Category Archives: Stem Cell Medicine


Century Therapeutics Announces Opening of Seattle Innovation Hub – Business Wire

PHILADELPHIA--(BUSINESS WIRE)--Century Therapeutics, developer of induced pluripotent stem cell (iPSC)-derived allogeneic cell therapies for cancer, today announced the opening of its Seattle-based Innovation Hub to develop next-generation product candidates that overcome barriers that have limited the effectiveness of cell therapies in solid tumor cancers.

The site will advance the companys novel iPSC science and allogeneic cell products by establishing expertise in data sciences and machine learning, synthetic biology, cancer biology and immuno-oncology. Centurys President of R&D, Hy Levitsky, M.D., will be based at the Seattle site, and together with Philadelphia-based Chief Scientific Officer Luis Borges, PhD., will oversee site operations and integration with the pipeline programs centered at Centurys Philadelphia headquarters.

The Innovation Hub supports not only Centurys continued pipeline growth and development, but also our expansion into Seattle, a center of excellence in cell therapies, said Lalo Flores, Chief Executive Officer of Century Therapeutics. I look forward to seeing the Century team grow, and am excited to have Luis and Hy leading the charge into this exciting new chapter.

Dr. Levitsky has extensive biotech industry experience, having previously served as Chief Scientific Officer at Juno Therapeutics in Seattle, as well as Head of Cancer Immunotherapy at Roche Pharma Research and Early Development. In addition, Dr. Levitsky earned his M.D. from Johns Hopkins University and has spent over 20 years on their faculty.

Dr. Borges has extensive cancer immunotherapy and cell therapy experience, having worked at Immunex, Amgen, Five Prime Therapeutics and Cell Medica, where as CSO he led the development of off-the-shelf CAR-cell therapies for the treatment of cancer in collaboration with the Baylor College of Medicine.

Centurys new Seattle Innovation Hub will provide the infrastructure needed to conduct in-depth analytics of product candidates in preparation for entry into the pipeline portfolio, said Dr. Levitsky.

The Hub will be key in realizing the potential of Centurys science to overcome limitations of first-generation cell therapies. Dr. Borges added, The Seattle and Philadelphia research laboratories will complement each other. With our current deep expertise in iPSC biology, immunology, cell and protein engineering, the new group in Seattle will help us transition to future generation products designed to have potent anti-tumor efficacy and robust safety windows.

About Century Therapeutics

Century Therapeutics is harnessing the power of stem cells to develop curative cell therapy products for cancer that overcome the limitations of first-generation cell therapies. Our genetically engineered, universal iPSC-derived immune effector cell products (NK, T, DC and macrophage) are designed to specifically target hematologic and solid tumor cancers. Our commitment to developing off-the-shelf cell therapies will expand patient access and provides an unparalleled opportunity to advance the course of cancer care. Century was launched in 2019 by founding investor Versant Ventures in partnership with Fujifilm and Leaps by Bayer. For more information, please visit http://www.centurytx.com.

Continued here:
Century Therapeutics Announces Opening of Seattle Innovation Hub - Business Wire

NanoSurface Bio Executes Exclusive License of Heart-on-Chip Technology Launched Into Space – Yahoo Finance

NanoSurface Biomedical announced today that it has executed an exclusive IP license agreement related to innovative heart-on-chip technology developed by researchers at the University of Washington (UW). An experimental system built from the same heart-on-chip technology was launched into space on Friday, March 6, 2020 at 11:50 PM EST aboard SpaceX's 20th resupply mission to the International Space Station (ISS) as part of the Tissue Chips in Space initiative conducted in partnership between the National Center for Advancing Translational Sciences (NCATS) and the ISS U.S. National Laboratory (ISS National Lab). NanoSurface will commercialize the heart-on-chip platform for use by pharmaceutical companies in preclinical drug development.

The heart-on-chip system will spend 30 days aboard the ISS as part of a series of experiments intended to study the effects of microgravity on human cells and tissues. "In space we are using the heart-on-chip system in microgravity conditions to help improve our understanding of the aging process and cardiac biology, but this heart-on-chip system also has enormous potential for accelerating the discovery of new medicines back here on Earth," said Deok-Ho Kim, an Associate Professor of biomedical engineering and medicine at Johns Hopkins University, the principal investigator for the heart-on-chip experiment aboard the ISS, and the scientific founder of NanoSurface Bio.

The heart-on-chip platform uses three-dimensional engineered cardiac tissues (3D ECTs) grown from human cardiomyocytes, or beating heart cells, derived from induced pluripotent stem cells (iPSCs). As the 3D ECTs beat, researchers can measure the amount of force generated by each contraction, and then evaluate how that force changes after treating the tissues with candidate drugs. 3D ECTs can be made from cells from either healthy individuals or individuals with diseases, offering great promise in predictive preclinical testing of candidate drugs for safety and efficacy.

"I am incredibly excited that the talented team at NanoSurface will be carrying this technology forward for use in the drug development industry," said Nathan Sniadecki, one of the inventors of the heart-on-chip technology and a professor of mechanical engineering at UW. Last year, Professor Sniadecki joined NanoSurfaces board of scientific advisors to guide the commercial development of the technology.

NanoSurface Bios execution of this exclusive license adds significant value to the portfolio of IP it has already licensed from researchers at UW. "It is well recognized that the drug development process is extremely slow and expensive. At NanoSurface we are eager to develop technologies that enable the use of human iPSC-derived cells and tissues in preclinical drug development, ultimately leading to better prediction of how drugs will affect patients in the clinic, lowering costs, and speeding life-saving medicines to market," said NanoSurface CEO Michael Cho.

About NanoSurface Biomedical

NanoSurface Biomedical is a biotechnology company based in Seattle, WA that develops stem cell-based assay technologies to accelerate drug development. NanoSurfaces structurally matured cardiac tissue models, assay instruments, and discovery services leverage human stem cell technology to help pharmaceutical companies predictively assess the safety and efficacy of candidate drugs early during preclinical development. NanoSurfaces mission is to help bring life-saving medicines to market in less time and at lower cost. To learn more, visit http://www.nanosurfacebio.com.

View source version on businesswire.com: https://www.businesswire.com/news/home/20200309005703/en/

Contacts

NanoSurface BiomedicalDirector of Sales & Marketing: Heejoon Choi, 800-913-4403 x702heejoon@nanosurfacebio.com

Continue reading here:
NanoSurface Bio Executes Exclusive License of Heart-on-Chip Technology Launched Into Space - Yahoo Finance

Beating Heart Cells Experiment Was Sent to the International Space Station for Investigations – Dual Dove

SpaceXs 20th and last resupply expedition to the International Space Station (ISS) under the current contract with NASA, took place on Friday. One of the new series of science experiments carried to the orbiting lab was small heart cells, as their potential to help people suffering from heart diseases is growing.

The mission launched on Friday night at 11:50 p.m. ET from Cape Canaveral Air Force Station in Florida, and it comprised supplies, various experiments, and materials for ongoing research. The Dragon spacecraft also transported the European Space Agencys (ESA) Bartolomeo, an exteriorcommercial research platform.

Among the payloads were two different experiments on cardiomyocytes, or beating heart cells, developed from pluripotent stem cells. Pluripotent stem cells are excellent because they can be transformed into multiple cell types in this case, they could end up being healthy heart cells for a patient suffering from heart disease.

The results of the investigations could be used to develop numerous healthy heart cells for children and adults with various heart diseases. They could also be utilized to understand heart health and the aging pattern in a much larger framework. The scientists for one of the experiments believe their analysis could even help astronomers lower the risks they experience throughout a long-term spaceflight, such as reduced heart function and irregular heartbeat.

Scientists already know that humans exposed to space experience changes similar to accelerated aging, so we hope the results can help us better understand and someday counteract the aging process, said Deok-Ho Kim, a primary scientist for one of the experiments, and associate professor of biomedical engineering and medicine at Johns Hopkins University.

For the following months, the heart cells will be studied at the space lab before returning to Earth.

Chunhui Xu, an associate professor of pediatrics at the Emory University School of Medicine, first found that stem cells grow at an accelerated pace in space by simulating the lack of gravity on Earth.

According to Xu, stem cell therapies to mend harmed heart cells need at least one billion cells for each patient, but they take a lot of time to develop on Earth. When her experiment utilizing simulated microgravity on Earth suggested promising moves toward a quick and safe process of generating cardiomyocytes, Xu realized that space was the ideal proving ground.

Herexperimentwas selected to be transported to the space station, and she and her colleagues hope they can confirm their observation performed on Earth and also find new insights.

After the cells get thawed, cell cultures will develop for 21 days in the Multi-Use Variable-Gravity Platform experiment module on the orbiting lab. After they return to Earth, researchers in California will prepare them and have them transported back to Emory in Atlanta so experts can analyze them.

In the meantime, during the experiment taking place in space, a control group of cells will be laid in a centrifuge in an altered gravity environment on Earth.

Weve worked together for years to bring basic and clinical science together, said Dr. Kevin Maher, director of the cardiac intensive care unit at Childrens Healthcare of Atlanta Heart Center and professor of pediatrics at Emory University, who is also one of the researchers working with Xu on the experiment.

There is a high request on the cells themselves, as they have to be pure and high quality. Residual stem cells that cannot transform into heart cells can trigger tumors, as per earlier research led by Xu. More mature cells have a more promising chance of becoming pure heart cells.

While conducting her ground-based experiment in which she simulated a lack of gravity, Xu discovered that the cells were more mature and even showed survival genes that could secure cell survival. Overall, these causes would enable cells to link with the heart tissue better and produce fewer issues.

The effects of their research could help create a more effective and cost-efficient method of developing the heart cells on Earth for those in need. Considering that heart disease is the leading cause of death in the United States and not only the cells have a huge potential to treat children and adults.

Known for her passion for writing, Paula contributes to both Science and Health niches here at Dual Dove.

View original post here:
Beating Heart Cells Experiment Was Sent to the International Space Station for Investigations - Dual Dove

Bias remains the biggest issue for women in medical research – Women’s Agenda

Before her presentation at the Kirby Institute to a crowd of 100 guests, I pulled Dr Louise Purton aside to ask her a few questions about her career to date as a medical researcher in Australia.

*************

Womens Agenda will soon be launching The Health Wrap, our weekly newsletter for women working in Health. Sign up here.

*************

Inevitably, we touched on the subject of gender discrimination. When I asked Dr Purton, who leads the Stem Cell Regulation Laboratory at the University of Melbourne, what has been the most frustrating challenge in her line of work she answered quickly. Bias, she said.

There is still a lot of bias against women researchers, and the only way to improve things is to implement 50/50 funding for men and women, as well as set measures specifically for people of colour.

Dr Purton was one of four women in research and medicine invited by UNSW Medicine to share their career trajectories and discuss the ways they have balanced work as a professional and as family members with domestic responsibilities.

PhD candidate Kerith-Rae Dias kicked off proceedings by describing her unique position as a clinical genomics and neuroscience researcher and first generation woman of colour. She spoke of the indelible mark her parents made on her as a young person growing up in Bombay.

My father was a marine engineer and over the dinner table, hed quiz me and my brother on things like what is gravity, or, what is the speed of light? Dias remarked.

The family emigrated to New Zealand, where Dias explained she learned to change her accent according to the groups she was moving within, another example of a successful professional woman code-switching in order to fit in.

Dias obtained her undergraduate and postgraduate degrees in Auckland, before moving to Western Australia to pursue her Masters in Forensic Science.

Since then, she has travelled to London and Montreal on research projects. When she returned from having her first child, her employer advised her that her role no longer existed. She later discovered that the role had simply been re-titled. And the new person in the role? A young man who was less qualified than Diaz, who was earning a salary that was 40% more than what shed received before she went on parental leave.

There is a clear lack of inclusive structures for returning mothers, Dias said. And we need that to change. The needle is finally moving, as long as we continue to build on mentoring and sponsoring opportunities for women.

Dr Clare Arnott is a cardiologist at RPA, and shares parenting duties with her husband who is also a cardiologist.She told audiences that recently, her husband was told by a male senior medical professional that he was a good researcher, but youll never be brilliant, because you have a working wife.

There was a sharp inhale from audiences. Dr Arnott is one of a minority of women in cardiology. In fact, in Australia, only 15% of cardiologists are women.

What does equality mean to me? Dr Arnott asked. Id hope my gender is irrelevant to the tables I get invited to. I want to be me based on merit.

My biggest challenges have been having a child during advanced training, learning how to advocate for myself and finally, asking for help. When I got back from having a child, there was no discussion about altered work systems. I was scared to ask for help. Even while in a short amount of time, I went through seven episodes of mastitis. I was embarrassed that I was putting others through stress.

She told audiences not to forget that we all have a right to be supported.She also credited her career to joining the Franklin Women Mentoring Program, where she was mentored by leading female professionals. You must ask yourself constantly; Why was I not invited? How am I making myself seen? Am I doing enough to be seen?

Dr Louise Purton flew up from Melbourne to deliver her presentation on her pathway to career success. At three, she was diagnosed with severe intestinal abscess. The antibiotics that were used to treat her eventually caused severe hearing loss, and it was not until she was 19 when she had her first cochlear implant.

Dr Purton spoke of the importance of fighting despite adversities.

Having hearing difficulties didnt stop me from doing what I wanted to do, she said. It didnt stop me from reaching the top, as hard as its been.

Dr Purton battled through funding loss, professional abuse, betrayals and bias at all levels. Yet she persevered.

Bias is the biggest reason why were not getting supported, she said. Its critical we call out the bias and discrimination when we see it.

Dr Purton also believes that having more than one director in organisations will be better. Id like to see diversity and inclusivity in leadership roles simultaneously. With that, you get more understanding for what people go through, and have people think outside the box.

When audiences asked the panel what they think men can do to contribute to improving the reality for women in medicine, the panel agreed that a range of strategies can be approached.

