Category Archives: Cell Biology

Dissecting the Unusual Biology of the SARS-CoV-2 Delta Variant – The Scientist

SARS-CoV-2 is fitter than ever. Its latest incarnation, the Delta (or B.1.617.2) variant, is the fastest-spreading form of the virus yet. First identified in India, which it swept through killing hundreds of thousands this spring, Delta has swiftly become the most dominant coronavirus variant worldwide. While its already driving rapid increases in hospitalizations and deathsoverwhelmingly in unvaccinated populationssimply by virtue of being more transmissible, it may also cause more severe disease than some previously dominating SARS-CoV-2 variants.

Meanwhile, emerging data suggest that when vaccinated people become infected with the Delta variant and develop symptomswhich, although increasing in frequency, remains exceedingly rare, officials reportthey might be as contagious as unvaccinated infected people. Those findings motivated the Centers for Disease Control and Prevention (CDC) to recommend in late July that in places with high transmission of the virus, even fully vaccinated people should wear masks indoors.

The Delta variants emergence isnt a shock to many researchers; spreading throughout the globe for a year and a half, SARS-CoV-2 was destined to adapt to humans and become better at infecting them. The mystery remains, however, exactly how Delta affects the human body differently than other variants. A flurry of recent research, little of which has undergone peer review, is yielding some clueson how the Delta variant might produce greater amounts of virus in peoples airways, for instance, which could make it more transmissible, and how it could cause more severe disease.

Scientists are also uncovering cause for hope. Although the antibodies induced by vaccines available in the US are slightly less effective against the Delta variant, the shots are largely still powerful protectors against severe cases of COVID-19. And according to preliminary data, breakthrough infections in vaccinated people resolve faster than in unvaccinated people.

Weve already seen a variety of other variants that have somebut maybe not allof the same characteristics that Delta has, such as increased transmission and a slight reduction in the effectiveness of vaccine-induced antibodies, says Richard Kennedy, an immunologist at the Mayo Clinic in Rochester, Minnesota. None of those are really a surprise. This [variant] just seems to have more of all of those.

Measuring SARS-CoV-2s transmissibility in the real world is notoriously difficult as human behavior is continuously changing, notes virologist Paul Bieniasz of Rockefeller University, but the fact that the Delta variant has so quickly displaced its dominating predecessor Alpha (B.1.1.7) is convincing evidence that its able to spread more rapidly in incompletely vaccinated populations. The UK governments Scientific Pandemic Influenza Group on Modelling, Operational sub-group (SPI-M-O) estimates that Delta is between 40 and 60 percent more transmissible than the Alpha variant and nearly twice as transmissible as the original virus first identified in China.

In explaining this heightened transmissibility, scientists point to not-yet-peer-reviewed data suggesting that infection with the Delta variant results in greater levels of virus accumulating more rapidly in human airways. In China, researchers tracked 62 people exposed to the virus who developed infections with the variant. They took daily measurements of the concentrations of viral RNA in swabs from the backs of the participants throats. Compared to 63 people infected with other variants, people with Delta infections tested positive sooner: four days after viral exposure compared to six. And when patients infected with Delta first tested positive, they carried around 1000 times more viral RNA. Although virologist Angela Rasmussen of the University of Saskatchewans Vaccine and Infectious Disease Organization cautions that viral RNA isnt necessarily a proxy for the amount of virus that is actually infectious, its probably a safe assumption that people are making more virus and shedding more virus when infected with the Delta variant, she says.

Perhaps the variant replicates faster inside cells or binds more tightly to human cells ACE2 receptor, which the viruss spike protein uses to enter cells, Kennedy speculates. But much attention has surrounded a particular mutation on the Delta variant called P681R, which suggests another potential mechanism. The mutation lies at the junction between two subunits of SARS-CoV-2s spike protein, at a site that allows the human cellular enzyme furin to cleave the spike into two parts. This cleavage, some experiments suggest, primes spike proteins in a way that allows them to fuse more readily with a host cells membrane, according to a detailed description in Nature.

The Delta variants P681R mutation seems to allow that cleavage to occur more efficiently than in the original SARS-CoV-2 strain. This could help the variant infect more cells in a given amount of time, and therefore, make more copies of itself overall. Having a more efficient furin cleavage site could lead to increased amounts of virus in the respiratory tract, Bieniasz says. The Alpha variantalso more transmissible than other versions of SARS-CoV-2 when it first emergedhas a similar mutation.

Some scientists are skeptical that the P681R mutation is to blame for the heightened viral load in people infected with Delta. Kei Sato, a virologist at the University of Tokyo, says that in his own experiments, he and his colleagues generally didnt observe much difference in replication or the ability to infect cells when comparing the Delta variant to a previously described form of the virus that lacks the P681R mutation. In any case, Bieniasz says hed be surprised if the Delta variants heightened transmissibility were caused by P681R alone. The variant carries a handful of other mutations on the spike protein, and more scattered around the genome. But there isnt really any information at this point that would tell you mechanistically what their contribution is to the increased fitness of Delta, he says.

The fact that the Delta variant has also triggered increasing numbers of infections in vaccinated people has generated a bevy of headlines, some of which have been criticizedfor their alarmist nature. The CDCs revised guidelines on mask-wearing were based in part on data from an outbreak in Massachusetts in the wake of large public gatherings. In July, officials identified 469 people in Barnstable County who tested positive, mostly for the Delta variant (participants had likely asked to be tested, and the majority had developed COVID-19 symptoms). Three-quarters were fully vaccinated, in part reflecting a relatively high vaccination rate in the area. Scientists stress that the vaccines still work as intended; they were designed to prevent severe disease, not infections. The Massachusetts study, for instance, reported only five hospitalizationsfour in fully vaccinated peopleand no deaths.

Nationwide, robust figures on the frequency of breakthrough cases are elusiveparticularly of asymptomatic or mild cases; the CDC only monitors instances that have progressed to hospitalization or death. According to an unpublished internal CDC document obtained by ABC News, breakthrough cases are reported for 0.096 percentor 153,000of the 156 million Americans who are fully vaccinated. A recent review of data from 25 states also estimated the rate of breakthrough cases to be well below 1 percent. According to that same review, the rates of hospitalizations and deaths with COVID-19 among the fully vaccinated range from effectively zero to 0.06 percent and 0.01, respectively.

Surprisingly, the Barnstable County study detected similar amounts of viral RNA in the specimens of vaccinated and unvaccinated participants. While thats not necessarily reflective of the peoples capacity to infect others, it is indisputable that in this outbreak, there seems to be transmission between previously vaccinated people, says Hana Akselrod, an infectious disease physician at George Washington University. The fact that participants had been gathering in bars and other crowed spaces probably created a high-risk setting for viral transmission, she adds.

Kennedy cautions that its hard to say how much of the increase in breakthrough infections is because of the Delta variants specific biology, rather than simply because its dominating a general surge in cases. But recent CDC data from Colorado does suggest vaccines are less effective against the variant; when it sped through Mesa County, the overall effectiveness of the available vaccines against symptomatic infection dropped to 78 percent, while in counties with lower Delta levels, it was 89 percent.

The variants fitness advantage could feasibly allow it to build up a high viral load before vaccinated immune systems crank out enough antibodies to quash it, Akselrod says. Anatomically speaking, its possible that while antibodies induced by current vaccines form high, protective levels in the lungswhere infection is associated with severe COVID-19 symptomsthey might be less able to access mucosal tissues in the nose and upper respiratory tract, allowing viral replication to occur there, Bieniasz speculates.

Another possible factor in establishing infections in vaccinated people is the Delta variants modest ability to evade vaccine-induced antibodies, Akselrod suggests. One yet-to-be-peer-reviewed paper posted to medRxiv in mid-July pitted antibodies isolated from 40 healthcare workers who had received either the Pfizer-BioNTech or Moderna mRNA vaccines against different variants. They found the Delta variant reduces the ability of antibodies to neutralize the virus by around 2.5-fold compared to a previously described isolateslightly more so than the Alpha variant but not as much as the Beta variant (B.1.351) first discovered in South Africa or the Gamma variant (P.1) first spotted in Brazil.

This is likely due to mutations to the Delta variants receptor binding domainthe patch on the spike protein that latches onto host cells ACE2 receptors and is targeted by vaccines, Kennedy says. What happens is the antibody either doesnt bind or doesnt bind as well, he explains. But the human immune system typically produces many different antibodies against individual viral proteinsnot to mention other kindsof immune machinerypreventing a complete reduction in vaccine effectiveness.

To wit: data from the Sisonke clinical trial in South Africa, reported in a news conference on Friday, suggest that even when it comes to Delta, the Johnson & Johnson vaccine is 71 percent effective against hospitalization and 95 percent effective against death. Rasmussen says she expects future variants to arise for which current vaccines are less protective, but for now, the vaccines are actually holding up very, very well against Delta.

In line with that idea is unpublished data from Singapore posted to medRxiv on July 31 that tracked Delta infections in 88 vaccinatedand mostly symptomaticpeople. Although their viral RNA loads for the first week of illness were similar to those recorded in a group of 130 infected unvaccinated people, they decreased much more quickly in the vaccinated group. And it showed that they had a huge spike in neutralizing antibodies immediately on testing positive, adds Rasmussen, who wasnt involved in the research.

Even so, several experts, including Rasmussen, Sato, and Bieniasz, agree with the CDCs recommendation that vaccinated people should wear masks in high-risk settings. No one wanted to be spending the summer sweating under masks, but this is the situation we have now, and we have to protect each other, Akselrod says. The primary use of masks is to prevent infections among unvaccinated people who are more contagious for longer and are most at risk for developing severe COVID-19 with Delta.

Its not clear if the elevated risk of hospitalization and death that someepidemiological studies have linked to Delta has more to do with characteristics of the populations in which Delta is circulating, or with the biology of the variant, Akselrod says. But some researchers have explored possible biological explanations. In a recent preprint, the University of Tokyos Sato and his colleagues report that when they allowed a SARS-CoV-2 isolate to infect modified monkey cells in vitro, the cells would stick together, forming connected structures known as syncytia, which have beenspotted in the lungs of people with COVID-19. The team suspected that SARS-CoV-2s furin cleavage site might be facilitating the observed clumping.

