Category Archives: Cell Biology

New analytic tool designed to help guide precision oncology discovery and treatments – Newswise

Newswise ANN ARBOR, Michigan Recent large-scale efforts to categorize the molecular data of multiple cancer types has yielded so much information that researchers now have a new question: How to turn all this data into meaningful information that guides cancer research and patient care.

A new analytic tool developed by University of Michigan Rogel Cancer Center researchers combines multiple data sets to help sift the signal from the noise.

Our idea was to combine three sources of data sets molecular data from both cancer cell lines and patients and drug profiling data to understand proper preclinical models that are most representative of these tumors, says Veerabhadran Baladandayuthapani, Ph.D., professor of biostatistics at the University of Michigan School of Public Health and senior author of a paper published in the Journal of Clinical Oncology Clinical Cancer Informatics that describes this new tool.

The tool, called TransPRECISE, uses data from 7,714 patient samples across 31 cancer types, collected as part of the Cancer Proteome Atlas. This is combined with 640 cancer cell lines from the MD Anderson Cell Lines Project and drug sensitivity data representing 481 drugs from the Genomics of Drug Sensitivity in Cancer model system.

The good thing is this is a very dynamic process. We can have this whole system set up in a computer. As new patients come in or new data comes in, you can keep adding it, says Rupam Bhattacharrya, M.Stat., a doctoral student and first author on the paper.

The tool builds on an earlier model the team had created, which they called PRECISE. With an eye toward precision medicine, they created a model to look at what changes occur to the molecular structure of individual patients individual tumors. TransPRECISE adds in data from cell lines and drug sensitivity, which will be helpful for researchers translating cancer cell biology into drug discovery.

Now that we have tens of thousands of tumors on these patients we can evaluate what might be the potential therapeutic efficiency of these drugs. The key idea was to develop an analytic tool to do that, says Baladandayuthapani, who is also director of the Rogel Cancer Centers cancer data science shared resource.

In the JCO Clinical Cancer Informatics paper, researchers validated the tool by comparing known drug responses and clinical outcomes in patient data. TransPRECISE identified the differences in proteins among individual tumors and accurately tied it back to actual patient outcomes. In addition, they looked at several pathways to predict potential drug targets. This yielded results that mirrored current treatment recommendations or targets being tested in clinical trials, such as ibruutinib for BRCA-positive breast cancer, and lapatinib for colon cancer.

We have so much data, how do we drill it down to make it more informative so an oncologist can understand? Our work would potentially help oncologists or researchers develop concrete hypotheses based on which mechanism is working, potentially bringing to the top drugs that might warrant more evaluation, Baladandayuthapani says.

The researchers have made publicly available a comprehensive database and visualization of the findings at https://bayesrx.shinyapps.io/TransPRECISE.

Additional authors: Min Jin Ha, Qingzhi Liu, Rehan Akbani, Han Liang

Funding: National Institutes of Health grants R21 CA220299-01A1, U54 CA224065, 3P50 CA070907-20S1, R01 CA160736, R01 CA194391, P30 CA46592, R01 CA1745486, U24 CA209851, U01 CA217842, P30 CA016672; Leukemia and Lymphoma Society, Cancer Prevention and Research Institute of Texas, National Science Foundation, University of Michigan Rogel Cancer Center, University of Michigan School of Public Health

Disclosure: None

Reference: Journal of Clinical Oncology Clinical Cancer Informatics, doi:10.1200/CCI.19.00140, published May 6, 2020

Resources:

University of Michigan Rogel Cancer Center, http://www.rogelcancercenter.org

Michigan Health Lab, http://www.MichiganHealthLab.org

Michigan Medicine Cancer AnswerLine, 800-865-1125

# # #

Go here to read the rest:
New analytic tool designed to help guide precision oncology discovery and treatments - Newswise

Imara to Present Data on IMR-687 in Sickle Cell Disease at the 25th Annual European Hematology Association (EHA) Congress – BioSpace

BOSTON, May 21, 2020 (GLOBE NEWSWIRE) -- Imara Inc. (Nasdaq: IMRA), a clinical-stage biopharmaceutical company dedicated to developing and commercializing novel therapeutics to treat patients suffering from rare inherited genetic disorders of hemoglobin, today announced that it will present interim data from the ongoing Phase 2a study of IMR-687 in patients with sickle cell disease at the 25th Annual European Hematology Association (EHA) Congress to be held virtually June 11-21, 2020.

The data will be presented by Biree Andemariam, M.D., Associate Professor at UConn School of Medicine and Director of the New England Sickle Cell Institute at UConn Health, and lead investigator for the trial. Dr. Andemariams presentation (Abstract #S290), titled IMR-687, A Highly Selective Phosphodiesterase 9 Inhibitor (PDE9I), Increases F-Cells and Fetal Hemoglobin in a Ph-2A Interim Analysis will be included in the oral abstract session, New Therapeutic Approaches for Sickle Cell Disease. The presentation will be available for on-demand viewing starting at 2:30 a.m. ET / 8:30 a.m. CEST on Friday, June 12, 2020, and will be accessible until October 15, 2020.

In January 2020, Imara completed enrollment in the IMR-687 Phase 2a clinical trial in sickle cell patients and the Company plans to report top-line data from the trial in the fourth quarter of 2020.