Challenge yourself by being around people who are different, people who look and think different from you, Dr Arnott said. Be inclusive and challenge each other. We need the support and advocacy from men too.

View original post here:
Bias remains the biggest issue for women in medical research - Women's Agenda

3D beating heart tissue experiment heads to Space Station – UW Medicine Newsroom

Note to editors and reporters: Live coverage on NASA Television of the SpaceX CRS-20 cargo launch carrying this experiment is scheduled at 8:30 p.m. EST, 11:30 p.m. PST March 6 and will be replayed twice on March 7. Coverage of the rendezvous with the International Space Station will be at 5:30 a.m. EST Monday, March 8, with installation at 8:30 a.m. All times are subject to change due if weather or launch conditions are unfavorable

MEDIARESOURCES:

Downloads:

Soundbites

Web-embeddable video

Photos of tissue loading

Soundbite Log

Otherresources:

Youtube link

Space exploration can take a toll on the human heart. Astronauts are at risk for changes in their cardiac function and rhythm. To learn how microgravity and other physical forces in space exact their effects on heart muscle, a Tissue Chips in Space project has now been packed and is awaiting launch to the International Space Station.

The experimental equipment consists of small, compact devices, a little bit larger than cell phone cases. The holders contain a row of tiny, 3-D globs of beating heart tissue grown from pluripotent stem cells, generated from human adult cells. The heart muscle tissue is supported between two flexible pillars that allow it to contract freely, in contrast to the rigid constraints of a Petri dish.

The devices also house a novel invention from the University of Washington. It automatically senses and measures the contractions of the heart tissues, and reduces the amount of time the astronauts will need to spend conducting this study.

The flexible pillars contain tiny magnets, explained UW graduate student Ty Higashi, one of the inventors. When the muscle tissue contracts, the position of the embedded magnets changes, and the motion can be detected by a sensor, he said. That information is then sent down to a laboratory on Earth.

This model will recapitulate, on a miniature scale, what might be happening to the architecture and function of heart muscle cells and tissues in astronauts during a space mission.

The project head is Deok-Ho Kim, a professor in bioengineering, who recently joined the Johns Hopkins University faculty in Baltimore. He and co-investigator, Nathan Sniadecki, a professor in mechanical engineering, began this study two years at the UW Medicine Institute for Stem Cell and Regenerative Medicine (ISCRM). Jonathan Tsui, a postdoc in bioengineering, Ty Higashi, a graduate student in mechanical engineering , and other members of the UW project team, continue the cross-country collaboration in Seattle. The team is working with several NASA and National Institutes of Health groups, and researchers at other universities, on this effort.

Sniadecki said that each of the tissues heading to the International Space Center contain about a half million heart cells.

They act like a full tissue, he explained. They contract, they beat and you can actually see them physically shorten in the dish. Were actually able to see little heart beats from these tissues.

The SpaceX shuttle delivering this scientific payload is expected to leave from Cape Canaveral no earlier than 8:50 p.m. PST (11:50 p.m. EST) Friday, March 6. The exact departure schedule depends on the weather and other factors.

Once on board, the experiment will run for 30 days before being returned to Earth for further analysis. A related space-based experiment will follow skyward later, to see if medications or mechanical interventions can offset what the heart muscle endures during extended space missions.

The space program is looking at ways to travel longer and farther, Sniadecki said. To do so, they need to think about protecting their crews. Having treatments or drugs to protect astronauts during their travel would make long term space travel possible.

Guarding against cardiac problems would be especially critical during space travel at distances never attempted before, such as a mission to Mars, said Sniadecki. This opportunity to really kind of push the frontier for space travel is every engineers dream.

He added, We also hope to gather information that will help in preventing and treating heart muscle damage in people generally, as well as in understanding how aging changes heart muscle.

Microgravity is known to speed up aging, and likely influence other cell or tissue properties. Because aging is accelerated in space, studies on the International Space Station is a way to more quickly assess this process over weeks, instead of years.

I think the medicine side of it is extremely helpful on Earth, too, because what we discover could potentially lead to treatments for counteracting aging, Sniadecki said.

This space medicine research project is funded by the National Center for Advancing Translational Sciences and the National Institute of Biomedical Imaging and Bioengineering. This heart tissue study is part of the national Tissue Chips in Space program.

Originally posted here:
3D beating heart tissue experiment heads to Space Station - UW Medicine Newsroom

Prof Ziad Mallat leads Cambridge effort to win 30m to tackle leading cause of heart attacks and strokes – Cambridge Independent

It is the worlds biggest killer and yet we dont fully understand the leading cause behind it.

Cardiovascular diseases claimed an estimated 17.9 million lives in 2016 31 per cent of all deaths around the globe.

And 85 per cent of these were due to heart attacks and stroke, most commonly caused by a blockage of the arteries known as atherosclerosis.

Now an international team led by a Cambridge professor of cardiovascular medicine is competing for a 30million prize from the British Heart Foundation to unravel its secrets.

If they beat the other three shortlisted teams in the charitys Big Beat Challenge, they will create the worlds first 3D map of atherosclerosis at single cell resolution, giving unparalleled insight into this hardening or blocking of the arteries.

Prof Ziad Mallat, of the Department of Medicine at the University of Cambridge, tells the Cambridge Independent:We are excited about the prospect of this. We hope we have assembled the right team.

Atherosclerosis is very debilitating. If it happens in the arteries that supply the brain, it causes stroke. If it happens in the arteries supplying the heart, it causes heart attacks.

It is really common across the world. Every five minutes in the UK there is one heart attack and one stroke.

Why is this having such a huge impact on the quality of life of people? We believe something is not being treated or understood.

Clinicians currently treat the risk factors for the disease, which include high blood cholesterol, high blood pressure and diabetes.

What we dont do is really treat what causes the disease, which is the malfunctioning of the immune system, says Prof Mallat.

When you have high blood pressure or cholesterol, this injures the arteries. Initially, the immune system sends immune cells to the injured vessel to try to heal the artery.

However, what we know is that most of the time the immune system doesnt operate properly and this prevents the healing, and so the disease progresses.

We have good understanding of how this happens in pre-clinical models, like mouse models, but very limited understanding of how it happens in humans.

We think this is what is preventing doctors and scientists from finding a treatment that would transform the way patients are treated.

Through their iMap, as they are calling it, Prof Mallat and the team of global experts he has assembled want to understand what is happening in the accumulations, known as plaques, that block the arteries and affect blood flow to the heart and other parts of the body. The plaques can be made up of fat, cholesterol, calcium and other substances.

These plaques obstruct the lumen [the interior space in the artery] and even burst into the lumen, leading to clot formation, which obstructs the blood flow. This causes the heart attacks and strokes, says Prof Mallat.

Our idea is to build the first 3D map of these fatty plaques, at

. We would like to know what each immune cell and each cell in the vessel wall is doing. What is its genetic make-up? What is its protein make-up? What is the fuel that it is using? Why, when the immune cell comes along to do a good job, does it stop doing it?

We want to interrogate each cell and work out how it is interacting and communicating with other cells.

Only with this 3D map of the plaques will we be able to understand what is happening inside. Once we have done this, we will be able to harness this knowledge to find new protective methodologies and therapies.

These therapies could harness the immune system, which raises the possibility of vaccinating against atherosclerosis.

If we understand how the immune cells react, we can use the information to re-educate them with vaccination, suggests Prof Mallat. If they are overreacting to fat components or protein components, we can educate them to make them do the right job when they see this in the arteries, to reduce the inflammation and limit the development of the disease.

The scale of this challenge, however, is vast and requires a multi-disciplinary approach.

It needs a lot of different expertise around the world, says Prof Mallat. You need good cardiologists, good molecular biologists, immunologists, mathematicians and computer scientists because the information will be huge and needs to be integrated together. You need people who know a lot about genomics, lipidomics and proteomics, so we have gathered world-leading experts in each of these areas to come together and look at this problem from every angle possible.

Among those helping Prof Mallat is Sarah Teichmann, from the Wellcome Sanger Institute at Hinxton, who is the co-founder of the global consortium working on the Human Cell Atlas a hugely ambitious and important project creating comprehensive reference maps of all human cells in the human body.

They are looking at the make-up of healthy organs, notes Prof Mallat. Some of the investigators are mapping some of the arteries and are looking at vascular cells like endothelial cells. It is intriguing but nobody else is looking at other cells in the artery. We are looking at both the healthy arteries and the diseased arteries. It is building on the work of the Human Cell Atlas.

Also on the team are experts from Imperial College London, Germany, France, Spain, the La Jolla Institute of Immunology in San Diego and from Icahn School of Medicine at Mount Sinai in New York.

Key to their work is the need for data and samples, and the group has multiple sources available.

We have organ donors from the Cambridge bio-repository and the clinical school at Mount Sinai, so we have access to healthy and diseased arteries from the same individuals.

We have access to blood from these individuals and to immune cells from other parts of the body, so we can compare what the immune cells are doing in different compartments.

The other source is from a cohort of thousands of individuals, through a collaboration with Professor Valentin Fuster in Madrid, who have been followed for more than 10 years, and they will be followed for another 10 years.

We have blood samples and microbiota from them. We also have access to imaging of their arteries. They are followed for cardiovascular outcomes, so if someone has a heart attack or stroke, it is documented.

We will be able to look at the ageing of the immune system in these individuals and how this correlates to changes in their arteries and the occurrence of disease.

All of this is being done at very high resolution, which has not been done before. Integrating the information from the genes, the proteins, the lipids and so on, to have a broad view, has never been possible.

There are parallels with the work being carried out at Cancer Research UK Cambridge Institute under Prof Greg Hannon, where the first virtual 3D tumour is being created using a multi-disciplinary team.

We are discussing with him how we can integrate some of the technologies he is developing. It will be fantastic to collaborate with him on this, says Prof Mallat.

What is known already is that our arteries are sensitive to changes in blood flow.

Even subtle perturbations in the micro-environment are sensed by the arteries and can be considered as a danger, explains Prof Mallat.

When it interprets this as a danger, it sends signals to the immune system to react. I would say this is happening almost continuously, and is aggravated of course when you have additional stimuli like high blood cholesterol or exposure to smoke.

While the use of imaging and monitoring of biomarkers is helping us diagnose atherosclerosis earlier, Prof Mallat describes this as not optimal, because we dont understand the disease in a comprehensive manner. A 3D map would aid diagnosis, prediction and prevention of disease, as well as opening up new therapeutic avenues.

Nobody knew 10 or 15 years ago that the immune system could play such a huge role in cancer, Prof Mallat points out. Now cancer immunotherapy is advancing enormously. We are convinced that atherosclerosis is highly motivated by the immune system but no-one is targeting the immune system to treat it. Thats why we want to understand it and we think this could really induce a revolution in our understanding and how we treat it.

Cambridge Cardiovascular to host events at Cambridge Science Festival

Visitors to Cambridge Science Festival will have a chance to find out more about the iMap project and the work of cardiovascular researchers.

Cambridge Cardiovascular, an umbrella group for the field, is involved in organising activities once again at this years festival, which runs from March 9 to 22.

At 6-7pm on Wednesday, March 18 at the Mill Lane lecture rooms in Cambridge, a talk titled More than a blocked pipe: The hardening of the arteries and their role in stroke and heart attacks will be delivered by Dr Nick Evans, of the Department of Medicine, and Prof Melinda Duer, of the Department of Chemistry.

At 6-7pm on Friday, March 20, also at Mill Lane lecture rooms, Dr Sanjay Sinha, of Cambridge Stem Cell Instituteand the Department of Medicine will discuss Mending broken hearts: stem cells for heart disease.

Then, from 11am to 4pm on Sunday, March 22, A View of the Heart will be on offer at the Cambridge Academy for Science and Technology, in Long Road, where cardiovascular scientists will help you explore the organ and visualise heartbeats.

Book at sciencefestival.cam.ac.uk.

The Big Beat Challenge

The British Heart Foundations 30million Big Beat Challenge is designed as the charitys moon-shot to propel our understanding of cardiovascular disease into a new era.

Some 75 applications were received from 40 countries following its launch in August 2018, and these have been whittled down to four, including the one led by Prof Mallat to map and treat atherosclerosis. The other ideas are:

Hybrid heart

Led by Jolanda Kluin, professor of translational cardiothoracic surgery at the University of Amsterdam in the Netherlands, this team plans to create a solution for heart failure by developing a soft robotic heart. They intend to design, build, test and implant a hybrid heart that consists of a soft robotic shell forming the soft artificial muscles and sensors to enable natural motion, and a tissue-engineered lining to make sure all the surfaces in contact with blood are safe. With wireless energy transfer, the vision is that this could replace the need for human heart transplantation.

Echoes

Led by Professor Frank Rademakers, chief medical technology officer at University Hospitals Leuven, Belgium, this team would develop wearable technology that can be used in daily life to capture more data than ever before. This information ranging from symptoms and physical activity to heart function and air quality could be used alongside genetic and healthcare data to transform diagnosis, monitoring and treatment of heart and circulatory diseases through the creation of a digital twin.

Cure heart

This project aims to provide a cure for inherited, killer heart muscle diseases. Led by Professor Hugh Watkins, BHF chair of cardiovascular medicine at the University of Oxford, these researchers will develop a treatment that targets and silences the faulty genes responsible for cardiomyopathies diseases of the heart muscle that can lead to sudden death at an early age. They intend to combine a deep understanding of underlying genetic mechanisms with new technologies, to stop the progression of the damage caused by genetic heart muscle diseases, or even reverse the damage.

Professor Sir Nilesh Samani, medical director at the British Heart Foundation, said: Heart and circulatory diseases remain the number one cause of death worldwide.

Were taking small steps forward every year but whats needed is a giant leap, which wont be achieved by a business-as-usual approach.

The Big Beat Challenge embodies our ambition to turbo-charge progress and could lead to its own man on the moon moment. I have absolutely no doubt the winning idea will define the decade in their area.

