How the Hippocampus Distinguishes True and False Memories

by Erica Moser

Image: iStock/metamorworks

Let’s say you typically eat eggs for breakfast but were running late and ate cereal. As you crunched on a spoonful of Raisin Bran, other contextual similarities remained: You ate at the same table, at the same time, preparing to go to the same job. When someone asks later what you had for breakfast, you incorrectly remember eating eggs.

This would be a real-world example of a false memory. But what happens in your brain before recalling eggs, compared to what would happen if you correctly recalled cereal?

In a paper published in Proceedings of the National Academy of Sciences, University of Pennsylvania neuroscientists show for the first time that electrical signals in the human hippocampus differ immediately before recollection of true and false memories. They also found that low-frequency activity in the hippocampus decreases as a function of contextual similarity between a falsely recalled word and the target word.

“Whereas prior studies established the role of the hippocampus in event memory, we did not know that electrical signals generated in this region would distinguish the imminent recall of true from false memories,” says psychology professor Michael Jacob Kahana, director of the Computational Memory Lab and the study’s senior author. He says this shows that the hippocampus stores information about an item with the context in which it was presented.

Researchers also found that, relative to correct recalls, the brain exhibited lower theta and high-frequency oscillations and higher alpha/beta oscillations ahead of false memories. The findings came from recording neural activity in epilepsy patients who were already undergoing invasive monitoring to pinpoint the source of their seizures.

Noa Herz, lead author and a postdoctoral fellow in Kahana’s lab at the time of the research, explains that the monitoring was done through intracranial electrodes, the methodology researchers wanted to use for this study. She says that, compared to scalp electrodes, this method “allowed us to more precisely, and directly, measure the neural signals that were generated in deep brain structures, so the activity we are getting is much more localized.”

Read the full story in Penn Today.

Michael Kahana is the Edmund J. and Louise W. Kahn Term Professor of Psychology in the School of Arts & Sciences and director of the Computational Memory Lab at the University of Pennsylvania. He is a member of the Penn Bioengineering Graduate Group.

Harnessing Artificial Intelligence for Real Biological Advances—Meet César de la Fuente

by Eric Horvath

In an era peppered by breathless discussions about artificial intelligence—pro and con—it makes sense to feel uncertain, or at least want to slow down and get a better grasp of where this is all headed. Trusting machines to do things typically reserved for humans is a little fantastical, historically reserved for science fiction rather than science. 

Not so much for César de la Fuente, PhD, the Presidential Assistant Professor in Psychiatry, Microbiology, Chemical and Biomolecular Engineering, and Bioengineering in Penn’s Perelman School of Medicine and School of Engineering and Applied Science. Driven by his transdisciplinary background, de la Fuente leads the Machine Biology Group at Penn: aimed at harnessing machines to drive biological and medical advances. 

A newly minted National Academy of Medicine Emerging Leaders in Health and Medicine (ELHM) Scholar, among earning a host of other awards and honors (over 60), de la Fuente can sound almost diplomatic when describing the intersection of humanity, machines and medicine where he has made his way—ensuring multiple functions work together in harmony. 

“Biology is complexity, right? You need chemistry, you need mathematics, physics and computer science, and principles and concepts from all these different areas, to try to begin to understand the complexity of biology,” he said. “That’s how I became a scientist.”

Read the full story in Penn Medicine News.

Center for Innovation & Precision Dentistry Positions Penn as a Leader in Engineering Health

by Devorah Fischler

Kathleen J. Stebe and Michel Koo urge “the academic community to adopt a coordinated approach uniting dental medicine and engineering to support research, training and entrepreneurship to address unmet needs and spur oral health care innovations.” (Image: Min Jun Oh and Seokyoung Yoon)

Penn’s Center for Innovation & Precision Dentistry (CiPD) is the first cross-disciplinary initiative in the nation to unite oral-craniofacial health sciences and engineering.

An institutional partnership formalizing the Center’s dual affiliation between the University of Pennsylvania School of Engineering and Applied Science and School of Dental Medicine makes CiPD unique.