About IMR-687

IMR-687 is a highly selective and potent small molecule inhibitor of PDE9. PDE9 uniquely degrades cyclic guanosine monophosphate (cGMP), an active signaling molecule that plays a role in vascular biology. Lower levels of cGMP are often found in people with sickle cell disease and beta-thalassemia and are associated with impaired blood flow, increased inflammation, greater cell adhesion and reduced nitric oxide mediated vasodilation.

Blocking PDE9 acts to increase cGMP levels, which are associated with reactivation of fetal hemoglobin, or HbF, a natural hemoglobin produced during fetal development. Increased levels of HbF in red blood cells have been demonstrated to improve symptomology and substantially lower disease burden in both patients with sickle cell disease and patients with beta-thalassemia.

About Imara

Imara Inc. is a clinical-stage biotechnology company dedicated to developing and commercializing novel therapeutics to treat patients suffering from rare inherited genetic disorders of hemoglobin. Imara is currently advancing IMR-687, a highly selective, potent small molecule inhibitor of PDE9 that is an oral, once-a-day, potentially disease-modifying treatment for sickle cell disease and beta-thalassemia. IMR-687 is being designed to have a multimodal mechanism of action that acts on red blood cells, white blood cells, adhesion mediators and other cell types. For more information, please visit http://www.imaratx.com.

Cautionary Note Regarding Forward-Looking Statements

Statements in this press release about future expectations, plans and prospects, as well as any other statements regarding matters that are not historical facts, may constitute forward-looking statements within the meaning of The Private Securities Litigation Reform Act of 1995. These statements include, but are not limited to, statements relating to the timing for reporting of data from the ongoing Phase 2a clinical trial evaluating IMR-687 in patients with sickle cell disease. The words anticipate, believe, continue, could, estimate, expect, intend, may, plan, potential, predict, project, should, target, will, would and similar expressions are intended to identify forward-looking statements, although not all forward-looking statements contain these identifying words. Actual results may differ materially from those indicated by such forward-looking statements as a result of various important factors, including: the impact of extraordinary external events, such as the risks and uncertainties resulting from the impact of the COVID-19 pandemic on the Companys business, operations, strategy, goals and anticipated milestones, including its ability to conduct and readout data from its ongoing Phase 2a clinical trial of IMR-687 in sickle cell disease ; and other factors discussed in the Risk Factors section of the Companys most recent Quarterly Report on Form 10-Q, which is on file with the Securities and Exchange Commission and in other filings that the Company makes with the Securities and Exchange Commission in the future. Any forward-looking statements contained in this press release speak only as of the date hereof, and the Company expressly disclaims any obligation to update any forward-looking statement, whether as a result of new information, future events or otherwise.

Media Contact:Krystle GibbsTen Bridge Communications508-479-6358krystle@tenbridgecommunications.com

Investor Contact:Michael Gray617-835-4061mgray@imaratx.com

Read the original:
Imara to Present Data on IMR-687 in Sickle Cell Disease at the 25th Annual European Hematology Association (EHA) Congress - BioSpace

Good News, We Can Activate The Cells That Keep Our Muscles From Wasting Away After 30 – ScienceAlert

Unfortunately, for those of us approaching or beyond 30 years of age - this is the point our muscles start to basically shrink away. You may think you've heard it all before, but a new review presents yet another reason we really should avoid slacking off when it comes to moving our behinds.

Amongst many other things, exercise has been shown to increase mitochondria- energy-generating organelles - within cells; it also increases blood supply, waste removal, and bone strength.

While many muscle degeneration studiesfocus on much older people, the slow decline of muscle tissue in our bodies actually begins in our 30s, with an average of between three to five percent muscle loss every year.

Thankfully, our muscles are able to regenerate, so with a bit of effort on our part, we can counteract this wastage.

Muscle growth, or regeneration after injury, involves satellite cells. When they divide into a new generation of cells, they can either become more satellite cells, or myoblasts - cells which then divide into muscle cells. When they're not used however, satellite cells go into an energy-saving dormancy.

Cellular biologist William Chen and colleagues from the University of Ottawa in Canada reviewed the literature on how these cells function, including human strength training and endurance exercise studies, as well as molecular and genetic studies on mice.

"It is evident that satellite cells are activated through various signalling pathways following exercise," the team found, with multiple studies of human training sessions indicating an increase in satellite cell proliferation.

Neglecting exercise causes problems, because like all cells, satellite cells become damaged and filled with cellular trash as the normal processes of life batter them about as they age. Even while dormant, when cellular activity is reduced, the process of keeping themselves in this state still produces cellular waste.

Thus, if satellite cells stay dormant for too long, the waste build-up can cause damage, preventing the cells from dividing and replacing themselves. Even worse, in ageing muscles, if they're not activated, satellite cells can get stuck in the dormant phase and lose all ability to divide and multiply.

This means they are no longer capable of replacing defunct muscle cells with shiny new healthy ones, or making more copies of themselves.

Healthy dormancy (quiescence) requires regular activation. (Chen et al, Open Biology, 2020)

"Physical inactivity becomes more detrimental with age as satellite cells accumulate higher levels of cellular waste and DNA damage during longer periods of deep quiescence," Chen and colleagues explain in their review.