The teams will prepare their final applications by June 14, with interviews in early September and a decision expected by the end of the year.

Read more

Our guide to the Cambridge Science Festival 2020

Can HIV be cured? Evelyn Trust-funded research at University of Cambridge probes viral latency

Inside the Cambridge lab in pole position to create a new coronavirus vaccine

Sanger Institute scientist helps unveil blueprint for extraordinary Human Cell Atlas

Prof Greg Hannon on taking over at the Cancer Research UK Cambridge Institute and creating the worlds first virtual reality tumour

Read more from the original source:
Prof Ziad Mallat leads Cambridge effort to win 30m to tackle leading cause of heart attacks and strokes - Cambridge Independent

Aspiration-assisted bioprinting for precise positioning of biologics – Science Advances

INTRODUCTION

Aggregated cells have been formed into spheroids (13), honeycombs (4), and strands (5) from a variety of different cell types and their cocultures. They have many advantages including the cellular capability to secrete extracellular matrix (ECM) components with an effective communication between cells in a native-like microenvironment (1). When cells are grown in an isolated fashion (i.e., cells in monolayers or cell-laden hydrogels), they usually do not facilitate native-like tissue microenvironment because of limited cell-cell and cell-ECM interactions (6). Three-dimensional (3D) cell aggregates, particularly tissue spheroids, are excellent candidates to mimic in vivo tissue microenvironments, which can be reconstituted to form reproducible complex tissues [such as bone (7) and pancreas (8)] and or tissue models [cancer (9)]. Furthermore, cocultured aggregates can be used as building blocks for fabricating scalable tissue complexes (2). Spheroids loaded with endothelial cells can also facilitate a denser tissue microenvironment, inherent ECM secretion, and prevascularized network (10). As a result, large-scale vascularized tissue complexes can be biofabricated using prevascularized cell aggregates as fundamental building blocks (7, 11), which allow more accurate representations of native tissues. Moreover, these tissue complexes stand to develop physiologically correct models for drug screening, disease modeling (i.e., cancer), and high-throughput screening (12). Therefore, various advantages of tissue spheroids make them a great candidate as building blocks for 3D bioprinting.

Despite these advantages, only a few methods including extrusion-based bioprinting (2, 11, 13, 14), droplet-based bioprinting (15), Kenzan (16), and biogripper approaches (4, 17) have been demonstrated for 3D bioprinting of spheroids made of cells, such as but not limited to human articular or nasal chondrocytes, human umbilical vein endothelial cells (HUVECs), human umbilical vein smooth muscle cells, etc. Extrusion-based bioprinting technique dispenses similar-sized spheroids suspended in a hydrogel ink through a glass nozzle. During bioprinting, spheroids in the hydrogel ink may self-aggregate inside the nozzle, leading to clogging issues (2). Although extrusion-based bioprinting of cellular aggregates in the form of strands can generate scalable tissues (3, 5), their use in high-precision applications, such as organ-on-a-chip platforms or microphysiological systems, is quite challenging. In droplet-based bioprinting, a single spheroid is loaded into a droplet during bioprinting, which enables the positioning of spheroids in 2D (15). Kenzan method uses an array of needles on which spheroids are skewered by a robotic arm; however, this method requires same-size spheroids since bioprinted spheroids have to fit properly in the needle array and smaller spheroids would be prone to fragment during insertion on the needle (16). Recently, the biogripper technique was introduced to manipulate defined micromolded tissue blocks. However, this technique enables bioprinting of only molded tissue blocks in the range of 600 m to 3.4 mm, where submillimeter scale is quite large for typical tissue building blocks (4, 17). All the existing techniques used in the literature suffer from the positional accuracy and precision of spheroid bioprinting; may induce substantial damage to biological, structural, and mechanical properties of spheroids; and are limited to the use of same-size spheroids for successful bioprinting.

Here, we present a new hybrid bioprinting approach through harnessing the power of aspiration forces, which enables us to pick and precisely bioprint a wide of range of biologics, with dimensions in an order-of-magnitude range (80 to 800 m), into/onto a gel substrate with minimal cellular damage. To better understand the response of biologics to the bioprinting process, we unveil the underlying mechanism explaining the physical behavior of viscoelastic spheroids and their interactions with physical governing forces during aspiration, lifting, and bioprinting. Compared to the abovementioned methods, the presented approach facilitated the bioprinting of spheroids in higher positional precision and accuracy, ~11 and ~15% with respect to the spheroid size, respectively. In addition, it enabled bioprinting in 3D with flexibility of bioprinting of nonuniform, any-size spheroids into (i) a functional gel as a scaffold-based approach or (ii) a sacrificial gel as a scaffold-free approach (Fig. 1). The newly developed aspiration-assisted bioprinting (AAB) platform, modified from a MakerBot Replicator 1 (<$1000) (see fig. S1), operates a custom-made glass pipette, which is used to pick up biologics and 3D bioprint them into or onto a gel substrate. AAB was coupled with microvalves for droplet-based bioprinting of functional or sacrificial hydrogels. To bioprint spheroids into a functional hydrogel (i.e., fibrin), fibrinogen and thrombin layers were printed via microvalve bioprinting to obtain fibrin constructs. Spheroids were then bioprinted into desired positions before the constructs were fully cross-linked. To bioprint spheroids onto a sacrificial hydrogel (i.e., alginate), microvalve bioprinting and aerosol cross-linking processes were used. The first step included the generation of sodium alginate droplets on a glass substrate. Second, calcium chloride (CaCl2) was fumed for instant cross-linking (18). Spheroids were then picked and 3D bioprinted onto the partially cross-linked alginate. Next, the print area was overlaid with sodium alginate droplets. Then, aerosol form of CaCl2 was applied again. The process was then repeated as many times as needed for building other layers. Next, bioprinted spheroids were maintained in the support gel temporally until partial fusion was realized. Last, the support gel was gently removed through decross-linking using a lyase solution.

In step 1, spheroids are picked from the cell media by a glass pipette, where required back pressure is set to lift spheroids. Afterward, spheroids can be bioprinted into sacrificial hydrogels (scaffold-free bioprinting) or functional hydrogels (scaffold-based bioprinting). In this regard, in step 2, microvalve bioprinting is used to bioprint a gel substrate, which can then be partially cross-linked using various different cross-linking schemessuch as but not limited to enzymatic, photo, and ionic cross-linkingas highlighted in step 3. Next, in step 4, spheroids are bioprinted precisely into designed positions, and spheroid bioprinting is repeated as many times as needed. Steps 2 to 4 can be repeated as needed. In step 5, bioprinted tissues are isolated from the support hydrogel (for scaffold-free bioprinting) or further grown in the functional hydrogel (for scaffold-based bioprinting). UV, ultraviolet.

The presented approach paves the way for bioprinting several tissue types and a wide range of spheroids in 2D and 3D in gel substrates with multiple applications presented throughout this paper, including (i) the development of physiologically relevant culture environments to demonstrate the collective angiogenic sprouting behavior of spheroids in a scaffold medium and (ii) fabrication of osteogenic tissues to decode the role of midterm osteogenic induction of stem cellbased spheroids (before bioprinting) on the mineralization and assembly behavior in a scaffold-free environment. In addition to spheroids, other living cells and tissue building blocks, including electrocytes from electric eel and tissue strands, can be bioprinted for a wide variety of applications, such as but not limited to tissue engineering, regenerative medicine, drug testing and pharmaceutics, disease modeling, microphysiological systems, biophysics, and biocomputing.

In the AAB process, the first step is to pick a spheroid (fig. S2) and then lift it and drag it rapidly outside the culture media by aspiration forces as shown in Fig. 2A. The spheroid should be captured with a minimum aspiration force to transfer but not break or damage it. During aspiration under an applied stress (), a spheroid with viscoelastic properties exhibits both viscous and elastic properties (19). It exhibits elastic properties like a solid at times shorter than relaxation time and like a fluid for t > , which can be described by the Maxwell modelddt+=ddt(1)where is the strain and is the modulus of elasticity of spheroids. During this process, two major impediments were experienced. First, spheroids were prone to fragment because of low cellular cohesivity and elasticity even under very low pressure levels. Second, detachment of spheroids from the pipette tip at the three-phase contact (air-liquid-tissue) was observed during lifting. The former can be mitigated by engineering of spheroids to attain better elastic properties [such as culturing them longer (20) or incorporating cells expressing higher cell-cell and cell-matrix adhesion or ECM proteins (21)], and the latter can be addressed by determining the minimum aspiration pressure (critical lifting pressure). A spheroid is lifted by pipette aspiration after overcoming gravity, buoyance force, hydraulic drag, and thermodynamic barrier at the interface. The main difficulty in lifting a spheroid is the binding energy caused by the surface tension at the interface between the culture media and air. The spheroid contributes to the lifting barrier in the form of a contact angle at the three-phase (tissue, air, and media) contact line. The energy barrier to lift a spheroid into air can be expressed asE=As1,2cos2d2(2)where As is the surface area of a spheroid, 1,2 is the surface tension between the media and air, and d is the contact angle at the three-phase contact line. During lifting, the three-phase contact angle (dynamic contact angle) is greater than the static contact angle because of dynamics effect (22). The dynamic contact angles of spheroids made of several types of cells were enumerated in table S1. By comparison, the thermodynamics barrier is about 500 times of gravity, 100 times of Stokes drag, and 5 times of Young-Dupre critical pressure. During lifting, the energy barrier can be estimated by adding a geometrical correction factor (m) as a result of slight changes in the spheroid geometry due to the aspiration of a tongue (see Fig. 2B). The initial contact length of the tongue (ht=0) of a spheroid ranges between Rp and 2Rp. Thus, m can be defined as followsm=14Rp24RS24Rp2=1Rp2RS2Rp21(RpRs)2(3)

In Eq. 3, Rp is the pipette tip radius and Rs is the spheroid radius. m can be approximated to 1 when the spheroid radius is much larger than the radius of the pipette tip. However, when lifting a small spheroid or when the radius of the pipette is comparable to that of the spheroid, m becomes much smaller. Consequently, the critical lifting pressure of a spheroid (PC) from the media can be expressed as follows (22)PC=mFmax/Ah=2mRS1,2cos2d2/(Rp2)(4)

The critical lifting pressure is proportional to the surface tension coefficient of the media-air interface, the radius of the spheroid, the dynamic contact angle at the three-phase contact line, and the tapered angle of the pipette (influences Rp, but this parameter was fixed).

(A) Time-lapse images during spheroid lifting process (at the interface of cell media and air). (B) A schematic showing physical parameters involved in lifting of a spheroid from the cell media. (C) SEM images, (D) surface tension (n = 5), and (E) the normalized collagen content of HUVEC, 3T3, 4T1, HDF, MSC/HUVEC, and MSC spheroids (compared to HUVEC spheroids) at day 2 (n = 4; *P < 0.05, **P < 0.01, and ***P < 0.001). (F) Critical lifting pressure to lift spheroids (in the range of 200 to 600 m in diameter) (n = 5). The experimental data spread under the theoretical curve, which was determined using the experimental data for 4T1 spheroids with parameters (d = 64 and s1,2 = 57.4 mN/m). Spheroids made of other cell types had lower theoretical critical lifting pressure values (data are not shown in the paper). For instance, the theoretical critical lifting pressure for HUVEC spheroids was 20% smaller than that of 4T1 spheroids. (G) Viscoelastic behavior of spheroids under aspiration (n = 3). Here, h denotes the advancement of spheroids inside a glass pipette. The aspiration experiment used a similar pipette as that in bioprinting. The aspiration pressure was determined according to the size of spheroids satisfying the condition that spheroids could be lifted from the cell media.

To demonstrate picking and lifting of spheroids, we fabricated a wide range of spheroid types with different viscoelastic and surface tension properties using HUVECs, mouse fibroblast cell line (3T3), mouse mammary carcinoma line (4T1), human dermal fibroblasts (HDFs), coculture of human mesenchymal stem cells (MSCs) and HUVECs, and MSCs. Various kinds of spheroids in a diameter range of 200 to 600 m were obtained using cell-repellent 96-well plates. Spheroids were fabricated at a cell density from 2500 to 10,000 per spheroid and cultured for 1 to 3 days (fig. S3A). During the first 24 hours, we closely monitored spheroid formation and observed that 3T3 and HDF spheroids became compact and withstood aspiration forces at the end of 20 hours, while others became compact at 24 hours, implying that these spheroids could be lifted anytime thereafter (fig. S4). To investigate the ultrastructure of spheroids, spheroids were analyzed by scanning electron microscopy (SEM), and SEM images demonstrated clear differences in surface morphology and compactness of different spheroids cultured for 2 days (Fig. 2C). The surface topology of MSC spheroids displayed a substantial amount of secreted ECM with smoother and more compact structure due to tight cell-cell and cell-ECM interactions. On the other hand, the surface topology of HUVEC spheroids displayed lesser ECM compound, which resulted in a pitted surface and tiny gaps between cells. To lift spheroids, spheroids were required to be compact and captured in the cell media and to withstand aspiration forces. Spheroids also have surface tension, with each cell being analogous to a liquid molecule (23, 24). Thus, surface tension of spheroids were investigated by a micropipette aspiration technique (19, 25) according to the Young-Laplace equation, which gave the relationship between the internal pressure of spheroids and cell media across a curved interface. According to the Young-Laplace equation, the surface tension coefficient can be estimated as =P2(1/Rp1/Rs), where P is the equilibrium aspiration pressure when the advancement of the spheroid inside the pipette was the same as the radius of the pipette tip. As shown in Fig. 2D, the surface tension of HUVEC, 3T3, 4T1, HDF, MSC/HUVEC, and MSC spheroids at 2 days of culture were measured to be ~14, 30, 37, 41, 51, and 66 mN/m, respectively. Surface tension of MSC-only spheroids was approximately five times higher than that of HUVEC spheroids. It is worth mentioning that Norotte et al. (26) used a different method to measure the surface tension of spheroids and obtained a similar result for HUVEC spheroids. While the surface tension of 4T1, HDF, and MSC/HUVEC spheroids increased over time, the surface tension of spheroids made from other cell types maintained similar properties during the 3-day culture (fig. S3B). This time frame, on the other hand, can vary for spheroids prepared using other techniques, such as but not limited to handing drop, microfluidic, rotator flask, and liquid overlay methods (27). Our results indicate that there is a positive correlation between the surface tension of spheroids and their compactness, confirming previous findings by Foty and Steinberg (28). In addition, the total collagen amount of spheroids was investigated using a hydroxyproline colorimetric assay kit since collagen is one of the major ECM components of spheroids (3). The measured collagen amount for 3T3, 4T1, HDF, MSC/HUVEC, and MSC spheroids at day 2 was ~1.4-, 1.4-, 1.5-, 2.8-, and 3.4-fold higher compared to that for HUVEC spheroids, respectively (Fig. 2E). The normalized collagen content in different spheroids showed a similar trend as compared to the compactness of spheroids; however, we did not observe any notable differences among the normalized collagen content for 3T3, 4T1, and HDF spheroids. A recent study reported a positive correlation between compactness of cell aggregates and their collagen expression (29).

