Parasite’s riff on essential enzyme highlights unique biology
Nicole Giese Rura | Whitehead Institute
September 18, 2018

Cambridge, Mass. — The primary currency of energy in cells—adenosine triphosphate (ATP)—is essential for their survival and without it, cellular processes would seize. In the apicomplexan Toxoplasma gondii (T. gondii), a parasite that Whitehead Member Sebastian Lourido studies, key components of the ATP synthase—the enzyme responsible for ATP production—have remained elusive. While investigating indispensable proteins with unknown functions, Lourido and Diego Huet, a postdoctoral researcher in Lourido’s lab, identified a critical component of the enzyme. While highly conserved from yeast to humans, it proved to be considerably different in T. gondii. The findings, published online September 11 in the journal eLife, underscore the unique biology of these parasites and highlight differences between them and their human hosts.

More closely related to plants than to animals, the single-celled apicomplexans are among the most common and deadly human pathogens. According to the World Health Organization, every year these diseases sicken hundreds of millions, kill hundreds of thousands—primarily children—and cost billions of dollars to treat. Species of apicomplexans cause malaria (Plasmodium spp.), cryptosporidiosis (Cryptosporidium spp.), and toxoplasmosis (T. gondii).

Using a CRISPR-based genetic screen that they had adapted to T. gondii, Lourido and Huet had previously identified about 200 genes in T. gondii that are fitness-conferring and specific to apicomplexans. Of that cadre, a few were localized to the mitochondria, where cells manufacture ATP, the cellular currency of energy. Because those genes have not been annotated previously, and the proteins encoded by them have no known function, Huet ran their protein sequences through a database that compared them to protein sequences with known structures.

One of the proteins came back with an interesting hit: it shares structural similarity, but not sequence similarity, with an integral part of the ATP synthase. Most of the protein subunits that compose the apicomplexan ATP synthase have been identified, but key components of the stator—a portion of the enzyme essential for its function—was not yet known.

When Huet experimentally removed the function of the stator subunit in T. gondii, the parasites’ growth stalled, their mitochondria were misshapen and shrunken, and energy production halted—all traits typical of interrupted ATP synthase function.

Because the apicomplexan ATP synthase varies so much from its hosts’ version, those differences, like the unusual stator, could serve as future drug targets. But for Lourido, who is also an assistant professor of biology at Massachusetts Institute of Technology (MIT), the unique stator protein emphasizes how unique and extraordinary apicomplexan organisms are compared to us and their other hosts.

This work was supported by the National Institutes of Health (NIH grants 1DP5OD017892, R21AI123746, and K99AI137218).

* * *

Sebastian Lourido’s primary affiliation is with Whitehead Institute for Biomedical Research, where his laboratory is located and all his research is conducted. He is also an assistant professor of biology at Massachusetts Institute of Technology.

* * *

Full Citation:

“Identification of cryptic subunits from an apicomplexan ATP synthase”

eLife, online September 11, 2018.  DOI: 10.7554/eLife.38097

Diego Huet (1) , Esther Rajendran (2) , Giel G van Dooren (2) , Sebastian Lourido (1,3*).

1. Whitehead Institute for Biomedical Research, Cambridge, United States

2. Research School of Biology, Australian National University, Canberra, Australia

3. Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States

Detangling DNA replication

Researchers identify an essential protein that helps enzymes relax overtwisted DNA so each strand can be copied during cell division.

Raleigh McElvery | Department of Biology
September 18, 2018

DNA is a lengthy molecule — approximately 1,000-fold longer than the cell in which it resides — so it can’t be jammed in haphazardly. Rather, it must be neatly organized so proteins involved in critical processes can access the information contained in its nucleotide bases. Think of the double helix like a pair of shoe laces twisted together, coiled upon themselves again and again to make the molecule even more compact.

However, when it comes time for cell division, this supercoiled nature makes it difficult for proteins involved in DNA replication to access the strands, separate them, and copy them so one DNA molecule can become two.

Replication begins at specific regions of the chromosome where specialized proteins separate the two strands, pulling apart the double helix as you would the two shoe laces. However, this local separation actually tangles the rest of the molecule further, and without intervention creates a buildup of tension, stalling replication. Enter the enzymes known as topoisomerases, which travel ahead of the strands as they are being peeled apart, snipping them, untwisting them, and then rejoining them to relieve the tension that arises from supercoiling.

