Category Archives: Cell Biology

UNM names 10 faculty to the rank of Distinguished Professor – UNM Newsroom

The University of New Mexicorecently announced the promotion and honor of 10faculty to the rank of Distinguished Professor. They include Lisa Broidy, Alexander Buium, Joseph Cook, Laura Crossey, Jeremy Edwards, Fernando Garzon, Kerry Howe, Mary Ann Osley, Nina Wallerstein and Douglas Ziedonis.

The rank of Distinguished Professor is the highest title that UNM bestows upon its faculty. It is awarded to those individuals who have demonstrated outstanding achievements, and are nationally and internationally renowned as scholars.

College of Arts and Sciences

Lisa Broidy

Lisa BroidyBroidys research focuses on how gender frames the structural, individual, and situational processes associated with violence and antisocial behavior. Building primarily from General Strain Theory and Developmental & Life Course Theories, her work contributes to the growing theoretical and empirical literature that account for gender differences in criminal involvement while also recognizing the significant heterogeneity that characterizes womens pathways into and out of crime.

She examines the relationship between gender and crime in both contemporary and historical contexts in the U.S. and cross-nationally. In investigating why women offend at much lower rates than men, her work suggests that the structural and social contexts women navigate limit their opportunities and motivations for serious offending. At the same time, her work illustrates that throughout the life course, girls and women confront a range of challenges that, for some, do prove criminogenic and can have both short and long-term consequences for their offending trajectories. Her work also examines the implications of these gendered processes for criminal justice policy and practice, particularly around female incarceration and domestic violence.

Alexandru Buium

Alexandru BuiumBuium was born in 1955, in Bucharest, Romania. He holds an M.S. from the University of Bucharest, Romania (1980) and a Ph.D. from the University of Bucharest, Romania (1983). From 1990 to 1995, he was a senior researcher at the Institute of Mathematics of the Romanian Academy. From 1995 to 1997, he was an associate professor at UNM. He has been a professor of mathematics at The University of New Mexico since 1997. He was awarded the Titeica Prize of the Romanian Academy of Science (1987), a Humboldt Fellow (1992/93), a Member of the Institute for Advanced Study, Princeton (1993/94) and a Fellow of the American Mathematical Society (class of 2016).

His visiting positions include at Columbia University (NYC), University of Paris 7 (Paris), Max Planck Institute (Bonn), Institute for Advanced Study (Princeton) and Institut des Hautes Etudes Scientifiques (Bures, France).

Buiums research areas include algebra, number theory and geometry. He has written several publications (6 research monographs and over 80 research papers) including the 2013 textbookMathematics: a Minimal Introduction, and the research monographsDifferential Function Fields and Moduli of Algebraic Varieties, Lecture Notes in Math(1986),Differential Algebraic Groups of Finite Dimension, Lecture Notes in Math(1992), andDifferential Algebra and Diophantine Geometry(1994).

Joseph Cook

Joseph CookAfter dropping out of high school in Silver City, Cook received his GED, and later B.S. in Biology at Western New Mexico University (1980), and M.S. (1982), and Ph.D. in Biology (1990) at UNM. He then moved to the University of Alaska and was promoted to Professor of Biology, Chief Curator, and Curator of Mammals and Cryogenic Collections at the University of Alaska Museum of the North. He later served as Professor (1990-2000) and Chair of the Department of Biological Sciences at Idaho State University (2000-2003). Subsequently, he returned to New Mexico as Professor of Biology and Curator of Mammals of the Museum of Southwestern Biology, where he also served as Director (2011-2017) and Curator of Genomic Resources (2007-2017). He was named Regents Professor in 2018.

Over the past two decades, he and his staff and students have built the UNM museum into the second largest collection of mammals worldwide, recently surpassing the British Museum in London. Critical biodiversity infrastructure, this resource is now the basis for >100 publications annually and is used widely in efforts to study emerging zoonotic pathogens, wildlife conservation, environmental pollutants, climate change, and the biological diversity of our planet. Cook has chaired national conservation committees (American Society of Mammalogists); led multiple international consortia and communities of practice (e.g., AIM-UP! Research Coordinating Network, Project Echos Museums and Emerging Pathogens in the Americas); was President of a national museum association (Natural Science Collections Alliance), and served on the National Academy of Sciences panel that reviewed U.S. bio-collection infrastructure.

Laura Crossey

Laura CrosseyCrossey works with aqueous and sedimentary geochemistry, and applications of low-temperature geochemistry to problems in hydrochemistry, diagenesis, geomicrobiology and geothermal processes. Her research approach combines field examination of modern environments (biogeochemistry of water and sediments) with laboratory analysis as well as core and outcrop evaluations applied to evaluate paleohydrology, spring sustainability and reservoir/aquifer characteristics.

She is an MSL Expert Consultant for the ChemCam Team, Mars Science Laboratory Rover. Other activities include geoscience outreach, K-12 outreach, and science education research as well as programs to increase the participation of under-represented groups in the science disciplines. She is a Fellow of both the American Association for the Advancement of Science (AAAS) and the Geological Society of America (GSA) and served as the Birdsall Dreiss Distinguished Lecturer for 2019 (sponsored by the Hydrogeology Division of GSA).

Jeremy Edwards

Jeremy EdwardsEdwards has worked at the interface of biology, bioinformatics, and engineering since the beginning of his scientific career. His graduate advisor was Dr. Bernhard Palsson, where he was the first person to take genome sequence information and develop predictive mathematical models of bacterial metabolism. His research started a significant global effort and many papers from his graduate work have over 800 citations. His graduate work sparked an intense interest in genomics technology and thus he worked with Dr. George Church at Harvard Medical School for his post-doctoral studies. He has worked on the development of genome technologies since that time.