Figure 2F denotes a linear relationship between the critical lifting pressure and diameter of spheroids (in the range of 200 to 600 m). When the diameter of spheroids increased, the critical lifting pressure also increased. The theoretical critical lifting pressure for each spheroid types at different diameters were determined using Eq. 4, where the maximum theoretical value belonged to 4T1 spheroids, which could be used as a baseline for other spheroid types to be tested during aspiration. In addition, the minimum critical lifting pressure among all spheroids types (from 200 to 600 m) was determined to be ~19 mmHg. The difference between the theoretical and experimental value of the critical lifting pressure was due to (i) the irregular shape of spheroids (elliptic or pancake-like) influencing the maximum energy barrier (30, 31) and (ii) the measurement inaccuracy of the pressure sensor (which was around 10%; see fig. S5, A and B). Another vital factor for successful bioprinting was to determine viscoelastic properties of spheroids, which were reflected by the relationship between stress and strain as a function of time. During the AAB process, spheroids were kept within a solid elastic regime. The operation time of AAB was defined by the elapsed time between spheroid picking and bioprinting, which was commonly less than 30 s. Therefore, spheroids were successfully bioprinted, while their relaxation time () was longer than the bioprinting operation time. For this reason, we plotted the aspiration curves of various spheroids (Fig. 2G). At the early stage of aspiration, spheroids deformed very quickly within the elastic regime. Then, after a relaxation time, accumulated stress was gradually released, and spheroids experienced a relaxation process and return to their original shapes. As shown in Fig. 2G, the relaxation time for HUVEC, 4T1, and MSC/HUVEC spheroids were around 3 to 5 min. The relaxation time for HDF spheroids could reach up to 18 min. Overall, these relaxation times were sufficient to assure successful bioprinting without permanent deformation to spheroids.

After lifting spheroids from the cell media and transferring them to the bioprinting stage, the back pressure was released, and the spheroids were fully recovered or, in other words, they returned to their original shapes as bioprinting time was less than the relaxation time. To bioprint a spheroid, the spheroid was partially submerged into a precross-linked or partially cross-linked gel substrate, as going deeper could result in their breakage due to penetration of the pipette tip into the spheroid. When the nozzle moved up, the spheroid was stuck to the gel because of adherence between the spheroid and the gel (Fig. 3A1), released from the pipette, and deposited into the gel. In AAB, successful bioprinting was determined by a constant adherence at the pipette tip (upward, Fup) and a variable bioprinting force at the interface of gel (downward, Fdown). To determine Fup, two angles were defined: 1=arccosRpRs, and 2 is the pipette taper angle. Fup can be expressed as Fup = 2Rps cos (1 + 2), where s is the surface tension of the spheroid. At the bottom of the spheroid, the maximum force pulling the spheroid downward due to the surface tension of the gel substrate, i.e., the maximum bioprinting force provided by the gel, can be expressed as Fdown=2Rsgcos2(12). Here, g is the surface tension of the gel, and 1 is the effective angle of wetting. For successful bioprinting, the maximum force pulling the spheroid downward needs to overcome the surface tension adherence at the tip of the pipette. Thus, successful printing, as shown in Fig. 3A2, needs to satisfy the condition Fdown > Fup. However, if a spheroid is not fully recovered (when the spheroid is submerged rapidly into the gel), there can be an extra stress (Fext) at the pipette tip. The extra stress may increase the difficulty of bioprinting since Fdown needs to overcome Fext + Fup.

(A1) A schematic showing critical parameters during bioprinting. (A2) An image from the traveling camera showing spheroid placement onto a gel substrate. Fluorescent images showing (A3) PSU and (A4) matrix patterns. GFP+ MSC spheroids were patterned onto (A5) COL I and (A6) gelatin-methacryloyl (GelMA). (A7 and A8) Images showing that eight fixed spheroids were bioprinted on top of each other in air without any gel support during bioprinting. DAPI, 4,6-diamidino-2-phenylindole. (B1) Time-lapse images of self-assembly process after bioprinting of 1-day cultured 3T3 spheroids at 0, 24, 48, and 72 hours (B2) and the normalized contact length and intersphere angle of fusing spheroids up to 24 hours. (B3) Cell viability of 3T3 spheroids that were not treated with bioprinting (control), after bioprinting inside the cell media (case 1), and after bioprinting into a gel substrate (case 2) (n = 3; *P < 0.05). CMTMR, 5-(and-6)-(((4-chloromethyl)benzoyl)amino) tetramethylrhodamine; CMFDA, 5-chloromethylfluorescein diacetate. (C1) Hematoxylin and eosin staining from the sagittal plane of a tail segment of electric fish showing stacked electrocytes in series. (C2) The SEM image of a single electrocyte. (C3) Calcein staining of bioprinted electrocytes. (D) A bioprinted cartilage tissue strand between pins. Photo credit: Bugra Ayan, Penn State University.

To verify the presented theoretical approach, we measured 1, 2, and 1 angles and obtained the surface tension of precross-linked gel [1% (w/v) alginate] from the literature (32). The results indicated that Fdown was ~32, 14, 12, 11, 9, and 7 times greater than Fup for HUVEC, 3T3, 4T1, HDF, MSC/HUVEC, and MSC spheroids, respectively, as enumerated in table S2. Surface tension of gel solutions can vary from gel to gel. In some gels [such as alginate (32)], surface tension is dependent on concentration; however, there are hydrogels where surface tension is not dependent on concentration, such as polyacrylamide (33). Therefore, we cannot make a general statement on the correlation between concentration and surface tension. In addition, concentration of the gel should also be compatible with microvalve bioprinting, as higher concentrations of precursor solutions are, in general, bioprintable as such concentrations can support droplet formation (34). For bioprinting into a gel substrate, the gel precursor solution should be flowable as well so that the solution can overlay the bioprinted spheroid quickly; therefore, we performed bioprinting on partially cross-linked or uncross-linked gel precursor.

Before performing the patterning and 3D bioprinting of spheroids, we first inspected the quality of positioning during bioprinting. In this regard, agarose beads (with near-perfect sphericity) and the motion stage of the 3D bioprinter were used as control groups. The positional precision and accuracy for the motion stage (empty pipettes), spherical agarose beads (ranging from 250 to 300 m in diameter), and 2-day cultured MSC/HUVEC spheroids (~300 m in diameter) were determined to be ~2 and 2.1 m, 14.4 and 10.4 m, and 34.7 and 45.9 m, respectively (fig. S5, C to E). The results indicate that the bioprinter motion stage had a small effect on the positional error during bioprinting and that bioprinting of spheroids was about two- and fourfold less precise and accurate compared to printing of agarose beads, respectively. This could be due to the viscoelastic nature, lesser sphericity, and slightly higher average diameter of tissue spheroids compared to more rigid agarose beads. We demonstrated the ability to bioprint spheroids precisely (~11% with respect to the average spheroid size) onto alginate through patterning of various shapes (fig. S6, A1 to A3), initials of Penn State University (PSU) (Fig. 3A3), and a matrix of spheroids of various shapes and dimensions (including irregular shapes) in the diameter range of 80 to 200 m (made of HUVEC, 3T3, and 4T1 cells) (Fig. 3A4). Moreover, we also demonstrated the bioprinting of green fluorescent protein (GFP)labeled MSC spheroids in the shape of triangle and ring into other gels, including type I collagen (COL I) and gelatin methacryloyl (GelMA), as shown in Fig. 3 (A5 and A6, respectively). To highlight the unprecedented precision of AAB, we bioprinted eight different-sized MSC spheroids on top of each other without any gel support (Fig. 3, A7 and A8) and also fabricated a hollow bridge shape with MSC spheroids using the same approach (fig. S6, B1 and B2). Moreover, we demonstrated another example of a hollow bridge configuration, where the middle spheroid in the second layer was supported by the underlying gel without any spheroid support underneath (fig. S7). This was accomplished by bioprinting the gel and spheroids alternatingly at each layer, which necessitated the precise control of gel thickness; however, the gel thickness was dependent on several factors, such as the droplet resolution, cross-linking time, and spreading behavior of droplets, which were not trivial to control at the same time using microvalve bioprinting. To overcome this fundamental limit, self-healing Bingham plastic support gels (35) could be used to freely move the spheroids inside gels, which may bring more flexibility in self-assembly of highly complex geometries including hollowed-out structures.

In addition, self-assembly of spheroids was investigated at predetermined time points. As shown in Fig. 3B1, 3T3 spheroids were bioprinted into fibrin with close proximity and then cultured for 72 hours. The two individual 3T3 spheroids bioprinted next to each other gradually fused to form a larger spheroid to minimize their surface energy, as described in a previous study (21). The normalized contact length and intersphere angle increased by 50% over the first 24 hours of culture (Fig. 3B2). To evaluate the role of bioprinting on the viability of spheroids, two cases were evaluated. In case 1, the viability of spheroids was determined immediately after lifting spheroids and depositing them into another cell medium reservoir. In case 2, the viability of spheroids was evaluated after bioprinting them into the gel substrate. Spheroids, which were not subjected to the bioprinting process, were used as a positive control group. The viability of spheroids in the positive control group and case 1 were determined to be ~94 and ~88%, respectively (Fig. 3B3). In case 2, cell viability was measured to be ~82%. The decrease in cell viability could be due to the aspiration force or dehydration during the rapid transfer or cell damage when spheroids were submerged into the hydrogel substrate; however, viability levels over 80% for bioprinting of spheroids could still be considered moderate, as viability of fabricated cells aggregates even without bioprinting could be in that range (36).

In addition to bioprinting of spheroids, the AAB system also enabled the bioprinting of other biologics. For example, we demonstrated the bioprinting of electrocytes isolated from electrogenic organs of electric fish. Because of the asymmetric functionality of their anatomy and their spatial arrangements in series configuration in an electric organ (similar to batteries connected in series; Fig. 3C1), electrocytes produce a considerable amount of electricity (37). In this study, five electrocytes (~400 m in diameter; Fig. 3C2) from weakly electric fish (Brachyhypopomus gauderio) were obtained according to the animal protocol (Institutional Animal Care and Use Committee #47870) and patterned next to each other onto agarose using AAB with a back pressure of ~20 to 25 mmHg (Fig. 3C3). To the best of our knowledge, this is the first bioprinting of electrocytes, which can be used in biofabrication of biological batteries for various applicationssuch as pacemakers, cochlear implants, and brain chipsin the future (38). In addition, these cells could be arranged in the form of biological circuits for biocomputing or as bioelectric interfaces for use in treating limb loss, musculoskeletal disorders, and body augmentation as well as cyborg organs interfacing electrical devices and biology (39). The presented approach showed the possibilities of bioprinting not only spheroids or single cells but also irregularly shaped aggregates. For example, Fig. 3D shows vertical bioprinting of tissue strands [described in our previous work (5)] into the space between pins, which can be used for fabrication of scalable tissues of muscle, fat, cartilage, nerves, blood vessels, etc.

To demonstrate other unique capabilities of the presented approach, heterogeneous tissue complexes were 3D bioprinted using spheroids of different sizes and types including tdTomato+ HUVEC spheroids (~155 m) and GFP+ MSC spheroids (~390 m) (Fig. 4, A and B). Spheroids were arranged into a pyramid structure to clearly visualize the heterogeneous architecture using confocal imaging, where alginate was used as a sacrificial gel and the first, second, and third layers constituted of HUVEC, MSC, and HUVEC spheroids, respectively. This example clearly demonstrates the unique deposition ability of the AAB technique, as the last HUVEC spheroid was accurately placed into the small and confined space between three adjacent MSC spheroids (~2.5-fold of HUVEC spheroid size) on the third layer. Alginate preserved the structural integrity of the bioprinted pattern during the initial stage of culture. Moreover, it enabled the bioprinted entities to stick in their precise locations, which were defined before the bioprinting process. To print the sacrificial alginate, we used a unipolar wave pulse with a dwell voltage (amplitude) of 5 V and a dwell time (valve opening duration) of 1000 s for actuating the microvalve dispenser, where a positive back pressure of ~103 kPa was used for driving the flow of sodium alginate inside the tubing. The reader is referred to fig. S8A for the optimization of dwell time and droplet volume. Afterward, sodium alginate layer was partially cross-linked with aerosol form of CaCl2 for 30 s, and spheroids were bioprinted as explained in Fig. 1. Patterned spheroids were partially fused and maintained their initial position after 48 hours of incubation. After 2 days of culture, alginate was gently dissolved [using an alginate lyase as explained in our earlier work (5)], and the bioprinted constructs maintained their integrity even after the sacrificial alginate was removed. As evident from the confocal images (Fig. 4, C1 to C6), spheroids were well connected in 3D. A similar arrangement was also presented with MSC spheroids of three different dimensions, where spheroids got larger as more layers were laid down (fig. S9, A to C). We also demonstrated a more complex pattern such as a 3D diamond pattern with MSC/HUVEC spheroids (Fig. 4, D to G). To the best of our knowledge, this is the first bioprinting of tissue spheroids with such a positional accuracy, ~15% with respect to the spheroid size, yielding highly intricate geometries and heterogeneous structures in 3D.