These topoisomerases are generally thought to be sufficient to allow replication to proceed. However, a team of researchers from MIT and the Duke University School of Medicine suggests the enzymes may require guidance from additional proteins, which recognize the shape characteristic of overtwisted DNA.

“We’ve known for a long time that topoisomerases are necessary for replication, but it’s never been clear if they were sufficient on their own,” says Michael Laub, an MIT professor of biology, Howard Hughes Medical Institute Investigator, and senior author of the study. “This is the first paper to identify a protein in bacteria, or eukaryotes, that is required to localize topoisomerases ahead of replication forks and to help them do what they need to do there.”

Postdoc Monica Guo ’07 and former graduate student Diane Haakonsen PhD ’16 are co-first authors of the study, which appeared online in the journal Cell on Sept. 13.

netbet sports betting

Although it’s well established that topoisomerases are crucial to DNA replication, it has now becoming clear that we know relatively little about the mechanisms regulating their activity, including where and when they act to relieve supercoiling.

These enzymes fall into two groups, type I and type II, depending on how many strands of DNA they cut. The researchers focused on type II topoisomerases found in a common species of freshwater bacteria, Caulobacter crescentus. Type II topoisomerases in bacteria are of particular interest because a number of antibiotics target them in order to prevent DNA replication, treating a wide variety of microbial infections, including tuberculosis. Without topoisomerases, the bacteria can’t grow. Since these bacterial enzymes are unique, poisons directed at them won’t harm human topoisomerases.

For a long time, type II topoisomerases were generally assumed adequate on their own to manage the overtwisted supercoils that arise during replication. Although researchers working in E. coli and other, higher organisms have pinpointed additional proteins that can activate or repress these enzymes, none of these proteins were required for replication.

Such findings hinted that there might be similar interactions taking place in other organisms. In order to understand the protein factors involved in compacting Caulobacter DNA — regulating topoisomerase activity specifically — the researchers screened their bacteria for proteins that bound tightly to supercoiled DNA. From there, they honed in on one protein, GapR, which they observed was essential for DNA replication. In bacteria missing GapR, the DNA became overtwisted, replication slowed, and the bacteria eventually died.

Surprisingly, the researchers found that GapR recognized the structure of overtwisted DNA rather than specific nucleotide sequences.

“The vast majority of DNA-binding proteins localize to specific locations of the genome by recognizing a specific set of bases,” Laub says. “But GapR basically pays no attention to the actual underlying sequence — just the shape of overtwisted DNA, which uniquely arises in front of replication forks and transcription machinery.”

The crystal structure of the protein bound to DNA, solved by Duke’s Maria Schumacher, revealed that GapR recognizes the backbone of DNA (rather than the bases), forming a snug clamp that encircles the overtwisted DNA. However, when the DNA is relaxed in its standard form, it no longer fits inside the clamp. This might signify that GapR sits on DNA only at positions where topoisomerase is needed.

An exciting milestone

Although GapR appears to be required for DNA replication, it’s still not clear precisely how this protein promotes topoisomerase function to relieve supercoiling.

“In the absence of any other proteins, GapR is able to help type II topoisomerases remove positive supercoils faster, but we still don’t quite know how,” Guo says. “One idea is that GapR interacts with topoisomerases, recognizing the overtwisted DNA and recruiting the topoisomerases. Another possibility is that GapR netbet sports betting appis essentially grabbing onto the DNA and limiting the movement of the positive supercoils, so topoisomerases can target and eliminate them more quickly.”

Anthony Maxwell, a professor of biological chemistry at the John Innes Centre who was not involved with the study, says the buildup of DNA supercoils is a key problem in both bacterial replication and transcription.

“Identifying GapR and its potential role in controlling supercoiling in vivo is an exciting milestone in understanding the control of DNA topology in bacteria,” he says. “Further work will be required to show how exactly these proteins cooperate to maintain bacterial genomic integrity.”

According to Guo, the study provides insight into a fundamental process — DNA replication — and the ways topoisomerases are regulated, which could extend to eukaryotes.

“This was the first demonstration that a topoisomerase activator is required for DNA replication,” she says. “Although there’s no GapR homolog in higher organisms, there could be similar proteins that recognize the shape of the DNA and aid or position topoisomerases.”