In just two years since CiPD was founded, the outcomes of this newly conceived research partnership have proven its value: microrobots that clean teeth for people with limited mobility, a completely new understanding of bacterial physics in tooth decay, enzymes from plant chloroplasts that degrade plaque, promising futures for lipid nanoparticles in oral cancer treatment and new techniques and materials to restore nerves in facial reconstructive surgery.

In addition, CiPD is training the next generation of dentists, scientists and engineers through an NIH/NIDCR-sponsored postdoctoral training program as well as fellowships from industry.

The center’s Founding Co-Directors, Kathleen J. Stebe, Richer & Elizabeth Goodwin Professor in Chemical and Biomolecular Engineering, and Michel Koo, Professor of Orthodontics in Penn Dental Medicine, published an editorial in the Journal of Dental Research, planting a flag for CiPD’s mission and encouraging others to mirror its method.

The two urge “the academic community to adopt a coordinated approach uniting dental medicine and engineering to support research, training and entrepreneurship to address unmet needs and spur oral health care innovations.”

Read the full story in Penn Engineering Today.

Michel Koo is a member of the Penn Bioengineering Graduate Group.

The Future of Medicine Rises in University City: University of Pennsylvania Opens New Multi-Disciplinary Research Labs in One uCity Square

by Holly Wojcik

One uCity Square

On September 14, Wexford Science & Technology, LLC and the University of Pennsylvania announced that the University has signed a lease for new laboratory space that will usher in a wave of novel vaccine, therapeutics, and engineered diagnostics research to West Philadelphia. Research teams from Penn are poised to move into 115,000 square feet of space at One uCity Square, the 13-story, 400,000 square foot purpose-built lab and office building within the vibrant uCity Square Knowledge Community being developed by Wexford. This is the largest lease in the building, encompassing four floors, and bringing the building to over 90% leased. The building currently includes industry tenants Century Therapeutics (NASDAQ: IPSC), Integral Molecular, Exponent (NASDAQ: EXPO), and Charles River Laboratories (NYSE: CRL).

The new University space will house Penn Medicine’s Institute for RNA Innovation and Penn Engineering’s Center for Precision Engineering for Health, underscoring the University’s commitment to a multi-disciplinary and collaborative approach to research that will attract and retain the best talent and engage partners from across the region. Penn’s decision to locate at One uCity Square reinforces uCity Square’s evolution as a central cluster of academic, clinical, commercial, entrepreneurial, and amenity spaces for the area’s innovation ecosystem, and further cements Philadelphia’s position as a top life sciences market.

Jonathan Epstein, MD, Executive Vice Dean and Chief Scientific Officer of Penn Medicine, shared his anticipation for the opportunities that lie ahead: “Penn Medicine is proud to build on its existing clinical presence in uCity Square and establish an innovative and collaborative research presence at the heart of uCity Square’s multidisciplinary innovation ecosystem. This strategic move underscores our commitment to accelerating advancements in biomedical research, industry collaboration, and equipping our talented teams with the resources they need to shape the future of healthcare.”

Locating the Penn Institute for RNA Innovation in the heart of the uCity Square community brings together researchers across disciplines who are already pursuing new vaccines and treatments, and better ways to deliver them. Their shared work will help to power the next phase of vaccine discovery and development.

Likewise, anchoring the work of Penn Engineering’s Center in the One uCity Square space will allow the School’s multi-disciplinary researchers and their collaborators to advance new clinical and diagnostic methods that will focus on intelligent therapeutics, genome design, diagnostics for discovery of human biology, and engineering the human immune shield.

“Penn Engineering has made a substantial commitment to precision engineering for health, an area that is not only important and relevant to engineering, but also critical to the future of humanity,” said Vijay Kumar, Nemirovsky Family Dean of Penn Engineering. “The space in One uCity Square will add another 30,000 square feet of space for our engineers to develop technologies that will fight future pandemics, cure incurable diseases, and extend healthy life spans around the world.”