(A) A schematic illustration of 3D bioprinting of a heterogeneous pyramid construct using different sizes and types of spheroids. (B) A photograph of the bioprinted three-layer heterogeneous pyramid. (C1 to C6) 3D reconstruction of confocal images of bioprinted pyramid of tdTomato+ HUVEC spheroids (first and third layers) and GFP+ MSC spheroids (second layer). (D) A schematic illustration of a diamond construct made of MSC/HUVEC spheroids. (E) A photograph showing 3D bioprinted diamond from the side camera. (F) Fluorescent images of the bioprinted diamond, which were stained with DAPI, CD31, and F-actin. (G1 to G4) Confocal images of the diamond construct (bottom view) (note that 1-day cultured HUVEC and 2-day cultured MSCs were used in these experiments). Photo credit: Bugra Ayan, Penn State University.

In this study, we have demonstrated the utilization of AAB in both scaffold-based and scaffold-free configurations and presented two unique applications demonstrating its potential in the development of physiologically relevant culture environments for studying angiogenic sprouting and fabrication of osteogenic tissues for tissue engineering purposes. To demonstrate the application of AAB in scaffold-based fabrication of physiologically relevant culture environments, we bioprinted HUVEC spheroids into fibrin hydrogel with a predetermined distance apart and studied their collective angiogenic sprouting behavior. This importantly signifies that angiogenic sprouting behavior can be tailored by varying the proximity of spheroids, hence directly affecting cell-cell signaling. To study the effect of distance on angiogenic sprouting, we deposited droplets of fibrinogen (6 mg/ml) [loaded with HDFs (0.5 million/ml)] and thrombin (2.4 U/ml) layer by layer (in 1:1 volume) to fabricate fibrin constructs, as such concentration ranges support angiogenesis of HUVECs (40). A positive back pressure of ~103 kPa was used for driving the flow of fibrinogen and thrombin solutions inside the tubing and dispenser. To dispense a ratio of 1:1 of fibrinogen and thrombin, we used dwell times of 500 and 700 s for obtaining the same droplet volumes at the microvalve dispensers, respectively. The reader is referred to fig. S8 (B and C) for the optimization of droplet generation for fibrinogen and thrombin solutions. HUVEC spheroids were then bioprinted into fibrin before complete gelation. The distances between the spheroids were maintained at 400 10, 800 13, and 3000 16 m (isolated spheroids used as a control group) (Fig. 5A1). Bioprinted constructs were cultured for a period of 7 days. Angiogenic sprouting from HUVEC spheroids at days 2, 5, and 7 was quantified using AngioTool software (41). Spheroids with close proximity to each other were seen to have higher total vessel length as compared to isolated spheroids right from the initial stages of culture. The combined length of sprouts was 13 and 11 mm for 400 and 800 m, respectively, as compared to the control group, which were around 6 mm (Fig. 5A2). A similar trend was also observed for the total vessel area. There was approximately 3- and 2.5-fold increase in the vessel area for 400- and 800-m distances on day 7 as compared to the control group, respectively. Although the total number of branching points or junctions was not significantly different among different groups at day 2, the sprouts branched more when spheroids were bioprinted close to each other, as compared to the control group (fig. S10). On day 7, the number of junctions for 400- and 800-m distances was 2.1- and 1.7-fold greater than those for the control group, respectively. The mean lacunarity refers to the amount of free space around the sprouted capillaries, and an increase in angiogenic sprouting results in a decrease in lacunarity (42). This yields a measurement of the sprouting activity, and the mean lacunarity was the highest for the control group compared to other two groups.

(A1) Epifluorescent images of bioprinted tdTomato+ HUVEC spheroids with varying distances (400 to 3000 m) apart on day 7. (A2) Graphical representation of various sprouting propertiesnamely, total vessels length, total number of junctions formed, vessel area, and mean lacunarityobtained at day 7 for bioprinted HUVEC spheroids (n = 3; ***P < 0.001). (B1) Epifluorescent images of bioprinted GFP+ and tdTomato+ HUVEC spheroids with varying distances (400 to 3000 m) apart on day 7 along with higher-magnification confocal images of the interface region in XY and YZ planes showing capillaries formed by both GFP+ and tdTomato+ HUVECs (indicated by white arrows). (B2) Directionality analysis demonstrating the direction and percentage of normalized number of sprouts on day 7 (n = 3; *P < 0.05, **P < 0.01, and ***P < 0.001) [note that green and red bars demonstrate the angles of interest (AOIs), which are [60, 60] for GFP+ HUVECs and [120, 240] for tdTomato+ HUVECs]. (B3) Confocal images at the interface of two spheroids showing capillaries formed by both GFP+ and tdTomato+ HUVECs. (C1) A schematic illustration of the directionality of sprouts from a HUVEC spheroid toward a spheroid of GFP+ HDF and MSC cocultures. (C2) The directionality analysis for different mixing ratios of HDF:MSC, including HDF (control), 2:1, 1:1, and MSC (control), on day 7 (n = 3; ***P < 0.001 shows significance between AOI and Other for each group, and #P < 0.05, ##P < 0.01, and ###P < 0.001 show significance among AOIs of different groups) [note that N/A represents the none applicability of the directionality analysis, as no sprouts were observed in the MSC-only group; no directionality analysis was performed for the 3000-m distance, as sprouting was not observed and HUVECs exhibited spreading only; 1-day cultured HUVECs were used in all experiments; and the critical lifting pressure for coculture HDF/MSC spheroids (2:1 and 1:1 ratio) was determined to be 28.7 and 29.1 mmHg, respectively, through interpolation of the critical lifting pressure values for HDF and MSC spheroids presented in Fig. 2F].

In our study, HUVEC spheroids bioprinted with 400- and 800-m distances apart were considered to be in close proximity. On the other hand, HUVEC spheroids bioprinted with 3000-m distance apart were far enough to be considered isolated spheroids (control group). Proximity between cells triggers a cascade of events that, in turn, influences cellular behavior, and to study these events, it is important to control localization under in vitro conditions (43). The presented approach offers the capability of controlling the distance between spheroids and thus aims to better understand this signaling between spheroids. Sprouting properties such as vessel length, junction formation, and vessel area were all enhanced when spheroids were close to each other as compared to when they were placed far apart. The presence of HDFs in the matrix surrounding HUVEC spheroids also reinforced the formation of stable sprouts. It is known that fibroblasts act as supporting cells and surround the capillary-like structures, which gradually develop by self-organization of endothelial cells and simultaneous degradation of fibrin matrix (44). Moreover, fibroblasts secrete various soluble growth factors such as vascular endothelial growth factor (VEGF), angiopoietin-1, platelet-derived growth factor (PDGF) (45, 46), which enhance and modulate the growth of angiogenic sprouts (47). Bioprinted HUVEC spheroids close to each other probably up-regulated the combined secretion of these growth factors in the vicinity of the spheroids, which led to enhanced sprouting properties. The sprouts from closely placed spheroids extended toward each other and gradually fused to form a capillary network.

To better understand whether the positioning of spheroids affected their collective angiogenic sprouting behavior, GFP+ and tdTomato+ HUVEC spheroids were bioprinted into fibrin gel (with same properties as discussed before) at 400-, 800-, and 3000-m distances apart, and cultured over a period of 7 days (Fig. 5B1). A directionality analysis was then performed to investigate the role of the presence of a nearby spheroid on the directionality of angiogenic sprouting. Thus, the angle made by each sprout with the horizontal line (0/180) was measured for all spheroids. The angle spans [60, 60] for GFP+ HUVEC and [120, 240] for tdTomato+ HUVEC spheroids, where sprouts primarily grew toward the other spheroid, were considered the angles of interest (AOIs), as highlighted in Fig. 5B2. On day 7, the percentage of normalized number of sprouts (where the total number of sprouts were normalized with respect to the total angle span) in AOI was significantly higher than that of the other angle span (indicated with Other) for 400- and 800-m distances, indicating a dominating directionality of angiogenic sprouting toward the other spheroid. The percentage of normalized number of sprouts for AOI was ~63 and 65 for GFP+ and ~59 and 61 for tdTomato+ HUVEC spheroids, for 400- and 800-m distances, respectively. Although the directionality of sprouts for all cases may not be very apparent on day 2 (initial stages of culture; fig. S11, A1 and A2), on day 5 (later stages of culture; figs. S11, B1 and B2, and S12), mature sprouts were more concentrated within AOI. In addition, for sprouts formed in directions falling outside the AOI (depicted in gray), we observed more random growth of sprouts over time. The sprouts for spheroids that were 3000 m apart were random and did not indicate any directionality from days 2 to 7 (Fig. 5B2, bottom, and fig. S12C). For 400-m distance, it was also observed that the GFP+ HUVECs suppressed the sprouting ability of tdTomato+ HUVECs day 2 onward, where higher numbers of GFP+ HUVEC sprouts were observed at the interface [see fluorescent images in fig. S11 (A1 to B1)]. Domination of GFP+ HUVEC sprouts could be due to the fact that GFP+ HUVECs in this experiment were from an earlier passage (passages 2 to 5) with respect to tdTomato+ HUVECs (passages 5 to 7) (48). In addition, we investigated whether sprouts from both spheroids interacted and thus performed confocal imaging at the interface close to the tdTomato+ HUVEC spheroid (Fig. 5B3). We noticed that some capillaries were formed by both GFP+ and tdTomato+ HUVECs, as shown by arrows, which could be due to vascular anastomosis or the migration of GFP+ HUVECs to the other side of the interface and their contribution to the capillary formation by tdTomato+ HUVECs spheroids. Overall, the results showed that HUVEC spheroids bioprinted close to each other influence each others sprouting behavior, which led to a well-organized network formation between the sprouting bodies.

In the abovementioned experiments, we investigated the role of multiple HUVEC spheroids on their collective sprouting behavior, where HDFs were bioprinted as a single-cell suspension in fibrin, had direct contact with HUVEC spheroids, acted as supporting cells, and aided in forming stable sprouts. To explore whether HDFs could still influence the sprouting behavior of HUVECs even when they were not in direct contact with HUVECs, we reconfigured the design of the experiments presented above. In this regard, a HUVEC spheroid was bioprinted along with a coculture spheroid of GFP+ HDFs and MSCs (with controlled distance apart), which enabled us to change the concentration of HDFs and study the differential effect of coculture composition on HUVEC sprouting. GFP+ HDF/MSC spheroids, in 2:1 and 1:1 ratios, were bioprinted next to tdTomato+ HUVEC spheroids in fibrin with the same properties as discussed before (Fig. 5C1). HDF and MSC (homocellular) spheroids were used as control groups. The distance between these spheroids were varied from 400 to 3000 m, as performed before. For the cases of 400 and 800 m, HUVECs were attracted toward spheroids of HDF, 2:1, and 1:1 groups and were seen arranging into sprouts over a period of 7 days (figs. S13 and S14). The directionality analysis on day 7 revealed that the percentage of normalized number of sprouts within AOI for HDF, 2:1, and 1:1 groups for 400-m distance were ~82, 80, and 67, respectively (Fig. 5C2). For 800-m distance, a similar directionality was observed, where the percentage of normalized number of sprouts within AOI for HDF, 2:1, and 1:1 groups were determined to be ~84, 81, and 76, respectively. On the other hand, spheroids in the MSC group (Fig. 5C2) or spheroids located apart with a 3000-m distance (fig. S15) did not induce sprouting day 2 onward; rather, HUVECs exhibited spreading behavior. On day 2, for the case of 800 m, although the tip cells from HUVEC spheroids were not observed to have any physical contact with HDFs at the interface of both spheroids, sprout-like structures were already formed (fig. S16A). These structures were directed toward HDFs, as the relevant directionality analysis revealed a significant difference between AOI and Other for HDF, 2:1, and 1:1 groups (fig. S16B). This could be due to the fact that HDFs communicated with HUVECs via paracrine signaling by the possible diffusion of secreted growth factors, such as VEGF, angiopoietin-1, and PDGF, as discussed before (45, 46). By day 5 when HDFs substantially migrated out of the coculture spheroids, HDFs interacted with HUVECs outside AOI, particularly within angle spans [60, 120] and [240, 300], and induced sprout formation in those directions (fig. S17). Overall, more stable sprouts were observed in HDF-involved spheroids, and the results revealed that better directionality of angiogenic sprouting could be attained when the density of HDFs increased in the coculture spheroid. The presence of MSCs, on the other hand, had a negative impact on angiogenic sprouting of HUVECs, which could be due to the inhibitory effect of MSCs on the angiogenic potential of HUVECs via cell-cell contact through modulation of the VE-cadherin/-catenin signaling pathway as described before (49, 50).

