Category Archives: Cell Biology

MSU’s Gregg Howe elected to the National Academy of Sciences – MSUToday

Michigan State University plant scientist Gregg Howe has been elected to the National Academy of Sciences, or NAS. Founded in 1863, the NAS is one of the oldest and most prestigious scientific membership organizations in the United States.

Howe is among 120 new members and 26 international members elected to the NAS in 2020 in recognition of their distinguished and continuing achievements in original research.

He joins 10 current and emeritus MSU faculty as members of NAS.

Professor Howe has made important contributions to our understanding of the complex biochemical mechanisms through which plants respond to challenges such as insect attack, said Stephen Hsu, senior vice president for research and innovation at MSU. His work informs fundamental questions in biology such as the evolutionary trade-off between defense and growth that are relevant to all organisms and also has applications to practical problems such as sustainable agriculture. Michigan State University is very proud of his accomplishments.

Howe, a University Distinguished Professor, MSU Foundation Professor and a member of both the MSU-DOE Plant Research Laboratory, or PRL, and the Plant Resilience Institute, is an internationally recognized leader in research on plant hormone biology and plant-insect interactions. Howe uses a combination of genetic, cell biological, molecular and biochemical analyses to study how plants use defensive compounds to protect themselves against herbivorous insects. His many honors and awards include selection as a fellow of American Association for the Advancement of Science and the American Society of Plant Biologists and being named a Clarivate Analytics Highly Cited Researcher for the past six years.

I am greatly honored to be elected into the National Academy of Sciences, said Howe, who also a professor in the Department of Biochemistry and Molecular Biology in the MSU College of Natural Science and an MSU AgBioResearch scientist. This recognition reflects the combined efforts of many talented students and collaborators over the years. I am also grateful for the very supportive research environment and terrific colleagues at MSU.

We are delighted about Gregg Howes election to the National Academy of Sciences, said Christoph Benning, PRL director. He has made outstanding contributions to science and the MSU community since his arrival here in 1997, and I congratulate him on behalf of the entire PRL community.

This years election brings the total number of active NAS members to 2,403 and the total number of international members to 501. International members are nonvoting members of the academy, with citizenship outside the United States.

For a complete list of the 2020 NAS cohort, available in July, visit the NAS website.

Read the original post:
MSU's Gregg Howe elected to the National Academy of Sciences - MSUToday

Advances in Research and Disease Modeling Expanding the Horizon of 3D Cell Culture Market – TMR BLOG

A paradigm shift from the conventional cell culture techniques to 3D cell culture is predicted to bring humongous growth for the 3D cell culture market across the forecast period of 2017-2025. From the validation of new targets and disease modeling to screening for safety and efficacy, 3D cell cultures bring the tremendous potential for the development of new drugs. They offer cutting-edge tools that assist in exploring the major factors of a particular disease.

In addition, 3D cell culture market gains mainly from the cancer segment as 3D models can mimic the interplay between the host environment and tumor for analyzing the efficacy of drug candidates in cancer patients. Greater biological relevance as compared to 2D monolayers is a prime advantage for an increase in the growth rate of the 3D cell culture market. In addition, 3D cell culture systems allow better cell interactions than done in 2D cell culture systems. Advances in microfabrication techniques, tissue engineering, and cell biology are supporting the development of varied 3D cell culture technologies.

The stem cell segment is anticipated to observe prominent growth during the forecast period of 2017-2025. Increased differentiation potential of stem cells under 3D cell culture may benefit the 3D cell culture market to a great extent. Pharmaceuticals and biotechnology sector expects to bring a larger revenue share throughout the forecast period owing to increasing demand for 3D cell culture systems for toxicity screening, drug discovery, and regenerative medicine.

Mergers, Acquisitions, and Partnerships Fuelling Market Growth

The advantages associated with the 3D cell culture system are resulting in frequent partnerships and acquisitions. Key players are involved in such activities to increase their footprint in the 3D cell culture market as well as speed up the process of new developments. zPREDICTA, a San Jose-based company, recently partnered with Labcorp for extending its customer base. zPREDICTAis a specialist in the development of tumor-specific 3D cell culture models and Labcorp is involved in operating a nationwide network of clinical laboratories. In another development, Nichirei Biosciences Inc. partnered with UPM Biomedicals to offer hydrogels that mimic the extracellular matrix (ECM) to encourage cell growth and differentiation. This development will help in bridging the gap between in vitro and in vivo studies by enabling 3D cell organoids for use in regenerative and personalized medicine. Similarly, Greiner Bio-One acquired Nano 3D Biosciences 3D cell culture technology.

New Insights and Developments Accelerating the Growth Rate

Disease modeling is an essential component for the discovery of a drug-related to a specific disease or disorder. With the help of 3D bioprinting and tissue engineering techniques, it is possible to unravel drug discovery. Therefore, this aspect can propel the growth rate of the 3D cell culture market.

For instance, the COVID-19 outbreak is motivating key players in the 3D cell culture market to come up with new developments and technologies for novel treatments that can tackle the deadly disease. For instance, the SARS-CoV-2 pandemic prompted Inventialifescience, an Australia based startup, to create 3D lung microtissues for drug testing. This is being developed through the companys bioprinting platform, Rastrum, which is being used to print human cells with cancer drug testing at a rapid rate.

To Get an Exhaustive Overview about the Competition in 3D Cell Culture Market, Request for a Report Brochure at https://www.transparencymarketresearch.com/sample/sample.php?flag=B&rep_id=35834

From the regional perspective, developed regions like North America and Europe are predicted to show an increasing growth curve across the forecast period, with increasing cancer cases and rising investment in research and development activities being the prime factors for growth. Asia Pacific also expects steady growth due to considerable demand for 3D cell culture systems in the developing regions.

Key players involved in the 3D cell culture market are ReproCELl, Thermo Fisher Scientific Inc., Global Cell Solutions, Hamilton Company, Kuraray Co. Ltd., and Merck.

Like Loading...

View original post here:
Advances in Research and Disease Modeling Expanding the Horizon of 3D Cell Culture Market - TMR BLOG

Researchers identify transcription factor that preserves the genomic integrity of germline – News-Medical.net

Reviewed by Emily Henderson, B.Sc.May 7 2020

The germline is the cell lineage of an organism that passes on its genetic material to its progeny. Genetic damage to the germline can cause developmental defects and even death of that same progeny. It is thought that biological mechanisms exist that ensure that aberrant germline cells are eliminated to maintain germline integrity, although the specific molecular basis for this is unknown.

In a new study published in Communications Biology, researchers from the University of Tsukuba identified the transcription factor Myc as a central molecular actor within the process of preserving the genomic integrity of the germline after DNA damage.

To achieve their goal, the researchers studied the fruit fly Drosophila to understand how germline integrity is maintained. This fly model presents a sterility syndrome called P-M (paternal-maternal) hybrid dysgenesis (HD), which results from a high rate of mutations and rearrangements in the DNA, leading to germline-loss and sterility.

At the molecular level, so-called P-elements are responsible for HD. P-elements are DNA segments of which the protein transposase. Transposases have an ability known as P-element mobilization wherein they move segments of DNA, which leads to mutations and DNA instability.

When male fruit flies carrying P-elements are crossed with females lacking P-elements, P-element mobilization and thus DNA damage occurs in their progeny, resulting in sterility.

The Drosophila P-M hybrid dysgenesis model has been known for decades, but the molecular basis of the resulting sterility is still not fully understood. The goal of our study was to further our understanding of the molecular mechanisms governing the elimination of damaged germline cells during reproduction.

Interestingly, in a separate set of experiments, we found that knockdown of the transcription factor Myc resulted in a similar germline-loss phenotype that we observe in hybrid dysgenesis. We wanted to know how Myc and hybrid dysgenesis were interconnected in the process of maintaining germline integrity."