So, giving these cells a regular prod through exercise, to remind them how to be active, helps more of them retain their full function.

However, "exercise alone does not prevent all age-related defects in skeletal muscle," the team wrote.

"The satellite cell population still diminishes to some degree over age... Regardless, lifelong resistance exercise should be pursued as the effects of [age-related skeletal muscle loss] are greatly diminished when compared with sedentary lifestyles."

The researchers point out the exact mechanisms of satellite cell replacement are not yet fully understood, nor is their behaviour during exercise. They recommend more research be focused in these directions.

With skeletal muscle making up 40 percent of our body in terms of weight, it makes sense that the condition of the bulk of these cells and their makers has huge impacts on our overall health.

Muscle strength is correlated with increased quality of life, particularly as we age. So this aspect of satellite cell biology is just one of many ways exercise helps to keep our muscles and the rest of us healthy.

This review was published in Open Biology.

Go here to see the original:
Good News, We Can Activate The Cells That Keep Our Muscles From Wasting Away After 30 - ScienceAlert

‘Senolytic’ therapy blunts liver tumor progression in animal models – News-Medical.Net

Reviewed by Emily Henderson, B.Sc.May 20 2020

"Senotherapy," a treatment that uses small molecule drugs to target "senescent" cells, or those cells that no longer undergo cell division, blunts liver tumor progression in animal models according to new research from a team led by Celeste Simon, Ph.D., a professor of Cell and Developmental Biology in the Perelman School of Medicine at the University of Pennsylvania and scientific director of the Abramson Family Cancer Research Institute. The study was published in Nature Cell Biology.

This kind of therapy is not something that has been tried before with liver cancer. And in our models, so-called 'senolytic' therapy greatly reduced disease burden, even in cases with advanced disease."

Celeste Simon, Ph.D., Professor of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania

Loss of the enzyme FBP1 in human liver cells significantly increases tumor growth. Previous research has shown FBP1 levels are decreased in stage 1 tumors, and further reduced as the disease progresses. In this study, Simon and her team used RNA-sequencing data to identify FBP1 as universally under-expressed in the most common form of liver cancer, hepatocelluar carcinoma, regardless of underlying causes like obesity, alcoholism, and hepatitis.

The loss of FBP1 in liver cells activates the neighboring hepatic "stellate cells"--which make up ten percent of liver mass--causing fibrosis (tissue scarring) and subsequent stellate cell senescence, both of which promote tumor growth. Researchers found that these senescent stellate cells can be selectively targeted by senolytics, including Navitoclax (already in clinical trials for other diseases, like hematological malignancies), in order to blunt tumor progression driven by liver cell-specific FBP1 loss.

The team provides the first genetic evidence for FBP1 as a bona fide metabolic tumor suppressor in the liver and that its loss in liver cells promotes the growth of tumors because of effects on other cells within the tumor microenvironment.

Using genetically engineered mouse models, the team eliminated FBP1and found the disease progressed more rapidly and tumor burden greatly increased in carcinogen-mediated, dietary, and other forms of hepatocellular carcinoma.

"The case with liver cancer is very dire, once you get beyond a certain stage there are limited, if any, treatments available," Simon said. "As obesity rates continue to increase and viral infections continue to be a problem, there is going to be an increasing surge of liver cancer which currently has few treatment options. And since FBP1 activity is also lost in renal cancer, FBP1 depletion may be generally applicable to a number of human cancers. What's unique about our senotherapy approach is that we are specifically targeting other cells in the liver tumor environment rather than the cancer cells themselves."

Next steps, according to researchers will be to begin to test these treatments in a clinical setting.

Excerpt from:
'Senolytic' therapy blunts liver tumor progression in animal models - News-Medical.Net

Assessing the Fallout From the Coronavirus Pandemic Cell Biology Cloud Computing Market 2020: Global Size, Supply-Demand, Product Type and End User…

Analysis of the Global Cell Biology Cloud Computing Market

The report on the global Cell Biology Cloud Computing market reveals that the market is expected to grow at a CAGR of ~XX% during the considered forecast period (2019-2029) and estimated to reach a value of ~US$XX by the end of 2029. The latest report is a valuable tool for stakeholders, established market players, emerging players, and other entities to devise effective strategies to combat the impact of COVID-19

Further, by leveraging the insights enclosed in the report, market players can devise concise, impactful, and highly effective growth strategies to solidify their position in the Cell Biology Cloud Computing market.

Research on the Cell Biology Cloud Computing Market Addresses the Following Queries

Get Free Sample PDF (including COVID19 Impact Analysis, full TOC, Tables and Figures) of Market Report @ https://www.marketresearchhub.com/enquiry.php?type=S&repid=2655551&source=atm

Competitive Landscape

The competitive landscape section offers valuable insights related to the business prospects of leading market players operating in the Cell Biology Cloud Computing market. The market share, product portfolio, pricing strategy, and growth strategies adopted by each market player is included in the report. The major steps taken by key players to address the business challenges put forward by the novel COVID-19 pandemic is discussed in the report.

Regional Landscape

The regional landscape section provides a deep understanding of the regulatory framework, current market trends, opportunities, and challenges faced by market players in each regional market. The various regions covered in the report include:

End-User Assessment

The report bifurcates the Cell Biology Cloud Computing market based on different end users. The supply-demand ratio and consumption volume of each end-user is accurately depicted in the report.