Now, his laboratory is in the NCI designated Cancer Research and Treatment Center at the University of New Mexico Health Sciences Center. He has a very active group of engineers, biologists, and chemists, all working together toward the development of ultrahigh-throughput DNA sequencing technology and computational biology.

UNM School of Engineering

Fernando Garzon

Fernando Garzon joined UNM in 2014 as a jointly-appointed faculty member with Sandia National Laboratories, coming from Los Alamos National Laboratory. He is currently the director of the Center for Microengineered Materials and is an Academic Alliance Professor and continues to conduct joint research with Sandia.

His research interests include low-environmental impact electro-synthesis of fuels, the development of advanced gas sensors, fuel-cell materials technology, upgrading of light hydrocarbons, advanced manufacturing of ceramic materials technology, solid-state ionic devices for reconfigurable electronics, and sensors with ultralow detection limits for uranium and arsenic groundwater contamination.

Garzon is a fellow and past president of the Electrochemical Society and received the Department of Energy Fuel Cell Program Research Award in 2009. He is also the winner of Scientific Americans Top 50 Science and Technology Achievements for 2003 award and received the LANL Fellows Prize for Research Leadership.

Kerry Howe

Kerry Howe has been the director of the Center for Water and the Environment since 2013, where he leads the $5 million National Science Foundation-sponsored Centers of Research Excellence in Science and Technology (CREST) Center for Water and the Environment project.

First funded in 2014, it was renewed for another $5 million over five years in 2020. Phase 1 of the CREST project focused on generating new knowledge about watersheds, treatment technologies for contaminated water, and interactions between water and energy production. Phase 2 is building on previous successes while expanding and redirecting the water-related research with new research questions, new partnerships with institutions, and a new emphasis on recruiting and retaining Native American students, a population that may be under-represented even among CREST centers.

Howe joined UNM in 2002 and is the recipient of awards including the Harrison Faculty Recognition Award, Stamm Outstanding Faculty Award and Regents Lecturer.

UNM Health Sciences Center

Mary Ann Osley

Mary Ann Osley is a professor of Molecular Genetics and Microbiology in the School of Medicine. She studies the processes that regulate the replication, transcription and repair DNA in chromosomes. Her work, which focuses on the role of histone proteins and chromatin in the model organism Saccharomyces cerevisiae, has important implications in the context of cancer genomics.

Her more recent work on cellular quiescence has important implications for how stem cells prevent aberrant proliferation as occurs in cancer cells. She has 63 peer-reviewed publications and has published in high-impact journals including Nature, Nature Cell Biology, Journal of Cell Biology, Molecular Cell Biology and Nucleic Acids Research.

She has received multiple grants from NIH including 3 R01 grants for her own research, and currently holds an NIH grant for her project Functional Analysis of Quiescence.

Nina Wallerstein

Nina Wallerstein is a professor of Public Health in the College of Population Health. She studies interventions in communities to promote improved health (health education, health promotion), alcohol prevention as well as other risky behaviors with an emphasis on adolescents, and methodologies for community-based participatory research.

Her work emphasizes empowerment-based, culture-centered interventions that have proven highly effective. Much of her work has engaged with the Jemez Pueblo, the Navajo Nation, and the Mescalero Apache community. Wallerstein has also worked internationally, especially in Brazil where she has formed sustained collaborations and promoted the adoption of community-based participatory research approaches throughout Brazil.

She has published more than 170 peer-reviewed articles and chapters, 7 authored, co-authored, or edited books including Community-Based Research for Health: Advancing Social and Health Equity, which is viewed as a field-defining work. She has been awarded more than $25 million in funding for her research, and currently has some $2.5 million in annual support.

Douglas Ziedonis

Douglas Ziedonis is a professor of Psychiatry in the School of Medicine and Executive Vice President of the UNM Health Sciences Center and the CEO of the UNM Health System. His research focuses on the intersection of mental illness and substance abuse and has been particularly impactful for the prevalence of tobacco use and associated health harms among schizophrenic patients.

His work has been continuously funded for over 25 years with 118 grants and has produced 328 publications/scholarly works including 146 original research articles, 21 invited articles, 9 books, 42 chapters in edited volumes, 20 behavioral therapy manuals, 12 organizational change and leadership development manuals, etc.

His work has been placed in the most prestigious journals in his field. Dr. Ziedonis research has not only examined methods of treating substance abuse that co-occurs with serious and persistent mental illness but has promoted organizational change within the medical and mental health provider communities to challenge widespread de facto acceptance of tobacco use among patients with some forms of mental illness.

Original post:
UNM names 10 faculty to the rank of Distinguished Professor - UNM Newsroom

Defined by their boundaries Cells and how to run them – The Economist

Aug 7th 2021

THE CHEMICAL reactions on which life depends need a place to happen. That place is the cell. All the things which biology recognises as indisputably alive are either cells or conglomerations of cells (viruses fall into disputable territory). Since the middle of the 19th century the cell has been seen as the basic unit of life.

Your browser does not support the

Enjoy more audio and podcasts on iOS or Android.

A cell requires something to keep its insides in and the outside out. That is the role of the cell membrane, a flexible film made largely of lipids. These are smallish tadpole-shaped molecules with heads that are comfortable in water and twin tails that shun it. When put into a watery solution they naturally form double layers in which the water-tolerant heads are on the outside and the water-wary bits on the inside. Some plant, fungal and bacterial cells employ more rigid structures, called cell walls, as further fortifications beyond their membranes. But it is the membrane which defines the cell.

What is more, the disposition of membranes determines what sort of cell it is. Some creatures use membranes chiefly to define their perimeters. These are called prokaryotes, and come in two varieties, bacteria and archaea. In others they are also used to create structures within cells, notably a nucleus to contain the DNA on which genes are written. Such cells may have ten or 20 times more membrane within them than they have defining their surfaces. They are called eukaryotic, Greek for truly nucleated. Creatures made from them are eukaryotes.