To demonstrate another major application of AAB, osteogenic tissues were bioprinted using MSC/HUVEC spheroids as building blocks in a scaffold-free configuration. After fabrication, two different osteogenic differentiation culture strategies were used, both with equal total exposure time of osteogenic induction. This was performed to decode the role of midterm osteogenic induction of stem cellbased spheroids (before bioprinting) on the mineralization and assembly behavior of generated tissues (fig. S18). In strategy no. 1, 2-day cultured spheroids were bioprinted into a triangle pattern (Fig. 6A). After bioprinting, triangle-shaped tissue complexes maintained their integrity and transformed into a more compact and dense structure after 3 days in proliferation culture conditions [growth media (GM)]. Spheroids in this triangle-shaped pattern self-assembled over time, where cells exhibited a viability of 83% after 5 days of incubation, and the assembled structure was further cultured for a period of 12 days in osteogenic media (OM) (Fig. 6, B and C). In general, MSCs cultured in OM express high levels of osteogenic markers (51). To confirm the osteogenic differentiation of bioprinted tissues, the early osteogenic differentiation marker, Runt-related transcription factor 2 (RUNX2), was used for immunohistochemical staining. At the same time, the presence of HUVECs was confirmed by CD31 staining. As shown in Fig. 6D, bioprinted tissues expressed osteogenic- and endotheliogenic-specific markers, as indicated by positive staining of RUNX2 and CD31. Calcium deposition of osteogenically differentiated tissues was also confirmed by Alizarin red staining. As shown in Fig. 6E, substantial calcium deposition was observed after 12 days of osteogenic induction, particularly at the core of the assembled tissue. As substantial contraction was observed after the fusion of MSC/HUVEC spheroids and the originally bioprinted triangular shape was not maintained (Fig. 6C) as well as the mineralization was not uniform throughout the tissue boundary (Fig. 6E), we also followed an alternative strategy (strategy no. 2) to preserve the bioprinted shape of osteogenic tissues (fig. S18). In this regard, MSC/HUVEC spheroids were first maintained in GM for 5 days, followed by inducting them with OM for 10 days. Next, spheroids were bioprinted and then kept for another 2 days in OM. In both strategies, the total exposure time to GM and OM was identical. At the end of culture (Fig. 6F), bioprinted tissues using strategy no. 2 exhibited strong expression of RUNX2 with more uniform distribution of mineralization demonstrated by Alizarin red staining, although RUNX2 staining was stronger in cores of spheroids (Fig. 6, G to J). Intensity analysis of RUNX2 revealed that RUNX2 staining was the strongest in the core of tissues or at the interface between spheroids and the weakest on the surface of the assembled tissues (Fig. 6, K and L, and fig. S19A), while CD31 staining was uniformly distributed throughout the histological sections of bioprinted tissues under both strategies (fig. S19B). A previous study demonstrated that HUVEC-mediated paracrine factors, including VEGF and the inflammatory mediator prostaglandin E2 (PGE2), promoted the osteogenesis of periodontal ligament stem cells under hypoxic conditions regulated by mitogen-activated protein kinase (MAPK) kinase/extracellular signalregulated kinase and p38 MAPK pathways (52). As the core of tissues and interface between spheroids were more hypoxic compared to the surface of assembled tissues, the osteogenic differentiation of MSCs could be further advanced in those regions by PGE2 and VEGF paracrine factors secreted by HUVECs. In addition, limited shape change was observed. This could be due to the fact that MSCs in MSC/HUVEC spheroids, cultured in OM for 10 days before bioprinting, were in the osteogenic differentiation pathway and their proliferation capability was diminished as reported in a previous study (53), and hence, limited contraction was observed in spheroids. Another advantage of strategy no. 2 is that the culture duration after bioprinting was reduced, where 2-day cultured tissues after bioprinting were still structurally stable. We also analyzed the expression levels of bone sialoprotein (BSP), type I collagen (COL1), alkaline phosphatase (ALP), RUNX2, and CDH2 (N-cadherin) genes for both strategies, and bioprinted tissues cultured in GM (labeled as 3D bioprinted tissue cultured with GM) and MSCs differentiated in 2D for 12 days (labeled as 2D MSCs cultured with OM) were used as control groups. As shown in Fig. 6M, expression levels of BSP, COL1, ALP, and RUNX2 genes for both strategies were similar to each other and significantly higher than those of control groups. In addition, the expression level of CDH2 gene (encoding N-cadherin protein) for all 3D bioprinted tissue groups were similar to each other and higher than that of the 2D control group. Overall, by changing the osteogenic induction window under the same total osteogenic induction duration, the shape of bioprinted tissues and uniformity of mineralization could be controlled, although no differences were detected in the expression levels of osteogenic genes.

Strategy no. 1: (A) Triangle-shaped tissue complexes were bioprinted using MSC/HUVEC spheroids and cultured for 3 days in GM and 12 days in OM. (B) Time-lapse images showing fusion of GFP+ spheroids up to day 15 (D15) after bioprinting. (C) An optical image showing the assembled tissue at day 15 after bioprinting. (D) Immunofluorescence staining (DAPI, CD31, F-actin, RUNX2, and DAPI + RUNX2) and (E) Alizarin red staining of the sectioned tissue. Strategy no. 2: (F) The final shape of the bioprinted tissue of osteogenic spheroids (cultured for 10 days in OM before bioprinting and 2 days in OM after bioprinting). Immunofluorescent images of (G) the bioprinted tissue and (H) confocal images of its histological sections stained for DAPI, CD31, and F-actin and (I) RUNX2 and DAPI + RUNX2. (J) Alizarin red staining of the tissue section. (K) Quantification of normalized RUNX2 intensity at different regions including the surface of assembled tissue, spheroid-spheroid interface, and core of spheroids (n = 50; **P < 0.01 and ***P < 0.001). (L) A representative heat map figure showing RUNX2/DAPI distribution in the surface of assembled tissue, spheroid-spheroid interface, and core of spheroids for strategy nos. 1 and 2. (M) BSP, COL1, ALP, RUNX2, and CDH2 gene expressions of 2D MSCs cultured in OM (control), 3D bioprinted tissues cultured in GM (control), and 3D bioprinted tissues cultured using strategy nos. 1 and 2 (n = 5; **P < 0.01 and ***P < 0.001).

Sodium alginate solution was made by dissolving 1% (w/v) sodium alginate (Sigma-Aldrich, UK) in deionized (DI) water. Calcium chloride solution was prepared by dissolving 4% (w/v) calcium chloride (CaCl2) (Sigma-Aldrich, St. Louis, MO) in DI water. For constructing fibrin scaffolds, fibrinogen (6 mg/ml) (Sigma-Aldrich) and thrombin (2.4 U/ml) (Sigma-Aldrich) were prepared for microvalve bioprinting. For bioprinting of electrocytes, agarose (A20070-100, Research Products International, IL) was dissolved in DI water to obtain a solution of 1% (w/v) agarose at 90C. COL I was extracted from rat tails according to a published protocol (54). GelMA was synthesized according to an established protocol (55). Detailed preparation methods of both materials can be found in the Supplementary Materials.

The mouse fibroblast cell line, 3T3, and the mouse mammary carcinoma line, 4T1, were obtained from the laboratory of A. Mastro, PSU (State College, PA). Culture media for 3T3 fibroblasts composed of Dulbeccos modified Eagles medium (DMEM) (Corning, Manassas, VA), supplemented with 10% fetal bovine serum (FBS) (Life Technologies, Grand Island, NY) and 1% penicillin-streptomycin (Corning). 3T3 cells were used at passages 22 through 27. 4T1 cells were grown in RPMI (Corning), 10% FBS, 1% penicillin-streptomycin. Passages 7 through 12 were used for 4T1 cells. HUVECs were purchased from Lonza and cultured in MCDB 131 base media (Corning) supplemented with 10% FBS (Corning), 1% glutamine (Gibco, Life Technologies), 0.5% bovine brain extract (Lonza, Walkersville, MD), heparin (10 U/ml) (Sigma-Aldrich), endothelial cell growth supplement (3 mg/ml) (Sigma-Aldrich), and 1% penicillin-streptomycin (Corning). HUVECs were used at passages 3 through 8. HUVECs were also transduced in house with EF1 tdTomato lentivector (Vectalys, Toulouse, France) to ease cell visualization for fluorescence microscopy according to the manufacturers instructions. GFP+ HUVECs were purchased from Angio-Proteomie (cAP-0001GFP; Boston, MA) and were used at passages 2 through 5. HDFs, obtained from N. Zavazavas laboratory at The University of Iowa (Iowa City, IA), were cultured in DMEM supplemented with 10% FBS (Corning), 1% glutamine (Gibco), 1% sodium pyruvate (Gibco), and 1% penicillin-streptomycin (Corning). HDFs were used at passages 7 through 12. GFP+ HDFs were purchased from Angio-Proteomie (cAP-0008-adGFP; Boston, MA) and were used at passages 2 through 6. MSCs were obtained from Lonza (Walkersville, MD) and RoosterBio (Frederick, MD) and cultured in SU-005 RoosterBasal-MSC (RoosterBio). Passages 4 through 8 were used for MSCs. GFP+ MSCs were purchased from Cyagen, cultured in SU-005 RoosterBasal-MSC (RoosterBio), and used at passages 2 through 6.

All cells were maintained at 37C in a 5% CO2 humidified atmosphere. Cell culture medium was changed every 2 to 3 days. Subconfluent cultures were detached from the flasks using a 0.25% trypsin0.1% EDTA solution (Life Technologies) and split to maintain cell growth.

Adherent cultures of each cell type were detached from the culture vessels with 0.25% trypsin-EDTA (Life Technologies, Grand Island, NY) solution. Trypsin was neutralized with appropriate GM, and cells were counted by a hemocytometer. Each cell type was then diluted to a concentration of 2500, 5000, and 10,000 cells in 200 l of appropriate GM. The cell suspension (200 l) was then pipetted into a single well of a U-bottom 96-well microplate with a cell-repellent surface (Greiner Bio-One, Monroe, NC). For fabrication of MSC/HUVEC spheroids made of 50,000 cells, MSCs and HUVECs were combined at a ratio of 92:8. The microplates were then incubated at 37C in a 5% CO2 humidified atmosphere. Spheroid formation was monitored daily on an EVOS FL cell imaging system (Life Technologies). For fabrication of GFP+ HDF/MSC coculture spheroids, 5000 cells were used in total, and GFP+ HDFs and MSCs were cocultured in ratios of 2:1 and 1:1 for 1 day. During the fabrication and culture of HUVECs spheroids, EGM-2MV medium (Lonza) was used.

Field-emission SEM (FEI Nova NanoSEM 630) was used to investigate the surface topography of fabricated spheroids and dissociated electrocytes (see the Supplementary Materials for dissociation of electrocytes). Spheroids were harvested after 2 days of culture. Spheroids and electrocytes were fixed in 4% paraformaldehyde (Sigma-Aldrich) overnight; samples were then carefully washed in phosphate-buffered saline (PBS) and dehydrated using graded ethanol solutions (25 to 100%). To ensure complete removal of water, samples were further dried in a critical point dryer (CPD300, Leica EM, Wetzlar, Germany). On complete dehydration, they were sputter-coated with iridium (Leica) and imaged at an accelerating voltage of 3 to 5 keV.

A total of 1000 2-day cultured spheroids of each type (with ~2500 cells per spheroid) were collected and homogenized in 100 l of distilled water. Then, 100 l of cell homogenate was transferred to a 2-ml pressure-tight vial, added with 100 l of 12 M hydrochloric acid, and hydrolyzed at 120C for 3 hours. The total collagen amount per spheroid type was quantified using a hydroxyproline colorimetric assay kit (BioVision Inc., CA) according to the manufacturers instructions. Collagen amount expressed by each spheroid type was determined using a PowerWave X-340 spectrophotometer (BioTek, Winooski, VT) at 560 nm, and the results were normalized to the collagen amount expressed by HUVEC spheroids at day 2. Experiments were repeated four times.

Bioprinting of spheroids was used with a MakerBot Replicator 1 3D printer (MakerBot, NY). The extrusion head was removed, and a holder for a pipette and two microvalve heads was 3D-printed using an Ultimaker 2 (Ultimaker) 3D printer. To control the 3D motion stage, a smoothie board (Uberclock, OR) was integrated. The reader is referred to the Supplementary Materials for the details on the construction of the AAB platform.

For fabrication of support constructs, microvalves (INKX0517500A, Lee Company, Bashville, TN) with 250-m nozzles (INZA3100914K, Lee Company) were integrated on the bioprinter head. To operate microvalves, a control board (IECX0501350A, Lee Company) was used, which was controlled using Arduino Uno (Arduino, Italy). To cross-link sodium alginate, a portable ultrasonic humidifier (CZHD20, Comfort Zone, China) was used to generate the aerosol form of CaCl2. Details about the bioprinter setupincluding the computer aided design model (fig. S1D), block diagram (fig. S1E), and computer interface (fig. S20)can be found in the Supplementary Materials.

Spheroids were collected into a petri dish from U-bottom 96-well plates, and then, their surface tension was measured using a micropipette aspiration technique, as explained in our recent work (8). Aspirated spheroids were monitored via an STC-MC33USB monochromatic camera (Sentech, Japan) equipped with 1-61448 and 1-61449 adaptor tubes (Navitar, Rochester, NY).

Spheroids fixed in 4% paraformaldehyde (Sigma-Aldrich) and rinsed in Dulbeccos PBS (DPBS) were stained with NucBlue ReadyProbes reagent (Life Technologies) to visualize the cell nuclei (Fig. 3, A3 and A4); actin cytoskeletal fibers were stained with ActinGreen 488 ReadyProbes reagent according to the manufacturers instructions. Imaging and 3D reconstruction of a heterogeneous pyramid structure (Fig. 4A) was performed using a Zeiss confocal microscope (LSM 880, Carl Zeiss AG, Oberkochen, Germany) using a 5 lens. The 3D reconstruction was obtained using Zen blue software (Carl Zeiss AG). A diamond structure (Fig. 4D) was first imaged through Zeiss Axiozoom (Carl Zeiss AG, Oberkochen, Germany) to capture the entire 3D structure and then further imaged on a confocal microscope (Olympus FV1000, Olympus America Inc., Center Valley, PA) to closely image the DAPI (4,6-diamidino-2-phenylindole), F-actin, and CD31 staining.