Regional and Country-level AnalysisThe report offers an exhaustive geographical analysis of the global Cell Biology Cloud Computing market, covering important regions, viz, North America, Europe, China, Japan, Southeast Asia, India and Central & South America. It also covers key countries (regions), viz, U.S., Canada, Germany, France, U.K., Italy, Russia, China, Japan, South Korea, India, Australia, Taiwan, Indonesia, Thailand, Malaysia, Philippines, Vietnam, Mexico, Brazil, Turkey, Saudi Arabia, U.A.E, etc.The report includes country-wise and region-wise market size for the period 2015-2026. It also includes market size and forecast by each application segment in terms of revenue for the period 2015-2026.Competition AnalysisIn the competitive analysis section of the report, leading as well as prominent players of the global Cell Biology Cloud Computing market are broadly studied on the basis of key factors. The report offers comprehensive analysis and accurate statistics on revenue by the player for the period 2015-2020. It also offers detailed analysis supported by reliable statistics on price and revenue (global level) by player for the period 2015-2020.On the whole, the report proves to be an effective tool that players can use to gain a competitive edge over their competitors and ensure lasting success in the global Cell Biology Cloud Computing market. All of the findings, data, and information provided in the report are validated and revalidated with the help of trustworthy sources. The analysts who have authored the report took a unique and industry-best research and analysis approach for an in-depth study of the global Cell Biology Cloud Computing market.The following players are covered in this report:AccentureAmazon Web ServicesBenchlingCisco SystemsDell EmcIBMDXC TechnologyOracleScaleMatrixIPERIONNovelBioCell Biology Cloud Computing Breakdown Data by TypePublic Cloud ComputingPrivate Cloud ComputingHybrid Cloud ComputingCell Biology Cloud Computing Breakdown Data by ApplicationGenomicsDiagnosticsClinical TrialsPharma ManufacturingOthers

Do You Have Any Query Or Specific Requirement? Ask to Our Industry [emailprotected] https://www.marketresearchhub.com/enquiry.php?type=E&repid=2655551&source=atm

Essential Findings of the Cell Biology Cloud Computing Market Report:

You can Buy This Report from Here @ https://www.marketresearchhub.com/checkout?rep_id=2655551&licType=S&source=atm

Excerpt from:
Assessing the Fallout From the Coronavirus Pandemic Cell Biology Cloud Computing Market 2020: Global Size, Supply-Demand, Product Type and End User...

Cell biologist to study coronavirus-related infection of respiratory cells – UC Riverside

Prue Talbot, a professor of cell biology at UC Riverside, has received a seed grant to study the COVID-19-related infection of respiratory cells.

She and her team will use the funds to test the hypothesis that electronic cigarettes and nicotine increase the ACE2 receptor on respiratory epithelium, providing more binding sites for the virus and increasing the possibility of infection.

We expect our work will help clarify if the use of tobacco products makes it more likely for an infection to occur, Talbot said.

Angiotensin converting enzyme-2, or ACE2, is a special receptor on some cells that has attracted the attention of drug companies as a potential target for addressing the novel coronavirus.

The $25,000, six-month grant from the California Tobacco-Related Disease Research Program of the University of California will support the research of three graduate students.

Follow this link:
Cell biologist to study coronavirus-related infection of respiratory cells - UC Riverside

Paying the Price of Protection – Newswise

Newswise Is the wanton killing of cells in autoimmune disease a case of mistaken identity, or does it arise from an important physiological service? The first is the commonly accepted view that autoimmune attack is a sort of mistake. But the latter view may be closer to the truth, according to a new model proposed by researchers at the Weizmann Institute of Science. Among other things, the model suggests a solution to the long-standing riddle of why some organs are susceptible to autoimmune diseases while others are not. The findings were published in the journal Immunity.

Yael Korem Kohanim, a doctoral student in the lab of Prof. Uri Alon in the Institutes Department of Molecular Cell Biology, who led the study, explains that autoimmune diseases can be divided into two types systemic ones like lupus that attack many organs in the body, and those like type 1 diabetes that affect just one organ. One of the riddles about this second, organ-specific type of autoimmune disease is why some organs get autoimmune diseases while others do not. The pancreas is an extreme example: the insulin-producing beta cells that make up 2-4% of the pancreas are highly prone, while the rest of the pancreas almost never gets an autoimmune disease.

Likewise, Hashimotos thyroiditis affects the thyroids of some 7% of the population, whereas the parathyroid glands right next to them are rarely affected by autoimmune syndromes. These organ-specific autoimmune diseases tend to follow a similar pattern, arising in children or young adults (unlike genetic diseases that appear at birth, or those of aging), and they involve the destruction of cells that secrete essential hormones. Immune cells called T cells somehow identify these endocrine cells as dangerous and eradicate them on contact.

Korem Kohanim, Prof. Alon, and the research group, including Dr. Avichai Tendler, Dr. Avi Mayo, and Prof. Nir Friedman in the Department of Immunology, asked: What if the T cells are meant to kill these cells all along?