This could open up a new field of drug research, she says, targeting activators like GapR to increase the efficacy of existing topoisomerase poisons to treat conditions like respiratory and urinary tract infections. After all, many topoisomerase inhibitors have become less effective due to antibiotic resistance. But only time will tell; there is still much to learn in order to untangle the complex process of DNA replication, along with its many twists and turns.

The research was funded by NIH grants, the HHMI International Predoctoral Fellowship, and the Jane Coffin Childs Memorial Fellowship.

New perspectives at the bench

Three MIT postdocs earn competitive Howard Hughes Medical Institute fellowships that support diversity in the sciences.

David Orenstein | Erika Reinfeld | Lisa Girard | Picower Institute for Learning and Memory
September 12, 2018

In recognition of their exceptional potential to be leaders in the life sciences, three MIT postdocs at the Koch, Picower, and Whitehead institutes at MIT are among 15 young researchers to earn Hanna Gray Fellowships from the Howard Hughes Medical Institute (HHMI), the Chevy Chase, Maryland-based organization has announced.

The early-career awards are given to individuals around the U.S. who are from racial, gender, ethnic, and other groups underrepresented in the life sciences. The fellowships will support their work with up to $1.4 million in funding over eight years, enough to last well into a tenure track position after they complete their postdoctoral studies.

“This program will help us retain the most diverse talent in science,” HHMI President Erin O’Shea said in the announcement. “We feel it’s critically important in academia to have exceptional people from all walks of life, all cultures, and all backgrounds — people who can inspire the next generation of scientists.”

MIT’s three Hanna Gray Fellows are Matheus Victor, who works in the lab of Li-Huei Tsai in the Picower Institute for Learning and Memory; Quinton Smith, who works in Sangeeta Bhatia’s lab at the Koch Institute for Integrative Cancer Research; and Jarrett Smith, who works in the David Bartel Lab at the Whitehead Institute for Biomedical Research.

Jarrett Smith

Jarrett Smith was always interested in science, but no one in his family had ever received a PhD, making biology research feel like an unlikely career path for him. Nevertheless, he followed his passion, which led him to his PhD program at the Johns Hopkins University School of Medicine. Despite his strong academic performance, Smith began graduate school with doubts about his ability to become a scientist. His mentors were incredible teachers, he says, but their self-assuredness could be intimidating.

“They were absolutely my role models, but I didn’t think of them as having gone through what I was going through. In the first few years, I felt like I had a lot of catching up to do,” Smith says.

Now, as a postdoc in David Bartel’s lab at the Whitehead Institute, he studies how cells respond to stress. When a cell is exposed to environmental stressors such as heat, UV radiation, or viral infection, proteins and RNAs in the cell may clump together into dense aggregates called stress granules. The exact function of stress granules and their potential role in disease are unknown, so Smith is investigating changes in the cell linked to their formation. His findings could shed light on a potential role for stress granules in cancer, infection, and neurodegenerative disease.

“I’m grateful for the support that the fellowship will provide during the formative years of my career,” Smith says. “This kind of opportunity gives you the confidence to set ambitious research goals and find out what you can accomplish.”

Quinton Smith

Quinton Smith fell in love with regenerative medicine the summer after his sophomore year of college. Working on an epidemiological study at the University of New Mexico Cancer Research Facility in his hometown of Albuquerque, Smith found himself extracting DNA from hundreds of mouthwash samples in order to answer overarching questions about the biological and behavioral factors that contribute to cancer.

The experience left him with a strong appreciation for the complexities of human disease, and an even stronger desire to translate his knowledge into better interventions for patients. To this end, he completed his PhD at Johns Hopkins University, exploring a variety of engineering techniques to control and probe the differentiation of pluripotent stem cells. In 2017 he joined the laboratory of Sangeeta Bhatia, the John J. and Dorothy Wilson Professor at MIT’s Institute for Medical Engineering and Science and Electrical Engineering and Computer Science and director of the Marble Center for Cancer Nanomedicine.

By then, the lab had already demonstrated success developing implantable “mini-livers” able to engraft and respond to regenerative stimuli in mice with damaged livers. However, the design is incomplete and Smith wants to incorporate a biliary tree to guide hepato-secreted bile acids that aid in the breakdown of fats but pose as a potential toxin to these therapeutic grafts. He is working to build such a tree with an unexpected tool — microfluidic vessels, forged by biomaterial-encapsulated acupuncture needles that, once removed, can be used to create a network of biliary-lined tunnels through which bile can flow.

