Category Archives: Cell Biology

Global Spatial Genomics and Transcriptomics Market: Focus on Product Type, Sample Type, Workflow, Application, End User, Region and Competitive…

New York, Nov. 25, 2020 (GLOBE NEWSWIRE) -- Reportlinker.com announces the release of the report "Global Spatial Genomics and Transcriptomics Market: Focus on Product Type, Sample Type, Workflow, Application, End User, Region and Competitive Landscape - Analysis and Forecast, 2020-2030" - https://www.reportlinker.com/p05989635/?utm_source=GNW Market Segmentation

Product Type Kits and Assays, Instruments, Software, and Services Sample Type Fresh Frozen Tissues, Fixed Frozen Tissues, Fixed-Formalin Paraffin Embedded Tissues, and Cultured Cells Workflow Spatial Imaging, Spatial Analysis, and Spatial Sequencing Application- Diagnostics, Drug Discovery and Development, Translational Research, Single-Cell Analysis, Cell Biology and Others End User Academic and Research Institutions, Biopharmaceutical and Biotechnological Companies, Contract Research Organizations and Other End Users

Regional Segmentation

North America U.S., Canada Europe Germany, France, Italy, U.K., Spain, and Rest-of-Europe Asia-Pacific China, Japan, India, Singapore, Australia, and Rest-of-Asia-Pacific (RoAPAC) Latin America Brazil, Mexico, and Rest-of-the-Latin America Rest-of-the-World

Growth Drivers

Increasing Prevalence of Various Types of Genetic Disorders Globally Technological Advancements in Sequencing Technologies Increasing Research Funding in the Field of Spatial Transcriptomics

Market Challenges

High Capital Requirement Hampering the Expansion of Global Reach Lack of Tools for Computational Analysis

Market Opportunities

Opportunity (by Product) Opportunity (by Technology) Expansion into New Research Application such as Spatial Metagenomics Expansion into Emerging Markets

Key Companies ProfiledNanoString Technologies, Inc., S2 Genomics, Inc., Flagship Biosciences, Inc., Akoya Biosciences, Inc. RareCyte, Inc., IONpath, Inc., Fluidigm Corporation, 10x Genomics, Inc., Visikol, Inc., Miltenyi Biotec, and BioSpyder Technologies

Key Questions Answered in this Report: What are the major market drivers, challenges, and opportunities in the global spatial genomics and transcriptomics market? What is the potential impact of biotechnological advancement in the diagnostic industry among the end users, such as researchers, pathologists, and laboratory technicians? What is the current market demand along with future expected demand for the global spatial genomics and transcriptomics market? How have spatial profiling helped cellular imaging and visualization to become a prominent tool for diagnostics in various clinical applications? What are the key development strategies which are implemented by the major players in order to sustain in the competitive market? How is each segment of the market expected to grow during the forecast period from 2020 to 2030 based each on segment?

Following are each segment type:o product typeo sample typeo workflowo applicationo end usero region, namely, North America, Europe, Asia-Pacific, Rest-of-the-World (ROW) Who are the leading players with significant offerings to the global spatial genomics and transcriptomics market? What is the expected market dominance for each of these leading players? Which emerging companies are anticipated to be highly disruptive in the future, and what are their key strategies for sustainable growth in the global spatial genomics and transcriptomics market?

Market OverviewHealthcare experts have found the spatial genomics and transcriptomics market to be one of the most rapidly evolving markets, which is predicted to grow at a CAGR of 23.20% during the forecast period of 2020-2030. The market is driven by certain factors, which include the increasing prevalence of various types of genetic disorders, such as cancer, neurological disorder, and rare diseases, inciting the development of high-resolution multiplex assays and instruments, technological advancements in sequencing technologies, and significant research funding in the field of spatial-based technology for executing R&D exercises.

The market is favored by the development of spatial profiling-based solutions for visualization and analysis of tissue microenvironment, tumor biology, and tissue biomarker. The gradual increase in the prevalence of neurological disorders and rare diseases globally has furthered the spatial genomics and transcriptomics market.

Furthermore, several contract research organizations are focusing on the development of spatial profiling-based services, which enables simultaneous in-situ spatial analysis of multiple biomarkers proteins or more than a hundred mRNAs from single formalin-fixed paraffin-embedded (FFPE) tissue or frozen tissue section.

Within the research report, the market is segmented on the basis of product type, sample type, workflow, application, end users, and region. Each of these segments covers the snapshot of the market over the projected years, the inclination of the market revenue, underlying patterns, and trends by using analytics on the primary and secondary data obtained.

Competitive LandscapeThe exponential rise in the application of precision medicine on the global level has created a buzz among companies to invest in the development of high-resolution multiplex diagnostics providing information on cellular interaction and tissue heterogeneity to understand disease biology and pathology. Due to technologically advanced solutions and intense market penetration, BioTechne Corporation has been a pioneer in this field and has been a significant competitor in this market.