Satoru Kobayashi, Study Corresponding Author and Professor

The researchers first investigated the number of germline cells at different embryonic stages in HD progeny and in normal flies that did not produce Myc.

They found that in both models the number of germline cells decreased at a similar stage of embryonic development, suggesting that both processes are connected. The researchers then followed the expression of Myc in HD progeny and found that it was reduced in germline cells before the number of germline cells decreased, suggesting that HD causes Myc downregulation to result in a germline-loss phenotype.

They then examined what happens if Myc continued to be expressed in HD progeny by overexpressing the transcription factor. While Myc overexpression in the HD germline led to increased germline cell numbers, the resulting germline exhibited a higher DNA mutation frequency and a lower capacity to develop into adulthood.

"These are striking results that show that Myc-dependent germline cell reduction serves to eliminate aberrant germline cells in which the genetic material has been damaged," says Professor Kobayashi. "We think that Myc is a central molecular actor in this process, serving as a quality-control during embryonic development."

Source:

Journal reference:

Ota, R & Kobayashi, S. (2020) Myc plays an important role in Drosophila P-M hybrid dysgenesis to eliminate germline cells with genetic damage. Communications Biology. doi.org/10.1038/s42003-020-0923-3.

Here is the original post:
Researchers identify transcription factor that preserves the genomic integrity of germline - News-Medical.net

What is the Secretome? – News-Medical.net

Biological organisms are incredibly complex machines made up of innumerable parts working in complementary ways. Understanding the complex, intricate interactions between tissues and cells is, therefore, key to a holistic understanding of the organism as a whole, and over the centuries that have encompassed the science of biology many advances in scientific knowledge have been made.

Image Credit: Kateryna Kon/Shutterstock.com

One recent area of research that has been gaining prominence in the understanding of the complex inter-and-intracellular dynamics of biological organisms is the science of secretomics, a type of proteomics concerned with the study of the secretome.

The secretome as a term was coined in 2004 by Tjalsma et al. It denotes all the factors secreted by the cell, along with the constituents of the secretory pathway. The term was revised in 2010 to include specifically the proteins which are secreted into the extracellular space.

Related terms include the matrisome (the subset which includes extracellular matrix proteins and associated proteins) receptome (membrane receptors) and the adhesome (proteins involved in cell adhesion.)

Cells such as endocrines and B-lymphocytes are two classes of cells that are specifically involved in the secretion of proteins. The liver and bone marrow are major secretory organs in the human body.

In humans, secreted proteins account for between 13 and 20% of the entire proteome (the entire set of proteins expressed in an organism, cell, tissue, or genome at any given time.) Included in the secretome are such things as growth factors, coagulation factors, adhesion molecules, cytokines, proteases, and chemokines.

A significant number (36%) of the 19670-human protein-coding genes transcribe secretory proteins. Other proteins coded for by these genes are membrane-spanning, intracellular (the main type 12631 genes encode for these) and 9% have at least one secreted protein product as well as membrane-spanning products. The secretome plays a major role in cell migration, signaling, and communication.

Several methods are used by researchers to study the secretome. These include:

Image Credit: science photo/Shutterstock.com

Each method has its distinct advantages and disadvantages for studying and identifying secretory proteins. A knowledge of them is advantageous for any study using secretomics.

The growing body of research on the secretome is proving more and more important to the field of modern medical research. Many medically important secreted proteins include coagulation factors, cytokines, and growth factors and they play a myriad of physiological and pathological roles within the body.

An understanding of the secretory pathways confers knowledge of how these secretory proteins play a role in the healthy biological processes of an individual.

All cells secrete proteins to varying degrees. The study and analysis of these proteins are providing valuable sources for new drugs and therapeutics. Not only this, but a large percentage of clinical blood diagnostic tests are directed at secretory proteins.

Medically important secretory proteins include:

The secretome is still poorly understood and represents a very recent area of research in the study of intercellular pathways in the body. Secretory proteins play a role which is becoming more apparent to medical scientists as the field of secretomics evolves.

Knowledge of how the secretome functions and which pathways are specifically involved is therefore of paramount importance and will no doubt contribute to the ongoing development of novel drugs and therapeutics.

More here:
What is the Secretome? - News-Medical.net

UB investigators uncover cellular mechanism involved in Krabbe disease – UB Now: News and views for UB faculty and staff – University at Buffalo…

A group of UB researchers have published a paper that clarifies certain cellular mechanisms that could lead to improved outcomes in patients with globoid cell leukodystrophy, commonly known as Krabbe disease.

The paper, titled Macrophages Expressing GALC Improve Peripheral Krabbe Disease by a Mechanism Independent of Cross-Correction, was published May 5 in the journal Neuron.

The research was led by Lawrence Wrabetz and M. Laura Feltri. Wrabetz and Feltri head the Hunter James Kelly Research Institute and both are professors in the departments of Biochemistry and Neurology in the Jacobs School of Medicine and Biomedical Sciences at UB.

The institute is named for the son of former Buffalo Bills quarterback Jim Kelly. Hunter Kelly died at age 8 in 2005 from complications of Krabbe disease.

Krabbe disease is a progressive and fatal neurologic disorder that usually affects newborns and causes death before a child reaches the age of 2 or 3.

Traditionally, hematopoietic stem cell transplantation, also known as a bone marrow transplant, has improved the long-term survival and quality of life of patients with Krabbe disease, but it is not a cure.

It has long been assumed that the bone marrow transplant works by a process calledcross-correction, in which an enzyme called GALC is transferred from healthy cells to sick cells.

Using a new Krabbe disease animal model and patient samples, the UB researchers determinedthatin reality cross-correctiondoes not occur. Rather, the bone marrow transplant helps patients through a different mechanism.

The researchers first determined which cells are involved in Krabbe disease and by which mechanism. They discovered that both myelin-forming cells, or Schwann cells, and macrophages require the GALC enzyme, which is missing in Krabbe patients due to genetic mutation.

Schwann cells require GALC to prevent the formation of a toxic lipid called psychosine, which causes myelin destruction and damage to neurons. Macrophages require GALC to aid with the degradation of myelin debris produced by the disease.

The research showed that hematopoietic stem cell transplantation does not work bycross-correction, but by providing healthy macrophages with GALC.

According to Feltri, the data reveal that improvingcross-correctionwould be a way to makebone marrow transplants and other experimental therapies such as gene therapy more effective.

Bone marrow transplantation and other treatments for lysosomal storage disorders, such as enzyme replacement therapy, have historically had encouraging but limited therapeutic benefit, says study first author Nadav I. Weinstock, an MD-PhD student in the Jacobs School. Our work defined the precise cellular and mechanistic benefit of bone marrow transplantation in Krabbe disease, while also shedding light on previously unrecognized limitations of this approach.

Future studies, using genetically engineered bone marrow transplantation or other novelapproaches,may one day build on our findings and eventually bridge the gap for effectively treating patients with lysosomal disease, he continues.

UB investigators included Daesung Shin, research assistant professor at the Hunter James Kelly Research Institute; Nicholas Silvestri, clinical associate professor of neurology, Jacobs School; Narayan Dhimal, PhD student; Chelsey B. Reed, MD-PhD student; and undergraduate student Oliver Sampson.

Also participating in the research were Eric E. Irons, MD-PhD student, and Joseph T.Y. Lau, a distinguished faculty member from the Department of Molecular and Cellular Biology at Roswell Park Comprehensive Cancer Center.

The research was funded by multiple grants from the National Institutes of Health awarded to Weinstock, Shin, Wrabetz and Feltri, and also supported by Hunters Hope.

Read the original:
UB investigators uncover cellular mechanism involved in Krabbe disease - UB Now: News and views for UB faculty and staff - University at Buffalo...