In addition to helping the viruses fuse with cells, its plausible that cleavage of the spike protein could also somehow cause infected cells to fuse with other cells close by. Notably, the team saw that syncytia induced by the Delta variantwhose P681R mutation may allow for more efficient cleavagewere much larger than those induced by a different form of SARS-CoV-2 that lacks the mutation. Interestingly, hamsters infected with SARS-CoV-2 viruses engineered to express the P681R mutation showed more signs of obstructed airways, and lost weight more quickly, than those infected with the other version of SARS-CoV-2suggesting the variant caused more severe disease. Sato hypothesizes that by causing larger syncytia to form, the Delta variant could be causing more severe tissue damage in the lung. I think that is the reason why the P681R-bearing virus is more pathogenic, he says.

Rasmussen isnt fully persuaded that the Delta variant is inherently more pathogenic. After all, hamsters are imperfect models of human COVID-19. But the P681R-driven syncytia formation might somehow explain why the virus is more transmissible, she says. Some other viruses are known to use such fused structures to spread from cell to cell, sometimes allowing more rapid spread across tissues.

Meanwhile, Akselrod says shes worried about a Delta-driven surge of mostly young, unvaccinated people wholl develop long-term symptoms known as long COVID. Amid a depleted healthcare workforce, the COVID-19 recovery clinic she directs is still struggling to care for long COVID patients who were infected with the Alpha variant in late 2020 or early 2021. To think of getting another giant wave of Delta on top of that is just very, very sad, she says. Its still unclear, though, whether long COVID may be proportionally more frequent with Delta, and whether even vaccinated people could develop long COVID. Thats a big scientific unknown right now, she says.

As researchers continue to chip away at questions about Delta, theyre also keeping in mind the new variants that are likely on the horizon. I think well see more variants that have increased transmissibility. We may or may not see variants that cause more severe disease. . . . Well undoubtedly see variants that have an increased ability to evade immune responses, Kennedy says. Well run out of letters in the Greek alphabet before we run out of variants.

Continue reading here:
Dissecting the Unusual Biology of the SARS-CoV-2 Delta Variant - The Scientist

Regenerative Medicine Market Size Worth $57.08 Billion By 2027: Grand View Research, Inc. – Markets Insider

SAN FRANCISCO, Aug. 12, 2021 /PRNewswire/ --The global regenerative medicine marketsize is expectedto reach USD 57.08 billion by 2027, growing at a CAGR of 11.27% over the forecast period, according to a new report by Grand View Research, Inc. Recent advancements in biological therapies have resulted in a gradual shift in preference toward personalized medicinal strategies over the conventional treatment approach. This has resulted in rising R&D activities in the regenerative medicine arena for the development of novel regenerative therapies.

Key Insights & Findings:

Read 273 page research report, "Regenerative Medicine Market Size, Share & Trends Analysis Report By Product (Cell-based Immunotherapies, Gene Therapies), By Therapeutic Category (Cardiovascular, Oncology), And Segment Forecasts, 2021 - 2027", by Grand View Research

Furthermore,advancements in cell biology, genomics research, and gene-editing technology are anticipated to fuel the growth of the industry. Stem cell-based regenerative therapies are in clinical trials, which may help restore damaged specialized cells in many serious and fatal diseases, such as cancer, Alzheimer's, neurodegenerative diseases, and spinal cord injuries. For instance, various research institutes have adopted Human Embryonic Stem Cells (hESCs) to develop a treatment for Age-related Macular Degeneration (AMD).

Constant advancements in molecular medicines have led to the development of gene-based therapy, which utilizes targeted delivery of DNA as a medicine to fight against various disorders. Gene therapy developments are high in oncology due to the rising prevalence and genetically driven pathophysiology of cancer. The steady commercial success of gene therapies is expected to accelerate the growth of the global market over the forecast period.

Grand View Research has segmented the global regenerative medicine market on the basis of product, therapeutic category, and region:

List of Key Players of Regenerative Medicine Market

Check out more studies related to Global Biotechnology Industry, conducted by Grand View Research:

Gain access to Grand View Compass, our BI enabled intuitive market research database of 10,000+ reports

About Grand View Research

Grand View Research, U.S.-based market research and consulting company, provides syndicated as well as customized research reports and consulting services. Registered in California and headquartered in San Francisco, the company comprises over 425 analysts and consultants, adding more than 1200 market research reports to its vast database each year. These reports offer in-depth analysis on 46 industries across 25 major countries worldwide. With the help of an interactive market intelligence platform, Grand View Research helps Fortune 500 companies and renowned academic institutes understand the global and regional business environment and gauge the opportunities that lie ahead.

Contact:Sherry JamesCorporate Sales Specialist, USAGrand View Research, Inc.Phone: 1-415-349-0058Toll Free: 1-888-202-9519Email: sales@grandviewresearch.comWeb: https://www.grandviewresearch.comFollow Us: LinkedIn| Twitter

Logo: https://mma.prnewswire.com/media/661327/Grand_View_Research_Logo.jpg

See more here:
Regenerative Medicine Market Size Worth $57.08 Billion By 2027: Grand View Research, Inc. - Markets Insider

Rewiring cell division to make eggs and sperm | Penn Today – Penn Today

To create eggs and sperm, cells must rewire the process of cell division.

Mitosis, the common type of cell division that human bodies use to grow everything from organs to fingernails and to replace aging cells, produces two daughter cells with the same number of chromosomes and approximately the same DNA sequence as the original cell. Meiosis, the specialized cell division that makes egg and sperm in two rounds of cell division, creates four granddaughter cells with new variations in their DNA sequence and half as many chromosomes in each cell. Meiosis uses most of the same cellular machinery as mitosis to achieve this very different outcome; only a few key molecular players prompt the rewiring from one type of division to another.

One such key player is the protein Meikin, which is found exclusively in cells undergoing meiosis. New research from Penns Michael Lampson and Jun Ma, collaborating with Whitehead Institute Member Iain Cheeseman and graduate student Nolan Maier, demonstrates how Meikin is elegantly controlled and sheds light on how the protein acts to serve multiple roles during different stages of meiosis.

The findings, which appear in the journal Developmental Cell, reveal that Meikin is precisely cut in half midway through meiosis. Instead of this destroying the protein, one half of the molecule, known as C-Meikin, goes on to play a critical role as a previously hidden protein actor in meiosis.

It was a pleasure working together to understand how some of the specialized meiotic functions that are necessary for making healthy eggs and sperm are controlled, says Lampson, a professor of biology in the School of Arts & Sciences.

Cells have this fundamental process, mitosis, during which they have to divide chromosomes evenly or it will cause serious problems like cancer, so the system has to be very robust, Maier says. Whats incredible is that you can add one or two unique meiotic proteins like Meikin and dramatically change the whole system very quickly.

During both mitosis and meiosis, sister chromatidscopies of the same chromosomepair up to form the familiar X shape of a chromosome. In mitosis, each chromatideach half of the Xis connected to a sort of cellular fishing line ,and these lines reel the chromatids to opposite ends of the cell, where the two new cells are formed around them.

In the first round of division in meiosis, however, the sister chromatids stick together, and one whole X is reeled into each new cell. Meikin helps to achieve this different outcome by ensuring that, while the chromosomes are being unstuck from each other in preparation for being pulled apart, each pair of sister chromatids stays glued together in the right place. Meikin also helps ensure that certain cellular machinery on the sister chromatids is fused so that they will connect to the same line and be reeled together to the same side of the cell.

More specifically, when chromosomes are first paired up, they are glued together by adhesive molecules in three regions: the centromere, or center of the X, where Meikin localizes; the region around the center; and the arms of the X. In the first round of meiosis, Meikin helps to keep the glue in the region around the center intact so the sister chromatids will stick together. Simultaneously, Meikin helps prime the center region to be unglued, while a separate process unglues the arms. This ungluing allows the chromosomes to separate and be prepared for later stages of meiosis.

Cheeseman and Maier initially predicted that Meikins role ended after meiosis I, the first round of meiotic cell division. In meiosis II, the second round of cell division, the cells being created should end up with only one sister chromatid each, and so the chromatids must not be kept glued together. Maier found that near the end of meiosis I Meikin is cleaved in two by an enzyme called Separase, the same molecule that cleaves the adhesive molecules gluing together the chromosomes. At first, this cleavage seemed like the end of Meikin and the end of this story.

However, unexpectedly, the researchers found that cells lacking Meikin during the second half of meiosis do not divide properly, prompting them to take another look at what happens to Meikin after it gets cleaved. They found that Separase cleaves Meikin at a specific point--carving it with the precision of a surgeons scalpelto create C-Meikin, a previously unknown protein that turns out to be necessary for meiosis II. C-Meikin has many of the same properties as the intact Meikin molecule, but it is just different enough to take on a different role: helping to make sure that the chromosomes align properly before their final division.

Theres a lot of protein diversity in cells that you would never see if you dont go looking for it, if you only look at the DNA or RNA. In this case, Separase is creating a completely different protein variant of Meikin than can function differently in meiosis II, says Cheeseman, who is also a professor of biology at the Massachusetts Institute of Technology. Im very excited to see what we might discover about other hidden protein forms in cell division.

Answering the question of Meikins role and regulation throughout meiosis required a close collaboration and partnership between Maier and Lampson lab senior research scientist Ma, the Lampson lab being experts on studying meiosis using mouse models. Working with mouse oocytes, or immature egg cells, Ma was able to reveal the behaviors and critical contributions of Meikin cleavage in meiotic cells in mice. Both labs credit the close exchange with helping them to get a deeper understanding of how cells rewire for meiosis.

Once cells have completed these specialized meiotic divisions, the researchers found that it was critical for oocytes to fully eliminate Meikin. They determined that, after meiosis II, C-Meikin is degraded by another molecule, the anaphase-promoting complex or APC/Cthis time for good. With Meikin gone and the rewiring of cell division reversed, eggs and sperm are ready for mitosis; should they fuse and form an embryo, that is the next cell division they will undergo. The researchers note that the way Meikin is regulated by being broken downfirst into C-Meikin and then completelymay help cells to organize their timing during meiosis. Breaking apart a protein is an irreversible step that creates a clear demarcation between before and after in a multi-step process.

The researchers hope that by uncovering the intricacies of meiosis, they may shed light on what happens when the creation of eggs and sperm goes wrong and perhaps contribute to the understanding of infertility.