Other key players in the market are NanoString Technologies, Inc., S2 Genomics, Inc., Flagship Biosciences, Inc., Akoya Biosciences, Inc. RareCyte, Inc., IONpath, Inc., Fluidigm Corporation, 10x Genomics, Inc., Visikol, Inc., Miltenyi Biotec, and BioSpyder Technologies.

On the basis of region, North America holds the largest share of spatial genomics and transcriptomics market due to improved healthcare infrastructure, rise in per capita income, and availability of state-of-the-art research laboratories and institutions in the region. Apart from this, Europe region is anticipated to grow at the fastest CAGR of 23.54% during the forecast period 2020-2030.

The market utilizes several technologies, such as barcoding, sequencing, mass cytometry, and microscopy, for the development of instruments and assay for spatial profiling of tissue section to gain an understanding of tissue microenvironment. Each solution offered by the leading players is the combination of next-generation omics tools for application in several clinical areas, such oncology, neurology, immunology, and pathology.

Countries Covered North America U.S. Canada Europe Germany France Spain U.K. Italy Rest-of-Europe Asia-Pacific China India Singapore Australia Japan Rest-of-Asia-Pacific Latin America Brazil Mexico Rest-of-Latin America Rest-of-the-WorldRead the full report: https://www.reportlinker.com/p05989635/?utm_source=GNW

About ReportlinkerReportLinker is an award-winning market research solution. Reportlinker finds and organizes the latest industry data so you get all the market research you need - instantly, in one place.

__________________________

Here is the original post:
Global Spatial Genomics and Transcriptomics Market: Focus on Product Type, Sample Type, Workflow, Application, End User, Region and Competitive...

Severe infections wreak havoc on mouse blood cell production | Imperial News – Imperial College London

Severe infections like malaria cause short and long-term damage to precursor blood cells in mice, but some damage could be reversed, find researchers.

A team led by researchers from Imperial College London and The Francis Crick Institute have discovered that severe infections caused by malaria disrupt the processes that form blood cells in mice. This potentially causes long-term damage that could mean people who have recovered from severe infections are vulnerable to new infections or to developing blood cancers.

The team also discovered that the damage could be reduced or partially reversed in mice with a hormone treatment that regulates bone calcium coupled with an antioxidant. The research could lead to new ways of preventing long-term damage from severe infections including malaria, TB and COVID-19.

The research is published today in Nature Cell Biology.

First author Dr Myriam Haltalli, who completed the work while at the Department of Life Sciences at Imperial, said: We discovered that malaria infection reprograms the process of blood cell production in mice and significantly affects the function of precursor blood cells. These changes could cause long-term alterations, but we also found a way to significantly reduce the amount of damage and potentially rescue the healthy production of blood cells.

Blood is made up of several different cell types, that all originate as haematopoietic stem cells (HSCs) in the bone marrow. During severe infection, the production of all blood cells ramps up to help the body fight the infection, depleting the HSCs.

Now, the team has shown how infections also damage the bone marrow environment that is crucial for healthy HSC production and function. They discovered this using advanced microscopy technologies at Imperial and the Crick, RNA analyses led by the Gottgens group at Cambridge University, and mathematical modelling led by Professor Ken Duffy at Maynooth University.

The mice developed malaria naturally, following bites from mosquitoes carrying Plasmodium parasites, provided by Dr Andrew Blagborough at Cambridge University. The researchers subsequently observed the changes in the bone marrow environment and the effect on HSC function.

Within days of infection, blood vessels became leaky and there was a dramatic loss in bone-forming cells called osteoblasts. These changes appear strongly linked to the decline in the pool of HSCs during infection.

Lead author Professor Cristina Lo Celso, from the Department of Life Sciences at Imperial, said: We were surprised at the speed of the changes, which was completely unexpected. We may think of bone as an impenetrable fortress, but the bone marrow environment is incredibly dynamic and susceptible to damage.

Reducing the pool of HSCs can have several consequences. In the short-term, it appears to particularly affect the production of neutrophils white blood cells that form an essential part of the immune system. This can leave patients vulnerable to further infections, with potentially long-term consequences for the functioning of the immune system.

In the long term, the pool of HSCs may remain below normal levels, which can increase the chances of the patient developing blood cancers like leukaemia.

By injecting fluorescent molecules (magenta) that would normally remain in circulation and taking a series of images over time, intravital microscopy revealed that infected mice had very leaky vessels with the contents of the bone marrow blood vessels, lined by endothelial cells (green), escaping into the surrounding tissue. The red boxes highlight the areas compared in the analysis and the white lines mark the bone.