AVROBIO and Magenta Therapeutics Announce Collaboration to Evaluate Targeted Antibody-Drug Conjugate as a Potential Conditioning Regimen for…

CAMBRIDGE, Mass.--(BUSINESS WIRE)--AVROBIO, Inc. (Nasdaq: AVRO) and Magenta Therapeutics (Nasdaq: MGTA) today announced a research and clinical collaboration agreement to evaluate the potential utility of MGTA-117, Magentas novel targeted antibody-drug conjugate (ADC) for conditioning patients before they receive one of AVROBIOs investigational lentiviral gene therapies.

The collaboration will combine AVROBIOs leadership in lentiviral gene therapies with Magentas expertise in ADC-based conditioning and is expected to further the two companies shared mission to enable patients to live free from disease. Under the collaboration, AVROBIO and Magenta will jointly evaluate MGTA-117 in conjunction with one or more of AVROBIOs investigational gene therapies. Magenta will retain all commercial rights to MGTA-117. AVROBIO will retain all commercial rights to its gene therapies and will be responsible for the clinical trial costs related to the evaluation of MGTA-117 with AVROBIOs gene therapies.

This agreement with Magenta springs from our strategic focus on maintaining technology leadership in gene therapy, said Geoff MacKay, AVROBIOs president and CEO. AVROBIO has always led by investing early in technological innovations that further the field of lentiviral gene therapy, such as plato, our proprietary platform designed to optimize the safety, potency and durability of our investigational lentiviral gene therapies. Were continually assessing new technologies that could be complementary to our plato platform to sustain our cutting-edge advantage and continue to evolve platos capabilities.

We believe targeted ADCs represent the next generation of medicines to prepare patients for gene therapy or transplant in a targeted, precise way. AVROBIOs investigational gene therapies complement our platform as well as our focus and commitment to patients. This partnership will allow Magenta to validate our conditioning platform in lentiviral gene therapy applications, said Jason Gardner, D.Phil., president and chief executive officer, Magenta Therapeutics. Weve selected ADCs as the preferred modality for our conditioning programs, as we believe they offer the most promising option for more patients. We have optimized our ADCs for gene therapy and transplant settings and look forward to collaborating with AVROBIO to evaluate MGTA-117 in specific gene therapy settings. Magenta will continue to develop MGTA-117 in other diseases, including blood cancers and genetic diseases.

MGTA-117, Magentas most advanced conditioning program, is a CD117-targeted antibody engineered for the transplant setting and conjugated to amanitin, a toxin in-licensed from Heidelberg Pharma. It is designed to precisely deplete only hematopoietic stem and progenitor cells and has shown high selectivity, potent efficacy, wide safety margins and broad tolerability in non-human primate models, suggesting that it may be capable of clearing space in bone marrow to support long-term engraftment and rapid recovery in humans. Magenta plans to complete IND-enabling studies this year.

AVROBIO currently uses a personalized conditioning regimen with precision dosing of busulfan, an extensively validated conditioning agent generally considered to be the gold standard for ex vivo lentiviral gene therapy, based on decades of general use and administration to hundreds of patients treated with lentiviral gene therapy candidates. The treating clinician uses therapeutic drug monitoring (TDM) to evaluate how quickly the patient metabolizes busulfan and adjusts the dose regimen accordingly with the goal of achieving the optimum result. AVROBIO has reported early clinical data with this precision conditioning regimen with TDM in its own clinical trials, adding to a body of data that suggest busulfan can effectively clear space in the patients bone marrow, where stem cells engraft, produce generations of daughter cells carrying the therapeutic gene and make the functional protein the patient needs to maintain cellular health.

About AVROBIO

Our mission is to free people from a lifetime of genetic disease with a single dose of gene therapy. We aim to halt or reverse disease throughout the body by driving durable expression of functional protein, even in hard-to-reach tissues and organs including the brain, muscle and bone. Our clinical-stage programs include Fabry disease, Gaucher disease and cystinosis and we also are advancing a program in Pompe disease. AVROBIO is powered by the plato gene therapy platform, our foundation designed to scale gene therapy worldwide. We are headquartered in Cambridge, Mass., with an office in Toronto, Ontario. For additional information, visit avrobio.com, and follow us on Twitter and LinkedIn.

About Magenta Therapeutics

Magenta Therapeutics is a clinical-stage biotechnology company developing medicines to bring the curative power of immune system reset through stem cell transplant to more patients with autoimmune diseases, genetic diseases and blood cancers. Magenta is combining leadership in stem cell biology and biotherapeutics development with clinical and regulatory expertise, a unique business model and broad networks in the stem cell transplant world to revolutionize immune reset for more patients. Magenta is based in Cambridge, Mass. For more information, please visit http://www.magentatx.com. Follow Magenta on Twitter: @magentatx.

AVROBIO Forward-Looking Statements

This press release contains forward-looking statements, including statements made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995. These statements may be identified by words and phrases such as aims, anticipates, believes, could, designed to, estimates, expects, forecasts, goal, intends, may, plans, possible, potential, seeks, will, and variations of these words and phrases or similar expressions that are intended to identify forward-looking statements. These forward-looking statements include, without limitation, statements regarding our business strategy for and the potential therapeutic benefits of our prospective product candidates, the design, commencement, enrollment and timing of ongoing or planned clinical trials, clinical trial results, product approvals and regulatory pathways, anticipated benefits of our gene therapy platform including potential impact on our commercialization activities, timing and likelihood of success, the expected benefits and results of our implementation of the plato platform in our clinical trials and gene therapy programs, the expected safety profile of our investigational gene therapies, and the potential and expected benefits of MGTA-117, Magentas investigational antibody-drug conjugate, including the ability of MGTA-117 to deplete hematopoietic stem and progenitor cells in order to clear space in bone marrow to support long-term engraftment in humans, as well as MGTA-117s potential application to AVROBIOs investigational gene therapies. Any such statements in this press release that are not statements of historical fact may be deemed to be forward-looking statements. Results in preclinical or early-stage clinical trials may not be indicative of results from later stage or larger scale clinical trials and do not ensure regulatory approval. You should not place undue reliance on these statements, or the scientific data presented.

Any forward-looking statements in this press release are based on AVROBIOs current expectations, estimates and projections about our industry as well as managements current beliefs and expectations of future events only as of today and are subject to a number of risks and uncertainties that could cause actual results to differ materially and adversely from those set forth in or implied by such forward-looking statements. These risks and uncertainties include, but are not limited to, the risk that any one or more of AVROBIOs product candidates will not be successfully developed or commercialized, the risk of cessation or delay of any ongoing or planned clinical trials of AVROBIO or our collaborators, the risk that AVROBIO may not successfully recruit or enroll a sufficient number of patients for our clinical trials, the risk that AVROBIO may not realize the intended benefits of our gene therapy platform, including the features of our plato platform, the risk that AVROBIO may not realize the intended benefit of MGTA-117 with respect to AVROBIOs investigational gene therapies, the risk that our product candidates or procedures in connection with the administration thereof will not have the safety or efficacy profile that we anticipate, the risk that prior results, such as signals of safety, activity or durability of effect, observed from preclinical or clinical trials, will not be replicated or will not continue in ongoing or future studies or trials involving AVROBIOs product candidates, the risk that we will be unable to obtain and maintain regulatory approval for our product candidates, the risk that the size and growth potential of the market for our product candidates will not materialize as expected, risks associated with our dependence on third-party suppliers and manufacturers, risks regarding the accuracy of our estimates of expenses and future revenue, risks relating to our capital requirements and needs for additional financing, risks relating to clinical trial and business interruptions resulting from the COVID-19 outbreak or similar public health crises, including that such interruptions may materially delay our development timeline and/or increase our development costs or that data collection efforts may be impaired or otherwise impacted by such crises, and risks relating to our ability to obtain and maintain intellectual property protection for our product candidates. For a discussion of these and other risks and uncertainties, and other important factors, any of which could cause AVROBIOs actual results to differ materially and adversely from those contained in the forward-looking statements, see the section entitled Risk Factors in AVROBIOs most recent Annual or Quarterly Report, as well as discussions of potential risks, uncertainties and other important factors in AVROBIOs subsequent filings with the Securities and Exchange Commission. AVROBIO explicitly disclaims any obligation to update any forward-looking statements except to the extent required by law.