The worlds prokaryotic cells vastly outnumber their eukaryotic cousins. Your own body has roughly as many single-celled prokaryotes living on and inside it (mostly in the gut) as it has eukaryotic cells making up muscles, nerves, bones, blood and so on. Some parts of Earths biosphere, such as the ocean floors, contain more or less nothing but prokaryotic life.

But almost everything you have ever looked at and recognised as aliveall the animals, plants, fungi and algaehas been composed of eukaryotic cells. Such cells are typically a lot larger than almost all prokaryotic ones and are capable of a far greater diversity in both form and function. Their versatility is seen in the wide range of shapes they take, from the conjoined starbursts of nerve cells to the creeping mutable blobbiness of amoebae.

Even prokaryotic cells, though, are big compared with the molecules they contain. A bacterium two millionths of a metre long encompasses around 3m protein molecules as well as the DNA which describes them, the RNA necessary to make use of those descriptions and the various smaller molecules that proteins stick together and break apart in the course of their duties (see previous Biology brief). The membrane of such a bacterium, moreover, contains around 20m lipid molecules.

But if you were to synthesise all the molecules found in that bacterium in a laboratory (quite possible, in theory) and pop them into a bacterium-sized bag you would not get a bacterium. You would get an itsy bitsy mess. A cell is not just a set of contents. It is also a set of processes running alongside each other. The only way to create a cell in which all the necessary processes are up and running is to start off with another such cell in which they are already doing so.

Feed a bacterium with the nutrients it needs and as it grows it will synthesise a copy of the DNA molecule on which its genome is stored. When it is big enough to have made a complete copy of that DNA it will split into two, with one DNA ending up in one cell, and the other in the other.

As it is for bacteria, so it is, mutatis mutandis, for all other life, for ever and ever, amen. Life is made of cells, and cells from pre-existing cells. The 30 trillion cells of which a human body is composed can in almost every case be traced back to the single fertilised egg which started it all (the exception is a condition known as chimerism in which two embryos fuse in the womb early on in development).

Of all the processes that continue from cell to cell as life goes on, none is more fundamental than those which provide lifes energy. These are completely dependent on the membranes in cells. Conditions on the two sides of a membrane will almost always be different; different molecules will be present in different concentrations. The laws of thermodynamics, though, take a dim view of different concentrations of something being next to each other. Small molecules and ions that are more frequent on one side of that membrane than the other will diffuse across it in an attempt to even things up. Proteins embedded in such membranes pump molecules in the opposite direction to maintain the distinction between inside and out.

It is by setting up a gradient of hydrogen ionshydrogen atoms with their electrons pulled offacross a membrane that living things put energy into a chemical form which they can use. This process depends on sets of proteins called electron-transport chains. These proteins are embedded in the membrane.

Electron-transport-chain proteins pass electrons to each other in a way that causes hydrogen ions on the inside of the membrane to get moved to the outside. The ions thus build up outside, which means that natures tendency to even out concentrations requires some of them to get back inside. This they do by means of a magnificent protein called ATP synthase, or just ATPase. Molecules of ATPase provide channels through the membrane which it is easy for the hydrogen ions to flow through. This flow yields usable energy, like the flow of water through a watermill.

That is not an idle metaphor. ATPase has several parts, one of which can rotate with respect to the others. As the ions flow through the protein they spin this rotor at a speed of 6,000rpm. If you could hear them at work they would be humming at something like the G two octaves below middle C. Another part of the molecule uses the kinetic energy of this spinning rotor to affix phosphate ions to a molecule called adenosine diphosphate (ADP), thus making adenosine triphosphate, or ATPcell biologys near-universal energy carrier.

In almost all instances where a cellular process requires energy, that energy is provided by breaking ATP back down into ADP. Adding an amino acid to a growing protein uses up roughly five ATPs. Synthesising membrane lipids costs about one ATP for every two carbon atoms used. A bacterium doubling in size uses about 10bn ATPs to build all the molecules it needs, meaning every one of the 10m or so ADP molecules the bacterium contains is turned into ATP and broken back down again 1,000 times during the process.

To keep the ATPase whirring, the cell requires a constant flow of electrons along its membrane-bound electron-transfer chains. There are two ways of creating such flows: respiration and photosynthesis.

Respiration breaks molecules of glucose down into carbon dioxide and water through a suite of reactions called the citric-acid cycle. A glucose-molecules worth of electrons typically pushes ten hydrogen ions across the membrane in which the respiratory electron-transfer chain is embedded. As they flow back through the ATPase they can generate 20 ATPs.

Photosynthesis uses the energy of sunlight to liberate electrons from water molecules, thus creating oxygen and also hydrogen ions ready for pushing across the membrane. Some of the ATP made this way powers a process that combines those ions with carbon-dioxide. A few more chemical reactions produce a sugar such as glucose, which then goes on to be built into all the other molecules from which life is made. Photosynthesis builds up the worlds biomass; respiration breaks it down.

In a prokaryotic cell the membrane in which electron-transfer proteins sit is that which surrounds the cell. In eukaryotic cells respiration takes place in intracellular structuresorganellescalled mitochondria. These consist of folded-up membranes rich in electron-transport chains. Containing lots of mitochondria (in humans, hundreds or thousands per cell is not uncommon) means such cells can generate a great deal of ATP. If all the membranes in your bodys mitochondria were joined and spread out flat they would cover several football fields.

Under a microscope, some mitochondria look a lot like bacteria. This is not a coincidence, it is a family resemblance. When Earth was a bit more than half its present age, which is to say around 2bn years ago, two prokaryotes, one from the archaea and one from the bacteria, contrived to merge. How, exactly, they did so is far from clear. But that merger created something truly novel: the first eukaryotic cell. Mitochondria are descendants of the bacterium involved, a descent demonstrated incontrovertibly by the fact they still have remnant genomes of their own which are distinctively bacterial. In human beings these little mitochondrial genomes are the only DNA not sequestered on chromosomes in the nucleus.