After determining in detail how severe infection affects the bone marrow environment and HSC function, the team tested a way to prevent the damage. Before infecting the mice, they treated them with a hormone that regulates bone calcium and an antioxidant to counter cellular oxidative stress, and then again after infection.

This process led to a tenfold increase in HSC function following infection compared to mice that received no treatment (around 20-40 per cent function compared to two percent function, respectively). Although this is not a complete recovery, the vast increase in function is a positive sign.

The team note that the requirement to start the hormone treatment before infection, combined with its expense and need to be refrigerated, make it unviable as a solution, especially in many parts of the world where severe infections like malaria and TB are prevalent.

However, they hope that proof that the impact of severe infection on HSC function can be significantly lessened will lead to the development of new treatments that can be widely administered.

Professor Lo Celso said: The long-term impacts of COVID-19 infection are just starting to be known. The impact on HSC function appears similar across multiple severe infections, suggesting our work on malaria could shed light on the possible long-term consequences of COVID-19, and how we might mitigate them.

Dr Haltalli concluded: Protecting HSC function while still developing strong immune responses is key for healthy ageing.

-

Manipulating niche composition limits damage to haematopoietic stem cells during Plasmodium infection by Myriam L.R. Haltalli et al. is published in Nature Cell Biology.

Go here to read the rest:
Severe infections wreak havoc on mouse blood cell production | Imperial News - Imperial College London

Synthetic Biology Market projected to expand at a CAGR of 26.3% from 2019 to 2027 – Murphy’s Hockey Law

Synthetic Biology Market: Introduction

Transparency Market Research has published a new report titled, Synthetic Biology Market. According to the report, the globalsynthetic biology marketwas valued atUS$ 4.96 Bnin2018and is projected to expand at a CAGR of26.3%from2019to2027.

In terms of product, the core product segment accounted for major share of the global synthetic biology market in2018. The segment is anticipated to witness strong growth from2019to2027. The core product segment is further sub-segmented into synthetic DNA, synthetic genes, synthetic cells, XNA (xeno nucleic acid), and chassis organisms. The synthetic DNA sub-segment accounted for major share of the global synthetic biology market due to the increasing research & developmental activities associated to this sub-segment and increased penetration in the market.

Request Brochure for Report https://www.transparencymarketresearch.com/sample/sample.php?flag=B&rep_id=421

Request for Analysis of COVID19 Impact on Synthetic Biology Market

https://www.transparencymarketresearch.com/sample/sample.php?flag=covid19&rep_id=421

Based on technology, the genome engineering segment held a major share in2018in synthetic biology market, due to its ability to make alterations to the genome of the living cell, and thereby gaining attention of the scientists and key players.

Based on application, the health care segment held a prominent share in2018in synthetic biology market due to increase in prevalence of various diseases, rise in key players, and expanding infrastructure as well as increasing focus of government in treatments and facilities in health care

Buy Synthetic Biology Market Report https://www.transparencymarketresearch.com/checkout.php?rep_id=421&ltype=S

Global Synthetic Biology Market: Key Players

Contact

Transparency Market Research,

90 State Street, Suite 700,

Albany, NY 12207

Tel: +1-518-618-1030

USA Canada Toll Free: 866-552-3453

Website: https://www.transparencymarketresearch.com/

Read more:
Synthetic Biology Market projected to expand at a CAGR of 26.3% from 2019 to 2027 - Murphy's Hockey Law

Catamaran Bio Launches with $42 Million Financing to Develop OfftheShelf CAR-NK Cell Therapies to Treat Solid Tumors – BioSpace

In assembling the founding team at Catamaran, we saw an opportunity to pioneer a highly differentiated approach to develop allogeneic cell therapies using CAR-NK cells, said Houman Ashrafian, Managing Partner, SV Health Investors and a founder of Catamaran. To date, the success of autologous CAR T-cell therapies in hematological malignancies has opened the door to the breakthrough potential of cell therapies for cancer, and Catamaran is now well positioned to improve upon this groundwork by developing off-the-shelf CAR-NK cell therapies capable of reaching solid tumors.

A novel approach to developing off-the-shelf cell therapies to address solid tumors

Catamarans TAILWIND Platform integrates proprietary capabilities to create novel, allogeneic CARNK cell therapies by harnessing the natural cancer-fighting properties of natural killer (NK) cells and enhancing them with the power of synthetic biology and innovative NK cell engineering and manufacturing. With the TAILWIND Platform, CAR-NK cells are programmed with NK cell-specific CAR architectures and potency-boosting switches to neutralize the hostile tumor microenvironment and enable efficacy against diverse cancer types, especially solid tumors. Additionally, the TAILWIND Platform includes proprietary, non-viral NK cell engineering technology for efficient modification of NK cells with customized genetic programs enabled by synthetic biology. Catamarans CAR-NK cell therapies use healthy donor cells that are engineered and manufactured for offtheshelf use, unlike current CAR-T cell therapies that use a patients own genetically modified T cells and require a customized, multi-week manufacturing process.