Magenta Therapeutics Forward Looking Statements

This press release may contain forward-looking statements and information within the meaning of The Private Securities Litigation Reform Act of 1995 and other federal securities laws, including, without limitation, statements regarding the research and clinical collaboration agreement between Magenta and AVROBIO, including the timing, progress and success of the collaboration contemplated under the agreement, the successful evaluation MGTA-117 in conjunction with one or more of AVROBIOs investigational gene therapies under the agreement, the anticipated cost allocation and other commercial terms under the agreement, Magentas strategy and business plan, as well as the future development, manufacture and commercialization between AVROBIO and Magenta. The use of words such as may, will, could, should, expects, intends, plans, anticipates, believes, estimates, predicts, projects, seeks, endeavor, potential, continue or the negative of such words or other similar expressions can be used to identify forward-looking statements. The express or implied forward-looking statements included in this press release are only predictions and are subject to a number of risks, uncertainties and assumptions, including, without limitation, risks set forth under the caption Risk Factors in Magentas most recent Annual Report on Form 10-K, as updated by Magentas most recent Quarterly Report on Form 10-Q and its other filings with the Securities and Exchange Commission, as well as risks, uncertainties and assumptions regarding the impact of the COVID-19 pandemic to Magentas business, operations, strategy, goals and anticipated timelines, including, without limitation, Magentas ongoing and planned preclinical activities, ability to initiate, enroll, conduct or complete ongoing and planned clinical trials, timelines for regulatory submissions and financial position. In light of these risks, uncertainties and assumptions, the forward-looking events and circumstances discussed in this press release may not occur and actual results could differ materially and adversely from those anticipated or implied in the forward-looking statements. You should not rely upon forward-looking statements as predictions of future events. Although Magenta believes that the expectations reflected in the forward-looking statements are reasonable, it cannot guarantee that the future results, levels of activity, performance or events and circumstances reflected in the forward-looking statements will be achieved or occur. Moreover, except as required by law, neither Magenta nor any other person assumes responsibility for the accuracy and completeness of the forward-looking statements included in this press release. Any forward-looking statement included in this press release speaks only as of the date on which it was made. We undertake no obligation to publicly update or revise any forward-looking statement, whether as a result of new information, future events or otherwise, except as required by law.

View source version on businesswire.com: https://www.businesswire.com/news/home/20200506005265/en/

Go here to see the original:
AVROBIO and Magenta Therapeutics Announce Collaboration to Evaluate Targeted Antibody-Drug Conjugate as a Potential Conditioning Regimen for...

Researchers shed new light on the structure of RNA – News-Medical.Net

Australian and US researchers have made a breakthrough in understanding the structure of a key genetic molecule, called RNA, and revealing for the first time how these changes impact RNA's function.

Publishing in the journal Nature, the research team developed a bioinformatics technique to resolve separate structures of RNA rather than viewing them as a 'blur' that averaged multiple structures. This underpinned their discovery that the structure of RNA can influence how cells function.

RNA is a DNA-like molecule that encodes genetic information. Certain viruses - including HIV and SARS-CoV2 - use RNA as their genetic material. The team were able to apply the techniques they developed to reveal how the structure of HIV's RNA genome influences which proteins the virus produces.

The international collaborative team was led by Walter and Eliza Hall Institute researcher Dr Vincent Corbin together with Mr Phil Tomezsko and Professor Silvi Rouskin at the Whitehead Institute for Biomedical Research, Boston (US). The research team also included the Institute's Computational Biology Theme Leader Professor Tony Papenfuss and mathematician and PhD student Mr Lachlan McIntosh.

At a glance

RNA is a molecule found in all living things that carries genetic information. RNA is an important regulator of how cells function, directly controlling which proteins are produced in cells, and can also switch genes on and off.

RNA molecules have a two-dimensional structure which influences how the genetic information contained within them can be accessed, said Dr Corbin, who led the project's bioinformatics research.

"The big question in RNA biology has been whether RNA molecules have a single, constant structure, or whether they can shift between different structures - and what this means for the function of a particular RNA molecule," he said.

"Our collaborators, led by Professor Silvi Rouskin, developed a technique for deciphering the structure of RNA molecules. We wanted to understand whether what we were detecting was a single structure of RNA, or an 'average' structure that blurs multiple different structures together.

"It's a bit like seeing red and yellow stripes, or blurring them together and thinking you can see orange," he said.

By developing a computational algorithm, the team were able to detect and measure the amount of different RNA structures.

We could detect these both in a test tube and in living cells, so we next looked at whether these structures influenced how RNA functioned."

Dr Vincent Corbin, Project Leader, Bioinformatics Research, Walter and Eliza Hall Institute

When RNA is produced in cells, it starts in a longer form that is 'spliced' or trimmed to remove unwanted parts.

"RNA splicing can influence how it encodes proteins," Dr Corbin said. "There are many examples of how altered RNA splicing influences how a cell functions - and in some cases, changes in RNA splicing have been associated with cancer or neurodegenerative diseases."

Certain viruses use RNA for their genome, including HIV and SARS-CoV2 (the coronavirus that causes COVID-19). In the case of HIV, RNA splicing influences which protein the virus produces - which changes at different stages of the virus's lifecycle.

"Using the HIV genome as a model system, we looked at whether RNA structure influences how HIV's RNA is spliced. We discovered that RNA structure was a critical determinant of RNA splicing in HIV, and influenced which viral proteins were produced," Dr Corbin said.

"This is the first clear evidence of how RNA structure can control RNA function. The techniques we have developed have opened up a new field of research into the role of RNA structure in regulating the function of cells."

Professor Papenfuss said the research showed how finely tuned biological systems are. "This study how very subtle changes in one tiny molecule can have big implications for the function of a virus. By using computational biology to unravel these changes, we've made a significant discovery about how viruses - and potentially human cells - function, which may underpin future discoveries about health and disease."

Source:

Journal reference:

Tomezsko, P. J., et al. (2020) Determination of RNA structural diversity and its role in HIV-1 RNA splicing. Nature. doi.org/10.1038/s41586-020-2253-5.

Read more from the original source:
Researchers shed new light on the structure of RNA - News-Medical.Net

The Role of Microphysiological Systems for Oncology and Stem Cell Research – News-Medical.net

Sponsored Content by PittconMay 6 2020

In this interview, Dr. Nancy Allbritton from The Allbritton Lab talks to News-Medical and Life Sciences about the revolutionary technology and techniques for the application of new technologies for oncology and stem cell research.

Dr. Nancy Allbritton provides insight into the organ-on-a-chip and the ability it has to monitor and control the environment at the cellular and tissue level is one of the most promising applications for microengineered systems. Dr Allbritton will be presenting her research and findings at Pittcon 2020 in Chicago.

Microphysical systems seek to replicate the smallest functioning unit of an organ. For intestine-on-a-chip, it might be large or small intestinal crypts. For a heart, it may be a section of contractile muscle. For the liver, it may be a liver lobule or group of lobules. It's a group of interconnected cells so that they are not just behaving as a single cell or just a small cluster, but they began to show higher-order behavior and functioning.