Spearheading the Penn Institute for RNA Innovation will be Drew Weissman, MD, PhD, the Roberts Family Professor for Vaccine Research, who along with Katalin Karikó, PhD, adjunct professor of Neurosurgery, discovered foundational mRNA technology that enabled the creation of vital vaccine technology, including the FDA-approved mRNA-based COVID-19 vaccines developed by Pfizer-BioNTech and Moderna.

In this new space at One uCity Square, Weissman and his research team and collaborators will further pursue their groundbreaking research efforts with a goal to develop new therapeutics and vaccines and initiate clinical trials for other devastating diseases.

In addition, two established researchers will join the Institute at One uCity Square: Harvey Friedman, MD, a professor of Infectious Diseases, who leads a team researching various vaccines. He will be joined by Vladimir Muzykantov, MD, PhD, Founders Professor in Nanoparticle Research, who focuses on several projects related to targeting the delivery of drugs, including mRNA, to create more effective, targeted pathways to deliver drugs to the vascular system, treating a wide range of diseases that impact the brain, lung, heart, and blood.

Dan Hammer, Alfred G. and Meta A. Ennis Professor in the Departments of Bioengineering and Chemical and Biomolecular Engineering in Penn Engineering and Director of the Center for Precision Engineering for Health, will oversee the Center’s innovations in diagnostics and delivery, cellular and tissue engineering, and the development of new devices that integrate novel materials with human tissues. The Center will bring together scholars from all departments within Penn Engineering and will help to foster increased collaboration with campus colleagues at Penn’s Perelman School of Medicine and with industry partners.

Joining the Center researchers in One uCity Square are Noor Momin, Sherry Gao, and Michael Mitchell. Noor Momin, who will join Penn Engineering in early 2024 as an assistant professor in Bioengineering, will leverage her lab’s expertise in cardiovascular immunology, protein engineering and pharmacokinetic modeling to develop next-generation treatments and diagnostics for cardiovascular diseases.

Read the full story in Penn Engineering Today.

Jonathan Epstein and Vladimir Muzykantov are members of the Penn Bioengineering Graduate Group.

Michael Mitchell is an Associate Professor in Bioengineering.

Bioengineering Faculty Member Named ‘Young Innovator’ for Creation of Multiple Myeloma Therapy

by Abbey Porter

Michael Mitchell

Michael J. Mitchell, Associate Professor in Bioengineering at the University of Pennsylvania School of Engineering and Applied Science, has been named a “Young Innovator of Cellular and Molecular Bioengineering” by Cellular and Molecular Bioengineering, the journal of the Biomedical Engineering Society (BMES).

The award recognizes faculty who are conducting some of the most innovative and impactful studies in the field of biomedical engineering. Recipients will present their research and be officially recognized at the BMES Annual Meeting in October.

Mitchell is being honored for creating an RNA nanoparticle therapy that stops the spread of the deadly bone marrow cancer multiple myeloma and helps to eliminate it altogether. Known for being difficult to treat, the disease kills over 100,000 people every year.

“We urgently need innovative, effective therapies against this cancer,” Mitchell says. “The nanotechnology we developed can potentially serve as a platform to treat multiple myeloma and other bone marrow-based malignancies.”

Mitchell, along with Christian Figuerora-Espada, a doctoral student in Bioengineering, previously published a study in PNAS describing how their RNA nanoparticle therapy stops multiple myeloma from moving through the blood vessels and mutating. In their current paper in Cellular and Molecular Bioengineering, which expands upon this RNA nanoparticle platform, they show that inhibition of both multiple myeloma migration and adhesion to bone marrow blood vessels, combined with an FDA-approved multiple myeloma therapeutic, extends survival in a mouse model of multiple myeloma.

Read more in Penn Engineering Today.