Keeping supply matched to demand

The researchers hypothesized that T cells might be kept on the payroll as an extra layer of protection to ensure that hormone levels stay within narrow limits. Hormones insulin, thyroid, cortisol tend to function in feedback loops; too little is as harmful as too much. When demand for the hormone rises for example, a demand for insulin when glucose is repeatedly sensed the cells not only step up production, they ramp up cell division to help meet that demand. But cell division carries risks, as a certain percentage of the new cells are likely to carry mutations. Most such mutations are harmless, but if one disrupts the cells delicate sensing machinery, the cell will misread the demand as high when it is actually low. The result is deadly: the cell will continue not only to pump out extra hormones, it will divide again and again to produce new cells with the same mutation, which will then divide again and produce even more of the hormone, soon causing severe disease.

T cells, which select their targets by recognizing small pieces of proteins that identify the cells they are meant to kill, could conceivably target the over-secreting cells in healthy organs. They act, in this case, as secret agents, removing cells that threaten to take over the organ and secrete too much hormone. In autoimmune diseases, the T cells might be primed to accomplish the same task but get overzealous, killing off non-mutant cells.

Was this hypothesis reasonable? Korem Kohanim, Prof. Alon, and the team delved into the literature and bioinformatics data on several single-organ autoimmune diseases, then creating a mathematical model for the functioning of healthy organs in which small numbers of T cells kept a low profile. In this model, the organs stay fit and productive as long as the T cells have a means of being highly selective, so that most of their targets would be the mutated cells.

This result agrees with the findings from research on the diseases showing that, in each one, the T cells identify proteins specifically connected to the production or secretion of the target cells hormones. In healthy organs, those T cells could use the same identification codes to target any cells that are overproducing the hormones. In other words, autoimmune diseases could be the result of a tradeoff: a layer of regulation preventing diseases linked to overproduction, while risking the opposite effect reduced production in some people.

The glands that refuse to pay the price

If other glands are not susceptible to autoimmune disease, does this mean they also forgo T-cell protection? The team went back to the literature and found that the parathyroid, for example, is highly prone to noncancerous growths called adenomas that are common in post-menopausal women, affecting as many as one in 50. These adenomas secrete huge amounts of hormone, causing a disease called hyperparathyroidism. Other examples, though less dramatic than parathyroid adenomas, supported the idea that a lack of T-cell intervention, as the model would suggest, could result in unchecked hormone secretion and cell growth.

The model explains a number of puzzling findings, says Korem Kohanim. For example, we looked at genetic sequences of T cells found in healthy people, and noted that some of them indeed have the exact same protein-identifying receptors as the T cells found in those with autoimmune diseases. The explanation has always been that the apparently healthy people have a mild form of the disease, or one in its initial stages. But the findings are more logical if you assume these auto-reactive T cells are meant to be there; that they are meant to keep us from getting diseases of cell division and hormone overproduction.

So far, says Korem Kohanim, the model provides a solution to a riddle that has long plagued researchers, though experimentation is needed to see if its claims are borne out. However, it has already garnered interest in the field and is generating discussion among top immunologists. The Alon lab will continue to collaborate on the project with the Friedman lab as they develop means of experimentally testing the models results.

We think that autoimmune diseases do not come out of nowhere. They are a malfunction, but one of a physiological system that is already in place, Korem Kohanim says.

We follow in the footsteps of pioneers at Weizmann Irun Cohen and Michal Schwartz and their students who emphasized the immune system as tending our bodies as well as fighting pathogens, Prof. Alon says, adding: I cant wait to see if this theory is fruitful in the sense of generating new experiments that will teach us about the mysteries of autoimmune diseases.

Prof. Uri Alons research is supported by the Sagol Institute for Longevity Research; the Jeanne and Joseph Nissim Center for Life Sciences Research; the Braginsky Center for the Interface between Science and the Humanities; the Kahn Family Research Center for Systems Biology of the Human Cell; the Zuckerman STEM Leadership Program; the Rising Tide Foundation; the estate of Olga Klein Astrachan; and the European Research Council. Prof. Alon is the incumbent of the the Abisch-Frenkel Professorial Chair.

Prof. Nir Friedmans research is supported by the David and Fela Shapell Family Institute for Preclinical Studies; the Dr. Dvora and Haim Teitelbaum Endowment Fund; the Pearl Welinsky Merlo Foundation for Scientific Progress Research Fund; the Florence Blau, Morris Blau, and Rose Peterson Fund; the Rising Tide Foundation; the Park Avenue Charitable Fund; Gertrude Comninos; the estate of Robert Einzig; and the estate of Emile Mimran. Prof Friedman is the incumbent ofthe Eugene and Marcia Applebaum Professorial Chair.

The Weizmann Institute of Science in Rehovot, Israel, is one of the worlds top-ranking multidisciplinary research institutions. The Institutes 3,800-strong scientific community engages in research addressing crucial problems in medicine and health, energy, technology, agriculture, and the environment. Outstanding young scientists from around the world pursue advanced degrees at the Weizmann Institutes Feinberg Graduate School. The discoveries and theories of Weizmann Institute scientists have had a major impact on the wider scientific community, as well as on the quality of life of millions of people worldwide.

See original here:
Paying the Price of Protection - Newswise

Superpower Discovered in Squids: They Can Massively Edit Their Own Genetics – SciTechDaily

Revealing yet another super-power in the skillful squid, scientists have discovered that squid massively edit their own genetic instructions not only within the nucleus of their neurons, but also within the axon the long, slender neural projections that transmit electrical impulses to other neurons. This is the first time that edits to genetic information have been observed outside of the nucleus of an animal cell.