I think one of the reasons microphysical systems are becoming increasingly important is that we've had a lot of breakthroughs in stem cell technology. The ability to grow human stem cells from different organs is now present, as well as the ability to create a tissue of differentiated cell types from primary stem cells or organ-specific cells from induced pluripotent stem cells.

Right now, it is the perfect time for this technology, because we have all the enabling microfabrication methods and stem cell biology innovations coming together. Another reason organs-on-chips are becoming important is the ability to grow human tissue. It's very hard to do experiments on the human population and get a good representation of the population, either because people aren't volunteering or because there just aren't enough people in that particular group of individuals. With the chip, you can begin to sample population-wide tissue variation. Organs on chips can also be a better way to test drugs. Rather than testing cells in a dish, which are usually tumor cells and very abnormal in their growth and other characteristics, organs-on-chips constructed from normal human cells can be used to get a more accurate representation of how drugs will affect humans. Additionally, they may outperform murine experiments in many ways since humans are not simply 70 kg mice.

We have been able to cure mice of all sorts of diseases, but much of this work hasnt translated to humans. So, while that was a good screening technology, a lot of drugs got through that were quite toxic to humans, and then a lot of drugs that may have worked well in humans, but were toxic to mice got blocked.

Image Credit:Shutterstock/royaltystockphoto.com

Microphysiologic devices should maximize the number of good drugs that could work well in humans. You can hopefully also get higher value information and create a much better drug pipeline. I think doing high-throughput screening on organ subunits on microfabricated devices can complement drug screening in humans. The idea is that you may be able to cut costs in several ways;

I believe you can also begin to develop human disease models. You can create mouse models of human disease, but they almost never completely recapitulate human disease. Even when it is a simple genetic mutation, the mice can often be asymptomatic or present with different symptoms and outcomes to that mutation. With the organ-on-a-chip, you can have functioning human tissue that mimics many of the symptoms of human diseases. Even more exciting, one can begin to put, for example, an intestine-on-chip coupled with the human gut microbiome or the gut flora and begin to understand how the tissue and microbiota interact.

This is important because our gut flora is very different from that of a mouse and other model organisms. We now know that intestinal microbes have a tremendous physiologic impact throughout our body including metabolism and mentation, our feeding behavior, and these are aspects of human behavior or physiology that can't be recapitulated in an animal model or cells in a dish.

Eventually, we'll be able to string human organs together as human organ-on-chip systems. For example, food is added to the gut model and absorbed, the nutrients travel to the liver, the liver metabolizes the absorbed compounds sending them out to the body as a whole including the brain and heart. The organs must be tied together to see the full effect in which the behavior or functioning of one organ impacts other connected organs. That will never replace a fully functioning human, but I think the expectation is that it may end up being a lot more accurate and, in some ways, cheaper than mice or other model mammalian systems.

The organ-on-a-chip systems will help us understand basic biology and the basic physiology of the human.

Microphysiologic devices depend on many fields of chemistry for their advancement including synthetic organic, polymer and analytical chemistry. We need more synthetic matrices to support cells and tissues including smart polymers and scaffolding materials that will support, direct and shape these organ systems. Right now, people often use matrices derived from native biologic materials, like collagen or matrigel, which are expensive and not fully defined. Polymer chemists and synthetic chemists are working hard to develop novel materials and matrices while analytical chemists, as well as engineers, are developing materials and methods for microdevice fabrication, sensor enhancements, and other device-related innovations.

Microphysiologic systems will clearly need an array of embedded and external sensors to monitor their health and well-being as well as pathophysiologic attributes. They will need robust, reliable (and often miniaturized) embedded sensors for oxygen consumption, glucose concentration, CO2 production, pH, and other chemical and physical attributes. Importantly the sensors should not perturb the system. These sensors will likely require external instrumentation or detection methods to monitor the embedded sensors, for example, RFID or optical readouts.

For engineering and chemistry, these are going to be huge areas where many can contribute to moving the field forward. For engineering, specifically, developing ways to make integrated systems that are efficient, low cost, manufacturable, shippable, self-contained, and can talk to each other will be important. This is an area where engineering and chemistry must work together in a team approach to move the field forward. It's going to take both disciplines to really advance the field.

Microfabricated technology refers to devices developed with micron-sized features. Cells have diameters on the order of 10 microns and many organ subunits span hundreds of microns so that micron-sized architectural features are required to recapitulate key features. For example, large intestinal crypts (the key physiologic subunit of the large intestine) is approximately 400 microns in length and 100 microns wide with the stem cell niche spanning tens of microns. Thus, microfabrication methods are perfect for rebuilding many of the key architectural features of the large intestine.

When it comes to rebuilding architectural features such as the lare intestine, microfabrication methods are perfect for this. Image Credit:Shutterstock/nobeastsofierece

We do intestine-on-a-chip, which is mostly the large intestine and have several platforms of varying complexity. The simplest is the human intestinal epithelial monolayers that have a stem and/or differentiated cells. These can be used to assess how the intestine transports and metabolizes drugs and nutrients or how the stem cells differentiate into mucus-producing, hormone-producing, or absorptive cell types. These systems are designed to be higher throughput model systems that are simple but do not possess the greatest possible information content. However, they are robust and reliable. We also have complex 3D tissues that replicate a wide range of physiologic behaviors as well as the architectural features of the human intestine. Importantly many of these systems will support the wide array of chemical and gas gradients found in the human intestine. They can also host the human microbiome so that a better understanding of the complex interplay between the human cells and microbes can be developed in both health and disease.

As with all of our model systems, we usually advise users of our systems to employ the simplest platform possible and then add in complexity as needed for the task at hand.

We have more complex platforms that are designed to replicate a lot of the different features of the human intestine, such as the architecture, cell migration behaviors, and stem cell fate decisions. These are shaped three-dimensional systems that exist as arrays of crypts (or microwells) covered with a monolayer of intestinal epithelial cells. The crypt array has a basal surface for a nutrient diffusion, and a lumen, like the inside of the intestine.

You can imagine the cells on the inside of the intestine, facing the waste or food, are very different from the cells that are at the base of the intestinal crypts. In these systems, the stem cells are found at the crypt or microwell base while the differentiated, mature cells such as absorptive cells are found facing the luminal surface, i.e. waste side. These systems are chemically and architecturally much more faithful duplicates of the human intestine than the simpler systems, but that also means they're a little harder to create, build, and maintain. With some of these three-dimensional systems, we create chemical gradients across the tissues, i.e. the long axis of the intestinal crypt, just as stem cell factors and food/bacterial metabolites exists in a gradient across your intestinal crypts. This microdevice permits very sophisticated micro-environmental control of these complex tissues.

We can do a lot of it, but we can't do it all. For example, with the chemical gradients, the stem cells that sit at the base of these microwells are exposed to very high growth factors, but about 400 microns away, so a few hair diameters, the cells don't see much of these factors. This chemical gradient mimics the signaling microenvironment of the intestine very closely. There are also all sorts of bacterial metabolites and products that are at very high concentration for the cells at the luminal surface, but low where the stem cells are down at the base of the microwells or crypts. We can also replicate these food and bacterial metabolite gradients by using purified compounds or actual bacteria on the luminal side of the intestine on a chip.

This 3D system begins to duplicate the human physiologic signals that control the cells behavior, but there are some things we cannot do yet. We don't have a blood supply, for example, or blood vessels going into our chip. Thats one of the next steps, but there is a lot of work to do to build it, yet still have a robust and reliable intestine-on-chip system. Our tissue is growing in complexity, but we still only have 1 to 2 tissues on the devices and a small subset of the bacteria found in the human intestine. One could imagine increasingly putting more and more tissue types. The intestine has epithelial cells, fibroblasts, muscle cells, neural cells, immune cells, all sorts of other cell types, and adding in all of those cell types would create a more normal physiologic organ. I think those are all goals for the future on which we will be moving forward.