Written by Greta Friar

The study was supported by grants from The Harold G & Leila Y. Mathers Charitable Foundation, the National Science Foundation (Grant 2029868), a Pilot award from the Global Consortium for Reproductive Longevity and Equity, the National Institutes of Health (GM126930 and GM122475), and the Henry and Frances Keany Rickard Fund Fellowship from the MIT Office of Graduate Education.

See the article here:
Rewiring cell division to make eggs and sperm | Penn Today - Penn Today

Stem Cell Scientists Explore the Latent Regenerative Potential of the Inner Ear – SciTechDaily

The organ of Corti, the hearing organ of the inner ear, contains rows of sensory hearing cells (green) surrounded by supporting cells (blue). Credit: Image by Yassan Abdolazimi/Segil Lab/USC Stem Cell

Scientists from the USC Stem Cell laboratory of Neil Segil have identified a natural barrier to the regeneration of the inner ears sensory cells, which are lost in hearing and balance disorders. Overcoming this barrier may be a first step in returning inner ear cells to a newborn-like state thats primed for regeneration, as described in a new study published inDevelopmental Cell.

Permanent hearing loss affects more than 60 percent of the population that reaches retirement age, said Segil, who is a Professor in the Department of Stem Cell Biology and Regenerative Medicine, and the USC Tina and Rick Caruso Department of Otolaryngology Head and Neck Surgery. Our study suggests new gene engineering approaches that could be used to channel some of the same regenerative capability present in embryonic inner ear cells.

In the inner ear, the hearing organ, which is the cochlea, contains two major types of sensory cells: hair cells that have hair-like cellular projections that receive sound vibrations; and so-called supporting cells that play important structural and functional roles.

When the delicate hair cells incur damage from loud noises, certain prescription drugs, or other harmful agents, the resulting hearing loss is permanent in older mammals. However, for the first few days of life, lab mice retain an ability for supporting cells to transform into hair cells through a process known as transdifferentiation, allowing recovery from hearing loss. By one week of age, mice lose this regenerative capacityalso lost in humans, probably before birth.

Based on these observations, postdoctoral scholar Litao Tao, PhD, graduate student Haoze (Vincent) Yu, and their colleagues took a closer look at neonatal changes that cause supporting cells to lose their potential for transdifferentiation.

In supporting cells, the hundreds of genes that instruct transdifferentiation into hair cells are normally turned off. To turn genes on and off, the body relies on activating and repressive molecules that decorate the proteins known as histones.In response to these decorations known as epigenetic modifications, the histone proteins wrap the DNA into each cell nucleus, controlling which genes are turned on by being loosely wrapped and accessible, and which are turned off by being tightly wrapped and inaccessible. In this way, epigenetic modifications regulate gene activity and control the emergent properties of the genome.

In the supporting cells of the newborn mouse cochlea, the scientists found that hair cell genes were suppressed by both the lack of an activating molecule, H3K27ac, and the presence of the repressive molecule, H3K27me3.However, at the same time, in the newborn mouse supporting cells, the hair cell genes were kept primed to activate by the presence of yet a different histone decoration, H3K4me1.During transdifferentiation of a supporting cell to a hair cell, the presence of H3K4me1 is crucial to activate the correct genes for hair cell development.

Unfortunately with age, the supporting cells of the cochlea gradually lost H3K4me1, causing them to exit the primed state. However, if the scientists added a drug to prevent the loss of H3K4me1, the supporting cells remained temporarily primed for transdifferentiation. Likewise, supporting cells from the vestibular system, which naturally maintained H3K4me1, were still primed for transdifferentiation into adulthood.

Our study raises the possibility of using therapeutic drugs, gene editing, or other strategies to make epigenetic modifications that tap into the latent regenerative capacity of inner ear cells as a way to restore hearing, said Segil. Similar epigenetic modifications may also prove useful in other non-regenerating tissues, such as the retina, kidney, lung, and heart.

Reference: 30 July 2021, Developmental Cell.DOI: 10.1016/j.devcel.2021.07.003

Additional co-authors of the study include Juan Llamas, Talon Trecek, Xizi Wang, andZlatka Stojanovain the Segil Lab at USC, and Andrew K. Groves at Baylor College of Medicine.

Sixty percent of this project was supported by federal funding from the National Institute on Deafness and Other Communication Disorders (R01DC015829, R01DC014832, T32DC009975, F31DC017376). Additional funding came from the Hearing Restoration Project at the Hearing Health Foundation.

See the rest here:
Stem Cell Scientists Explore the Latent Regenerative Potential of the Inner Ear - SciTechDaily

Astellas and Minovia Therapeutics Announce Strategic Collaboration for Novel Mitochondrial Cell Therapy Programs – Business Wire

TOKYO & HAIFA, Israel--(BUSINESS WIRE)--Minovia Therapeutics, Ltd. (CEO; Natalie Yivgi-Ohana, Ph.D., Minovia) and Astellas Pharma Inc. (TSE: 4503, President and CEO: Kenji Yasukawa, Ph.D., Astellas) today announced a worldwide strategic collaboration and license agreement for the research, development, and commercialization of novel cell therapy programs for diseases caused by mitochondrial dysfunction.

Through this strategic collaboration, Astellas and Minovia aim to accelerate the creation of allogeneic mitochondrial cell therapy programs. The two companies will jointly research cell therapy program candidates comprised of cells derived from Astellas proprietary genetically-engineered, induced pluripotent stem cells and augmented with Minovias proprietary MAT platform technology. The goal of these programs will be to treat diseases caused by mitochondrial dysfunction, through the transfer of healthy mitochondria to restore the patients tissues.

Minovia is a leading company in the field of mitochondrial cell therapy that utilizes mitochondrial transfer to deliver healthy mitochondria to a patients diseased cells. Minovia has a unique technology platform called Mitochondrial Augmentation Therapy (MAT), where the patients own cells are isolated, loaded with healthy mitochondria obtained from a healthy donor, and then re-infused back into the patient. Minovia is currently conducting research, development and clinical studies with MAT in mitochondrial diseases.

Astellas is engaged through its US subsidiary companies, Astellas Institute for Regenerative Medicine (AIRM) and Universal Cells Inc., to advance allogeneic, off-the-shelf, differentiated cell therapy programs derived from pluripotent stem cells1. The new collaboration with Minovia extends Astellas capabilities in mitochondrial biology, and follow its recent acquisition of Mitobridge, Inc. and Nanna Therapeutics Limited2,3.

We are excited and honored to collaborate with Astellas, stated Minovias Co-founder and Chief Executive Officer, Natalie Yivgi-Ohana, Ph.D. We share with Astellas both their passion for mitochondrial science and their commitment to patients in need of new therapies. As Minovia continues the development of Mitochondrial Augmentation Therapy, we believe this partnership is critical to accelerate the development of off-the-shelf, allogeneic cell therapy programs for the many patients living with mitochondrial diseases caused by mitochondrial dysfunction.

Naoki Okamura, Representative Director, Corporate Executive Vice President, Chief Strategy Officer and Chief Financial Officer, at Astellas said, "We, at Astellas, have positioned mitochondrial biology as one of the Primary Focuses of our research and development strategy to develop therapies for patients with unmet medical needs. One of the aspirations of this Primary Focus is to establish a mitochondrial cell therapy platform. Minovia is pioneering mitochondrial cell therapy and has unique technologies for enhancing delivery of healthy mitochondria to the patients cells. This strategic collaboration with Minovia will accelerate and expand our pipeline of treatment options for patients with diseases with underlying mitochodrial dysfunction.

Under the terms of the agreement, Minovia receives an upfront cash payment of $20M USD. Through the joint research program with Minovia, if Astellas develops and commercializes product candidates for diseases caused by mitochondrial dysfunction, Minovia is eligible to receive up to $420M USD per product in future development, regulatory and commercial milestone payments from Astellas.

*1: R&D Meeting (December 10, 2020). Available at: https://sw4503.swcms.net/ja/ir-library/ir-meetings/inframe/main/014/teaserItems1/07/linkList/0/link/RDmeeting2020_pre_jp.pdf *2: Astellas Corporate website Accelerating the discovery and development of novel drugs that target mitochondrial functions.. Available at: https://www.astellas.com/jp/en/stories/science/mitobridge *3: Astellas Corporate website Primary Focus - Mitochondria Biology. Available at: https://www.astellas.com/jp/en/partnering/primary-focus#Mitochondria-Biology

About MinoviaMinovia Therapeutics, Ltd. is a clinical stage company and the first to use a mitochondrial cell therapy approach with the hope of bringing life-changing therapies to patients living with mitochondrial diseases, through their proprietary Mitochondrial Augmentation Therapy (MAT) platform. Minovia has an on-going autologous cell therapy program in clinical development, utilizing MAT for the future potential treatment of primary and secondary mitochondrial diseases. For more information, please visit our website at https://www.minoviatx.com.

About AstellasAstellas Pharma Inc. is a pharmaceutical company conducting business in more than 70 countries around the world. We are promoting the Focus Area Approach that is designed to identify opportunities for the continuous creation of new drugs to address diseases with high unmet medical needs by focusing on Biology and Modality. Furthermore, we are also looking beyond our foundational Rx focus to create Rx+ healthcare solutions that combine our expertise and knowledge with cutting-edge technology in different fields of external partners. Through these efforts, Astellas stands on the forefront of healthcare change to turn innovative science into value for patients. For more information, please visit our website at https://www.astellas.com/en.

Cautionary NotesIn this press release, statements made with respect to current plans, estimates, strategies and beliefs and other statements that are not historical facts are forward-looking statements about the future performance of Astellas. These statements are based on managements current assumptions and beliefs in light of the information currently available to it and involve known and unknown risks and uncertainties. A number of factors could cause actual results to differ materially from those discussed in the forward-looking statements. Such factors include, but are not limited to: (i) changes in general economic conditions and in laws and regulations, relating to pharmaceutical markets, (ii) currency exchange rate fluctuations, (iii) delays in new product launches, (iv) the inability of Astellas to market existing and new products effectively, (v) the inability of Astellas to continue to effectively research and develop products accepted by customers in highly competitive markets, and (vi) infringements of Astellas intellectual property rights by third parties. Information about pharmaceutical products (including products currently in development) which is included in this press release is not intended to constitute an advertisement or medical advice.

