluting 1:1 with serum-free RPMI and then layering onto Histopaque (Sigma-Aldrich no. 10771) before centrifuging at 400 RCF for 30 min. The interface containing the PBMCs was then collected using a transfer pipette, diluted with 50 ml of serum-free RPMI, and centrifuged for 5 min at 500 RCF. Cells were washed with again with cRF10 and then either used fresh or stored at 80C until further use.

Frozen devil PBMC was thawed and cultured in cRF10 at 35C with 5% CO2 for 2 hours; cells were then washed in FACS buffer and counted, and 3 105 PBMCs were used per sample. DFT2.JV cells were removed from culture flasks and counted, and 2 105 cells were used per sample. Samples were incubated with 50 l of normal goat serum (Thermo Fisher Scientific, catalog no. 01-6201) diluted 1:200 in FACS buffer for 15 min at 4C, and 50 l of anti-CD200 serum diluted 1:100 was added (1:200 final) for 30 min at 4C. Cells were then washed two times and stained with 50 l of anti-mouse IgG AF647 diluted to 1 g/ml in FACS buffer for 30 min at 4C. The cells were then washed two times, stained with DAPI (Sigma-Aldrich, catalog no. D9542) to identify live cells, and analyzed on the BD FACSCanto II. PBMC and DFT cells were run separately, and then PBMC and DFT2 were mixed at a ratio of 10:1 by volume for the combined samples (N = 1 per treatment) (fig. S4A). The experiment was repeated (N = 1 per treatment), except that PBMCs and DFT cells were mixed at a 5:1 ratio (fig. S4B).

DFT1.C5065 and DFT2.JV cells were labeled with 5 M CTV and cultured for 3 days at 37C. On the day of the assays, peripheral blood from one devil was collected and stored at ambient temperature for less than 3 hours. One hundred microliters of whole blood was aliquoted into 15-ml tubes and stored at ambient temperature while DFT cells were prepared. The media on CTV-labeled DFT cells were decanted, and the cells were detached from the flask by incubating in 2.5 ml of TrypLE Select for 5 min at 37C. The cells were washed with cRF10, resuspended in cRF10, and counted. DFT cells were then diluted to 1 104 cells/ml in cRF10, and 100 l was aliquoted into appropriate 15-ml tubes containing 100 l of whole blood. One microliter of purified anti-CD200 (0.5 g per tube) was diluted into the appropriate tubes and incubated for 15 min at ambient temperature. Next, anti-mouse IgG AF647 (0.5 g per tube) was added to each tube. Note: 0.5 l (0.5 g) of concentrated secondary antibody was accidentally added directly to the tube for the data shown in the top row, middle column of fig. S5A; for all other tubes, the secondary antibody was diluted 1:20 in PBS and 10 l was added to each tube. The cells were then incubated for 15 min at ambient temperature. The cells were then diluted in 1 ml of ammonium chloride RBC lysis buffer [150 mM NH4Cl, 10 mM KHCO3, and 0.1 mM EDTA disodium (Na2-2H2O)] and mixed immediately gently pipetting five times. The cells were incubated at ambient temperature for 10 min, then diluted with 5 ml of PBS, and centrifuged 500 RCF for 3 min. Some tubes contained residual RBCs, so the pellet was vigorously resuspended in 5 ml of RBC lysis buffer, incubated for 5 min, diluted with 5 ml of cold FACS buffer, and centrifuged 500 RCF for 3 min. The cells were then resuspended in 250 l of FACS buffer and stored on ice until analysis on a Beckman Coulter MoFlo Astrios (N = 1 per treatment). Data were analyzed in FCS Express version 6 (fig. S5).