Our intestinal-on-chip technologies can be used to understand drug transport and metabolism by humans (as opposed to mice or tissue-cultured tumor cells). Additionally, the microbes in the human intestine or the human intestinal epithelium can convert drugs into their active form or into a toxic metabolite. Along that line, one could begin to do rapid screens as to how drugs are modified, metabolized and transported across the intestinal epithelium. Importantly many drugs cause intestinal dysfunction or side effects and screens can be performed for the impact of the drugs on the intestinal cells themselves and the impact on intestinal barrier function, e.g. leakiness.

As an example, it's now also clear that in cancer chemotherapy, the gut and the bacteria within the gut play a huge role in how well the chemotherapy is working. How this works is not well understood, so I think there's going to be a big push to use these systems to understand how we can make anti-cancer drugs that are more effective and less toxic on the gut and other biological systems. We are only beginning to scratch the surface.

Image Credit:Shutterstock/ nobeastsofierce

I dont think microphysiological systems can ever totally replace animal models. It's the law in the US to test drugs on animals. We are also a long way off from having a full human-on-a-chip with all of the different organ systems in place and interconnected, whereas a mouse or other animal system is already there. I think it's more likely that the chips will reduce the number of animals used so that you can get higher value information, thereby complementing rather than replacing animal models. The chips will also enable insights as to how humans might ultimately respond differently from animal model systems.

On-going work in the organ-on-chip area is to demonstrate that these devices actually mimic and replicate human responses (and many have already been shown to mimic human physiology when animal models failed). Overall the future for organ-on-chip technologies appears quite bright and will undoubtedly grow in future importance and impact.

There are already devices that demonstrate many of the complex disease phenotypes. For example, blood vessels on-chip have replicated blood vessel diseases such as atherosclerosis and tumor metastases. There are some amazing heart-on-chip devices with functioning cardiac tissue subunits that accurately recapitulate the impact of cardiovascular drugs, so I think we are making good progress. I think that we will see a growing number of impressive and highly predictive disease models as time goes by.

Near term, our big goal is to really make an intestine-on-a-chip that fully replicates the human small and large intestines. We're working hard at putting a human microbiome or normal intestinal bacteria on our intestine-on-chip. In addition to chemical gradients, gas gradients such as oxygen exist across the intestinal crypts which we're working hard at replicating. We really want to add in more tissue types, such as the immune system, fibroblasts, and nervous tissue. We've got quite a bit of work to do to make a fully functioning replica so we are also starting to team up with other people, particularly liver-on-a-chip people so that our intestine-on-a-chip can absorb food and then send it to their liver-on-chip to create a fully functioning liver-intestinal model to recapitulate food digestion, metabolism, and detoxification.

A big part of our work is to make the systems robust and reliable as well as easy to use for biologists and clinical investigators. That may sound trivial, but it's not. A lot of great devices remain isolated in the inventors' lab due to their high degree of complexity. When biologists try to use these complex devices, there are just too many failure points. Even to make one of these systems shippable will be a challenge, because how do you give it to FedEx, yet arrive in good condition across the world? There are a lot of challenges with scalability, manufacturing, robustness, and reliability that my lab, in particular, is interested in tackling to make sure that these organ-on-chips get out into the real world and fulfill their potential.

To find out more please visit http://allbritton.web.unc.edu/

Dr. Allbritton is a Professor in the Department of Bioengineering and the Frank & Julie Jungers Dean of Engineering at the University of Washington in Seattle. She has been the scientific founder of four companies and enjoys international travel.

Link:
The Role of Microphysiological Systems for Oncology and Stem Cell Research - News-Medical.net

Cell Biology Cloud Computing Market Development, Market Trends, Key Driven Factors, Segmentation And Forecast To 2020-2026| Accenture, Amazon Web…

Latest Report On Cell Biology Cloud Computing Market including Market Landscape, and Market size, Revenues by players, Revenues by regions, Average prices, Competitive landscape, market Dynamics and industry trends and developments during the forecast period.

The global Cell Biology Cloud Computing market is broadly analyzed in this report that sheds light on critical aspects such as the vendor landscape, competitive strategies, market dynamics, and regional analysis. The report helps readers to clearly understand the current and future status of the global Cell Biology Cloud Computing market. The research study comes out as a compilation of useful guidelines for players to secure a position of strength in the global market. The authors of the report profile leading companies of the global Cell Biology Cloud Computing market, Also the details about important activities of leading players in the competitive landscape.

Key companies operating in the global Cell Biology Cloud Computing market include: , Accenture, Amazon Web Services, Benchling, Cisco Systems, Dell Emc, IBM, DXC Technology, Oracle, ScaleMatrix, IPERION, NovelBio

Get PDF Sample Copy of the Report to understand the structure of the complete report: (Including Full TOC, List of Tables & Figures, Chart) :

https://www.qyresearch.com/sample-form/form/1664672/global-cell-biology-cloud-computing-market

The report predicts the size of the global Cell Biology Cloud Computing market in terms of value and volume for the forecast period 2020-2026. As per the analysis provided in the report, the global Cell Biology Cloud Computing market is expected to rise at a CAGR of xx % between 2020 and 2026 to reach a valuation of US$ xx million/billion by the end of 2026. In 2020, the global Cell Biology Cloud Computing market attained a valuation of US$ XX million/billion. The market researchers deeply analyze the global Cell Biology Cloud Computing industry landscape and the future prospects it is anticipated to create

Segmental Analysis

The report has classified the global Cell Biology Cloud Computing industry into segments including product type and application. Every segment is evaluated based on growth rate and share. Besides, the analysts have studied the potential regions that may prove rewarding for the Cell Biology Cloud Computing manufcaturers in the coming years. The regional analysis includes reliable predictions on value and volume, thereby helping market players to gain deep insights into the overall Cell Biology Cloud Computing industry.

Global Cell Biology Cloud Computing Market Segment By Type:

, Public Cloud Computing, Private Cloud Computing, Hybrid Cloud Computing

Global Cell Biology Cloud Computing Market Segment By Application:

,Genomics, Diagnostics, Clinical Trials, Pharma Manufacturing, Others

Competitive Landscape

It is important for every market participant to be familiar with the competitive scenario in the global Cell Biology Cloud Computing industry. In order to fulfil the requirements, the industry analysts have evaluated the strategic activities of the competitors to help the key players strengthen their foothold in the market and increase their competitiveness.

Key companies operating in the global Cell Biology Cloud Computing market include: , Accenture, Amazon Web Services, Benchling, Cisco Systems, Dell Emc, IBM, DXC Technology, Oracle, ScaleMatrix, IPERION, NovelBio

Key questions answered in the report:

Enquire for Customization in the report @ https://www.qyresearch.com/customize-request/form/1664672/global-cell-biology-cloud-computing-market

TOC

1 Market Overview of Cell Biology Cloud Computing1.1 Cell Biology Cloud Computing Market Overview1.1.1 Cell Biology Cloud Computing Product Scope1.1.2 Market Status and Outlook1.2 Global Cell Biology Cloud Computing Market Size Overview by Region 2015 VS 2020 VS 20261.3 Global Cell Biology Cloud Computing Market Size by Region (2015-2026)1.4 Global Cell Biology Cloud Computing Historic Market Size by Region (2015-2020)1.5 Global Cell Biology Cloud Computing Market Size Forecast by Region (2021-2026)1.6 Key Regions Cell Biology Cloud Computing Market Size YoY Growth (2015-2026)1.6.1 North America Cell Biology Cloud Computing Market Size YoY Growth (2015-2026)1.6.2 Europe Cell Biology Cloud Computing Market Size YoY Growth (2015-2026)1.6.3 China Cell Biology Cloud Computing Market Size YoY Growth (2015-2026)1.6.4 Rest of Asia Pacific Cell Biology Cloud Computing Market Size YoY Growth (2015-2026)1.6.5 Latin America Cell Biology Cloud Computing Market Size YoY Growth (2015-2026)1.6.6 Middle East & Africa Cell Biology Cloud Computing Market Size YoY Growth (2015-2026)1.7 Coronavirus Disease 2019 (Covid-19): Cell Biology Cloud Computing Industry Impact1.7.1 How the Covid-19 is Affecting the Cell Biology Cloud Computing Industry