The study, led by Isabel C. Vallecillo-Viejo and Joshua Rosenthal at the Marine Biological Laboratory (MBL), Woods Hole, is published this week in Nucleic Acids Research.

The longfin inshore squid, Doryteuthis pealeii, long established as a research organism for fundamental biological studies. Credit: Elaine Bearer

The discovery provides another jolt to the central dogma of molecular biology, which states that genetic information is passed faithfully from DNA to messenger RNA to the synthesis of proteins. In 2015, Rosenthal and colleagues discovered that squid edit their messenger RNA instructions to an extraordinary degree orders of magnitude more than humans do allowing them to fine-tune the type of proteins that will be produced in the nervous system.

But we thought all the RNA editing happened in the nucleus, and then the modified messenger RNAs are exported out to the cell, says Rosenthal, senior author on the present study. Now we are showing that squid can modify the RNAs out in the periphery of the cell. That means, theoretically, they can modify protein function to meet the localized demands of the cell. That gives them a lot of latitude to tailor the genetic information, as needed. The team also showed that messenger RNAs are edited in the nerve cells axon at much higher rates than in the nucleus.

Top, schematic of squid anatomy showing the location of the giant axon, an unusually large neural projection that partly controls the squids jet propulsion system, used for very fast movement, attacks and escapes. Below, schematic of a neuron, showing the location of the nucleus where all RNA editing was previously thought to occur, and the axon, where local RNA editing was identified in squid. Credit: Vallecillo-Viejo et al, Nucl. Acids Res., 2020.

In humans, axon dysfunction is associated with many neurological disorders. Insights from the present study could accelerate the efforts of biotech companies that seek to harness this natural RNA editing process in humans for therapeutic benefit.

Scientists from Tel Aviv University and The University of Colorado at Denver collaborated with MBL scientists on the study.

Previously, Rosenthal and colleagues showed that octopus and cuttlefish also rely heavily on mRNA editing to diversify the proteins they can produce in the nervous system. Together with squid, these animals are known for strikingly sophisticated behaviors, relative to other invertebrates.

Reference: Spatially regulated editing of genetic information within a neuron by Isabel C Vallecillo-Viejo, Noa Liscovitch-Brauer, Juan F Diaz Quiroz, Maria F Montiel-Gonzalez, Sonya E Nemes, Kavita J Rangan, Simon R Levinson, Eli Eisenberg and Joshua J C Rosenthal, 23 March 2020, Nucleic Acids Research.DOI: 10.1093/nar/gkaa172

See the original post:
Superpower Discovered in Squids: They Can Massively Edit Their Own Genetics - SciTechDaily

Novel insights on SARS-CoV-2 cell entry may inform further vaccine and treatment development – News-Medical.Net

A research group from Harvard Medical School reported two cryo-electron microscopy structures derived from a single preparation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein in their paper available on the preprint server bioRxiv*. Understanding the nuances of prefusion and postfusion conformations shed light on viral cell entry and may, in turn, guide the development of vaccines and drugs.

Novel Coronavirus SARS-CoV-2 Colorized scanning electron micrograph of a VERO E6 cell (purple) exhibiting elongated cell projections and signs of apoptosis, after infection with SARS-COV-2 virus particles (pink), which were isolated from a patient sample. Image captured at the NIAID Integrated Research Facility (IRF) in Fort Detrick, Maryland. Credit: NIAID

The current pandemic of coronavirus disease 2019 (COVID-19) reached nearly every corner of our planet. Due to its high case-fatality rate and catastrophic economic and social repercussions, both SARS-CoV-2 and COVID-19 became the top research priority of scientists around the world.

Membrane fusion represents a pivotal early step for all enveloped viruses in order to enter host cells and establish the infection. Albeit this is an energetically favorable process, there are high kinetic barriers when two membranes approach each other before fusion, primarily due to hydration repulsive forces.

The fusion protein for coronaviruses is the S-protein that decorates the surface of the viral particle as an extensive crown, which is how these viruses were named in the first place. The protein also instigates the neutralizing antibody response, which makes it an important target in vaccine development endeavors.

In short, the viral S-protein forms a trimer and catalyzes fusion between viral and target cell membranes, which is the first step of infection. This is why a research group decided to appraise distinct conformational states of SARS-CoV-2 S-protein in order to further inform the development of vaccines and therapeutics.

In this study, a fully wild-type form of SARS-CoV-2 S-protein was expressed in human embryonic kidney 293 (HEK-293) cells, which are widely used in cell biology research. Cryogenic electron microscopy was then used to derive two structures representing S-protein's prefusion and postfusion states both derived from a single preparation previously solubilized in detergent.

The cell-cell fusion assay was utilized to quantify the fusion activity mediated by SARS-CoV-2 S-protein. Structure determination was carried out by rounds of three-dimensional classification, refinement, and masked local refinement. All experiments were performed with a Biacore 3000 system, which is used for real-time biomolecular interaction analysis by surface plasmon resonance technology.