See the article here:
Astellas and Minovia Therapeutics Announce Strategic Collaboration for Novel Mitochondrial Cell Therapy Programs - Business Wire

Growing demand for Cell Culture-based Vaccine to propel the global Cell Culture Market – BioSpace

In molecular and cellular biology, cell culture acts as a significant tool for biological research. Today, most biotechnology products are primarily reliant on the mass culturing of animal cell lines. Cell cultures have found applications in diverse areas and serve as a model system for numerous research efforts.

An increase in funding from the government for cell-based research is significantly triggering the growth of the global cell culture market. In addition to this, cell culture techniques are widely used as an alternative to current egg-based strategies for the development of cell-based vaccines. With an increase in communicable diseases and the risk of a pandemic, cell culture has potential applications in the development of viral vaccines. Thus, cell culture technology is been extensively used in the development of U.S-licensed vaccines such as vaccines against rubella, smallpox, chickenpox, hepatitis, rotavirus, and polio. All such factors coupled with the growing prevalence of chronic diseases leading to a surge in demand for monoclonal antibodies and growing use of single-use technologies are escalating the growth of the global market.

Growing demand for cell culture products including equipment, reagents, and cell culture medium from academic and research institutes is fuelling the growth of the market. Moreover, the growing demand for 3D cell culture, major players focusing to expand the business in untapped regions, and the launch of new cell cultures are strengthening the growth of the global market. In March 2021, Thermo Fisher Scientific announced launch of new Gibco Human Plasma-like Medium (HPLM). It mimics the metabolic profile of human plasma and is the first cell culture medium that is especially designed to enable the researchers to get a realistic view of cell growth in the human body. Furthermore, growing demand for Covid-19 vaccine due to the rapid spread of deadly coronavirus will generate ample opportunities for the growth of the global market during the forecast period.

The global cell culture market is segregated based on product, application, end-user, and region. The product segment is bifurcated into consumables and instruments. The application segment is divided into stem cell technology, tissue engineering & regenerative medicine, drug screening & development, cancer research, and others. The end-user comprises pharmaceutical & biotechnology companies, research institutes, and others.

Regionally, North America is estimated to dominate the global cell culture market during the forecast period. This is majorly due to increase in cell culture-based vaccine approvals, growing demand from research institutes, and the growing prevalence of chronic diseases. Asia Pacific is predicted to grow rapidly during the forecast period. This is attributed to the increase in government funding for research purpose and the growing demand for cell culture products.

Get Sample of this Research Report for more Insights https://www.zionmarketresearch.com/sample/cell-culture-industry

Thermo Fisher Scientific, Becton, Dickinson and Company, Lonza Group AG, Danaher Corporation, GE Healthcare, Merck KGaA, Sartorius AG, HiMedia Laboratories, Eppendorf, and Corning Incorporated are some of the prominent players that are operating in the global cell culture market.

About Us

Zion Market Research is an obligated company. We create futuristic, cutting edge, informative reports ranging from industry reports, company reports to country reports. We provide our clients not only with market statistics unveiled by avowed private publishers and public organizations but also with vogue and newest industry reports along with pre-eminent and niche company profiles. Our database of market research reports comprises a wide variety of reports from cardinal industries. Our database is been updated constantly in order to fulfill our clients with prompt and direct online access to our database. Keeping in mind the clients needs, we have included expert insights on global industries, products, and market trends in this database. Last but not the least, we make it our duty to ensure the success of clients connected to usafter allif you do well, a little of the light shines on us.

Contact Us

Zion Market Research

244 Fifth Avenue, Suite N202

New York, 10001, United States

Tel: +49-322 210 92714

USA/Canada Toll-Free No.1-855-465-4651

Email: sales@zionmarketresearch.com

Website: https://www.zionmarketresearch.com

See the article here:
Growing demand for Cell Culture-based Vaccine to propel the global Cell Culture Market - BioSpace

Researchers Find Molecular Switch That Regulates Fat Burning in Mice – Newswise

Newswise BOSTON Linked to serious health problems including cancer, diabetes and cardiovascular disease, obesity affects more than a third of adults in the United States. Presently, there are few safe and effective nonsurgical therapeutic interventions available to patients with obesity.

Now, a multi-disciplinary team of researchers has demonstrated that a metabolic regulatory molecule called Them1 prevents fat burning in cells by blocking access to their fuel source. Led by microscopy experts at Beth Israel Deaconess Medical Center (BIDMC) and metabolism experts at Weill Cornell Medicine and NewYork-Presbyterian, the study may contribute to the development of a new type of obesity treatment. The teams findings were published June 9 in Nature Communications.

To help explain how the protein Them1 turns off heat production, BIDMCs cell biology and microscopy expert, Susan Hagen, PhD, associate vice-chair for research in the Department of Surgery at BIDMC, and Yue Li, PhD, a postdoctoral researcher in her laboratory, used light and electron microscopy to observe Them1 in action in mouse brown fat cells grown in the laboratory.

Them1 is an interesting molecule, said Hagen. If you inhibit or block its expression, metabolism increases and that reduces body weight.

The experiments showed that when the cells are stimulated to burn fat, a chemical modification causes Them1 molecules to spread out, or diffuse, throughout the cell. This frees the cellular powerhouses called mitochondria to efficiently turn the cells fat stores into energy. But when the stimulation stops, Them1 molecules quickly reorganize into a structure called a biomolecular condensate. Situated between the mitochondria and the fats they use as fuel, the condensed Them1 molecules limit energy production.

It turned out to be so incredibly interesting, said Hagen, who is also director of Microscopy and Histology Core Facilities at BIDMC and associate professor of surgery at Harvard Medical School. We asked other microscopy experts whether they had ever seen anything like the unusual images we found in resting cells. Using very sophisticated electron microscopy techniques, we were able to show for the first time, as far as we know what the bimolecular condensate looks like in electron microscopy.

The study explains a new mechanism that regulates metabolism, said David Cohen, chief of the Division of Gastroenterology and Hepatology at Weill Cornell Medicine and NewYork-Presbyterian/Weill Cornell Medical Center and the Vincent Astor Distinguished Professor of Medicine at Weill Cornell Medicine. Them1 hacks the energy pipeline and cuts off the fuel supply to the energy-burning mitochondria. Humans also have brown fat and produce more Them1 in cold conditions, so the findings may have exciting implications for the treatment of obesity.

Cohen and Hagen, both members of the Harvard Digestive Diseases Center, have been collaborators since 1983. The current study supported in part by a five-year, multi-PI grant from the National Institutes of Health also included collaborators with expertise in structural biology from Emory University.

This was the most fun I have ever had in science in my life, Hagen added. Including multiple primary investigators with different expertise gives you the power of doing things that you could never do on your own.

Co-authors included Yue Li, Samaksh Goyal, Lay-Hong Ang, and Mahnoor Baqai of BIDMC; Norihiro Imai, Hayley T. Nichols, Tibor I. Krisko of Weill Cornell; Blaine R. Roberts, Matthew C. Tillman, Anne M. Roberts, and Eric A. Ortlund of Emory University.

This work was supported by the National Institutes of Health (R01 DK 103046, R01 DK0488730 and NIHT32DK007533), the Harvard Digestive Disease Center (P30 DK034854) and the National Institutes of Health shared-instrumentation grant program for the High Pressure Freezer (S10 OD019988-01), the Pinnacle Research Award from the AAASLD Foundation, Weill Cornell Department of Medicine Pre-Career Award, and an American Heart Association Postdoctoral Fellowship, and a Research Science Institute/Center for Excellence in Education Summer Research Fellowship.

The authors declare no competing interests.

About Beth Israel Deaconess Medical Center Beth Israel Deaconess Medical Center is a patient care, teaching, and research affiliate of Harvard Medical School and consistently ranks as a national leader among independent hospitals in National Institutes of Health funding. BIDMC is the official hospital of the Boston Red Sox. For more information, visitwww.bidmc.org.

Beth Israel Deaconess Medical Center is a part of Beth Israel Lahey Health, a health care system that brings together academic medical centers and teaching hospitals, community and specialty hospitals, more than 4,000 physicians and 35,000 employees in a shared mission to expand access to great care and advance the science and practice of medicine through groundbreaking research and education.

Read the original here:
Researchers Find Molecular Switch That Regulates Fat Burning in Mice - Newswise

New study shines light on how plant cells perceive and respond to mechanical forces – News-Medical.net

Minuscule tunnels through the cell membrane help cells to perceive and respond to mechanical forces, such as pressure or touch. A new study in the journal Science is among the first to directly investigate what one type of these mechanosensitive ion channels is doing in the tip-growing cells in moss and pollen tubes of flowering plants, and how.

Biologists led by Elizabeth Haswell at Washington University in St. Louis discovered that so-called PIEZO channels are not found along the plasma membrane in plant cells as they are in animal cells.

Instead, they observed that PIEZO channels have retreated into the plant cell, an unexpected discovery. PIEZO channels are found deeper within the cell, in the membranes of vacuoles -- the large, intracellular organelles that help maintain cell turgor and fulfill a number of other roles in the plant cell.

PIEZO channels in plants play a dramatic and critical role in regulating the shape of the vacuole and how much membrane there is."

Elizabeth Haswell, Professor of Biology in Arts & Sciences and a Howard Hughes Medical Institute-Simons Faculty Scholar

"This is the first example of PIEZO channels involved in regulating organelle morphology," she said. "The data we present could lead to new lines of investigation for both plant and animal PIEZO homologs."

As the name suggests, mechanosensitive ion channels are paths, or tunnels, through cell membranes that respond to mechanical forces. Under certain forces a channel opens, allowing the flow of ions across the membrane.

In humans, PIEZO channels are essential for life; without them, cell development halts. They are recognized for their role in perceiving light touch, shear force and compressive force. Dysfunction in PIEZO channels has been linked to multiple human diseases.

PIEZO channels were first identified in plant genomes in 2010. After a decade of research on animal homologs, this new research shines a spotlight on plant cells and explores how they differ from animal cells. Other research teams have recently shown that PIEZO channels are involved in mechanical sensing in plant roots.

The researchers made their initial discoveries using the tip-growing cells of a somewhat atypical model plant, spreading earthmoss (Physcomitrium patens).

But the scientists were able to extend their findings beyond moss to cells from other distantly related plants, including in pollen tubes in a classic model, the flowering plant Arabidopsis thaliana.

"Mosses are one of the groups that comprise the bryophytes, which are the second largest land plant lineage," said Ivan Radin, a research scientist in the Haswell laboratory and first author of the new paper.