Catamaran is focused on expanding the frontier of cell therapies to treat solid tumors and provide transformative benefit to cancer patients. We are doing this by creating allogeneic cell therapies that harness the innate cancer-fighting power of NK cells and enhancing them with new biologically-powerful attributes from our leading-edge technologies all originating from our custom-built TAILWIND Platform for designing, engineering and manufacturing off-the-shelf CAR-NK cell therapies, said Vipin Suri, PhD, MBA, Chief Scientific Officer of Catamaran.

During Catamarans stealth period, the start-up team assembled key components of the TAILWIND Platform and related intellectual property, including a set of potency-boosting cellular switches to enable therapeutic action in the immunosuppressive tumor microenvironment of solid tumors, and it generated early proof of concept using a non-viral transposon system to efficiently deliver large genetic cargos into NK cells. Based on this early work, the company has rapidly advanced two lead CAR-NK cell therapy programs to lead optimization stage.

With its holistic and cutting-edge approach, Catamaran stands out in the rapidly-evolving NK cell field with a platform that addresses the full complement of capabilities necessary to develop CAR-NK cell therapies, while focusing on the high-impact technologies of synthetic biology and innovative gene delivery systems that can enable these new cell therapies to offer extraordinary value in the field of cancer treatment, said Maina Bhaman, Partner, Sofinnova Partners.

Scientific founders and leadership team

Catamarans scientific founders are pioneers in NK cell biology, engineering, manufacturing and clinical application and are proven innovators in the cell therapy field:

Additional founders of Catamaran are Kevin Pojasek, PhD, and Tim Harris, PhD, through their roles as venture partners with SV Health Investors.

The leadership team at Catamaran Bio has deep expertise in cell therapy research and product development, and the team includes: Vipin Suri, PhD, MBA, Chief Scientific Officer, who has more than 20 years of biopharmaceutical experience, including as a co-founder of Obsidian Therapeutics and Serien (formerly Raze) Therapeutics, and earlier in R&D roles at GSK, Pfizer and Wyeth; Mark Boshar, JD, Chief Operating Officer, who has more than 25 years of leadership experience spanning legal, business development, financings and operations for biotechnology companies, including as VP, Legal Affairs at Rubius Therapeutics, Associate General Counsel at Millennium Pharmaceuticals, a senior advisor to a range of venture-backed start-up companies, and earlier as a life sciences attorney with WilmerHale; Chris Carpenter, MD, PhD, Chief Medical Officer, who has 20 years of clinical and laboratory experience in oncology, including as CMO of Rubius Therapeutics, SVP and Head of Cancer Epigenetics Discovery at GSK, and roles at Merck and Harvard Medical School/Beth Israel Deaconess Medical Center; Celeste Richardson, PhD, Senior VP of Research, who has 16 years of experience in research and drug discovery in biotechnology and pharmaceutical companies, including at Obsidian Therapeutics and Novartis; and Bharat Reddy, PhD, MPhil, MA, Senior Director of Business Development, who has served as director of business development at bluebird bio, as well as roles at SV Health Investors and ClearView Healthcare Partners.

Catamaran is positioned to open up new territory for cancer treatments with highly potent CAR-NK cell therapies, and we are confident in the experienced leadership team and the scientific expertise that is propelling the companys research and development, said Caroline Gaynor, Principal, Lightstone Ventures.

Concurrent with the Series A financing, Maina Bhaman of Sofinnova Partners, Caroline Gaynor of Lightstone Ventures and Rob Woodman of Takeda Ventures join Houman Ashrafian and Kevin Pojasek on the Catamaran board of directors.

About Catamaran Bio

Catamaran Bio is developing novel, off-the-shelf CAR-NK cell therapies designed to treat a broad range of cancers, including solid tumors. Our proprietary capabilities enable us to harness the natural cancer-fighting properties of NK cells and enhance and tailor their effectiveness with the power of synthetic biology and innovative non-viral cell engineering. We are using our TAILWINDTM Platform, an integrated suite of technologies, to specifically address the end-to-end methods of engineering, processing and manufacturing NK cells and rapidly advance our pipeline of CAR-NK cell therapy programs.

Our team combines experienced biopharmaceutical leadership with founding scientists who are pioneers in NK cell biology, engineering, manufacturing and clinical application. Catamaran is backed by leading financial and corporate investors, including SV Health Investors, Sofinnova Partners, Lightstone Ventures, Takeda Ventures and Astellas Venture Management. For more information, please visit http://www.catamaranbio.com and follow us on LinkedIn and @CatamaranBio on Twitter.