The ability to bolster cutting-edge research is but one benefit of Smith’s HHMI award. He and Bhatia praise the seeds the program plants within the academic environment, providing much-needed visibility that achievements in STEM are, and will continue to be, driven by scientists from all walks of life.

“I think this program is directing a shift in what a researcher looks like, offering a motivating exposure to the next generation of students who have the potential to impact science regardless of their background,” Smith says. “This is an incredible and humbling opportunity to explore my passion for the promise of regenerative medicine, and to have the resources to tinker and extend my creativity around translational research.”

Matheus Victor

As a graduate student at Washington University in St. Louis, Matheus Victor, a native of Recife, Brazil, who came to Florida at the age of 15, learned that in the U.S., Latino immigrants are rare among scientific researchers. But there he was, pursuing his dreams to become a neuroscientist. The realization inspired him to lead a Latin American student group at WashU and to conduct outreach activities including creating bilingual curricula for local students.

“I was so privileged to be in a top tier graduate program pursuing my interest,” he said. “How many people get to pursue an interest? We live in a world where you have to earn money and you have to feed your family.”

Now he’s a new postdoc at MIT’s Picower Institute for Learning and Memory in the lab of Institute director and Picower Professor Li-Huei Tsai, with a prestigious fellowship that will support him as he embarks on two investigations of the role of specific cell types in brain aging and cognitive decline.

In one, he plans to turn human induced pluripotent stem cells into microglia, an immune cell of the nervous system increasingly implicated in Alzheimer’s disease, and implant them in the brains of mice where the original microglia have been removed. With this chimera model Victor can test how microglia with different genetic variations act in a mammalian brain to see how those variations might contribute to disease pathology.

In the other, he is interested in studying how inhibitory interneurons change in the aging brain. The neurons are of particular interest because they are the source of a crucial brain rhythm that is notably reduced in Alzheimer’s disease. Understanding more about how they function and falter could help explain that important change.

Jarrett Smith receives Hanna Gray Fellowship from HHMI
Greta Friar | Whitehead Institute
September 12, 2018

Cambridge, Mass — Jarrett Smith, postdoctoral researcher in David Bartel’s lab at the Whitehead Institute, has been announced as a recipient of the Howard Hughes Medical Institute (HHMI)’s 2018 Hanna Gray fellowship. The fellowship supports outstanding early career scientists from groups underrepresented in the life sciences. Each of this year’s fifteen awardees will be given up to $1.4 million dollars in funding over the course of their postdoctoral program and beginning of a tenure-track faculty position.

“This program will help us retain the most diverse talent in science,” said HHMI President Erin O’Shea. “We feel it’s critically important in academia to have exceptional people from all walks of life, all cultures, and all backgrounds – people who can inspire the next generation of scientists.”

For Smith, who began his postdoctoral training in the Bartel lab in January, finding out he got the fellowship was a defining moment.

“I’m grateful for the support that the fellowship will provide during the formative years of my career,” Smith says. “This kind of opportunity gives you the confidence to set ambitious research goals and find out what you can accomplish.”

In the Bartel lab, Smith studies how cells respond to stress. When a cell is exposed to environmental stressors such as heat, UV radiation, or viral infection, proteins and RNAs in the cell may clump together into dense aggregates called stress granules. Several diseases are associated with altered stress granule formation, but the exact function of stress granules and their potential role in disease are unknown. Smith is investigating changes in the cell linked to their formation. His findings could shed light on a potential role for stress granules in cancer, viral infection, and neurodegenerative disease.

Growing up, Smith was always interested in science but no one in his family had ever received a PhD, making biology research feel like an unlikely career path for him. Nevertheless, he followed his passion, which led him to a PhD program at the Johns Hopkins University School of Medicine. Despite his strong academic performance, Smith began graduate school with doubts about his ability to become a scientist. His mentors were incredible teachers but their self-assuredness could be intimidating.

“They were absolutely my role models, but I didn’t think of them as having gone through what I was going through. In the first few years, I felt like I had a lot of catching up to do,” Smith said.

Smith says he was frequently inspired and guided by his graduate school mentor, Geraldine Seydoux. Under her tutorship he became more confident in his abilities.

“I try to pick mentors who are the kind of scientist I aspire to be,” Smith said.

With that tenet in mind, he set his sights on David Bartel’s lab for his postdoctoral research. He had heard that Bartel was a great mentor and knew the Bartel lab had expertise in all of the research techniques he wanted to learn. Since arriving at Whitehead Institute, Smith says he has experienced support not only from Bartel, but from the entire lab as well.