"When we can show that the same thing happens both in moss and a flowering plant, as we did here, the most likely conclusion is that the process is ancestral -- it's at least as old as the land plants are," Radin said, noting that land plants colonized Earth about a half a billion years ago.

Radin became the Haswell laboratory's de facto moss specialist with coaching from co-author Magdalena Bezanilla, a professor of biological sciences at Dartmouth University. Bezanilla previously worked with Washington University's Ralph Quatrano, emeritus dean and the Spencer T. Olin Professor Emeritus of Biology, who was an early adopter of moss.

"The more time passes, the more we love it," Radin said. "Moss proved to be an exceptionally good model."

As a next step in this research, scientists in the Haswell laboratory are now conducting additional experiments to show how external and internal forces directly affect PIEZO channels in moss cells.

"Plant PIEZO channels are likely to be controlled by membrane tension in plants the same way they are in animals," Haswell said. The scientists are also exploring the evolution of these channels in algae.

Now they know where PIEZO channels are found in the cell, Haswell and her team are poised to find out what these proteins are doing in the vacuoles.

"We are looking at how PIEZO channel activation results in membrane elaboration and how it is regulated," Haswell said. "We want to know how the localization evolved and what it does in other cell types. We plan to compare and contrast the structure and function with the animal channels and in organisms across the green lineage."

Source:

Journal reference:

Radin, B., et al. (2021) Plant PIEZO homologs modulate vacuole morphology during tip growth. Science. doi.org/10.1126/science.abe6310.

See the original post here:
New study shines light on how plant cells perceive and respond to mechanical forces - News-Medical.net

Architecture of cellcell junctions in situ reveals a mechanism for bacterial biofilm inhibition – pnas.org

Significance

Pseudomonas aeruginosa bacteria form antibiotic-tolerant biofilms that pose significant challenges in clinical settings. Overcoming these challenges requires fundamental insights into how biofilms are formed, combined with innovative strategies to disrupt biofilms. Electron cryotomography in situ data presented here reveal the arrangement of the key P. aeruginosa adhesin CdrA at biofilm cellcell junctions. Guided by our imaging data, we raised and characterized a CdrA-specific nanobody binder capable of disrupting these cellcell junctions, thereby increasing the efficacy of antibiotic-mediated bacterial killing in biofilms. Together these data provide a pathway for developing effective alternative bacterial infection treatment strategies.

Many bacteria, including the major human pathogen Pseudomonas aeruginosa, are naturally found in multicellular, antibiotic-tolerant biofilm communities, in which cells are embedded in an extracellular matrix of polymeric molecules. Cellcell interactions within P. aeruginosa biofilms are mediated by CdrA, a large, membrane-associated adhesin present in the extracellular matrix of biofilms, regulated by the cytoplasmic concentration of cyclic diguanylate. Here, using electron cryotomography of focused ion beammilled specimens, we report the architecture of CdrA molecules in the extracellular matrix of P. aeruginosa biofilms at intact cellcell junctions. Combining our in situ observations at cellcell junctions with biochemistry, native mass spectrometry, and cellular imaging, we demonstrate that CdrA forms an extended structure that projects from the outer membrane to tether cells together via polysaccharide binding partners. We go on to show the functional importance of CdrA using custom single-domain antibody (nanobody) binders. Nanobodies targeting the tip of functional cell-surface CdrA molecules could be used to inhibit bacterial biofilm formation or disrupt preexisting biofilms in conjunction with bactericidal antibiotics. These results reveal a functional mechanism for cellcell interactions within bacterial biofilms and highlight the promise of using inhibitors targeting biofilm cellcell junctions to prevent or treat problematic, chronic bacterial infections.

Prokaryotic cells including bacteria and archaea are frequently found in nature as part of surface-attached, multicellular communities called biofilms (13). Biofilms constitute the majority of bacterial biomass on Earth (1), representing a fundamental mode of bacterial existence. While bacterial biofilms may prove beneficial to eukaryotes as host-associated microbiomes (4, 5), the formation of pathogenic bacterial biofilms is associated with the establishment of serious chronic antibiotic-tolerant infections (6).

Recently, important advances have been made in understanding early events in biofilm formation (7); however, the molecular mechanisms underlying how mature biofilms are formed and stabilized are still poorly understood. One of the hallmarks of mature biofilms is the presence of an extracellular polymeric substance (EPS) matrix that binds bacterial cells together into a sessile community, promoting antibiotic tolerance and providing protection from other predatory organisms (811). The EPS matrix of biofilms is a complex mixture of molecules, consisting of proteins, polysaccharides, and extracellular DNA (12). Comprehending the spatial arrangement of molecules in the EPS matrix of biofilms has been problematic (13) due to the inherent difficulty associated with high-resolution microscopic imaging inside the tissue-like environment of a biofilm. As a result, mechanisms of cellular tethering and the architecture of cellcell junctions within biofilms are incompletely understood at the fundamental molecular level.

Nevertheless, elegant optical microscopy studies on Vibrio cholerae biofilms have provided clues to the internal organization of the EPS matrix, revealing that the proteins required for mature biofilm formation (RbmA, Bap1, and RbmC) fail to accumulate at the cell surface in the absence of an exopolysaccharide (called VPS) and that loss of RbmA function dramatically alters biofilm architecture (14, 15). In vitro studies of V. cholerae proteins have revealed an exopolysaccharide-dependent (RbmA) adhesin oligomerization pathway (16), and other studies suggest that direct interactions between the RbmA adhesin and glycans on partner cells lead to cellcell adhesion (17). In Escherichia coli, an auto-aggregating adhesin known as Antigen 43 (Ag43) has been proposed to mediate cellcell interactions in biofilms by a Velcro-like mechanism in which two Ag43 molecules from apposing cells dimerize to bind cells to each other (18). In both of these comparatively well-characterized bacterial species, direct visualization of the EPS matrix has thus far not been performed at the macromolecular scale, and the spatial arrangement of molecules at cellcell junctions remains unclear.

Pseudomonas aeruginosa is a human pathogen of critical concern, posing a significant challenge in hospital settings due to its ability to form antibiotic-tolerant biofilms (1921). Cellcell interactions in the EPS matrix of P. aeruginosa biofilms are facilitated by the expression of a 220 kDa adhesin (Fig. 1A) known as CdrA in a cyclic diguanylate (c-di-GMP)dependent manner (22). Under high cytoplasmic c-di-GMP concentrations, CdrA expression is increased, and the mature CdrA protein is tethered to the outer membrane of P. aeruginosa cells through its membrane protein partner CdrB (22, 23). In these conditions, CdrA promotes cellular aggregation and biofilm formation by directly binding to polysaccharides in the EPS matrix of biofilms, such as the Psl or Pel polysaccharides (22, 24). When cytoplasmic c-di-GMP concentrations are lower, CdrA is cleaved and released into the extracellular milieu by the action of a periplasmic protease, promoting biofilm disaggregation (22).

CdrAB expression results in the appearance of 70-nm-long, matchstick-shaped protrusions on the surface of P. aeruginosa cells. (A) Schematic representation of the CdrA sequence highlighting previously determined and predicted functional regions including a TPS (two-partner secretion) domain, putative N-terminal cleavage site, and the known carboxyl-terminal cleavage site. (B) PAO1 (Left) and PAO1 psl (Right) induced to express CdrAB and stained with fluorescent (Alexa 488) lectin to label mannose-containing polysaccharides. (C) Floccules formed by CdrAB expression before (Left) and after agitation in the presence of mannose (Right). (D) A slice through an electron cryotomogram of a P. aeruginosa PAO1 cell expressing CdrAB. (E) Three-dimensional segmentation of the boxed area in D (red dashed line). The outer membrane of the cell (purple) and matchstick-shaped cell surface molecules (green) are shown. (F) Cropped and magnified view of the boxed region from the tomographic slice shown in D (solid yellow line) with matchstick-shaped protrusions indicated (red arrowheads). (G) Length quantification of cell-surface matchstick-shaped protrusions; lengths are measured in electron cryotomograms (71 2 nm [SD, n = 108] from 12 tomograms). Refer also to SI Appendix, Fig. S1 and Movie S1.

In this study, we have performed electron cryotomography (cryo-ET) of focused ion beam (FIB)milled tissue-like multicellular specimens to image intact cellcell junctions of P. aeruginosa bacteria, revealing the arrangement of CdrA in the extracellular matrix of P. aeruginosa biofilms. We have supplemented our in situ imaging with electron cryomicroscopy (cryo-EM) and native mass spectrometry (MS) experiments on biochemically purified CdrA protein, which together show that CdrA forms an extended structure at the outer membrane, forming cellcell junctions via polysaccharide binding partners. Finally, single-domain antibodies (nanobodies) raised against purified CdrA protein provided a valuable probe to test and verify all our hypotheses experimentally on wild-type bacterial biofilms and were found to limit wild-type biofilm formation and potentiate the activity of antibiotics against mature biofilms. These data identify CdrA as a promising candidate for the development of therapeutics.

To study the molecular mechanism by which CdrA tethers cells to the EPS matrix within biofilms of P. aeruginosa, we employed an inducible expression system in which both CdrA and CdrB are expressed. A mutation in the carboxyl-terminal part of CdrA (in the sequence TAAG, described in ref. 23) prevents the cleavage and release of CdrA into the extracellular environment, locking the protein in a biofilm-promoting state. Upon expression of the biofilm-promoting CdrA adhesin and its membrane anchor CdrB, P. aeruginosa cells formed biofilm-like floccules in solution (SI Appendix, Fig. S1A). Floccules formed by a psl strain that lacked the Psl polysaccharide were smaller than in the corresponding wild-type PAO1 strain and could be disrupted by vigorous agitation alone. Floccules formed by wild-type PAO1 were extensively stained by a mannose-sensitive lectin, unlike the corresponding psl strain (Fig. 1B), and could only be fully disrupted by agitation when the medium was supplemented with an excess of D-mannose sugar (Fig. 1C and SI Appendix, Fig. S1B), demonstrating that cellcell clumping mediated by CdrAB is polysaccharide-dependent in wild-type PAO1 P. aeruginosa, confirming previous reports (22, 24). Cryo-ET of single cells expressing CdrAB, disassembled from cell clumps by vigorous vortexing in the presence of excess D-mannose, showed protrusions emanating from the P. aeruginosa cell surface (Fig. 1 DF and SI Appendix, Fig. S1 CE and Movie S1). These 3-nm-wide protrusions projected roughly orthogonally to the outer membrane of P. aeruginosa and had a broad tip, resulting in a matchstick-like appearance. The length of these matchstick-shaped protrusions was 71 2 nm (SD, n = 108), as measured in three-dimensional cryo-ET data (Fig. 1G).