Spheroids were removed from the U-bottom well plates and subjected to the bioprinting process. Spheroids directly taken from plates were used as a positive control group. Viability was assessed using a LIVE/DEAD viability assay kit (Life Technologies, Grand Island, NY). Briefly, spheroids were washed twice in DPBS and then placed in the dye solution consisting of 1 M calcein acetoxymethyl and 1.6 M ethidium homodimer-1 in PBS. Live cells were able to take up and retain the calcein dye, resulting in bright green fluorescence of their cytoplasm. The ethidium homodimer could only enter dead cells where it binds to nucleic acids, producing a bright red fluorescence. Spheroids were imaged at 20-m z-stack step size on the Olympus FV1000 confocal microscope with a 60-m aperture setting resulting in approximately five to seven images per spheroid. Each image was then analyzed using ImageJ software (National Institutes of Health, USA).

Spheroids made of 3T3 cells were collected from U-bottom 96-well plates and then washed with DPBS twice. CellTracker Orange CMTMR [5-(and-6)-(((4-chloromethyl)benzoyl)amino) tetramethylrhodamine] (Thermo Fisher Scientific) and CellTracker Green CMFDA (5-chloromethylfluorescein diacetate) (Thermo Fisher Scientific, MA) were used according to the manufacturers instructions. Spheroids were bioprinted with close proximity, and then, images were captured at 0, 24, 48, and 72 hours using EVOS FL Cell Imaging System. The contact length and contact angle of two spheroids were measured every 4 hours up to 24 hours.

Images of sprouting GFP+ and tdTomato+ HUVEC spheroids at day 7 were taken on the Zeiss Axiozoom microscope at a magnification of 20 to capture the spheroids bioprinted at various distances. Images of sprouts at days 2 and 5 were taken using the EVOS FL Cell Imaging System. Sprouting behavior of spheroids was analyzed using AngioTool (41). To obtain the cross-sectional view of the capillary-like structures formed by GFP+ and tdTomato+ HUVEC spheroids at day 7, samples were imaged using a 40 oil lens on the Olympus FV1000 confocal microscope. For the study with GFP+ HDF/MSC coculture and HUVEC spheroids, samples were imaged on the EVOS microscope using fluorescent (4) channels at days 2 and 5. Images of those samples on day 7 were taken using fluorescent (16 and 40) and phase (16) channels by the Axiozoom microscope.

To induce osteogenic differentiation, MSC/HUVEC spheroids were bioprinted and cultured in two different strategies in a custom culture media made of 92% osteogenic differentiation media (Cell Applications Inc., San Diego, CA) and 8% HUVEC culture media. To confirm the morphology of bioprinted tissues undergoing osteogenic differentiation, tissues were sectioned and stained with RUNX2, CD31, F-actin, and DAPI. Cross-sectioned samples were washed three times with DPBS, fixed in 4% paraformaldehyde for 60 min, permeabilized in 0.2% Triton X-100 for 30 min, and blocked with 2.5% normal goat serum (NGS) for 60 min at room temperature. To visualize osteogenic and endothelial-specific genes, the samples were incubated with mouse anti-RUNX2 primary antibody (1:100 in 2.5% NGS) and rabbit anti-CD31 primary antibody (1:100 in 2.5% NGS) for 60 min; washed three times with DPBS; and incubated with goat anti-mouse Alexa Fluor 488 secondary antibody (Molecular Probes; 1:250 in 2.5% NGS) to label RUNX2, goat anti-rabbit Alexa Fluor 647 secondary antibody (Molecular Probes; 1:250 in 2.5% NGS) to label CD31, Alexa Fluor 568 phalloidin (Molecular Probes; 1:1000 in 2.5% NGS) to label filamentous actin, and DAPI (1:1000 in 2.5% NGS) to visualize cell nuclei for 60 min. The stained samples were washed three times with DPBS and imaged by an Olympus FV10i-LIV Confocal Laser Scanning Microscope (Olympus America Inc.) and analyzed using ImageJ software. In addition to sectioned samples, bioprinted tissues were also stained, as a whole-mount sample, as explained above, and fluorescent images were taken on the Zeiss Axiozoom microscope.

To confirm the calcium deposition, cross-sectioned samples were fixed in 4% paraformaldehyde, washed three times with distilled water, and incubated with 2% Alizarin red S stain solution for 30 min at room temperature. Stained samples were washed three times with distilled water and imaged using the EVOS microscope.

To quantify the intensity of RUNX2 and CD31, areas of interest on confocal images were selected using ImageJ. Fifty regions of interest were determined at the surface of the assembled tissue, spheroid-spheroid interface, and core of spheroids on fluorescent images. Each box was used for quantification of the fluorescence intensity. The intensity of RUNX2 and CD31 was normalized by the intensity of DAPI. A representative heat map was generated for the RUNX2/DAPI intensity.

Real-time polymerase chain reaction (PCR) was performed to quantify the gene expression levels of BSP, COL1, ALP, RUNX2, and CDH2. The primers of the measured mRNA genes were as follows: BSP (forward, AAC GAA GAA AGC GAA GCA GAA and reverse, TCT GCC TCT GTG CTG TTG), COL1 (forward, ATG ACT ATG AGT ATG GGG AAG CA and reverse, TGG GTC CCT CTG TTA CAC TTT), ALP (forward, AGC TGA ACA GGA ACA ACG TGA and reverse, CTT CAT GGT GCC CGT GGT C), RUNX2 (forward, GGT TAA TCT CCG CAG GTC ACT and reverse, CAC TGT GCT GAA GAG GCT GTT), CDH2 (forward, GAG CAG TGA GCC TGC AGA TTT T and reverse, TGC TCA GAA GAG AGT GGA AAG CT), and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) (forward, ATG GGG AAG GTG AAG GTC G and reverse, GGG GTC ATT GAT GGC AAC AAT A). Real-time PCR was analyzed by using SsoFast EvaGreen Supermix (Bio-Rad, USA), and all results were normalized using GAPDH.

All data were presented as means SD and analyzed by Minitab 17.3 (Minitab Inc., State College, PA, USA) using one-way analysis of variance (ANOVA) to test for significance when comparing the data. Post hoc Tukeys multiple-comparison test was used to determine the individual differences among the groups. Differences were considered significant at *P < 0.05, **P < 0.01, and ***P < 0.001. For directionality analysis, t test was used to compare the results between AOI and Other (where differences were considered significant at *P < 0.05, **P < 0.01, and ***P < 0.001), and ANOVA (with Tukeys multiple comparison test) was used to compare the results among different groups (where differences were considered significant at #P < 0.05, ##P < 0.01, and ###P < 0.001). Repeated-measures ANOVA (with Tukeys multiple-comparison test) was conducted to compare the results among different days of same groups, and differences were considered significant at *P < 0.05, **P < 0.01, and ***P < 0.001.

See original here:
Aspiration-assisted bioprinting for precise positioning of biologics - Science Advances

Molecular Signature of Young-Onset Parkinson’s Disease Is… : Neurology Today – LWW Journals

Article In Brief

A unique molecular structureevident in induced pluripotent stem cells taken from people with young-onset Parkinson's diseasesuggests that the defects may be present throughout patients' lives, and that they could therefore be used as diagnostic markers.

Induced pluripotent stem cells (iPSCs) taken from patients with young-onset Parkinson's disease (YOPD) and grown into dopamine-producing neurons displayed a molecular signature that was corrected in vitro, as well as in the mice striatum, by a drug already approved by the US Food and Drug Administration (FDA), a study published in the January 27 online edition of Nature Medicine found.

Although the patients had no known genetic mutations associated with PD, the neurons grown from their iPSCs nonetheless displayed abnormally high levels of soluble alpha-synucleina classic phenotype of the disease, but one never before seen in iPSCs from patients whose disease developed later in life. Surprisingly, for reasons not yet understood, the cells also had high levels of phosphorylated protein kinase C-alpha (PKC).

In addition, the cells also had another well-known hallmark of PD: abnormally low levels of lysosomal membrane proteins, such as LAMP1. Because lysosomes break down excess proteins like alpha-synuclein, their reduced levels in PD have long been regarded as a key pathogenic mechanism.

When the study team tested agents known to activate lysosomal function, they found that a drug previously approved by the FDA as an ointment for treating precancerous lesions, PEP005, corrected all the observed abnormalities in vitro: it reduced alpha-synuclein and PKC levels while increasing LAMP1 abundance. It also decreased alpha-synuclein production when delivered to the mouse striatum.

Unexpectedly, however, PEP005 did not work by activating lysosomal function; rather, it caused another key protein-clearing cellular structure, the proteasome, to break down alpha-synuclein more readily.

The findings suggest that the defects seen in the iPSCs are present throughout patients' lives, and that they could therefore be used as diagnostic markers. Moreover, the drug PEP005 should be considered a potentially promising therapeutic candidate for YOPD and perhaps even for the 90 percent of PD patients in whom the disease develops after the age of 50, according to the study's senior author, Clive Svendsen, PhD, director of the Cedars-Sinai Board of Governors Regenerative Medicine Institute and professor of biomedical sciences and medicine at Cedars-Sinai.

These findings suggest that one day we may be able to detect and take early action to prevent this disease in at-risk individuals, said study coauthor Michele Tagliati, MD, FAAN, director of the movement disorders program and professor of neurology at Cedars-Sinai Medical Center.

But the study still raises questions regarding the biological mechanisms, and certainly does not warrant off-label prescribing of PEP005 at this time, said Marco Baptista, PhD, vice president of research programs at the Michael J. Fox Foundation, who was not involved with the study.

Repurposing PEP005 is a long way away, Dr. Baptista said. This is not something that neurologists should be thinking about prescribing or recommending to their patients.

Accumulation of alpha-synuclein has been seen in iPSC-derived dopaminergic cultures taken from patients with known genetic defects, but such defects account for only about 10 percent of the PD population. In those without known mutations, on the other hand, no defects in iPSC-derived dopamine-producing neurons have been seen. Until now, however, such studies had been conducted only in patients who had developed PD after age 50.

My idea was why to look in young-onset patients, said Dr. Svendsen.

The idea paid off more richly than he expected. We were shocked to find a very, very prominent phenotype, a buildup of alpha-synuclein, in the neurons of these patients who are genetically normal, Dr. Svendsen said. None of the controls had a buildup of synuclein, and all but one of the early PD patients had a twofold increase in it.

The signature is so consistent, he said, that it offers a natural model that can be interrogated to further understand its workings.

Because high levels of PKC were also seen, Dr. Svendsen said, We picked a bunch of drugs known to reduce PKC. We found one, PEP005, which is actually extracted from the milkweed plant, and it completely reduced synuclein levels almost to normal in dopaminergic neurons. And it also increased dopamine levels in those cells, so we got two for one.

After observing the effects of PEP005 in vitro, We put it into the mouse brain and found it reduced synuclein in vivo, Dr. Svendsen said. But we had to infuse it right into the brain. We're now trying to work out how to get it across the blood-brain barrier more efficiently.

To determine how PEP005 lowers cellular levels of alpha-synuclein, his group tested whether it was activating the lysosome, but found to their surprise that it did not do this until after the synuclein had already been degraded.

Then we asked whether it could be the proteosome, which also breaks down proteins but normally doesn't break down synuclein, Dr. Svendsen said. But when we applied PEP005, it did activate the proteasome. So we think that might be the mechanism.

Because the drug is currently applied externally, Dr. Svendsen said, the next step will be to see if it crosses the blood-brain barrier when applied to the skin of mice, and whether that results in a lowering of synuclein levels in dopaminergic neurons.

Justin Ichida, PhD, the Richard N. Merkin assistant professor of stem cell biology and regenerative medicine at the USC Keck School of Medicine, said the findings are quite important in the field. The potential diagnostic tools they made could be important in clinical care. And identifying a drug that may very effectively reverse the disease in neurons is a very important discovery.

He wondered, however, whether the increase in alpha-synuclein is truly specific to Parkinson's neurons or if it would also be seen in iPSC neurons from patients with Alzheimer's disease or amyotrophic lateral sclerosis.

I wonder if alpha-synuclein accumulating is a sign of PD in a dish or is a consequence of neurodegeneration or impaired protein degradation in general, Dr. Ichida said. That's a key question if you want to use this molecular signature as a diagnostic tool.

He also questioned if proteins other than alpha-synuclein, such as tau, would also be seen to accumulate in the iPSCs of YOPD patients.

If one of the protein-clearance mechanisms in the cell is working poorly, you would imagine that other things would also accumulate, Dr. Ichida said.

In response, Dr. Svendsen said that while some proteins other than alpha-synuclein were reported in the paper at increased levels, We did not look at tau specifically, but are in the process of looking right now. It could be that synuclein and some other proteins are somehow altered to evade them from being degraded by the lysosome, or that there is a general lysosomal problem.

Patrik Brundin, MD, PhD, director of the Center for Neurodegenerative Science and Jay Van Andel Endowed Chair at Van Andel Research Institute in Grand Rapids, MI, called the paper very interesting and thought-provoking. If these findings hold up, they could shift our understanding of young-onset PD. They imply that there is a strong genetic component that has not been picked up in prior genetic studies.

Dr. Brundin said he would like to see the results replicated in another lab using different sets of reagents. It is so intriguing and rather unexpected that one wonders if the observations really apply, as the study states, to 95 percent of all YOPD.

He also questioned whether all the young-onset PD patients are similar. Clearly the iPSCs studied here are not monogenetic PD, so they must be very diverse genetically and still all have the same alpha-synuclein change.

Dr. Brundin also asked why the abnormalities seen in YOPD neurons have not previously been seen in older cases of PD. Is there a specific cutoff regarding age-of-onset when these purposed genetic differences apply? he asked.

Dr. Svendsen responded: We don't know why the YO have this phenotype or exactly what the cut off is. We have, however, looked at one adult-onset case that did not show this phenotype. Also, one of our YO cases did not show this phenotype. Thus some patients even with early onset may not have it. We are currently testing many more cases from older-onset patients.

Dr. Brundin also wanted to know whether non-dopaminergic neurons have the same deficits described in the study.