Carl June to Receive 2024 Breakthrough Prize in Life Sciences

by Meagan Raeke

Image: Courtesy of Penn Medicine

CAR T cell therapy pioneer Carl June, the Richard W. Vague Professor in Immunotherapy in the Perelman School of Medicine and director of the Center for Cellular Immunotherapies (CCI) at Penn Medicine’s Abramson Cancer Center, has been named a winner of the 2024 Breakthrough Prize in Life Sciences for the development of chimeric antigen receptor (CAR) T cell immunotherapy, a revolutionary cancer treatment approach in which each patient’s T cells are modified to target and kill their cancer cells. The invention sparked a new path in cancer care, harnessing the power of patients’ own immune systems, a once-elusive goal that brought fresh options for those who could not be successfully treated with conventional approaches.

Founded in 2012, the Breakthrough Prizes are the world’s largest science awards, with $3 million awarded for each of the five main prize categories. June is the sixth Breakthrough Prize laureate from Penn, which joins Harvard and MIT among the institutions whose researchers have been honored with the most Breakthrough Prizes.

“This award is not only a testament to Dr. June’s outstanding contributions to science, but also a shining example of the caliber of discoveries and research which Penn faculty set their sights upon,” said Penn President Liz Magill. “We are immensely proud to have Dr. June as a member of the Penn academic community, and we know that CAR T cell therapy is just the first chapter in an inspiring and lifesaving new era of medicine.”

June is internationally recognized for his role in pioneering the CAR T cell therapy, which led to the first FDA-approved personalized cellular therapy, for children and young adults with the blood cancer known as acute lymphoblastic leukemia, in August of 2017—a step which has spurred five additional approvals of the technique in other blood cancers. June joined Penn in 1999, building momentum for Penn to become a global hub for cell and gene therapy. Gene-modified T cells engineered in June’s lab to retrain a patient’s own immune cells to attack cancer were used in the first clinical trial of CAR T cell therapy in 2010. Some of the earliest children and adults treated have experienced long-lasting remissions of 10 years or more. In addition to the FDA approvals that have made the therapy commercially available to patients across the world, thousands more have benefited from clinical trials testing these transformative treatments, including for the treatment of solid tumors and even autoimmune diseases like lupus.

“Dr. June’s tireless commitment to advancing T cell immunotherapy research has been life-changing for many patients affected by cancer, who have lived longer, fuller lives, thanks to the discoveries made in his lab,” said J. Larry Jameson,executive vice president of the University of Pennsylvania for the Health System and dean of the Perelman School of Medicine. “We are proud to see one of Penn’s most esteemed scientists recognized for the impact of his foundational work to develop a new class of cancer immunotherapy treatment.”

Read the full story in Penn Today.

June is a member of the Penn Bioengineering Graduate Group. Read more stories featuring June on the BE Blog here.

A Suit of Armor for Cancer-fighting Cells

by Nathi Magubane

Chimeric antigen receptor T cell (CAR T) therapy has delivered promising results, transforming the fight against various forms of cancer, but for many, the therapy comes with severe and potentially lethal side effects. Now, a research team led by Michael Mitchell of the School of Engineering and Applied Science has found a solution that could help CAR T therapies reach their full potential while minimizing severe side effects. (Image: iStock / Meletios Verras)

In recent years, cancer researchers have hailed the arrival of chimeric antigen receptor T cell (CAR T) therapy, which has delivered promising results, transforming the fight against various forms of cancer. The process involves modifying patients’ T-cells to target cancer cells, resulting in remarkable success rates for previously intractable forms of cancer.

Six CAR T cell therapies have secured FDA approval, and several more are in the pipeline. However, these therapies come with severe and potentially lethal side effects, namely cytokine release syndrome (CRS) and neurotoxicity. These drawbacks manifest as a range of symptoms—from high fever and vomiting to multiple organ failure and patient death—posing significant challenges to broader clinical application.

Now, a research team led by Michael Mitchell, associate professor in the School of Engineering and Applied Science at the University of Pennsylvania, has found a solution that could help CAR T therapies reach their full potential while minimizing severe side effects. Their findings are published in the journal Nature Materials.

“Addressing CRS and neurotoxicity without compromising the therapeutic effectiveness of CAR T cells has been a complex challenge,” says Mitchell.