To establish the identity of the matchstick-shaped protrusions, molecules in the outer membrane of P. aeruginosa cells from the inducible CdrAB expression strain were stripped and biochemically purified (Materials and Methods and SI Appendix, Fig. S2). The major component from the purification revealed a protein running at 150 kDa as a single band on a gel (Fig. 2 A, Inset and SI Appendix, Fig. S2A). Proteomic peptide fingerprinting analysis of the purified sample confirmed its identity as CdrA (SI Appendix, Fig. S2B). Next, cryo-EM of the MS-verified, purified CdrA specimen revealed matchstick-shaped filamentous particles on the grid (Fig. 2A and Movie S2). A visual inspection of these particles suggested that they closely resembled the matchstick-shaped protrusions observed in whole-cell cryo-ET (Fig. 1 DF). To probe this further, cryo-ET data of the purified sample was collected and quantified, showing that the matchstick-shaped protrusions were 71 1-nm (SD, n = 75) long (Fig. 2B), again indicating that they corresponded to the same cryo-EM density observed on the P. aeruginosa cell surface (Fig. 1 DG). To assess whether the carboxyl-terminal TRRG mutation alters the architecture of CdrA, we purified particles secreted by cells expressing wild-type CdrA (Fig. 2C) and found that native CdrA, in the same manner as the mutant, forms elongated matchstick-shaped particles, while no protein was detected in the uninduced control (Fig. 2C).

Cell-surface matchstick-shaped protrusions correspond to a mature form of CdrA. (A) Cryo-EM micrograph of protein purified from the surface of cells expressing CdrAB. Structures resembling matchstick-shaped protrusions are indicated (red arrowheads). (Inset) SDS-PAGE of the purified protein. (B) Length quantification of purified protein resembling matchstick-shaped protrusions, measured in electron cryotomograms (71 1 nm [SD, n = 75] from five tomograms). Refer also to SI Appendix, Fig. S2 and Movie S2. (C) Cryo-EM micrograph of protein purified from the surface of cells expressing wild-type CdrAB. Matchstick-shaped structures are indicated (red arrowheads). (Lower) SDS-PAGE of purified wild-type CdrA protein from an induced cdrA/pCdrAB strain (+) versus an uninduced cdrA/pCdrAB control ().

To determine the oligomerization state of CdrA in solution, the purified CdrA (TRRG mutant) protein was analyzed using native MS (SI Appendix, Fig. S2C), which showed that CdrA exists as monomers in solution. Thus, the matchstick-shaped densities observed in our cryo-ET data (Fig. 1 DF) correspond to a single copy of CdrA protein projecting from the outer membrane into the extracellular environment. The measured masses of CdrA (163,286 3 Da and 164,047 1 Da) determined by native MS agree with previous work showing that the protein undergoes proteolytic processing into a mature functional form (22, 23).

We next wished to understand how CdrA mediates cellcell interactions within the extracellular matrix of P. aeruginosa biofilms by direct visualization of cellcell junctions using high-resolution cryo-ET. We used the inducible CdrAB expression strain to promote flocculation of P. aeruginosa cells (SI Appendix, Fig. S1) and deposited these floccules onto cryo-EM grids. While P. aeruginosa cells at the edges of the cell clump could be observed (Fig. 3A), the multicellular, tissue-like specimen was too thick for direct cryo-EM imaging. To visualize the internal arrangement of the cellcell junctions, thin lamellae of these specimens were produced by cryo-FIB milling (SI Appendix, Fig. S3 A and B), which supported high-resolution imaging using cryo-ET.

FIB milling and cryo-ET of P. aeruginosa cellcell junctions. (A) Cryo-EM of biofilm-like floccules produced by P. aeruginosa cells expressing CdrAB. Cryo-EM views of the multicellular aggregate. Red arrowheads indicate cells observed at the edge of the cellular aggregate (yellow box). (B and C) Cryo-ET slices through FIB-milled specimens revealing cells (red asterisks) and cellcell junctions (white asterisks). For clarity, white arrowheads indicate ice contamination. Refer also to SI Appendix, Fig. S3.

In electron cryotomograms of cryo-FIBmilled lamellae, we observed P. aeruginosa bacterial cells in close proximity with each other (Fig. 3 B and C). A visual inspection of the cellcell junctions between the P. aeruginosa cells revealed matchstick-shaped protrusions corresponding to CdrA molecules (Fig. 4 A and B and SI Appendix, Fig. S3C). These CdrA molecules were projecting outward, away from the bacterial outer membrane and extending between P. aeruginosa cells into the intercellular space (Fig. 4C and Movie S3). The length of these CdrA molecules was 71 1 nm (SD, n = 20), in line with the observations of CdrA on single P. aeruginosa cells and also of CdrA molecules after purification (Figs. 1, 2, and 4 D and E and Movie S4). While CdrA has been shown to self-interact in the absence of polysaccharides (25), in our data, direct CdrA:CdrA linkages were never observed, nor was a Velcro-like side-by-side configuration of CdrA molecules from apposing cells seen. The lack of direct CdrA stacking, in conjunction with the known interactions between CdrA and polysaccharides (22, 24), and the ability of the monosaccharide mannose to disrupt CdrA-mediated cell clumps (Fig. 1C and SI Appendix, Fig. S1B) are all consistent with a scenario in which CdrA molecules extend out of the bacterial cell surface to tether cells through interactions with polysaccharide binding partners known to be abundant in the EPS matrix.

CdrA extends into the EPS matrix to mediate cellcell interactions. (A and B) Cryo-ET slice (A) and corresponding three-dimensional segmentation (B) of a cellcell junction within a P. aeruginosa PAO1 biofilm-like floccule. The multicellular specimen was processed by cryo-FIBmilling into thin lamellae suitable for high-resolution cryo-ET. Cell outer membranes (blue and purple) and CdrA (green) are highlighted in B. (C) Enlarged view of the boxed region in A. Comparison of CdrA at cellcell junctions within biofilm-like floccules to (D) CdrA on the surface of single cells and (E) biochemically purified CdrA in solution. CdrA is highlighted with red arrowheads. Refer also to SI Appendix, Fig. S3 and Movies S3 and S4.

To assess whether these CdrA-mediated cellcell junctions might be specifically targeted, we made use of single-domain llama antibodies (or nanobodies), which have recently been shown to be a powerful tool for structural and cell biology (26). A panel of nanobodies was raised against purified CdrA protein, and based on the ability of the nanobodies to interfere with flocculation of P. aeruginosa bacteria in our inducible expression system (SI Appendix, Fig. S1A, further discussed in SI Appendix, Fig. S5), one positive nanobody binder was selected for further experiments. This nanobody (termed NBCdrA) was coupled with 5-nm gold nanoparticles (NBCdrA-gold) and incubated with P. aeruginosa cells expressing CdrA localized to the cell surface. NBCdrA-gold labeling of these cells revealed a shell of gold particles specifically around cells expressing CdrAB, 70 nm from the cell surface (Fig. 5A). No specific NBCdrA-gold labeling was observed in control cells in which CdrA was absent from the outer membrane (Fig. 5B), confirming that the cell-surface 71-nm matchstick-shaped protrusions correspond to CdrA molecules.

Nanobody binding to CdrA on cells and in vitro. (A and B) Cryo-EM micrographs of (A) cells with induced CdrAB expression or (B) control cells, labeled with a NBCdrA-gold conjugate (red arrowheads). Cropped and magnified views of the boxed areas in each panel are shown. (C) Native mass spectra of CdrA alone (lower spectrum) or CdrA bound to the CdrA-specific nanobody NBCdrA (upper spectrum) shows binding in a 1:1 ratio. (D) A series of native MS experiments with increasing NBCdrA concentrations were performed, yielding a binding curve. Each data point shows the relative fractional intensity of NBCdrA binding peaks over total peak intensity (labeled as fractional abundance) versus NBCdrA concentration. SD (error bars) was calculated from the average of five observed charge states in three independent experiments. Refer also to SI Appendix, Fig. S4.

As a further verification, we performed native MS of a sample containing both purified CdrA and NBCdrA and observed the formation of a 1:1 complex (Fig. 5C). To probe nanobody-binding, we performed a series of native MS experiments with a constant CdrA concentration titrated against increasing NBCdrA concentrations. These native experiments were used to estimate a binding affinity of NBCdrA with CdrA of 0.75 0.26 M (SD) (Fig. 5D and SI Appendix, Fig. S4 A and B). These binding experiments in native MS validated our microscopic observation of NBCdrA-gold binding to CdrA on cells (Fig. 5 A and B). Furthermore, the 1:1 binding observed in MS, together with the localization of NBCdrA-gold 70 nm from the outer membrane, strongly suggest that a region at the broad N-terminal tip of cell-surface CdrA is specifically targeted by NBCdrA.

Given the mechanistic scenario suggested by our cellular (Fig. 1), in vitro (Fig. 2), and in situ imaging (Figs. 3 and 4) in which CdrA extends into and likely tethers cells to the EPS matrix, we next set out to determine whether this function of CdrA could be blocked by the targeted use of nanobodies to inhibit P. aeruginosa biofilm formation. First, we assessed whether the anti-CdrA nanobody (NBCdrA), the nanobody shown to bind to the broad tip of CdrA molecules (Fig. 5 A and B), could disrupt CdrA-mediated cellcell adhesion and flocculation of P. aeruginosa in our inducible CdrAB expression system. In line with our expectation, the addition of NBCdrA disrupted preformed cell clumps, causing the cells to return to a planktonic state, forming a cloudy culture (SI Appendix, Fig. S5A). This effect could be quantified by measuring the mass of cell clumps, which confirmed that cellular aggregation decreased with increasing NBCdrA concentration. In contrast, another CdrA nanobody found in our screen (NBCdrA-2) had no effect on CdrAB-mediated flocculation (SI Appendix, Fig. S5A), potentially due to its lower binding affinity for CdrA (SI Appendix, Fig. S5 BD). These observations suggest that a specific region at the tip of CdrA molecules 70 nm away from the cell (as indicated by NBCdrA-gold labeling) is critical for CdrA-mediated cellcell tethering, and blocking this region with NBCdrA leads to disruption of cellcell junctions.