1.7.1.1 Cell Biology Cloud Computing Business Impact Assessment Covid-19

1.7.1.2 Supply Chain Challenges

1.7.1.3 COVID-19s Impact On Crude Oil and Refined Products1.7.2 Market Trends and Cell Biology Cloud Computing Potential Opportunities in the COVID-19 Landscape1.7.3 Measures / Proposal against Covid-19

1.7.3.1 Government Measures to Combat Covid-19 Impact

1.7.3.2 Proposal for Cell Biology Cloud Computing Players to Combat Covid-19 Impact 2 Cell Biology Cloud Computing Market Overview by Type2.1 Global Cell Biology Cloud Computing Market Size by Type: 2015 VS 2020 VS 20262.2 Global Cell Biology Cloud Computing Historic Market Size by Type (2015-2020)2.3 Global Cell Biology Cloud Computing Forecasted Market Size by Type (2021-2026)2.4 Public Cloud Computing2.5 Private Cloud Computing2.6 Hybrid Cloud Computing 3 Cell Biology Cloud Computing Market Overview by Type3.1 Global Cell Biology Cloud Computing Market Size by Application: 2015 VS 2020 VS 20263.2 Global Cell Biology Cloud Computing Historic Market Size by Application (2015-2020)3.3 Global Cell Biology Cloud Computing Forecasted Market Size by Application (2021-2026)3.4 Genomics3.5 Diagnostics3.6 Clinical Trials3.7 Pharma Manufacturing3.8 Others 4 Global Cell Biology Cloud Computing Competition Analysis by Players4.1 Global Cell Biology Cloud Computing Market Size (Million US$) by Players (2015-2020)4.2 Global Top Manufacturers by Company Type (Tier 1, Tier 2 and Tier 3) (based on the Revenue in Cell Biology Cloud Computing as of 2019)4.3 Date of Key Manufacturers Enter into Cell Biology Cloud Computing Market4.4 Global Top Players Cell Biology Cloud Computing Headquarters and Area Served4.5 Key Players Cell Biology Cloud Computing Product Solution and Service4.6 Competitive Status4.6.1 Cell Biology Cloud Computing Market Concentration Rate4.6.2 Mergers & Acquisitions, Expansion Plans 5 Company (Top Players) Profiles and Key Data5.1 Accenture5.1.1 Accenture Profile5.1.2 Accenture Main Business and Companys Total Revenue5.1.3 Accenture Products, Services and Solutions5.1.4 Accenture Revenue (US$ Million) (2015-2020)5.1.5 Accenture Recent Developments5.2 Amazon Web Services5.2.1 Amazon Web Services Profile5.2.2 Amazon Web Services Main Business and Companys Total Revenue5.2.3 Amazon Web Services Products, Services and Solutions5.2.4 Amazon Web Services Revenue (US$ Million) (2015-2020)5.2.5 Amazon Web Services Recent Developments5.3 Benchling5.5.1 Benchling Profile5.3.2 Benchling Main Business and Companys Total Revenue5.3.3 Benchling Products, Services and Solutions5.3.4 Benchling Revenue (US$ Million) (2015-2020)5.3.5 Cisco Systems Recent Developments5.4 Cisco Systems5.4.1 Cisco Systems Profile5.4.2 Cisco Systems Main Business and Companys Total Revenue5.4.3 Cisco Systems Products, Services and Solutions5.4.4 Cisco Systems Revenue (US$ Million) (2015-2020)5.4.5 Cisco Systems Recent Developments5.5 Dell Emc5.5.1 Dell Emc Profile5.5.2 Dell Emc Main Business and Companys Total Revenue5.5.3 Dell Emc Products, Services and Solutions5.5.4 Dell Emc Revenue (US$ Million) (2015-2020)5.5.5 Dell Emc Recent Developments5.6 IBM5.6.1 IBM Profile5.6.2 IBM Main Business and Companys Total Revenue5.6.3 IBM Products, Services and Solutions5.6.4 IBM Revenue (US$ Million) (2015-2020)5.6.5 IBM Recent Developments5.7 DXC Technology5.7.1 DXC Technology Profile5.7.2 DXC Technology Main Business and Companys Total Revenue5.7.3 DXC Technology Products, Services and Solutions5.7.4 DXC Technology Revenue (US$ Million) (2015-2020)5.7.5 DXC Technology Recent Developments5.8 Oracle5.8.1 Oracle Profile5.8.2 Oracle Main Business and Companys Total Revenue5.8.3 Oracle Products, Services and Solutions5.8.4 Oracle Revenue (US$ Million) (2015-2020)5.8.5 Oracle Recent Developments5.9 ScaleMatrix5.9.1 ScaleMatrix Profile5.9.2 ScaleMatrix Main Business and Companys Total Revenue5.9.3 ScaleMatrix Products, Services and Solutions5.9.4 ScaleMatrix Revenue (US$ Million) (2015-2020)5.9.5 ScaleMatrix Recent Developments5.10 IPERION5.10.1 IPERION Profile5.10.2 IPERION Main Business and Companys Total Revenue5.10.3 IPERION Products, Services and Solutions5.10.4 IPERION Revenue (US$ Million) (2015-2020)5.10.5 IPERION Recent Developments5.11 NovelBio5.11.1 NovelBio Profile5.11.2 NovelBio Main Business and Companys Total Revenue5.11.3 NovelBio Products, Services and Solutions5.11.4 NovelBio Revenue (US$ Million) (2015-2020)5.11.5 NovelBio Recent Developments 6 North America Cell Biology Cloud Computing by Players and by Application6.1 North America Cell Biology Cloud Computing Market Size and Market Share by Players (2015-2020)6.2 North America Cell Biology Cloud Computing Market Size by Application (2015-2020) 7 Europe Cell Biology Cloud Computing by Players and by Application7.1 Europe Cell Biology Cloud Computing Market Size and Market Share by Players (2015-2020)7.2 Europe Cell Biology Cloud Computing Market Size by Application (2015-2020) 8 China Cell Biology Cloud Computing by Players and by Application8.1 China Cell Biology Cloud Computing Market Size and Market Share by Players (2015-2020)8.2 China Cell Biology Cloud Computing Market Size by Application (2015-2020) 9 Rest of Asia Pacific Cell Biology Cloud Computing by Players and by Application9.1 Rest of Asia Pacific Cell Biology Cloud Computing Market Size and Market Share by Players (2015-2020)9.2 Rest of Asia Pacific Cell Biology Cloud Computing Market Size by Application (2015-2020) 10 Latin America Cell Biology Cloud Computing by Players and by Application10.1 Latin America Cell Biology Cloud Computing Market Size and Market Share by Players (2015-2020)10.2 Latin America Cell Biology Cloud Computing Market Size by Application (2015-2020) 11 Middle East & Africa Cell Biology Cloud Computing by Players and by Application11.1 Middle East & Africa Cell Biology Cloud Computing Market Size and Market Share by Players (2015-2020)11.2 Middle East & Africa Cell Biology Cloud Computing Market Size by Application (2015-2020) 12 Cell Biology Cloud Computing Market Dynamics12.1 Industry Trends12.2 Market Drivers12.3 Market Challenges12.4 Porters Five Forces Analysis 13 Research Finding /Conclusion 14 Methodology and Data Source 14.1 Methodology/Research Approach14.1.1 Research Programs/Design14.1.2 Market Size Estimation14.1.3 Market Breakdown and Data Triangulation14.2 Data Source14.2.1 Secondary Sources14.2.2 Primary Sources14.3 Disclaimer14.4 Author List

About Us

QYResearch always pursuits high product quality with the belief that quality is the soul of business. Through years of effort and supports from huge number of customer supports, QYResearch consulting group has accumulated creative design methods on many high-quality markets investigation and research team with rich experience. Today, QYResearch has become the brand of quality assurance in consulting industry.