The experiment above was repeated with the following modifications. DFT cells were labeled with 5 M CFSE and incubated for 2 days at 37C. On the day of the assays, fresh blood was collected from two devils. Purified anti-CD200 and NMS were labeled with Zenon mouse IgG AF647 (Thermo Fisher Scientific no. Z25008) and blocked with the Zenon blocking agent. A total of 1 104 CFSE-labeled DFT cells were diluted directly into 100 l of whole blood in 15-ml tubes, and 12 l (2-l antibody, 5-l labeling agent, and 5-l blocking agent) of Zenon AF647labeled purified NMS or anti-CD200 was added directly to the cells. The cells were incubated for 30 min at ambient temperature. The cells were then gently resuspended in 2.5 ml of RBC lysis buffer and incubated for 10 min at ambient temperature. The cells were diluted with 10 ml of PBS and centrifuged 500 RCF for 3 min. The cells were resuspended in 1.5 ml of RBC lysis buffer and incubated for another 10 min to lyse residual RBCs. The tubes were then resuspended in 9 ml of cRF10 and centrifuged 500 RCF for 3 min. The cells were resuspended in 350 l of cold FACS buffer containing DAPI (200 ng/ml) and stored on ice until analysis on a Beckman Coulter MoFlo Astrios (N = 1 per treatment for n = 2 devils) (Fig. 6, A to D).

The anti-human PD-L1 nanobody (KN035) (24) protein sequence was reverse-translated and as a double-stranded DNA gBlock (Integrated DNA Technologies) (table S5). The sequence was modified to include DNA extension that overlaps FAST vectors. The signal peptide from hamster IL-2 (also in pAF92) was added to the nanobody to increase secretion efficiency in CHO cells. The gBlock was inserted into a NotI-HF and Sma Idigested mCitrine FAST vector with NEB HiFi DNA Assembly Master Mix (NEB no. E2621). Transformation, purification of plasmid DNA, and sequencing were performed as described above.

ExpiCHO cells (Thermo Fisher Scientific no. A29127) for high-yield protein production were maintained at 37C with 8% CO2 with constant shaking at 200 rpm in ExpiCHO Stable Production Medium (SPM) (Thermo Fisher Scientific no. A3711001). ExpiCHO cells were added to a six-well plate at 3 105 cells per well in ExpiCHO SPM and cultured overnight. The next day, 2-g plasmid DNA was added to 100 l of PBS in a microcentrifuge tube. PEI was diluted to 60 g/ml in PBS and incubated at room temperature for 5 min. Diluted plasmid DNA was added to 100 l of PEI solution to achieve a 3:1 PEI:DNA ratio and incubated at room temperature for 15 min. During this time, ExpiCHO cells were transferred to 15-ml centrifuge tubes, washed with PBS at 300g for min, resuspended in 3-ml OptiPRO serum-free media (Thermo Fisher Scientific no. 12309019), and returned to the six-well plate. The PEI:DNA solution was then added directly to cells and incubated overnight. The next day, plates were inspected for fluorescence, and the media were removed and replaced with ExpiCHO SPM supplemented with hygromycin (1 mg/ml). Media were changed every second day until selection was complete. Once selection was complete, the cells were moved to 50-ml TPP TubeSpin bioreactor tubes (Sigma-Aldrich no. Z761028) and maintained at 4 106 to 6 106 cells/ml in ExpiCHO SPM with hygromycin (0.2 mg/ml). Supernatant was collected 2 weeks after transfection and stored at 4C.

CHO cells expressing either human PD-L1 or human CD80 fused to miRFP670 (table S3) were plated at 100,000 cells per well into a U-bottom 96-well plate and centrifuged at 300g for 5 min, and the supernatant was discarded. Two hundredmicroliter supernatant containing secreted PD-L1 nanobody was added to CHO cell lines either neat or diluted in 1:10 and 1:100 in FACS buffer. Cells were incubated at 4C for 30 min before being washed in FACS buffer for analysis on a Beckman Coulter FACSCanto II (Fig. 6F).

N. Howlader, A. M. Noone, M. Krapcho, D. Miller, A. Brest, M. Yu, J. Ruhl, Z. Tatalovich, A. Mariotto, D. R. Lewis, H. S. Feuer, K. A. Cronin, SEER Cancer Statistics Review, 19752016 (National Cancer Institute, 2016).

E. Gasteiger, C. Hoogland, A. Gattiker, S. Duvaud, M. R. Wilkins, R. D. Appel, A. Bairoch, in The Proteomics Protocols Handbook, J. M. Walker, Ed. (Humana Press, ed. 1, 2005), pp. 571607.

View original post here:
A novel system to map protein interactions reveals evolutionarily conserved immune evasion pathways on transmissible cancers - Science Advances