To explore the effect of the nanobodies on wild-type P. aeruginosa bacteria, a custom microfluidics flow system was used to cultivate P. aeruginosa biofilms. Combining the flow system with continuous fluorescence microscopy imaging, we found that the formation of P. aeruginosa PA14 biofilms was significantly delayed upon the addition of NBCdrA (Fig. 6), showing that NBCdrA can also interfere with the function of native CdrA molecules on wild-type cells. These biofilm inhibitory effects were not observed with NBCdrA-2 (SI Appendix, Fig. S5E), confirming that NBCdrA-2 does not interfere with the adhesive function of CdrA in the EPS matrix and demonstrating again that targeted binding of NBCdrA to the tip of functional CdrA is sufficient to inhibit biofilm formation in wild-type P. aeruginosa bacteria.

Nanobody-mediated disruption of CdrA inhibits biofilm formation. (A) Biofilms of P. aeruginosa PA14 expressing TFP (teal fluorescent protein) were cultivated in the presence or absence of NBCdrA and monitored over 10 h under microfluidics flow cell conditions. Representative images are shown. (B) Quantification of fluorescence in the experiments described in A. Each time point represents three images per time point from at least three independent experiments (error bars show SD). Statistical significance was assessed using the Students t test at all times points (****P < 0.0001 at all times at or after 2.5 h post treatment). Refer also to SI Appendix, Fig. S5.

NBCdrA access to cell-surface CdrA is likely to be greater in immature, developing biofilms in which cellcell junctions have not yet formed and in which diffusion deep into a multicellular specimen is not needed. We next investigated whether preformed, mature P. aeruginosa biofilms could also be disrupted by NBCdrA. Using the same flow setup, we found that bacterial cells in preformed PA14 biofilms were efficiently killed when an otherwise sublethal colistin antibiotic treatment was supplemented with NBCdrA (Fig. 7 A and B and Movie S5). The efficacy of bacterial killing was substantially increased when NBCdrA was added earlier, during biofilm development, before the addition of colistin (Fig. 7 A and B), in line with our data demonstrating the inhibitory effect of NBCdrA on developing biofilms (Fig. 6). PAO1 biofilms were similarly susceptible to colistin-mediated bacterial killing when pretreated with NBCdrA, although a higher concentration of nanobody was required to achieve a similar effect (Fig. 7C and SI Appendix, Fig. S6). These results using wild-type P. aeruginosa biofilms demonstrate the key role of the CdrA protein in mediating cellcell interactions and highlight the importance of these interactions for effective biofilm formation, which directly promote tolerance of those biofilms to antibiotic treatment.

Nanobody-assisted bacterial killing in preexisting, mature biofilms. (A) Biofilms were grown in a microfluidics flow setup either in the presence (NBCdrA pretreatment) or absence (No-nanobody or NBCdrA treatment) of NBCdrA. After 10 h, NBCdrA was added to the NBCdrA treatment condition, and all samples were treated with PI (propidium iodide) and a sublethal dose of colistin (1 g/mL) and imaged for a further 10 h. Fluorescence microscopy images with TFP (blue) for live cells or PI staining (red) for dead cells are shown. (B) Quantification of fluorescence in the experiments described in A, following the addition of colistin. The ratio of PI to TFP fluorescence was calculated over three replicate experiments (error bars denote SD). Statistical significance was assessed using the Students t test for all time points (****P < 0.0001, for times after 6 to 7 h versus the no-nanobody control). (C) Quantification of fluorescence in the experiments performed on PAO1 strain of P. aeruginosa, following the addition of colistin. The ratio of PI to green fluorescent (GFP) fluorescence was calculated over three replicate experiments (error bars denote SD). Statistical significance was assessed using the Students t test for all time points (****P < 0.0001) for times after 10 h versus the no-nanobody control). Refer to SI Appendix, Fig. S6 for images.

Our results establish that functional cell-surface CdrA forms an extended structure, projecting into the EPS matrix, to mediate cellcell interactions. This arrangement is consistent with CdrA acting as a molecular tether, in which copies of CdrA anchored on the cell surface at their C termini may be glued at their N-terminal adhesive regions by secreted polysaccharides, known to be abundant in the EPS matrix of P. aeruginosa and previously shown to interact with CdrA (13, 24). Our results demonstrate that CdrA is a major adhesin mediating biofilm formation in wild-type P. aeruginosa and show that adhesins with similar roles, such as LecB (27), cannot effectively compensate for its disruption. The difference in the sensitivity of different P. aeruginosa strains (PA14 and PAO1) to NBCdrA treatment (Fig. 7) suggests variability in the contribution of CdrA or its accessibility during biofilm formation.

Nevertheless, while the exact contribution of different adhesins in P. aeruginosa biofilm development will require detailed experiments to delineate, the prominent role of CdrA-mediated cellcell junctions is highlighted by our experiments. Our reconstituted cell flocculation system used a nonLapG-cleavable mutant of CdrA, which was important for in situ imaging using cryo-ET. Future innovative assays will be needed to replicate structural studies with wild-type CdrA protein at cellcell junctions. The wild-type protein, natively secreted from cells, had the same overall architecture as the carboxyl-terminal nonLapG-cleavable mutant of CdrA (Fig. 2C). We predict that this mutation at the carboxyl-terminal membrane anchor 70 nm away from the adhesive tip of CdrA will have little or no effect on its adhesive function, as indicated by the ability of CdrA(TRRG) to form cellcell junctions in our assays.

This paradigm of a bacterial adhesin interacting with the EPS matrix is functionally distinct from either the alternative Velcro-like mechanism of cellcell adhesion proposed for E. coli Ag43 (18) or from the more nuanced case of V. cholerae in which multiple adhesins and exopolysaccharides appear to regulate cellcell interactions (14, 16). In both E. coli and V. cholerae, the spatial organization of cellcell junctions in biofilms has not yet been observed experimentally at high resolution, and further research will be needed to understand the arrangement of these junctions at the molecular level. In our in situ cryo-ET data, while CdrA proteins were observed, extracellular polysaccharides could not be resolved. It therefore remains to be described how polysaccharides are arranged in the EPS matrix in relation to bacterial cells and other matrix molecules. While it is possible that direct CdrA:CdrA interactions may additionally stabilize P. aeruginosa cell junctions in some strains (25), these were not observed in our data.

Recently, latest generation optical microscopy techniques have proven invaluable in providing novel insights into bacterial biofilm formation (14, 15, 28). In this study, we have leveraged the latest cryo-ET techniques to reveal the arrangement of cellcell junctions that are of key importance in biofilm formation. This study highlights the utility of FIB milling and cryo-ET to deliver high-resolution insights into tissue-like multicellular specimens, which have traditionally been considered beyond the reach of structural cell biology. With increasing throughput and automation of these methods (29, 30), structure determination of molecules in cellcell junctions may become practical and a possible extension of this work in the future.

Moreover, we also show that biofilm formation in wild-type P. aeruginosa bacteria can be inhibited by targeting the filamentous CdrA adhesin with a nanobody binder that interacts with the tip of the functional protein in a 1:1 ratio. Although gold-labeled nanobody was concentrated 70 nm away from the outer membrane of P. aeruginosa bacteria at the tip of CdrA adhesins, identifying the exact epitope of nanobody-binding to CdrA will require higher resolution structural data. Our results are consistent with the scenario in which nanobody-binding precludes the interaction between CdrA and polysaccharides in the EPS matrix (Fig. 8). This nanobody was further shown to promote the activity of an antibiotic in killing bacterial cells in a preformed wild-type P. aeruginosa biofilm. The strategy of neutralizing biofilm adhesion mechanisms may also be applicable to similar large cell-surface adhesins such as the filamentous hemagglutinin of Bordetella pertussis (31) and the repeat-in-toxin domain-containing adhesins such as SiiE of Salmonella enterica and LapA from Pseudomonas fluorescens (3234). Such an approach may also be useful in treating bacteria whose adhesins utilize alternative proposed cellcell adhesion mechanisms, such as Ag43 of E. coli (18) or RbmA of V. cholerae (16).

CdrA-mediated cellcell junctions. Schematic depiction of the mechanism of biofilm inhibition by targeting CdrA revealed in this study. Refer also to Movie S5.

While neutralizing therapies now represent an important treatment route for many diseases, they remain relatively underexploited in the field of antimicrobials (35, 36). In the context of the increasing challenges posed by the emergence of antimicrobial resistance (37, 38), our results highlight how specific adhesins on the surface of bacterial cells may serve as promising targets for biofilm inhibition or for the prevention of chronic infections in a medical setting.

Refer also to SI Appendix, Supplementary Text for further details.

For CdrAB(TRRG) expression, an arabinose-inducible pMQ72 plasmid system was used, as described in ref. 23, and transformed into the PAO1 pslBCD or PAO1 cdrA strains, described in ref. 22. The strains PAO1 psl pMQ72-cdrAB(TRRG) and PAO1 cdrA pMQ72-cdrAB(TRRG) were tested alongside a PAO1 cdra pMQ72 empty vector control. Cultures were induced with arabinose to induce flocculation. To disaggregate cells, floccules were briefly vortexed with (for strains with wild-type Psl) or without (for the Psl deletion strain) the addition of 0.5% mannose (weight/volume). To stain for Psl-containing sugars, floccules were pelleted and resuspended in 80 g/mL Alexa488-conjugated Narcissus pseudonarcissus lectin (Glycomatrix) for 30 min at 4 C, washed three times with phosphate-buffered saline (PBS), and deposited on a glass slide ahead of imaging. Microscopy of floccules was performed using a Zeiss Axio Imager M2 microscope.

To purify CdrA, subcultures of PAO1 pslBCD pMQ72-cdrAB(TRRG) were plated on lysogeny broth agarsupplemented with gentamicin and arabinose to induce CdrAB expression and incubated overnight at 37 C. The following day, cells were scraped from the plates into PBS, vortexed to shear CdrA from the outer membrane, and centrifuged to remove cells. The supernatant was precipitated with polyethyleneglycol 6,000 and centrifuged to remove contaminating proteins. The resulting soluble CdrA sample was dialyzed and purified by anion exchange chromatography. Eluted fractions were analyzed by sodium dodecyl sulfatepolyacrylamide gel eletrophoresis (SDS-PAGE), and fractions containing CdrA were further purified by size exclusion chromatography.