All the mitochondria in all the eukaryotes in the world date back to that merger. Similarly, chloroplaststhe organelles of photosynthesis found in plants and algaedate back to a later event in which a eukaryote engulfed a photosynthetic bacterium. Many eukaryotes remained single-celled, and do so to this day. But others began forming colonies which permitted division of labour between cells and encouraged the development of specialised body parts called organs. Which are the subject of next weeks Biology brief.

In this series on the levels of life1 Biologys big molecules2 Cells and how to power them*3 Making organs4 The story of a life5 What is a species, anyway?6 Finding living planets

This article appeared in the Schools brief section of the print edition under the headline "Layers of power"

Link:
Defined by their boundaries Cells and how to run them - The Economist

CFI invests $3.9 M in McGill research – McGill Newsroom

McGills researchers will soon acquire the highly specialized tools they need to innovate in their fields thanks to funding from the Government of Canada through the Canada Foundation for Innovation (CFI)s John R. Evans Leaders Fund (JELF). The Honourable Franois-Philippe Champagne, Minister of Innovation, Science and Industry, made the announcement today of $77 million across 50 institutions in Canadathrough the program. In total, twenty-one McGill research projects have received a combined $3.9 million in federal grants through three rounds of JELF. McGill recipients will also receive additional funds from the provincial government and the university toward the total project budget for their research endeavors.

Every profession requires the best tools and systems to improve outcomes, and research is no exception, says Martha Crago, Vice-Principal (Research and Innovation). McGills researchers are working on highly innovative solutions across the research spectrum: in healthcare, for the environment and with industry. Thanks to the new technologies and infrastructure acquired through the JELF program, McGills expertise will be augmented, and new possibilities generated for the benefit of all Canadians.

Thanks to the JELF investment, Professor of Electrical and Computer Engineering, AJung Moon, will welcome a humanoidan autonomous mobile robotto the McGill Responsible Autonomy & Intelligent System Ethics (RAISE) lab. Her research group will also acquire an integrated digital projector, RGB-D sensor, and a motion capture system as well as an additional 7-DOF robotic arm. RAISE lab will use these new tools to investigates the impact of interactive or collaborative robots (cobots) on individuals and society. Collaborative robotics is one of the fastest-growing sub-sectors of robotics today, and it offers a promising long-term investment for Canada's economy, says Moon. However, robot influence on humans can be pre-programmed, one-sided, and deployed at scale. This can expose us to new types of harm, such as systematic manipulation of our actions and decisions. This research will help establish empirically-grounded guidelines to ensure responsible design and deployment of collaborative robotic systems in Canada and abroad.

With the investment from the CFI, biology professor Fiona Sopers lab will gain three unique tools to help quantify the contributions of plants in controlling the effects of climate change. The new acquisitions include the Acetylene Reduction Assay by Cavity ring-down laser Absorption Spectroscopy (ARACAS) systemone of the first of its kind in Canadawhich will measure nitrogen fixation in tropical plants, one of the most essential processes for the health and productivity of whole ecosystems. This infrastructure will be complemented by a portable photosynthesis system, as well as a multi-mode microplate-reader. These versatile instruments can be used to complete greenhouse growth chamber-based experiments, to analyze plant samples collected in the field, and to conduct in situ measurements in tropical ecosystems. The funding will also upgrade existing greenhouse lighting infrastructure in the McGill Phytotron that also houses Co2 -controlled growth chambers central to Sopers research.

McGill will also acquire a new state-of-the-art microscopy system, which Professor Jackie Vogel will use to advance understanding on the causes of cancer and birth defects. Vogels lab studies key events early in cell division (mitosis). While mitosis has been studied for hundreds of years, most of the research has focused on the last stage of the process, when the cell cleaves to form two identical cells. The earliest events in cell division remain relatively mysterious. With this new microscope, Vogel will be able to detect the fast movement of molecules within living cells and distinguish molecules that are very close together without damaging the cell with the phototoxic effects of intense light.

View a complete list of CFI JELF-funded projects:

https://www.mcgill.ca/research/channels/news/cfi-invests-4M-mcgill-research

Founded in Montreal, Quebec, in 1821, McGill University is Canadas top ranked medical doctoral university. McGill is consistently ranked as one of the top universities, both nationally and internationally. Itis a world-renownedinstitution of higher learning with research activities spanning two campuses, 11 faculties, 13 professional schools, 300 programs of study and over 40,000 students, including more than 10,200 graduate students. McGill attracts students from over 150 countries around the world, its 12,800 international students making up 31% of the student body. Over half of McGill students claim a first language other than English, including approximately 19% of our students who say French is their mother tongue.

Read the original:
CFI invests $3.9 M in McGill research - McGill Newsroom

Study finds novel approach to attenuate mitochondrial dysfunction that drives diet-induced obesity – News-Medical.Net

A team of University of California, Irvine, scientists have discovered a novel pharmacological approach to attenuate the mitochondrial dysfunction that drives diet-induced obesity. The results of their study were published recently in the journal, EMBO Molecular Medicine.

Consuming a high-fat diet can lead to obesity and metabolic disorders such as diabetes and fatty liver. Palmitate, a fat abundant in a Western diet, triggers metabolic dysfunction by causing excessive mitochondrial fission within cells. Mitochondria play a crucial role in a cell's energy production, but also coordinate cell stress responses. Too much mitochondrial fission impairs their function, undermining metabolism and increasing toxic by-products associated with insulin resistance in some tissue types.