We don't know which neurons specifically have the protein deficit as we cannot do single-cell proteomics, Dr. Svendsen answered. It could be a little in all cells or a lot in a small set. Immunocytochemistry is not quantitative but showed that it is more likely a general increase in synuclein and not specific to dopaminergic neurons.

While the findings in iPSCs suggest that the abnormal levels of alpha-synuclein must be present at birth, Dr. Brundin said, I do not know how to reconcile the present findings with genetic data.

The absence of previously described mutations in the YOPD patients means only that more work must be done to uncover the genetic underpinnings, Dr. Svendsen said.

We're just at the tip of the iceberg with understanding the genome, he said. It's such a bizarrely complex beast. Perhaps there are a thousand different proteins interacting to stop the synuclein from being degraded. In 10 years, we probably will be clever enough to see it. We know it must be there. Now the genome guys will go after it.

Dr. Baptista from the Michael J. Fox Foundation said he agreed with the view that there must be genetic alterations underpinning the defects seen in the iPSCs.

Just because we call something non-genetic could simply reflect the current ignorance of the field, he said. I think the discoveries are simply difficult to make.

He added that he wished that the main comparator in the study was not healthy controls, and that there were more older-onset iPSCs to compare against YOPD patients' samples.

Dr. Svendsen said it could be that the iPSCs from older-onset patients might yet be found with additional study to display abnormalities similar to those seen in YOPD.

Right now we only see it in young onset, he said. We may need to leave the cultures longer to see in the older-onset patients. We are doing those experiments now.

Drs. Tagliati and Svendsen disclosed that an intellectual patent is pending for diagnostic and drug screening for molecular signatures of early-onset Parkinson's disease. Dr. Ikeda is a co-founder of AcuraStem Inc. Dr. Brundin has received commercial support as a consultant from Renovo Neural, Inc., Lundbeck A/S, AbbVie, Fujifilm-Cellular Dynamics International, Axial Biotherapeutics, and Living Cell Technologies. He has also received commercial support for research from Lundbeck A/S and Roche and has ownership interests in Acousort AB and Axial Biotherapeutics. Dr. Baptista had no disclosures.

See the original post:
Molecular Signature of Young-Onset Parkinson's Disease Is... : Neurology Today - LWW Journals

How industry hopes to take on COVID-19 – Bioprocess Insider – BioProcess Insider

The biopharma space has stepped up its efforts to both prevent and treat the coronavirus (SARS-CoV-2) that is threatening to bring the world to its knees.

A month is a very long time when it comes to infectious diseases. The first cases and deaths from the novel coronavirus (COVID-19) led to a response to contain the virus, but the difficulties of containment and the nature of international travel means cases and deaths have become global.

The latest statistics place the number of cases at 95,483 and deaths at 3,286 across 84 countries, though by the time you are reading this the number is likely to have skyrocketed.

So as the world tilters on the edge of a pandemic, we take a look at how industry is responding. There is no specific treatment for the virus, nor a vaccine, but a proactive response is seeing the pharma industry throw everything in its arsenal at attempting to stymie this global threat.

First off, vaccines. As the World Health Organization (WHO) states it can take a number of years for a new vaccine to be developed, it has not stopped companies and academia stepping up their R&D efforts.

Both Sanofi and J&J have separately teamed up with the US Department of Health and Human Services (HHS) to expediate vaccine development.

Sanofi Pasteur aims to reverse engineer proteins isolated from the virus to produce DNA sequences, which will then be mass produced using Sanofi Pasteurs baculoviral expression system and formulated into a vaccine that elicits an immune response. Well that is the aim.

Johnson & Johnsons unit Janssen Pharmaceutical, meanwhile, is reviewing products in development for Middle East Respiratory Syndrome (MERS) or Severe Acute Respiratory Syndrome (SARS), to identify promising candidates for the novel coronavirus, and aims to upscale production and manufacturing capacities, leveraging its AdVac and PER.C6 technologies.

Another Big Vaccine company, GlaxoSmithKline, has teamed with Chinese biotech Clover Biopharmaceuticals to help develop a preclinical protein-based vaccine candidate. GSK will provide its pandemic adjuvant system for further evaluation of Clovers S-Trimer, a trimeric SARS-CoV-2 spike (S)-protein subunit vaccine candidate produced using a mammalian cell-culture based expression system.

Inovio Pharmaceuticals has also entered the race, and like GSK has teamed up with a Chinese company. Together with Beijing Advaccine Biotechnology and a grant of up to $9 million from the Coalition for Epidemic Preparedness Innovations (CEPI), Inovio hopes to bring its DNA vaccine candidate INO-4800 rapidly into clinical trials. VGXI a subsidiary of GeneOne Life Science has been selected to manufacture the DNA vaccine from its facilities in The Woodlands, Texas.

Thegenome sequence for 2019-nCoVwas published on January 10, 2020, a VGXI spokesperson recently toldBioprocess Insider. This DNA sequence information is used by Inovio and their collaborators at the Wistar Institute to design a synthetic DNA plasmid for manufacturing at VGXI. No viral particles or proteins are involved in the manufacturing process. When delivered as a vaccine, the DNA plasmid can elicit a protective immune response.

RNA vaccines are also being investigated. Moderna Therapeutics recently shipped the first batch of its investigational messenger RNA vaccine mRNA-1273 to the National Institute of Allergy and Infectious Diseases (NIAID) for use in a Phase I study. The vaccine is designed to train the immune system to recognize cells invaded by the coronavirus.

Moderna also received a grant from CEPI, as has CureVac, which is looking to use its mRNA vaccine platform to expedite a candidate into trials. CureVacs technology and mRNA platform are especially suitable to rapidly provide a response to a viral outbreak situation like this, said CureVac CTO Mariola Fotin-Mleczek. Currently, we are in the process of developing a vaccine that, after successful preclinical tests, could be tested rapidly in humans in a clinical study.

But industry could be pipped to the clinical trial post by academia, with Israels MIGAL Research Institute claiming to be sitting on a human vaccine against COVID-19 as a by-product of a vaccine it has developed against avian coronavirus Infectious Bronchitis Virus (IBV).

From research conducted at MIGAL, it has been found that the poultry coronavirus has high genetic similarity to the human COVID-19, and that it uses the same infection mechanism, a fact that increases the likelihood of achieving an effective human vaccine in a very short period of time, the Institute says.

According to MIGALs Biotechnology group leader Chen Katz, the vaccine is based on a new protein expression vector, which forms and secretes a chimeric soluble protein that delivers the viral antigen into mucosal tissues by self-activated endocytosis a cellular process in which substances are brought into a cell by surrounding the material with cell membrane, forming a vesicle containing the ingested material causing the body to form antibodies against the virus.

Other pharma companies are looking to treat coronavirus, rather than prevent.

Regeneron has teamed with the HHS to use its VelociSuite technologies to identify and validation and develop preclinical candidates and bring them to development, having followed a similar approach to advance its investigational Ebola treatment REGN-EB3.

The tech platform includes the VelocImmune mouse technology, a genetically modified strain in which genes encoding mouse immune system proteins have been replaced by their human equivalents.

The life-saving results seen with our investigational Ebola therapy last year underscore the potential impact of Regenerons rapid response platform for addressing emerging outbreaks, said George Yancopoulos, Regeneron CSO. Our unique suite of technologies expedites and improves the drug discovery and development process at every stage, positioning Regeneron to respond quickly and effectively to new pathogens.

Meanwhile this week, Takeda announced it is looking to a therapy to target COVID-19 based on polyclonal hyperimmune globulin (H-IG). The candidate, TAK-888, aims to concentrate pathogen-specific antibodies from plasma collected from recovered patients. Initially, due to a lack of current donors, the firm will produce the therapy in a segregated area within its manufacturing facility in Georgia.

The Japan-headquartered firm will also review its current pipeline for any other viable candidates to take on COVID-19.

Such an approach has aided Gilead Sciences efforts. The firm has begun two Phase III clinical studies of its antiviral candidate remdesivir, developed (though never approved) to treat Ebola virus. It has also shown promise against other infectious diseases including Marburg, MERS and SARS.

This is an experimental medicine that has only been used in a small number of patients with COVID-19 to date, so Gilead does not have an appropriately robust understanding of the effect of this drug to warrant broad use at this time, Gilead said.

With about 1,000 patients set to be tested with remdesivir, Gilead has turned to a stockpile manufactured in response to Ebola to address present coronavirus needs, and in anticipation of expanded use is manufacturing two formulations of remdesivir, in both liquid and freeze-dried forms, while upping capacity and production internally and externally.

According to San Marinos Bioscience Institute SpA, a regenerative medicine center and stem cell production facility, mesenchymal stem cells could potentially be treatment for the novel coronavirus by improving lung microenvironment, inhibiting immune system overactivation, promoting tissue repair, protecting lung alveoli epithelial cells, preventing pulmonary fibrosis, and improving lung function.

The company, citing the Chinese open repository for scientific researchers chinaXiv.org , says at least 14 trials are taking place in China using stem cells to treat coronavirus patients after positive animal testing showed stem cells might be able to repair the severe organ damage caused by the virus.

The firm even reports that a critically ill 65-year-old Chinese woman infected with SARS-CoV-2, whose conditions significantly improved after the infusion of mesenchymal stem cells.

If mesenchymal stem cells do prove to be the solution to the potential coronavirus crisis, Bioscience Institute alludes to the advantage that they are obtained from fat cells.

That means that everyone can utilize his/her cells, eliminating any contamination or rejection risk, said Giuseppe Mucci, CEO of Bioscience Institute.

But expanding them to the quantity needed for infusion, that corresponds to at least 1 million cells per kg of weight, takes 2 to 3 weeks. That is why it is useful to cryopreserve a personal reserve of mesenchymal stem cells, that would allow to access an early, more successful, treatment.

Read the original post:
How industry hopes to take on COVID-19 - Bioprocess Insider - BioProcess Insider

Cell Therapy Market Size, Share & Trends Analysis Report By Use-type, By Therapy Type, By Region And Segment Forecasts, 2020 – 2027 – Yahoo…

Cell Therapy Market Size, Share & Trends Analysis Report By Use-type (Research, Commercialized, Musculoskeletal Disorders), By Therapy Type (Autologous, Allogeneic), By Region, And Segment Forecasts, 2020 - 2027

New York, March 05, 2020 (GLOBE NEWSWIRE) -- Reportlinker.com announces the release of the report "Cell Therapy Market Size, Share & Trends Analysis Report By Use-type, By Therapy Type, By Region And Segment Forecasts, 2020 - 2027" - https://www.reportlinker.com/p05868803/?utm_source=GNW

The global cell therapy market size is expected to reach USD 8.8 billion by 2027 at a CAGR of 5.4% over the forecast period. Cellular therapies hold a great therapeutic promise across various clinical applications. This has resulted in substantial global investments in research and clinical translation. Moreover, rapid advances in stem cell research hold the potential to fulfill the unmet demand of pharmaceutical entities, biotech entities, and doctors in disease management. These factors have boosted revenue growth for the market.

Currently, there are a limited number of FDA-approved commercial stem and non-stem cell therapies in the market.Furthermore, LAVIV (Azficel-T), manufactured and commercialized by Fibrocell Technologies, witnessed revenue wind-down in the past years.

Key developers are making substantial investments in the adoption of advanced technologies to address the aforementioned challenges.

The introduction of proprietary cell lines is recognized as the primary means by which a single cell can be exploited for the production of a robust portfolio of candidates. Companies are leveraging new technologies not only for the expansion of their product portfolio but also for establishing out-licensing or co-development agreements with other entities to support their product development programs.

For instance, MaxCyte has more than 40 high-value cellular therapy partnership programs within immune-oncology, regenerative medicine, and gene editing, including fifteen clinical-stage programs. Increase in the number of collaborations between entities for product commercialization is anticipated to accelerate market revenue to a major extent in the coming years.

In Asia Pacific, the market is anticipated to witness significant growth over the forecast period.This is attributed to rising awareness cellular therapies among patients and healthcare entities in chronic disease management.

In addition, availability of therapeutic treatment at lower prices is also driving the regional market. Japan is likely to witness fast growth over the forecast period attributed to increasing research activities on regenerative medicine.

Further key findings from the report suggest: The clinical-use segment accounted for low revenue share due to stringent regulations and non- commercial viability of some products However, the expanding knowledge over the commercial potential of cellular therapies is anticipated to result in the commercialization of a large number of products in the coming years On the contrary, the research-use segment accounted for the largest revenue share in 2019 owing to increase in research activities to explore the potential of the therapy in substantially improving disease management Furthermore, an increase in funding to explore the potential of these therapies has contributed to the large share of the research segment Allogenic therapies dominated the revenue share in 2019 owing to relatively lower relapse rates and growth in stem cell banking activities This is due to the high price and a large number of companies involved in the development of allogenic therapies Moreover, several companies are preparing to shift their business towards allogeneic therapy product development, resulting in significant revenue growth in this segment Autologous therapies are estimated to grow at the fastest pace during the forecast period Lack of donors and low affordability of allogeneic therapies are two key factors contributing to the increase in adoption of autologous therapies Considering the growing share of the cell therapy market in the biopharma industry, the companies are striving to gain a competitive advantage Vericel Corporation, JCR Pharmaceuticals Co. Ltd., MEDIPOST, and Osiris Therapeutics, Inc. are some key players operating in the market These companies are engaged in the expansion of their product portfolio, either through product development or acquisition of other players operating in the space.Read the full report: https://www.reportlinker.com/p05868803/?utm_source=GNW

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

__________________________

Story continues

Clare: clare@reportlinker.comUS: (339)-368-6001Intl: +1 339-368-6001

Original post:
Cell Therapy Market Size, Share & Trends Analysis Report By Use-type, By Therapy Type, By Region And Segment Forecasts, 2020 - 2027 - Yahoo...