View source version on businesswire.com: https://www.businesswire.com/news/home/20201123005485/en/

View original post here:
Catamaran Bio Launches with $42 Million Financing to Develop OfftheShelf CAR-NK Cell Therapies to Treat Solid Tumors - BioSpace

Real Time Spying on the Symphony of Cellular Signals That Drive Biology – SciTechDaily

To visualize cellular signals within a neuron, researchers scattered reporters in clusters (green) across the cell. They then identified the signal each cluster represented (multiple colors).Credit: C. Linghu, S. Johnson et al./Cell 2020

A new imaging technology lets scientists spy on the flurry of messages passed within cells as they do . . . potentially everything.

Until now, most scientists could visualize only one or two of these intracellular signals at a time, says Howard Hughes Medical Institute InvestigatorEd Boyden of the Massachusetts Institute of Technology. His teams new approach could make it possible to see as many signals as you want in real time, at once, Boyden says giving researchers a more detailed view of cells internal discussions than ever before.

In tests with neurons, the researchers examined five signals involved in processes such as learning and memory, Boyden and his colleagues report today (November 23, 2020) in the journal Cell. You could apply this technology to all sorts of biological mysteries, he says. Every cell works due to all the signals inside it. Because signaling contributes to all biological processes, a better means to study it could illuminate a host of diseases, from Alzheimers to diabetes and cancer.

The teams new approach is a breakthrough, says Clifford Woolf, a neurobiologist at Harvard Medical School who was not involved with the work. He plans to use it to examine how pain-sensing neurons become more sensitive in injury or illness. With the new imaging technology, he says we can take apart whats happening in cells in a way that just has not been possible before.

Give a computer or a human brain information, and it will crackle with electrical impulses as it prepares a response. Within cells, these impulses result in spurts of multiple molecular signals. Boyden describes this process as a group conversation. Signals within a cell are like a set of people trying to decide what to do for the evening: they take into account many possibilities, and then decide what to collectively do, he says.

These cellular discussions are what prompt, for example, a neuron to encode a memory or a cell to turn cancerous. Despite their importance, scientists still dont have a strong grasp of how these signals work together to guide a cells behavior.

To see cell signaling in action, scientists typically introduce genes encoding sensors connected to fluorescent proteins. These molecular reporters sense a signal and then glow a specific color under the microscope. Researchers can use a different color reporter for each signal to tell the signals apart. But finding sets of reporters with colors that a microscope can differentiate is challenging. And a typical cellular conversation can involve dozens of signals or more.

Changyang Linghu and Shannon Johnson, scientists in Boydens lab, got around this limitation by affixing reporters to small, self-assembling proteins that act like LEGO bricks. These small proteins clicked together, forming clusters that were randomly scattered across the cell like little islands. Each cluster, which appears under the microscope as a luminescent dot, reports only one type of cellular signal. Its like having some islands with thermometers to report temperature and other islands with barometers measuring pressure, Johnson says.

In experiments with neurons, the team created clusters that each glowed upon detection of one of five different signals, including calcium ions and other important signaling molecules. After imaging the live cells, the researchers attached molecular labels to the glowing dots to identify the reporters located there. Using computer analyses, the team turned the dots magenta, yellow, and other colors, depending on whether they represented calcium or another signal. This let them see which signals were switching on and off across a cells interior.

By monitoring so many signals at once, the team was able to figure out how each signal related to one another. Teasing apart such relationships could help scientists understand complex processes like learning, Linghu says.

He likens a cell to an orchestra and its signals to a symphony. Its difficult to fully appreciate a symphony by listening to just a single instrument, he says. Because the new technique lets scientists observe multiple signals at the same time, we can understand the symphony of cellular activities.

Boydens team estimates it may be possible to detect as many as 16 signals with their technology, but improvements in genetic engineering techniques could raise that number significantly. Potentially, you could look at dozens, hundreds, or even more signals, he says. The next challenge, Boyden says, is getting sensors for all of those signals into a cell.

Reference: Spatial multiplexing of fluorescent reporters for dynamic imaging of signal transduction networks by Changyang Linghu, Shannon L. Johnson et al., 23 November 2020, Cell.DOI:: 10.1016/j.cell.2020.10.035

View original post here:
Real Time Spying on the Symphony of Cellular Signals That Drive Biology - SciTechDaily

Post-doctoral Fellow in Computational Stem Cell Biology job with THE UNIVERSITY OF HONG KONG | 234737 – Times Higher Education (THE)

Work type:Full-timeDepartment:School of Biomedical Sciences (22600)Categories:Academic-related Staff

Applications are invited for appointment asPost-doctoral Fellow in Computational Stem Cell Biology (several posts) in the School of Biomedical Sciences(Ref.:502657), to commence as soon as possible for one to three year(s), with the possibility of renewal subject to satisfactory performance.