He says that unwanted interactions between CAR T and immune cells called macrophages drive the overactivation of macrophages, which in turn result in the release of toxic cytokines that lead to CRS and neurotoxicity.

“Controlling CAR T-macrophage interactions in vivo is difficult,” Mitchell says. “So, our study introduces a materials engineering-based strategy that involves incorporating a sugar molecule onto the surface of CAR T cells. These sugars are then used as a reactive handle to create a biomaterial coating around these cells directly in the body, which acts as a ‘suit of armor,’ preventing dangerous interactions with macrophages.”

First author Ningqiang Gong, a postdoctoral researcher in the Mitchell Lab, elaborates on the technique, “We attached this sugar molecule to the CAR T cells using metabolic labeling. This modification enables the CAR T cells to attack cancer cells without any hindrance.”

“When symptoms of CRS begin to manifest, we introduce another molecule—polyethylene glycol (PEG)—to create the suit of armor, which effectively blocks dangerous interactions between these engineered T cells, macrophages, and the tumor cells themselves,” Gong says.

Read the full story in Penn Today.

Carl June on the Boundless Potential of CAR T Cell Therapy

by Meagan Raeke

Carl June, at the flash mob celebration of the FDA approval of the CAR T cell therapy he developed, in August 2017. (Image: Courtesy of Penn Medicine Magazine)

For most of modern medicine, cancer drugs have been developed the same way: by designing molecules to treat diseased cells. With the advent of immunotherapy, that changed. For the first time, scientists engineered patients’ own immune systems to recognize and attack diseased cells.

One of the best examples of this pioneering type of medicine is CAR T cell therapy. Invented in the Perelman School of Medicine by Carl June, the Richard W. Vague Professor in Immunotherapy, CAR T cell therapy works by collecting T cells from a patient, modifying those cells in the lab so that they are designed to destroy cancerous cells, and reinfusing them into the patient. June’s research led to the first FDA approval for this type of therapy, in 2017. Six different CAR T cell therapies are now approved to treat various types of blood cancers. Carl June, at the flash mob celebration of the FDA approval of the CAR T cell therapy he developed, in August 2017. (Image: Courtesy of Penn Medicine Magazine)

CAR T cell therapy holds the potential to help millions more patients—if it can be successfully translated to other conditions. June and colleagues, including Daniel Baker, a fourth-year doctoral student in the Cell and Molecular Biology department, discuss this potential in a perspective published in Nature.

In the piece, June and Baker highlight other diseases that CAR T cell therapy could be effective.

“CAR T cell therapy has been remarkably successful for blood cancers like leukemias and lymphomas. There’s a lot of work happening here at Penn and elsewhere to push it to other blood cancers and to earlier stage disease, so patients don’t have to go through chemo first,” June says. “Another big priority is patients with solid tumors because they make up the vast majority of cancer patients. Beyond cancer, we’re seeing early signs that CAR T cell therapy could work in autoimmune diseases, like lupus.”

As for which diseases to pursue as for possible future treatment, June says, “essentially it boils down to two questions: Can we identify a population of cells that are bad? And can we target them specifically? Whether that’s asthma or chronic diseases or lupus, if you can find a bad population of cells and get rid of them, then CAR T cells could be therapeutic in that context.”

“What’s exciting is it’s not just theoretical at this point. There have been clinical reports in other autoimmune diseases, including myasthenia gravis and inflammatory myopathy,” Baker says. “But we are seeing early evidence that CAR T cell therapy will be successful beyond cancer. And it’s really opening the minds of people in the field to think about how else we could use CAR T. For example, there’s some pioneering work at Penn from the Epstein lab for heart failure. The idea is that you could use CAR T cells to get rid of fibrotic tissue after a cardiac injury, and potentially restore the damage following a heart attack.”

Baker adds, “there’s no question that over the last decade, CAR T cell therapy has revolutionized cancer. I’m hoping to play a role in bringing these next generation therapies to patients and make a real impact over the next decade. I think there’s potential for cell therapy to be a new pillar of medicine at large, and not just a new pillar of oncology.”