“Jarrett’s graduate experience with P granules in nematodes brings much appreciated expertise to our lab, and we are all excited about what he will discover here on stress-granule function,” Bartel says. “Receiving this fellowship is a well-deserved honor, and I am very happy for him.”

Smith noted that he is netbet sports betting appdeeply grateful for the community he’s found at Whitehead Institute. However, he also noted that throughout his scientific career he has typically been the only black person in the room. One of the joys of applying for the fellowship was meeting the rest of the candidates, a diverse and impressive group of scientists, he says. He looks forward to seeing the other fellows again at meetings hosted by the HHMI.

“I’ve never really had a scientific role model that shared those experiences or that I could identify with in that way,” Smith says, but he hopes that future aspiring scientists won’t have to go through the same experience. His brother-in-law recently began an undergraduate major in biology. Smith enjoys being there to answer his questions about school work or life as a researcher.

“I’d never ask him if he thinks of me as a role model,” Smith says, laughing. “But I’m glad that I have the chance to help people who—like I did—might question whether they could be successful in the sciences.” With the support of the fellowship and his lab, and an exciting research question he is eager to tackle, Smith has never been more certain that he belongs right where he is.

A Summer of Science

Victor Rivera-Santana, a chemistry major at the University of Puerto Rico at Mayagüez, visited MIT Biology for 10 weeks to investigate protein form and function.

August 30, 2018

Victor Rivera-Santana grew up on the western edge of Puerto Rico, in what he refers to as an “atmosphere of science.” His mother is a professor of animal science at the University of Puerto Rico at Mayagüez, and in elementary school he would attend her lectures about the effects of environment and hormones on animal behavior. Three years ago, Rivera-Santana enrolled there as an undergraduate, and has been studying chemistry ever since — with the exception of this past summer, when he became a full-fledged member of the MIT Biology Department for 10 weeks during MIT’s Summer Research Program in Biology (MSRP­-Bio).

Rivera-Santana remembers being drawn to basic research because of its simple, pure, and noble nature, stemming from the creativity of the researcher. “Science almost always has an application, so the fact that researchers in basic science are not looking for an application per se doesn’t mean their work won’t have one in the future,” he says. “The researcher fulfills his or her own curiosity, and afterwards someone else can find a way to put that into practice in society.”

A rising senior, Rivera-Santana chose chemistry because he enjoyed analyzing the minute building blocks of life, but wasn’t sure which field he would ultimately pursue. With chemistry, he could engineer the major to encompass biology and physics as well, which would give him “a taste of everything.”

At first he didn’t know what post-graduation life might hold. However, two weeks into the MSRP-Bio program he’d made up his mind: a PhD. “I like the people, I like the passion, and most importantly I like the research — everything is so interesting it’s hard to pick,” he says.

Rivera-Santana applied to MSRP-Bio early last January because he had it on good authority from three independent sources that this was the program for him. First a good friend and former MSRP-Bio student suggested it, then his professor, and finally his father.

He had two main expectations coming in. First, that everyone would be intimidating and aloof. “Boy was I wrong,” he says. “The MIT faculty are really accessible and engage you as a potential researcher. You can stop them as they’re walking down the hall, or ask them questions during the scheduled Q&A sessions.”

Second, he expected everyone would be hardworking, irrespective of their area of focus. “I was very pleased to find that’s the case,” he says. “I have not met one person at MIT who would not go the extra mile to do their job correctly.”

Rivera-Santana worked in Thomas Schwartz’s lab, investigating an aggregate of proteins known as the nuclear pore complex (NPC), which is embedded in the nuclear envelope and controls the passage of proteins, RNAs, and even ribosomal subunits between the cytoplasm and the nucleus. Although the NPC is vital to cellular survival, its structure is not yet fully understood.

The Schwartz lab goes bit by bit, studying each of its components and their interactions with one another. Rivera-Santana concentrated on one NPC protein in particular, Nup93, parsing its role and design. He hopes this work will eventually help scientists understand the complex as a whole, “because as the name says, it’s complex.”

Rivera-Santana studied four different variants of Nup93, working to express each variant by itself in bacteria. Most of his days in lab went by “either very slowly or very quickly.” He would spend the beginning of the week growing the bacteria to express his proteins — a relatively low-key process since the bacteria “essentially take care of themselves.” The latter half of the week, though, when he began the purification process, was more fast-paced. It involved extracting and purifying the proteins from the bacterial cytosol, while at the same time taking steps to prevent the proteins from becoming damaged, such as keeping them at low temperatures by performing the purification steps in the cold room.