The selected candidates will work in a company incorporated by the University of Hong Kong that was established to administer and support the Universitys innovation endeavors. They will conduct research in a computational lab and closely work with experimental lab(s).The specific project will be discussed and determined between each candidate and the Principal Investigator together.

Applicants should have a Ph.D. degree in Bioinformatics, Genetics, Computational Biology or other relevant quantitative disciplines, and in-depth experience in at least one of the programming languages for scientific computing, for example, Python, R, Perl, C/C++ and JAVA.Research experience in stem cells, regenerative medicine, cancer, heart disease, liver disease, immunology or drug screening would be an advantage.Preference will be given to those with expertise in bioinformatics and computational biology, and with strong interests in translational research utilizing next-generation sequencing and single-cell sequencing to study any of the above research areas.The ability to work independently, participate in highly collaborative projects, and contribute intellectually to research development are requisites.Applicants should also be highly motivated individuals with good verbal and written communication skills in English.

A competitive salary commensurate with qualifications and experience will be offered.

The University only accepts online applications for the above posts. Applicants should apply online and upload an up-to-date C.V.Review of applications will start from December 4, 2020 and continue untilDecember 30, 2020, or until the posts are filled, whichever is earlier.

Continued here:
Post-doctoral Fellow in Computational Stem Cell Biology job with THE UNIVERSITY OF HONG KONG | 234737 - Times Higher Education (THE)

Prachee Avasthi Honored with 2020 WICB Junior Award for Excellence in Research – Newswise

Newswise Prachee Avasthi was selected by the Women in Cell Biology (WICB) of the American Society for Cell Biology for the 2020 WICB Junior Award for Excellence in Research.Avasthi is an associate professor of Biochemistry and Cell Biology at Geisel School of Medicine at Dartmouth College, though she noted that the work recognized by this award was done at the University of Kansas Medical Center, where she was until recently.

Prachee Avashthis nominator, Wallace Marshall, University of California, San Francisco, called her a star. He wrote: She has proven a willingness to use any approach necessary to pursue the most important questions, and a complete fearlessness to go against prevailing dogma. At the same time, she has proven a highly effective mentor for her trainees, and a role model for both junior and senior investigators alike. In my opinion, she perfectly represents the qualities that the ASCB WICB Junior Award for Excellence in Research seeks to encourage.

Avasthi is an associate professor of Biochemistry and Cell Biology at Geisel School of Medicine at Dartmouth College, though she noted that the work recognized by this award was done at the University of Kansas Medical Center, where she was until recently.

Using a unicellular green alga as a model system, her lab uses chemical biology, biochemistry, genetics, and quantitative live-cell imaging to uncover novel mechanisms regulating assembly of the cilium. That work led to investigation of the intersection of the microtubule and actin cytoskeleton, as well as fundamental actin dynamics and function.

She stays busy outside the lab as well. An advocate for improved publication practices, she serves on the boards of directors for ASAPbio and eLife. She also founded New PI Slack, the online peer-mentoring community for junior faculty, and is on the steering committee of Rescuing Biomedical Research.

Avasthi said, I am quite stunned to receive this award! The past winners are leaders in their fields such that none of the previous selections surprised me. Im routinely inspired by the creativity and brilliance of others, so it means a lot to me that the colleagues I so respect see the beauty that I see in the science were uncovering. I am incredibly honored and am thrilled for my lab members, whose hard work is being recognized by this award. They deserve this.

Her talk at Cell Bio Virtual 2020 will be on Cytoskeletal Diversity, Flexibility, and Functions.

More here:
Prachee Avasthi Honored with 2020 WICB Junior Award for Excellence in Research - Newswise

New study on CRISPR: the stake of unintended consequences in embryos – BioNews

23 November 2020

A recent paper published in the journalCell revealed the cautionary finding that unwanted changes are introduced after modifying genesin human embryos with CRISPR/Cas9. The study, led by Dr Dietrich Egli, assistant professor of developmental cell biology at Columbia University Vagelos College of Physicians and Surgeons, tested theeffects of CRISPR-based genome editingon embryos carrying a mutationin a gene called EYS (eyes shut homolog) which could lead to hereditary blindness. It shows that applying this potent approachto repair a blindness-causing gene in the formation of an early embryo discards the whole chromosome, or a considerable portion of it, and that the loss of the chromosome is widespread.

CRISPR-based genome editing has revolutionised molecular life sciences. It allows scientists to perform accurate modifications in the genomes of living tissues and may lead to new medical therapies such as innovative cancer treatments and curing hereditary illnesses. In October 2020, CRISPR discoverers (Professors Emmanuelle Charpentier of Max Planck Institute for Infection Biology, Germany, and Jennifer Doudna of University of California, Berkeley) were jointly awarded the Nobel Prize in chemistry.