Elegant genetic studies in mice show that maintaining mitochondrial networks in a fused state can overcome high fat diet-induced obesity. Our study uses a small molecule to re-shape mitochondria in multiple tissues simultaneously, reversing obesity and correcting metabolic disease even though mice continue to consume the unhealthy diet."

Aimee Edinger, UCI Chancellor's Fellow, professor of developmental & cell biology and senior author

In their new study, Professor Edinger and her team utilized their patented water-soluble, orally bioavailable, synthetic sphingolipid SH-BC-893 to inhibit endolysosomal trafficking proteins required for mitochondrial fission. The study was conducted using in vitro experiments and a high-fat diet-induced obesity mouse model. The researchers observed that SH-BC-893 prevented mitochondrial dysfunction in the liver, brain, and white adipose tissue of mice consuming a Western diet. As a result, circulating levels of critical metabolic hormones, leptin and adiponectin, were normalized leading to weight loss, improved glucose handling, and reversal of fatty liver disease despite continued access to high-fat food.

"Imbalances in the hormones leptin and adiponectin that accompany obesity create an uphill battle for people trying to lose weight. Having too much leptin can increase appetite while too little adiponectin activity is linked to many metabolic diseases. How or why is not really clear, but the state of the mitochondria may be an important link between these hormones and obesity," said Elizabeth Selwan, a former graduate student researcher in UCI's Department of Developmental and Cell Biology and co-lead author of the study.

The study's findings suggest that SH-BC-893 could be a promising therapy for managing diet-induced obesity. The authors found the drug to be safe and effective in the mouse model and plan on further investigating the compound for possible use in human patients.

"This compound works through a novel mode of action if it is safe and effective in humans, it would offer a new weight loss strategy that could also be combined with other treatments," said Professor Edinger.

Source:

Journal reference:

Jayashankar, V., et al. (2021) Drug-like sphingolipid SH-BC-893 opposes ceramide-induced mitochondrial fission and corrects diet-induced obesity. EMBO Molecular Medicine. doi.org/10.15252/emmm.202013086.

Go here to see the original:
Study finds novel approach to attenuate mitochondrial dysfunction that drives diet-induced obesity - News-Medical.Net

On Heels of $17 Billion SPAC Deal, Ginkgo Inks Collab with Synthetic Biology Firm – BioSpace

California synthetic biology firm Antheia has joined forces with Boston-based biotech companyGinkgo Bioworksto strengthen its efforts to develop and produce essential medicines to treat a wide range of diseases.

Antheia is looking to leverage Ginkgo's cell programming technology and high throughput enzyme design and screening capabilities to support its pipeline of critical active pharmaceutical ingredients (APIs) and key starting materials (KSMs).

Antheia creates complex, plant-inspired therapies through its whole-cell engineering approach to reconstruct complex molecules in yeast. As the company moves its activities to a more commercial scale, it continually invests in strain optimization strategies to ensure the highly efficient production of its target products.

Since most of the world's medicines come from nature, Antheia has developed a more efficient and lower-risk method of creating nature-inspired drugs that are easier, faster, and more efficient to manufacture. In addition to making essential medications more available, this method also prevents the wastage of natural resources and overharvesting.

"Antheia is at the cutting edge of synthetic biology innovation, and its whole-cell engineering platform is capable of producing entire classes of medicines that were previously inaccessible. We are thrilled that Ginkgo's platform can support innovators like Antheia as they create next generation manufacturing technologies for essential medicines," said Barry Canton, co-founder and chief technology officer at Ginkgo Bioworks, in astatement.

The partnership comes on the heels of Ginkgo's newly-inked$17 billion mergerwith Soaring Eagle Acquisition Corp a deal that is expected to add some $15 billion in estimated value to the former when it finalizes in the third quarter of 2021, subject to shareholder and regulatory clearances. Its completion also makes Ginkgo Bioworks a public company.

Ginkgo specializes in enzyme discovery, strain improvement, mammalian cell engineering, and cultured ingredients development, among others. Its solutions serve a wide range of industries, including pharmaceuticals, industrials, food, chemicals, and more.

Ginkgo's products are developed through two core technologies: Ginkgo Codebase (referring to its libraries of cell, enzymes, and genetic programs) and Ginkgo Foundries (referring to its automated biological factories).

Its partnership with Antheia makes it even more possible for critical medicine to be produced on-demand under more efficient and environment-friendly conditions. In addition, biomanufacturing pharmaceuticals have a greater advantage over traditional production methods in terms of cost, quality control, sustainability, and supply chain resilience and agility.

"Antheia is committed to using synthetic biology to enable more equitable access to essential medicines. By partnering with Ginkgo Bioworks, a global leader in organism engineering, we are greatly increasing our ability to develop essential medicines at the speed and scale necessary to drive change in global pharma supply chains," noted Kristy Hawkins, co-founder and CSO at Antheia.

Read more here:
On Heels of $17 Billion SPAC Deal, Ginkgo Inks Collab with Synthetic Biology Firm - BioSpace

FGI Night of Stars to Honor Established Designers and Other Talents – Yahoo Lifestyle

OUT AND ABOUT (HOPEFULLY): Just getting outdoors can be a celebration of sorts, but the Fashion Group International has grander plans for the return of its Night of Stars event this fall.

News that the gala is scheduled to be held in-person was considered a win in itself when revealed in May. Now organizers are detailing their all-out gala plans.

More from WWD

Slated for Oct. 13 at Cipriani South Street at Casa Cipriani, the 37th annual red carpet is being touted as The Evolutionaries. Honorees will include a mix of international designers and other enterprising talents.

Tommy Hilfiger will be the leading star, so to speak, as the recipient of the Superstar and Lifetime Achievement award. Monse founders and Oscar de la Rentas creative duo Fernando Garcia and Laura Kim will be honored as Vanguards.