Read the full story at Penn Medicine Today.

An Improved Delivery System for mRNA Vaccines Provides More Powerful Protection

by Devorah Fischler

(From left to right) Xuexiang Han, Michael Mitchell and Mohamad-Gabriel Alameh

The COVID-19 vaccine swiftly undercut the worst of the pandemic for hundreds of millions around the world. Available sooner than almost anyone expected, these vaccines were a triumph of resourcefulness and skill.

Messenger RNA vaccines, like the ones manufactured by Moderna or Pfizer/BioNTech, owed their speed and success to decades of research reinforcing the safety and effectiveness of their unique immune-instructive technology.

Now, researchers from the University of Pennsylvania School of Engineering and Applied Science and the Perelman School of Medicine are refining the COVID-19 vaccine, creating an innovative delivery system for even more robust protection against the virus.

In addition to outlining a more flexible and effective COVID-19 vaccine, this work has potential to increase the scope of mRNA vaccines writ large, contributing to prevention and treatment for a range of different illnesses.

Michael Mitchell, associate professor in Penn Engineering’s Department of Bioengineering, Xuexiang Han, postdoctoral fellow in Mitchell’s lab, and Mohamad-Gabriel Alameh, postdoctoral fellow in Drew Weissman’s lab at Penn Medicine and incoming assistant professor in the Department of Pathology and Laboratory Medicine at the Perelman School of Medicine, recently published their findings in Nature Nanotechnology.

mRNA, or messenger ribonucleic acid, is the body’s natural go-between. mRNA contains the instructions our cells need to produce proteins that play important roles in our bodies’ health, including mounting immune responses.

The COVID-19 vaccines follow suit, sending a single strand of RNA to teach our cells how to recognize and fight the virus.

Read the full story in Penn Engineering Today.

The Physics of Fat Droplets Reveal DNA Danger

by Devorah Fischler

Fat is a normal and necessary part of the body. Fat cells store and release energy, as well as play significant roles in hormonal regulation and immunity.

Engineers and scientists at the University of Pennsylvania are the first to discover fat-filled lipid droplets’ (pictured above in green) surprising capability to indent and puncture the nucleus, the organelle which contains and regulates a cell’s DNA.

In recent decades, a concerning rise in metabolic illnesses – such as cardiovascular disease, high blood pressure and diabetes – has focused scientific attention on the biology and chemistry of fat, resulting in a wealth of information about how fat cells work.

But fat cells and their metabolic activities are only part of the story.

Fat-filled lipid droplets, tiny spheres of fat many times smaller than fat cells, are a growing subject of scientific interest. Found inside many different cell types, these lipid particles have long been little understood. Studies have begun to illuminate these droplets’ participation in metabolic functions and cellular protection, but we still know next to nothing about the physical nature of fat.

Now, researchers at the University of Pennsylvania School of Engineering and Applied Science have looked beyond biochemistry to publish groundbreaking work on the physics of these droplets, revealing them to be a potential threat to a cell’s nucleus. In the August issue of the Journal of Cell Biology, they are the first to discover fat-filled lipid droplets’ surprising capability to indent and puncture the nucleus, the organelle which contains and regulates a cell’s DNA.

The stakes of their findings are high: a ruptured nucleus can lead to elevated DNA damage that is characteristic of many diseases, including cancer.

The study was led by Dennis E. Discher, Robert D. Bent Professor in the Department of Chemical and Biomolecular Engineering, Bioengineering, and in Mechanical Engineering and Applied Mechanics, Irena Ivanovska, Ph.D. research associate in Penn’s Molecular and Cell Biophysics Lab, and Michael Tobin, Ph.D. candidate in the Department of Bioengineering.

“Intuitively, people think of fat as soft,” says Discher. “And on a cellular level it is. But at this small size of droplet— measuring just a few microns rather than the hundreds of microns of a mature fat cell—it stops being soft. Its shape has a much higher curvature, bending other objects very sharply. This changes its physics in the cell. It can deform. It can damage. It can rupture.”

Read the full story in Penn Engineering Today.