However, like most innovative techniques, there are currently technical challenges. For example, it is possible to produce so-called off-target effects, where edits are performed in the wrong area. Researchers are still unsure as to how this might affect patients. Another concern is mosaicism, where some cells carry the edit but others do not. Such changes performed to sperm,egg and embryos can be passed to subsequent generations. In the second international summit on human genome editing, there was broad agreement among the experts in attendance that these risks are high.

Despite these serious concerns, in December 2018, Dr He Jiankui shocked the world by announcing that the first babies had been born with altered genomes (see BioNews 978). His work has attracted a backlash from the international scientific community and various governments. Dr He has been sentenced to three years in jail and fined for performing 'illegal medical practices'.

The new research indicates that CRISPR genome editing is currently not ready for clinical application to correct mutations in this early phase of human development. These findings should deter premature clinical use of genome editing on embryos. Thus, using CRISPR to edit the genomes of embryos is a far-off reality.

Due to the serious ethical concerns, the US government does not allow the use of federal funds to perform research on human embryos. The experiment was sponsored by private funding (the New York Stem Cell Foundation and the Russell Berrie Foundation programme). In Australia, section 15 of the Prohibition of Human Cloning for Reproduction Act 2002 prohibits a person from altering the genome of a human embryo in such a manner that the change is heritable by its descendantsandthe person intended this to be so. The penaltyfor this offence is imprisonment for 15 years.

We need to guide responsible and ethical research to achieve safe and effective use. In November 2020, the members of the International Society for Stem Cell Research (ISSCR) task force were charged with revising the2016 ISSCR Guidelines (the Guidelines for Stem Cell Research and Clinical Translation). The ISSCR is the largest stem cell organisation in the world. As a contribution to the developing and controversial stem cell field, this organisation has developed guidelines that address the global diversity of ethical, legal, ethical, cultural and political perspectives related to stem cell research and its translation to clinical application. The guidelines underscore widely shared principles that call for rigour, oversight and transparency. Strict adherence to these principles assures that such cutting-edge research is being conducted with integrity and that innovative medical treatments are evidence-based. Recent advances in this field include innovations in genome editing, organoidsand chimeras. Responding to these various developments in science, the updates will encompass a broader and more expansive scope of research and clinical endeavour, imposing rigour on every stage of the study, addressing the cost of regenerative medicine products and stressing the need for precise and effective public communication.

The persuasive ISSCR Guidelines have been adopted by some scientists, clinicians and institutions around the world. While mere guidelines do not supersede local laws, they could inform the interpretation as well as the development of local laws and provide guidance for research practices not covered by the law. As these guidelines will be updated soon, it is important that they do not encourage the clinical application of the CRISPR approach on genome-editing human embryos for the time being.

Read the original here:
New study on CRISPR: the stake of unintended consequences in embryos - BioNews

Basic concepts lay the foundation for personalized immunotherapy – News-Medical.Net

Reviewed by Emily Henderson, B.Sc.Nov 23 2020

Personalized Immunotherapy for Tumor Diseases and Beyond introduces personalized immunotherapy with multi-dimensional models of analysis to determine the best plan for the immunotherapy of patients.

The book introduces readers to some basic concepts which lay the foundation for personalized immunotherapy: the development of a major histocompatibility complex (MHC), the genome profile of T cells and tumor cells, and genome-wide association studies. Chapters also cover special topics such as new immunoassay methods related to personalized immunotherapy and targeted immunotherapy which are geared towards familiarizing readers with current research practices.

Focusing on the central theme of personalized immunotherapy, the authors provide a wealth of information about T-cell screening, tumor neoantigen cloning, primary tumor cell culture for T-cell cloning, bioinformatics strategies for understanding T-cell and primary tumor cell biology and function, and new developments in research on adoptive T-cell immunotherapy.

These developments include T-cell gene therapy and T-cell gene editing, transgenic T-cells for increasing affinity to tumor cells such as CAR T-cells and TCR T-cells, and the systematic modeling of polyclonal specific T-cells and biobank technology.

Personalized Immunotherapy for Tumor Diseases and Beyond is an ideal handbook for medical professionals and students involved in personalized medicine, immunology, and oncology. General readers interested in the new developments in these fields will also benefit from the information provided.

Go here to see the original:
Basic concepts lay the foundation for personalized immunotherapy - News-Medical.Net

Star-Shaped Brain Cells May Hold the Key to Why and How We Sleep – SciTechDaily

Astrocytes in the brain expressing a fluorescent calcium indicator captured with a two-photon microscope. Credit: Image by Ashley Ingiosi, courtesy of Current Biology

A study published in the journal Current Biology suggests that star-shaped brain cells known as astrocytes could be as important to the regulation of sleep as neurons, the brains nerve cells.