To get a jump on the celebration, there will be a pre-party the night before the first of its kind on Oct. 12 that will be hosted by Garcia and Kim at the de la Renta boutique on Madison Avenue. Limited tickets will be available and the gathering will benefit the Fashion Group Foundation for its educational mission.

Fausto Puglisi, creative consultant for Roberto Cavalli, will be on hand at the annual awards event to receive the Fashion Star award. Luxury skin care specialist Augustinus Bader, a scientist, physician and authority in stem cell biology and regenerative medicine, is this years Beauty honoree.

Gabriela Hearst will be saluted with the Sustainability award, while Norma Kamali will be celebrated with the Founders Award. The designer expanded her portfolio by launching a home collection a few months ago.

Stylist Law Roach, who has worked with Cardi B, Zendaya, Celine Dion and other celebrities, will receive the Fashion Provacateur award from FGI. The Retail award will be given to the cereal and sneaker-selling Kith founder Ronnie Fieg. And Mastercards chief marketing and communications officer Raja Rajamannar will receive the Tech and Brand Innovation award.

Story continues

Provided coronavirus concerns do not result in substantial federal, state or local restrictions, Night of Stars will mark FGIs first IRL red carpet in two years. In adherence with CDC restrictions and guidelines, FGI is investigating prescreening for vaccinations, on-site testing and touchless entry to be sure that our guests feel protected and experience the luxury evening to which they are accustomed, according to James DAdamo, chairman of FGIs board.

While organizers have yet to reveal the presenters, the annual dinner has attracted power players in music, entertainment and fashion in years past. Artist and illustrator Ruben Toledo has once again created exclusive artwork for the invitations and a commemorative journal for the black-tie event.

FGIs president and chief executive officer Maryanne Grisz said, In a year of transformation, evolution and emotion, we are delighted to be together as a community once again. FGI celebrates these incredible visionaries, who, through their creativity, innovation and drive, represent true leadership, a deeper meaning around the event and where we are going as an industry.

Best of WWD

Sign up for WWD's Newsletter. For the latest news, follow us on Twitter, Facebook, and Instagram.

Go here to read the rest:
FGI Night of Stars to Honor Established Designers and Other Talents - Yahoo Lifestyle

Study uncovers molecular mechanisms that promote cell-to-cell adhesion and communication – News-Medical.net

A study led by Northwestern Medicine investigators has identified the molecular mechanisms within protein complexes that promote cell-to-cell adhesion and communication, according to findings published in the Proceedings of the National Academy of Sciences.

Sergey Troyanovsky, PhD, professor of Dermatology and of Cell and Developmental Biology, was senior author of the study.

Cadherins and catenins are proteins that form multiprotein complexes, helping bind cells together and stabilize cell-cell interactions, thereby forming different tissues. Those complexes, called cadherin-catenin complex (CCC), form clusters, but exactly how other CCC-associated proteins are recruited into these clusters and how they affect the overall clustering process has up until now remained understudied, according to Troyanovsky.

Using mass spectrometry and cross-linking, a proteomics approach which involves chemically "linking" two or more neighboring molecules by a covalent bond, the investigators discovered that most CCC-associated proteins interact with CCCs outside of adherens junctions, or protein complexes that include cadherin receptors. Furthermore, structural modeling revealed that there is limited space for CCC-associated proteins to form clusters in the first place.

Next, the investigators analyzed two example CCC-associated proteins essential for cell polarity and cell proliferation, and found that each protein formed separate CCC-associated clusters.

A431 cells expressing EcDn were briefly stained and then immediately imaged simultaneously in green and red channels.

The findings suggest that protein-driven CCC clustering plays a role in cell-to-cell adhesion, as well as enables cells to communicate with each other by synchronizing their signaling networks.

Different proteins which associate with CCC can sort cadherin into different populations of CCC clusters, and that's important because it's actually a mechanism of how signaling units can be equalized in different cells."

Sergey Troyanovsky, PhD, Member of the Robert H. Lurie Comprehensive Cancer Center of Northwestern University

Troyanovsky said the findings may also improve the understanding of the intracellular mechanisms that give rise to skin diseases, as most skin diseases are caused by defects in cell-to-cell adhesion within the epidermis.

Source:

Journal reference:

Troyanovsky, R.B., et al. (2021) Sorting of cadherincatenin-associated proteins into individual clusters. PNAS. doi.org/10.1073/pnas.2105550118.

Visit link:
Study uncovers molecular mechanisms that promote cell-to-cell adhesion and communication - News-Medical.net

Berkeley Lights and Bayer announce a multi-year agreement aimed at revolutionizing the discovery of next-generation traits – Yahoo Finance

Agreement debuts Berkeley Lights rapid high-throughput functional screening capability to accelerate and expand the discovery and development of Bayer Crop Sciences seeds and traits product pipeline

EMERYVILLE, Calif., Aug. 11, 2021 (GLOBE NEWSWIRE) -- Berkeley Lights, Inc. (Nasdaq: BLI) a leader in the functional characterization of live biology, announced a multi-year agreement with Bayer to develop and perform high-throughput functional screening workflows aimed at accelerating and expanding the discovery of novel traits. Terms of the agreement were not disclosed.

Berkeley Lights will leverage its platform to screen individual variants of bioactives for Bayer in a massively high-throughput manner. The outcome will be a significant acceleration of the agricultural leaders pipeline for discovery and development of novel traits. This agreement marks the first application of Berkeley Lights technology for use in the agricultural sector.

After evaluating the technology landscape, it became clear that the Berkeley Lights Platform is uniquely positioned to enable Bayer Crop Science to deliver on our commitment for world-class innovation and standards in sustainability for farmers, consumers and the environment, said Brianna White, Head of Trait Design and Science for Bayer. Our agreement with Berkeley Lights to develop and perform high-throughput functional screening workflows will enable us to accelerate and expand our trait discovery program.