This study has identified a structure in the vicinity of the fusion peptide called the fusion peptide proximal region (FPPR), which prompts the fusogenic structural rearrangements of S-protein. Nonetheless, certain questions are open regarding membrane fusion, since the regions near the viral membrane are still not evident in the reconstructions even in the structures obtained by the Harvard authors with a full-length S construct.

It has been shown how spontaneous structural transformation to the postfusion state under mild conditions is independent of target cells. The prefusion trimer (3.1-Angstrom resolution) forms a steadfastly packed structure with three receptor-binding domains, substantially different from recently published structures of a stabilized S-ectodomain trimer.

Conversely, the postfusion conformation (3.3-Angstrom resolution) is a robust tower-like trimer decorated by N-linked glycans (i.e., oligosaccharides attached to a nitrogen atom) along its long axis with more or less even spacing suggesting a possible association with mechanism protecting the virus from unfavorable external conditions and host immune responses.

Whether other viral proteins (such as M-protein) may aid in stabilizing the spike by interacting with the heptad repeat 2 remains an intriguing question. "We still need a high-resolution structure of the intact S-protein in the context of the membrane and other viral components to answer the various open questions," explain study authors.

The most unanticipated finding from the current study is the notion that the kinetic barrier for the conformational transition relevant for viral entry is surprisingly low for this S-protein. Hence, further investigation is needed to elucidate whether this observation relates directly to successful membrane fusion or infection, and perhaps effective human-to-human transmission.

A model for structural rearrangements of SARS-Cov-2 S protein. (A) Structural changes independent of a target cell. We suggest that both the prefusion and postfusion spikes are present on the surface of mature virion and the ratio between them may vary (diagram of virion). The postfusion spikes on the virion are formed by S2 after S1 dissociates in the absence of ACE2. (B) ACE2-dependent structural rearrangements. Structural transition from the prefusion to postfusion conformation inducing membrane fusion likely proceeds stepwise as follows: 1) FPPR clamps down RBD through CTD1 in the prefusion S trimer, but it occasionally flips out of position and allows an RBD to sample the up conformation. 2) RBD binding to ACE2 creates a flexible FPPR that enables exposure of the S2 cleavage site immediately upstream of the adjacent fusion peptide (FP). Cleavage at the S2 site, and perhaps also the S1/S2 site, releases the structural constraints on the fusion peptide and initiates a cascade of refolding events in S2, probably accompanied by complete dissociation of S1. 3) Formation of the long central three-stranded coiled-coil and folding back of HR2. 4) Formation of the postfusion structure of S2 that brings the two membranes together, facilitating formation of a fusion pore and viral entry.

It seems that a safe and effective vaccine represents the only viable option to reduce or eliminate the threat of SARS-CoV-2 adequately. The early round of vaccine candidates played with various forms of the S-protein, primarily inspired and modeled on those designed against the original SARS-CoV and MERS-CoV viruses.

However, some sobering insights surfaced from the development of SARS-CoV vaccines, suggesting that certain S-protein-based immunogens generate harmful immune responses to lungs or liver in animal models, as well as an immune enhancement or antibody-dependent enhancement of infectivity that makes the infection even more acute.

"It will be critical to define structural determinants that distinguish the ineffective or deleterious responses from the protective responses, to refine next-generation vaccine candidates," caution study authors. "Refined immunogens will be particularly critical if SARS-CoV-2 becomes seasonal and returns with antigenic drift, as do influenza viruses", they add.

Even though virus-encoded enzymes (such as proteases or RNA-dependent RNA polymerase) are outstanding therapeutic targets, fusion inhibitors that inhibit conformational changes of S-protein may also be budding drug candidates. Such fusion inhibitors may even be superior, as (akin to antibodies) they do not need to cross cell membranes in order to reach their target.

Furthermore, S-protein functions uniquely without any obvious cellular homologs, making it a more probable target for highly specific inhibitors with fewer side effects when compared to viral enzyme inhibitors. For example, the same research group has recently identified several small-molecule fusion inhibitors against HIV envelope spike, which was guided by a neutralizing antibody.

"Our work, which represents one of many complementary studies, may guide our responses to the spread of SARS-CoV-2 in a more rational way than would have been possible even a few months ago", conclude study authors. These insights may prove very useful for combined strategy in developing both the vaccine and an efficacious drug.

medRxiv publishes preliminary scientific reports that are not peer-reviewed and, therefore, should not be regarded as conclusive, guide clinical practice/health-related behavior, or treated as established information.

See more here:
Novel insights on SARS-CoV-2 cell entry may inform further vaccine and treatment development - News-Medical.Net

Agilex Biolabs Presents the Australian Advantage, including the World’s Largest Rebate on Clinical Trial Spend, at BIO Korea 2020 – Business News Asia

Agilex Biolabs, Australias largest specialist bioanalytical laboratory for biotech clinical trials, is presenting the Australian Advantage including the worlds most attractive rebate on clinical trials costs, at BIO Korea 2020.

The presentation by Kurt Sales (B.Sc; B.Sc (MED) Hons; M.Sc, Ph.D, PGCM)Director, Immunoassay at Agilex Biolabs, also detailed how well the Australian clinical trials industry had managed during the COVID-19 crisis with most sites, CROs, and labs including Agilex Biolabs, staying open.