Led by researchers at Washington State Universitys Elson S. Floyd College of Medicine, the study builds new momentum toward ultimately solving the mystery of why we sleep and how sleep works in the brain. The discovery may also set the stage for potential future treatment strategies for sleep disorders and neurological diseases and other conditions associated with troubled sleep, such as PTSD, depression, Alzheimers disease, and autism spectrum disorder.

What we know about sleep has been based largely on neurons, said lead author and postdoctoral research associate Ashley Ingiosi. Neurons, she explained, communicate through electrical signals that can be readily captured through electroencephalography (EEG). Astrocytesa type of glial (or glue) cell that interacts with neuronsdo not use electrical signals and instead use a process known as calcium signaling to control their activity.

It was long thought that astrocyteswhich can outnumber neurons by five to onemerely served a supportive role, without any direct involvement in behaviors and processes. Neuroscientists have only recently started to take a closer look at their potential role in various processes. And while a few studies have hinted that astrocytes may play a role in sleep, solid scientific tools to study their calcium activity have not been available until recently, Ingiosi said.

To delve deeper into astrocytes role in sleep, she and her coauthors used a rodent model to record astrocytes calcium activity throughout sleep and wake, as well as after sleep deprivation. They used a fluorescent calcium indicator that was imaged via tiny head-mounted microscopes that looked directly into the brains of mice as they moved around and behaved as they normally would. This indicator allowed the team to see calcium-driven fluorescent activity twinkling on and off in astrocytes during sleep and waking behaviors. Their one-of-a-kind methodology using these miniature microscopes allowed the team to conduct the first-ever study of astrocytes calcium activity in sleep in freely behaving animals.

The research team set out to answer two main questions: do astrocytes change dynamically across sleep and wake states like neurons do? And do astrocytes play a role in regulating sleep need, our natural drive to sleep?

Looking at astrocytes in the frontal cortex, an area of the brain associated with measurable EEG changes in sleep need, they found that astrocytes activity changes dynamically across the sleep-wake cycle, as is true for neurons. They also observed the most calcium activity at the beginning of the rest phasewhen sleep need is greatestand the least calcium activity at the end of the rest phase, when the need for sleep has dissipated.

Next, they kept mice awake for the first 6 hours of their normal rest phase and watched calcium activity change in parallel with EEG slow wave activity in sleep, a key indicator of sleep need. That is, they found that sleep deprivation caused an increase in astrocyte calcium activity that decreased after mice were allowed to sleep.

Their next question was whether genetically manipulating astrocyte calcium activity would impact sleep regulation. To find out, they studied mice that lacked a protein known as STIM1 selectively in astrocytes, which reduced the amount of available calcium. After being sleep deprived, these mice did not sleep as long or get as sleepy as normal mice once allowed to sleep, which further confirmed earlier findings that suggest that astrocytes play an essential role in regulating the need for sleep.

Finally, they tested the hypothesis that perhaps astrocyte calcium activity merely mirrors the electrical activity of neurons. Studies have shown that the electrical activity of neurons becomes more synchronized during non-REM sleep and after sleep deprivation, but the researchers found the opposite to be true for astrocytes, with calcium activity becoming less synchronized in non-REM sleep and after sleep deprivation.

This indicates to us that astrocytes are not just passively following the lead of neurons, said Ingiosi. And because they dont necessarily display the same activity patterns as neurons, this might actually implicate a more direct role for astrocytes in regulating sleep and sleep need.

More research is needed to further unravel the role of astrocytes in sleep and sleep regulation, Ingiosi said. She plans to study astrocytes calcium activity in other parts of the brain that have been shown to be important for sleep and wake. In addition, she would like to look at astrocytes interactions with different neurotransmitters in the brain to start to tease out the mechanism by which astrocytes might drive sleep and sleep need.

The findings of our study suggest that we may have been looking in the wrong place for more than 100 years, said senior author and professor of biomedical sciences Marcos Frank. It provides strong evidence that we should be targeting astrocytes to understand why and how we sleep, as well as for the development of therapies that could help people with sleep disorders and other health conditions that involve abnormal sleep.

Reference: A Role for Astroglial Calcium in Mammalian Sleep and Sleep Regulation by Ashley M. Ingiosi, Christopher R. Hayworth, Daniel O. Harvey, Kristan G. Singletary, Michael J. Rempe, Jonathan P. Wisor and Marcos G. Frank, 24 September 2020, Current Biology.DOI: 10.1016/j.cub.2020.08.052

Support for the study came from the National Institutes of Health.

More:
Star-Shaped Brain Cells May Hold the Key to Why and How We Sleep - SciTechDaily