We are excited to support Bayer in the discovery and development of novel traits, said Eric Hobbs, PhD, chief executive officer of Berkeley Lights. This agreement is an example of Berkeley Lights executing on our commitment to deploy our technologies into new, large market segments. Our proprietary approach to high-throughput functional screening is applicable to a variety of sectors and applications even beyond agriculture, including antibody, therapeutic protein, and enzyme engineering for pharmaceutical, life science, and industrial products.

Story continues

About Berkeley LightsBerkeley Lights is a leading digital cell biology company focused on enabling and accelerating the rapid development and commercialization of biotherapeutics and other cell-based products for our customers. The Berkeley Lights Platform captures deep phenotypic, functional, and genotypic information for thousands of single cells in parallel and can also deliver the live biology customers desire in the form of the best cells. Our platform is a fully integrated, end-to-end solution, comprising proprietary consumables, including our OptoSelect chips and reagent kits, advanced automation systems, and application software. We developed the Berkeley Lights Platform to provide the most advanced environment for rapid functional characterization of single cells at scale, the goal of which is to establish an industry standard for our customers throughout their cell-based product value chain.

The Berkeley Lights Platform is FOR RESEARCH USE ONLY. Not for use in diagnostic procedures.

Forward-Looking StatementsTo the extent that statements contained in this press release are not descriptions of historical facts regarding Berkeley Lights or its products, they are forward-looking statements reflecting the current beliefs and expectations of management. Such forward-looking statements involve substantial known and unknown risks and uncertainties that relate to future events, and actual results and product performance could differ significantly from those expressed or implied by the forward-looking statements. Berkeley Lights undertakes no obligation to update or revise any forward-looking statements. For a further description of the risks and uncertainties relating to the Companys growth and evolution, including its ability to accelerate, expand and/or revolutionize the discovery of novel traits and to expand its technology approach to high-throughput functional screening into new sectors and applications, see the statements in the "Risk Factors" sections, and elsewhere, in our filings with the U.S. Securities and Exchange Commission.

Berkeley Lights Press Contactchristy.nguyen@berkeleylights.com

Investor Contactir@berkeleylights.com

Continue reading here:
Berkeley Lights and Bayer announce a multi-year agreement aimed at revolutionizing the discovery of next-generation traits - Yahoo Finance

Immune-Onc Therapeutics Announces FDA Clearance of IND Application to Initiate First-In-Human Trial of IO-108, a Novel Antagonist Antibody Targeting…

PALO ALTO, Calif.--(BUSINESS WIRE)--Immune-Onc Therapeutics, Inc. (Immune-Onc), a clinical-stage cancer immunotherapy company developing novel biotherapeutics targeting immunosuppressive myeloid checkpoints, today announced that the U.S. Food and Drug Administration (FDA) has cleared the companys Investigational New Drug (IND) application for IO-108, a novel antagonist antibody targeting Leukocyte Immunoglobulin-Like Receptor B2 (LILRB2, also known as ILT4) for the treatment of solid tumors. Preclinical data presented at the 2020 Society for Immunotherapy of Cancers annual meeting demonstrate that IO-108 functions as a myeloid checkpoint inhibitor. IO-108 reprograms immune-suppressive myeloid cells toward a pro-inflammatory phenotype, leading to enhanced innate and adaptive anti-tumor immunity.

"The clearance of the IO-108 IND represents another major milestone for Immune-Onc as we progress our pipeline of novel myeloid checkpoint inhibitors targeting the LILRB family of immune inhibitory receptors, said Charlene Liao, Ph.D., chief executive officer of Immune-Onc. We are highly encouraged by the strength of the preclinical data of IO-108 and are pleased that our expertise in LILRB biology and translational sciences enables us to advance this asset into the clinic. We look forward to initiating the trial to further understand the role of LILRBs in cancer, and to test the potential of IO-108 in treating patients with advanced solid tumors.

The Phase 1, multicenter, dose-escalation study will consist of a monotherapy cohort and a combination therapy cohort to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of IO-108 alone and in combination with pembrolizumab, an anti-PD-1 antibody. Biomarkers will be assessed to enable a mechanistic understanding of clinical data and inform future trials. This study may also provide an opportunity to identify preliminary efficacy signals. After determination of the recommended Phase 2 dose, Immune-Onc plans to evaluate the efficacy, safety, and tolerability of IO-108 in combination with pembrolizumab and as monotherapy in indication-specific expansion cohorts.

IO-108 binds to LILRB2 with high affinity and specificity and blocks the interaction of LILRB2 with multiple ligands that are involved in cancer-associated immune suppression including HLA-G, ANGPTLs, SEMA4A, and CD1d. In preclinical studies, treatment of various primary human immune cell systems containing myeloid cells with IO-108 results in enhanced pro-inflammatory responses to multiple stimuli that are relevant to anti-tumor immunity. As a single agent, IO-108 reverses the anti-inflammatory myeloid cell phenotype that results from tumor conditioning and promotes the differentiation of monocytes into pro-inflammatory dendritic cells. Moreover, IO-108 potentiates the effect of PD-1 blocking antibodies on CD4+ T cell activation in co-cultures with allogeneic macrophages. In mouse models IO-108 inhibits the growth of solid tumors, which is associated with enhanced T cell responses. Together these data demonstrate that IO-108 has the potential to provide additive or synergistic benefit in combination with standard-of-care immunotherapies and/or immunogenic therapies for solid tumors that are both resistant and sensitive to T-cell checkpoint inhibitors.