The presentation is at 13.50 on Friday 22, May 2020 via the virtual conference platform.https://www.biokorea.org/index.asp

Agilex Biolabs also launched a News Video Update on the COVID-19 impact on clinical trials in Australia. Watch here.https://youtu.be/vZuHAYZ-GiE

Agilex Biolabs specializes in bioanalysis of small molecules and biologics for PK, immunogenicity, biomarkers and immunological pharmacodynamics assessments.

Agilex Biolabs, the only FDA-inspected lab of its type in the region, is located in Adelaide, South Australia in a science and biotech specialist hub.

Agilex Biolabs CEO Jason Valentine said: Aglilex Biolabs has just expand its labs by more than 30% to accommodate biotech demand from APAC and the USA.

He said more than 38% of Agilex Biolabs biotech clients already come from the APAC region primarily from China and South Korea.

APAC clients are particularly attracted by Agilex Biolabs FDA-inspected status, and the more than 40% rebate on clinical trial spend that applies in Australia, he said.

Our world-class bioanalytical facilities have OECD GLP Recognition with NATA (Australian Government OECD GLP Compliance monitoring authority) and ISO 17025 Accreditation for global recognition.

Agilex Biolabs specialises in bioanalysis of small molecules and biologics for PK, immunogenicity, biomarkers and immunological pharmacodynamics assessments utilising LC-MS/MS, immunoassay (Mesoscale, Gurolab, Luminex) and flow cytometry (BD FACSymphony A3, 20 colour cell analyser).

Agilex also offers pharmacodynamics services that include immunobiology services using the latest state-of-the-art technology to support immunology, cell biology and mode of action assays, including: Immunophenotyping Receptor occupancy Cytokine release assays (whole blood or PBMC stimulation assays) and cytokine/biomarker profiling PBMC assays and cellular mechanism of action assays (eg: ADCC)

The FDA-inspected facilities have more than 65 dedicated laboratory staff, and annually support more than 80 clinical trials. This year they will analyse more than 60,000 samples for pharma/biotechs from US, Europe and APAC.

Please Book a Briefing with us before you start your next clinical trial.https://calendly.com/agilexbiolabs/15min

Australia: +61 8 8302 8777 | China: +86 21 8036 9483 | South Korea: +82 80 812 1255 | USA: +1 800 247 1909

About Agilex Biolabshttps://www.agilexbiolabs.com/

Agilex Biolabs, Australias leading bioanalytical laboratory, has more than 20 years experience in performing regulated bioanalysis, including quality method development, method validation and sample analysis services. We have successfully supported hundreds of preclinical and clinical trials around the world where customers choose Australia for the streamlined regulatory process and access to the worlds most attractive R&D rebate of more than 40% on clinical trial work conducted in Australia.

We offer services for both small molecules and biologics for PK, immunogenicity (PD) and biomarker bioanalysis utilising the two platforms of LC-MS/MS and Immunoassay.

Agilex Biolabs operates a fully quality-assured laboratory ensuring that, within the principles of GLP, assays are validated to the latest FDA/EMA guidance and study samples are assayed and reported to the sponsors desired format using WATSON LIMS. Laboratory certifications include OECD GLP and ISO/IEC17025.

Our highly experienced team consists of over 65 dedicated laboratory staff with over 15 years average industry experience across senior scientists. Expertise includes development of robust compliant PK and PD assays de novo or by method transfer.

Our laboratory is fitted with 7 SCIEX tandem mass spectrometers, 6 API 4000 and 1 QTRAP 5500 as our standard platform for LC-MS/MS analysis. For immunoassay, we employ the state-of-the-art systems of Meso Scale Discovery (MSD) and Gyrolab xPlore.

Agilex Biolabs owns a suite of validated biosimilar assays that have already supported biosimilar studies executed in the region. These assays include Avastin/Bevacizumab, Lucentis/Ranibizumab, Actemra/Tocilizumab, Xolair/Omalizumab, Herceptin/Trastuzumab and Solaris/Eculizumab. We would like to discuss how we can support companies with biosimilar pipelines looking to conduct trials in the region. Our validated biosimilar assays require less setup saving time and money.

Agilex Biolabs also have a suite of biomarker assays that have been developed to support studies in the areas of endocrinology (estrone, estradiol, progesterone, testosterone and others).

Recently, Agilex Biolabs has added a comprehensive cannabinoid assay to its suite of validated assays to support clinical trials. The fully validated assay includes the five cannabinoids THC, CBD, CBN and the two THC-metabolites OHTHC and COOHTHC.

Our LC-MS/MS experience includes NCEs, sugars, nucleotides, enantiomers, steroids, prodrugs, peptides, immunosuppressants, nanoparticles, neurotransmitters, oligonucleotides and polymeric mixtures.

Our biologics experience entails PK analysis using colorimetric, fluorescence or chemiluminescence detection for recombinant or fusion proteins, monoclonal antibodies, ADCs, immunogenicity testing, biomarker analysis and PBMC blood stimulation assays.

See us featured in Endpointshttps://tinyurl.com/uqmkzcu

Media Contact:Kate NewtonMedia@AgilexBiolabs.com

[Got news tip or press release? Email us at editor@businessnewsasia.com]

Go here to see the original:
Agilex Biolabs Presents the Australian Advantage, including the World's Largest Rebate on Clinical Trial Spend, at BIO Korea 2020 - Business News Asia