See the original post:
Cell Biology Cloud Computing Market Development, Market Trends, Key Driven Factors, Segmentation And Forecast To 2020-2026| Accenture, Amazon Web...

Broad Foundation brings together stem cell scientists, engineers and physicians at University of Southern – Mirage News

The Broad Foundation brings together stem cell scientists, engineers and physicians at USC and beyond

Developing new stem cell therapies requires more than a solo biologist having a eureka moment alone in the lab. Real progress relies on collaborations between biologists, engineers and physicians. Thats why The Eli and Edythe Broad Foundation has continued its support of two strategic initiatives: innovation awards bringing together teams of engineers and scientists from USC and Caltech, and clinical research fellowships for physician-scientists.

Engineering new approaches: The Broad Innovation Awards

For the fifth consecutive year, the Broad Innovation Awards are providing critical funding to USC-affiliated faculty members pursuing multi-investigator research collaborations related to stem cells. For the first year, these collaborations are also drawing on the expertise of biomedical engineers from Caltech. Each award provides $200,000 of funding for a one-year project.

Were very excited to be joining our colleagues at USC in pioneering new approaches to advancing stem cell research, said Stephen L. Mayo, chair of the Division of Biology and Biological Engineering at Caltech. Were thankful to The Broad Foundation for supporting cross-town collaborations between scientists with different expertise but common goals.

With support from a Broad Innovation Award, Andy McMahon, the director of the Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, is collaborating with Caltech biomedical engineer Long Cai to leverage a new technology for understanding chronic kidney disease. The technology, called seqFISH, provides information about genetic activity taking place in intact tissueenabling the study of the interactions between cells in their native environments.

Dr. Cais seqFISH technology will provide an unprecedented insight into the cellular interplay underlying chronic kidney disease caused by a maladaptive response to acute kidney injury, said McMahon, who is the W.M. Keck Provost and University Professor of Stem Cell Biology and Regenerative Medicine, and Biological Sciences, as well as the chair of the Department of Stem Cell Biology and Regenerative Medicine at USC. We aim to better understand this maladaptive responsewhich is more common in malesin order to find new targets for preventing the progression to chronic kidney disease.

A second Broad Innovation Award brings together USC Stem Cell scientist Rong Lu and Caltech synthetic biologist Michael Elowitz. Their team will study the spatial organization of blood-forming stem and progenitor cells, also called hematopoietic stem and progenitor cells (HSPCs), which reside in the bone marrow. By pinpointing the locations of specific HSPCs, the scientists may find clues to explain why certain HSPCs are so dominantreplenishing the majority of the bodys blood and immune cells after a disruption such as a bone marrow transplantation.

Spatial advantages may be the primary drivers of what we refer to as the clonal dominance of certain HSPCs, said Lu, a Richard N. Merkin Assistant Professor of Stem Cell Biology and Regenerative Medicine, Biomedical Engineering, Medicine, and Gerontology at USC. Understanding the spatial competition between HSPCs could help improve bone marrow transplantation and provide new insights into aging and the development of diseases such as leukemiawhich are associated with clonal dominance.

Elowitz added: Thanks to the Broad Innovation Award and this exciting collaboration with Rong Lu, we will be able to bring a new, synthetic biology approach to record cell histories and read them out in individual cells within their native spatial context, providing new insights into fundamental questions in blood stem cell development.

A third Broad Innovation Award brings together three collaborators at USC: Michael Bonaguidi, an assistant professor of stem cell biology and regenerative medicine, biomedical engineering, and gerontology; Robert Chow, a professor of physiology and neuroscience, and biomedical engineering; and Jonathan Russin, an assistant professor of neurological surgery and associate surgical director for the USC Neurorestoration Center. Their project focuses on finding new approaches to treating epilepsy by studying neural cells called astroglia. These cells perform a variety of key functions that support the health of neurons in the brain, and they may also play a role in modulating epileptic seizures.

Although adults dont tend to generate many new brain cells, humans do produce a limited number of new astroglia, said Bonaguidi. We will examine these newborn astroglia at the single-cell level to better understand their role in epileptic patients, and to lay the groundwork for identifying new treatments.

The doctors are in: The Broad Clinical Research Fellowships

The Broad Clinical Research Fellowships are also entering their fifth consecutive year. These fellowships support stem cell research by physician-scientists and residents who intend to practice medicine in California.

These fellowships provide a very special opportunity for our medical residents to engage deeply in laboratory research, as a complement to their extensive training in patient care, said Laura Mosqueda, Dean of the Keck School of Medicine of USC. This valuable research experience gives them a much more complete perspective on how to meet the challenges of finding the best possible treatments for their patients.

A USC resident physician in general surgery, Kemp Anderson will spend his fellowship studying necrotizing enterocolitis, a very serious intestinal infection that affects nearly 10 percent of premature infants. Specifically, he will explore how a molecule involved in cellular communication, called farnesoid X receptor, or FXR, might contribute to this disease.

If FXR plays a role in compromising intestinal barrier function in these premature infants, then altering the activity of FXR could potentially yield treatment modalities for necrotizing enterocolitis, avoiding the morbidity and mortality associated with surgical intervention, said Anderson, who is performing the research under the mentorship of Christopher Gayer and Mark Frey at Childrens Hospital Los Angeles (CHLA). Im deeply appreciative of the benefactors and the selection committee for awarding me the Broad Clinical Fellowship, as it is allowing me devoted time to focus on this important project, and to become a more well-rounded physician through this academic pursuit.

Brittany Rocque, a resident physician in general surgery, will use her fellowship to seek better ways to predict, detect and diagnose immune rejection in patients who have undergone liver transplantation. Nearly 60 percent of pediatric patients and at least 15 percent of adult patients reject their liver transplants, and this can currently only be confirmed through an invasive surgical biopsy. Rocque is utilizing the technology Imaging Mass Cytometry to identify and analyze the types of immune cells involved in rejection.

My project has the potential to provide a noninvasive option to assess rejection in transplanted patients, and to expand our understanding of immune rejection, said Rocque, who is being co-mentored by Juliet Emamaullee and Shahab Asgharzadeh at CHLA. Im greatly looking forward to applying my passion for transplantation surgery in the context of basic science, and enhancing my appreciation for the nuances of research, thanks to the Broad Clinical Research Fellowship.

A hematology-oncology fellow who will be transitioning to a junior faculty position at USC next year, Caitlin ONeill will study a condition known as clonal hematopoiesis or CH, a phenomenon common in the aging population. CH involves genetic mutations that cause the expansion of a particular population of blood cells without leukemia or related malignancies. CH increases risks for certain health conditions including heart disease.

During her Broad Clinical Research Fellowship, ONeill will look at one mutation seen in patients with CH: a mutation to the gene called Tet methylcytosine dioxygenase 2, or TET2. ONeill will explore if this mutation promotes blood clots, inflammation and heart disease.

The goal is to inform therapies to prevent heart disease and leukemic progression in aging patients with CH, said ONeill, who is working with co-mentors Casey OConnell and Rong Lu at USC. Im very happy to be working on this project, with support from the Broad Clinical Research Fellowship, during my transition to becoming a faculty member at USC.

See the original post here:
Broad Foundation brings together stem cell scientists, engineers and physicians at University of Southern - Mirage News