ABOUT LILRB2 (ILT4)

LILRB2, also known as ILT4, is expressed mostly on myeloid cells, including monocytes, dendritic cells, macrophages, and neutrophils. In solid tumors, interaction of LILRB2 with tumor microenvironment (TME) relevant ligands, including HLA-G, ANGPTLs, SEMA4A, and CD1d, makes myeloid cells pro-tumorigenic (tolerating or promoting tumor growth) and promotes tumor immune evasion.

ABOUT IMMUNE-ONC THERAPEUTICS, INC.

Immune-Onc Therapeutics, Inc. (Immune-Onc) is a clinical-stage cancer immunotherapy company dedicated to the discovery and development of novel myeloid checkpoint inhibitors for cancer patients. The company aims to translate unique scientific insights in myeloid cell biology and immune inhibitory receptors to discover and develop first-in-class biotherapeutics that disarm immune suppression in the tumor microenvironment.

Immune-Onc has a promising pipeline with a current focus on targeting the Leukocyte Immunoglobulin-Like Receptor subfamily B (LILRB) of myeloid checkpoints. Immune-Oncs focused platform approach has led to the development of several promising therapeutics across various stages of development, including IO-108 in Phase 1 clinical development for solid tumors and IO-202, a first-in-class antibody targeting LILRB4 (also known as ILT3), in Phase 1 clinical development for the treatment of acute myeloid leukemia (AML) and chronic myelomonocytic leukemia (CMML). The company also plans to evaluate IO-202 in solid tumors. The U.S. Food and Drug Administration granted IO-202 Orphan Drug Designation for treatment of AML in October 2020. Additional assets in Immune-Oncs pipeline include IO-106, a first-in-class anti-LAIR1 antibody, and multiple undisclosed programs for solid tumors and hematologic malignancies.

The company has strategic research collaborations with The University of Texas, Albert Einstein College of Medicine, and Memorial Sloan Kettering Cancer Center, and has invested in proprietary models, assays, and tools to interrogate the biology and translate this cutting-edge research into the development of novel therapies. Headquartered in Palo Alto, California, Immune-Onc has assembled a diverse team with deep expertise in drug development and proven track records of success at leading biotechnology companies. For more information, please visit http://www.immune-onc.com and follow us on Twitter and LinkedIn.

Originally posted here:
Immune-Onc Therapeutics Announces FDA Clearance of IND Application to Initiate First-In-Human Trial of IO-108, a Novel Antagonist Antibody Targeting...

UPMC-Pitt Researchers Receive Grant to Advance Liver Organoids – UPMC & Pitt Health Sciences News Blog – UPMC

Share this on:

Researchers from the University of Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine and Carnegie Mellon Universitys (CMU) Computational Biology Department have been awarded almost $1.5 million from the National Science Foundation (NSF) to develop new approaches for producing liver organoids tiny, lab-grown human organs that have potential for disease modeling, drug discovery and transplantation.

Mouse models are often used for research on human diseases, but there is a gap between studies in cells and animals and translation to humans, said principal investigator Dr. Mo Ebrahimkhani, associate professor of pathology and bioengineering at Pitt and member of the Pittsburgh Liver Research Center and the McGowan Institute. Organoids help fill that gap because they can capture the complexity of our human organs and tissues.

Dr. Mo Ebrahimkhani

To grow these mini organs, researchers start with induced pluripotent stem cells (iPSCs), which are made from adult cells. iPSCs can turn into almost any cell type, and given the right signals at the right time, they can grow into complex 3D organoids of multiple tissue types.

Traditionally, organoid development is directed by adding a cocktail of chemicals at different times. But this trial-and-error approach is hard to optimize and can introduce variability among batches, hampering reproducibility and scalability of organoid production and making it difficult to compare findings across labs. Another problem is that development can stall, leaving cells with immature or abnormal characteristics or lacking blood vessels, which are important for transplantation applications.

These challenges prompted the NSFs call for research proposals that apply multidisciplinary approaches to understand processes of cellular differentiation during organ development and improve production of mature, functional cells or organoids.

Ebrahimkhani, in collaboration with Dr. Samira Kiani, co-principal investigator and associate professor of pathology at Pitt, and Dr. Ziv Bar-Joseph, professor of computational biology and machine learning who heads the Systems Biology Group at CMU, received funding to develop liver organoids that can be produced consistently and reliably. The researchers aim to program iPSCs with genetic instructions that guide cells to produce liver tissues with specified structure and function, rather than relying on external addition of growth factors.

We will develop programmable organoids with genetic circuits and switches, allowing them to sense the cell types and states, so they know when its the right time to turn on and turn off their programs, Ebrahimkhani explained.

The cells can be frozen, stored and shipped to different labs, he said. This will improve reproducibility, reliability and sharing of the science.

To generate genetic programs for liver organoids, the researchers aim to understand the processes that control organ development by combining two different tactics.

To improve lab-grown liver organoids, researchers will use computational analyses and genetic tools to understand and guide organ development. Credit: Emma Br

Our lab uses synthetic biology to manipulate genetic pathways in cells. By building these pathways, we understand the process, said Ebrahimkhani. Its like building a car from all the individual components. As I build the car, I understand how it functions.

Bar-Josephs lab will use computational analyses to reconstruct models that help explain how cells differentiate and interact with one another during organ development, an approach termed systems biology.

Systems biology is like trying to understand the car by taking it apart and looking at its individual components, said Bar-Joseph. In this research, we aim to combine synthetic biology and systems biology to understand organ development.

As part of the grant, the researchers will also develop an outreach program to help educate the public about stem cell and organoid research. They will create videos and a website on the Tomorrow Life platform, a science communication filmmaking initiative directed by Kiani.

Although the grant will focus on liver organoids, the researchers say that their findings could also inform the development of other types of human organoids, such as brain, kidney or retina.

Read this article:
UPMC-Pitt Researchers Receive Grant to Advance Liver Organoids - UPMC & Pitt Health Sciences News Blog - UPMC