Antibodies to CdrA were raised in a llama by intramuscular immunization with purified protein using Gerbu LQ#3000 as the adjuvant. Total RNA was extracted from blood, and VHH complementary DNAs were generated by RT-PCR. The pool of VHH-encoding sequences was amplified by two rounds of nested PCR and cloned into the SfiI sites of the phagemid vector pADL-23c. Electro-competent E. coli TG1 cells were transformed with the recombinant pAD-23c vector, and the resulting TG1 library stock was infected with M13K07 helper phage to obtain a library of VHH-presenting phages. Phages displaying VHHs specific for CdrA were enriched via two rounds of bio-panning on biotinylated CdrA, and individual phagemid clones were picked. VHH-displaying phages were recovered by infection with M13K07 helper phage and tested for binding to CdrA by enzyme-linked immunosorbent assay (ELISA). Phage binders were ranked according to the ELISA signal and grouped according to CDR3 sequence identity.

To express nanobodies, nanobody phagemids were first transformed by heat shock into WK6 E. coli cells. Subcultures of transformants were prepared and induced with isopropyl -D-thiogalactoside and incubated overnight. The following day, cells were pelleted, resuspended in a lysing buffer, and homogenized. His-tagged nanobodies were purified by Ni-NTA affinity chromatography and size exclusion chromatography.

Data were collected on a Q-Exactive ultrahigh mass range mass spectrometer (Thermo Fisher Scientific) and analyzed using Xcalibur 4.2 (Thermo Fisher Scientific) and UniDec (39) software packages. Nanobody-binding experiments were performed by adding increasing amounts of NBCdrA to a constant 2 M CdrA. The resulting calculated binding affinity (KD) from native MS was determined by employing a nonlinear regression curve fitting for one-site specific binding in GraphPad Prism 8.0.

For protein identification, tryptic peptides were obtained by digesting the protein sample with trypsin as described in ref. 40. Peptides separated using a chromatographic system connected to an Eclipse Tribrid Orbitrap mass spectrometer (Thermo Fisher Scientific). Raw data files were processed for protein identification using MaxQuant (version 1.6.3.4) and searched against the UniProt database.

NBCdrA was dialyzed into 20 mM Tris pH 7.4, 150 mM NaCl, and incubated with a 2.5 molar excess of Ni-NTA 5-nm gold (Nanoprobes) for 30 min at room temperature. Unbound gold was separated from NBCdrA-gold using a PD-10 desalting column. To label cells, NBCdrA-gold was incubated with PAO1 cdrA pMQ72-cdrAB(TRRG) floccules followed by the addition of mannose ahead of cryo-EM sample preparation.

To assess the efficacy of nanobody-mediated floccule disaggregation, PAO1 cdrA pMQ72-cdrAB(TRRG) bacterial floccules were incubated with NBCdrA or NBCdrA-2. Floccules were pelleted at 300 g for 5 min. The supernatant was removed, and the remaining cells were pelleted at 15,000 g for 15 min. The two pelleted fractions were weighed, and floccule mass was calculated as a percentage of total cell mass.

To determine the effect of nanobodies on the formation of wild-type biofilms, a previously described flow system was used (41). P. aeruginosa PA14 or PAO1 was visualized via fluorescence of the teal fluorescent protein (TFP) or green fluorescent protein (GFP), respectively. Biofilms were grown and visualized in microfluidics chambers generated through soft lithography techniques (42, 43). Bacteria were prepared as described previously (44). Bacterial cultures were allowed to attach for 1 h without flow, after which the flow rate was set to 1.0 L/min and imaged for 10 to 12 h, as indicated.

For the biofilm inhibition assays, the biofilm medium was supplemented either with no-nanobody, 0.3 mg/mL NBCdrA (PA14 cells), or 0.8 mg/mL NBCdrA (PAO1 cells) as indicated and imaged for 10 h postinoculation. To test the impact of colistin on a preformed biofilm, bacteria were prepared, inoculated, and grown in microfluidics chambers as described above. After 10 h of allowing the biofilm to establish in the absence of any treatment, KA medium was switched with KA medium supplemented with propidium iodide (PI) and 1.0 g/mL colistin +/ NBCdrA, as indicated and imaged for an additional 10 h of incubation.

Cryo-EM samples were prepared by depositing 2.5 L of bacterial floccules, disaggregated bacterial floccules (single cells), or NBCdrA-goldlabeled cells onto freshly glow discharged Quantifoil grids. Samples were fixed with 1% paraformaldehyde applied directly on the grid and manually blotted. Next, buffered fiducial gold was added, and the grid was blotted and plunge-frozen into liquid ethane using a Vitrobot Mark IV (Thermo Fisher Scientific).

Cryo-FIB milling of plunge-frozen biofilm-like floccules was performed as described previously (45) on a Scios DualBeam FIB/scanning electron microscope (SEM) microscope (FEI/Thermo Fisher Scientific) equipped with a Quorum PP3010T cryo-FIB/SEM preparation system. The loading stage and milling procedure were adapted, with minor alterations, from ref. 46. Grids were sputter-coated with platinum before milling and coated with a layer of organometallic platinum. Ion beam current for milling was reduced stepwise while adjusting the stage tilt as described in ref. 45. Final polishing of the lamellae resulted in 150- to 300-nm-thick lamellae.

Tilt series data were collected on a Titan Krios microscope (Thermo Fisher Scientific) operating at 300 kV fitted with a Quantum energy filter (Gatan) and direct electron detector (Gatan) using SerialEM software (47). A dose-symmetric tilt scheme was employed for cryo-ET data (48). Untilted video frame stacks of purified protein were collected on the same microscope using EPU software (Themo Fisher Scientific). Cryo-EM images of NBCdrA-goldlabeled cells were collected on a Talos Arctica 200 kV cryo-ET (Thermo Fisher Scientific).

Tilt series alignment was carried out using the eTOMO graphical user interface in the IMOD software (47). CTF (contrast transfer function) parameters for the aligned stacks were estimated using CTFFIND (49), and data were reconstructed using Tomo3D (50). Contrast in cryo-ET data were enhanced via the tom_deconv deconvolution as described in ref. 51 or via bandpass and Laplacian filtering as implemented in Fiji (52). Segmentation of image data were performed manually in IMOD (47).

All study data are included in the article and/or supporting information.

T.A.M.B. is a recipient of a Sir Henry Dale Fellowship, jointly funded by the Wellcome Trust and the Royal Society (202231/Z/16/Z). T.A.M.B. thanks the Vallee Research Foundation and the John Fell Fund for support. G.A.O. acknowledges support from the NIH (R37-AI83256), and C.V.R. acknowledges funding from the Medical Research Council (MR/N020413/1). J.H. is funded by the Edward Penley Abraham Cephalosporin Fund, and Protein Production United Kingdom is supported by the Rosalind Franklin Institute Engineering and Physical Sciences Research Council Grant EP/S025243/1. We thank Adam Costin for help with cryo-EM imaging, Carey Nadell for providing the microfluidics chambers, and Wanda Kukulski for facilitating FIB milling experiments. We would like to acknowledge the MRC Laboratory of Molecular Biology Electron Microscopy Facility for access to cryo-FIB sample preparation.

Author contributions: C.E.M. and T.A.M.B. designed research; C.E.M., J.R.B., S.K.-L., D.B.M., P.C.H., J.H., M.R.W., L.M.E., J.B., A.N.M., C.J.H., and T.A.M.B. performed research; J.R.B., S.K.-L., P.C.H., J.H., M.R.W., L.M.E., J.B., R.J.O., C.V.R., and G.O. contributed new reagents/analytic tools; C.E.M., J.R.B., S.K.-L., D.B.M., L.M.E., J.B., R.J.O., C.V.R., G.A.O., and T.A.M.B. analyzed data; and C.E.M. and T.A.M.B. wrote the paper.

The authors declare no competing interest.

This article is a PNAS Direct Submission.

This article contains supporting information online at https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.2109940118/-/DCSupplemental.

Go here to see the original:
Architecture of cellcell junctions in situ reveals a mechanism for bacterial biofilm inhibition - pnas.org

A single dose of Covishield vaccine induced high neutralizing antibody and T cell responses – Sri Lankan study – nation.lk – The Nation Newspaper

A study in Sri Lanka has found that 93.4% individuals developed antibodies to a single dose of the Covishield vaccine, the made-in-India variant of Oxford-AstraZenecas AZD-1222 formulation, while 97.1% developed neutralizing antibodies.

With the initiation of the COVID-19 vaccination program, Health Care Workers in Sri Lanka were the first to be vaccinated with Covishield vaccine.

Therefore, the Allergy, Immunology and Cell Biology Unit, Department of Immunology Molecular and Molecular Medicine investigated antibody responses to the SARS-CoV-2, neutralizing antibody responses, antibody responses to the variants, along with T cell responses.

The full article is available here at Nature Communications, which has an impact factor of 14.9:Immune responses to a single dose of the AZD1222/Covishield vaccine in health care workers | Nature Communications

93.4% individuals developed antibodies to a single dose of the vaccine and 97.1% developed neutralizing antibodies. There was no difference in immune responses in younger individuals compared to those >60 years of age.

The vaccine induced potent T cell responses with 67.1% developing T cell responses. However, the antibody responses to the beta SARS-CoV-2 virus variant was low. Antibodies to delta was not measured, as delta was not a problem during the time the study was conducted (February and March 2021).

The research team included scientists from the Allergy, Immunology and Cell Biology Unit, Department of Immunology Molecular and Molecular Medicine, Colombo Municipality Council and University of Oxford.

The funding for this study was provided by the World Health Organization, UK Medical Research Council and the Foreign and Commonwealth Office and the Chinese Academy of Medical Sciences (CAMS) Innovation Fund for Medical Science (CIFMS), China.

The group is also studying immune responses to other vaccines such as Sputnik, Moderna and Sinopharm and how the immunity to these different vaccine persist and compare with natural infection.

The most recent preprint on the immune responses to the Covishield vaccine at 16 weeks is available here:Immune responses to a single dose of the AZD1222/Covishield vaccine at 16 weeks in individuals in Sri Lanka | medRxiv

Source: USJ

View post:
A single dose of Covishield vaccine induced high neutralizing antibody and T cell responses - Sri Lankan study - nation.lk - The Nation Newspaper