“Purifying is the really challenging part, but it’s also the most fun,” Rivera-Santana says. “I had to work at four degrees Celsius, and I’m from Puerto Rico so you can just imagine how I bear the temperature,” he adds.

Looking back, his most exciting summer experience was purifying his first protein. “I just felt this bundle of joy well up inside me,” he says. “When I ran it through the gel to check its identity and I saw that beautiful blob of ‘ink’ that told me I had my protein, I just felt so happy.”

While Rivera-Santana thoroughly enjoyed his experiences in lab, he was also thrilled to meet other budding researchers and explore Boston’s museums and brick buildings. His proudest moment was cooking his first meal for his new MSRP-Bio friends (a classic Puerto Rican dish: rice, beans, chicken, and plantains). He enjoys putting smiles on people’s faces, “especially when those grins are full of food.” After all, MSRP-Bio isn’t just about being at MIT; it’s also about meeting people and being part of the community.

He also learned he could live by himself, thousands of miles from his family. And he’s prepared to do it again next summer, perhaps in the same lab. “I’m definitely considering doing MSRP-Bio again,” he says. “And I’m certainly also thinking about MIT for graduate school.”

Until then, the three things he’ll miss the most — in no particularly order — are his MSRP-Bio cohort, his lab mentor, and the tasty East Coast cherries.

Photo credit: Raleigh McElvery
Exploring cancer metabolism

Matthew Vander Heiden seeks new cancer treatments that exploit tumor cells’ abnormal metabolism.

Anne Trafton | MIT News Office
August 28, 2018

Nearly 100 years ago, the German chemist Otto Warburg discovered that cancer cells metabolize nutrients differently than most normal cells. His discovery launched the field of cancer metabolism research, but interest in this area waned; by the 1970s most cancer scientists had shifted their focus to the genetic mutations that drive cancer development.

In the past decade or so, interest in cancer metabolism has resurged, and the first drugs that target cancer cells’ abnormal metabolism were approved to treat leukemia in 2017.

“Cancer metabolism is a very sophisticated field at this point,” says Matthew Vander Heiden, an associate professor of biology at MIT. “We have a lot better understanding of what nutrients cancer cells use and what determines how those nutrients are used. This has led to different ways to think about drugs.”

Vander Heiden, who is also a member of MIT’s Koch Institute for Integrative Cancer Research, is one of the people responsible for the recent surge in cancer metabolism research. As a graduate student and postdoc, he published some of the first studies of how cancer cells alter their metabolism, and now his lab at MIT is devoted to the topic.

“All of the time that I was in grad school and working as a postdoc, I was never working in a lab that was dedicated to studying metabolism. So my vision, if someone gave me a job, was to set up a lab that could really be built in a way that would allow us to ask questions about metabolism,” he says.

Metabolism and cancer

Vander Heiden grew up in a small town in Wisconsin, and unlike most of his high school classmates, he headed out of state for college, to the University of Chicago. He was interested in science, so decided on a pre-med track. A work-study job in a plant biology lab led him to discover that he also enjoyed doing research.

“At that point I already had this idea I was going to go to medical school, but then the idea of MD/PhD came up, and I ended up going down that path,” Vander Heiden says.

While in the MD/PhD program at the University of Chicago Medical School, he worked in the lab of Craig Thompson, now president of Memorial Sloan Kettering Cancer Center. At that time, Thompson was studying the biochemical regulation of apoptosis, the programmed cell death pathway. For his PhD thesis, Vander Heiden investigated the function of a protein called Bcl-x, which is a regulator of apoptosis found in the membranes of mitochondria — cell organelles responsible for generating energy.

“That project really got me thinking about how the mitochondria work and how metabolism works,” Vander Heiden recalls. “At the time, I came to the realization that we don’t understand cell metabolism anywhere near as well as we thought we did, and someone should really study this.”

After finishing his degrees, he spent five years doing clinical training, then decided to pursue research in cancer metabolism.

“Altered metabolism has been known about in cancer for 100 years, but few people were studying it,” Vander Heiden says. “The challenge was finding a lab that would allow me to study metabolism and cancer, which in 2004-2005 was not such an obvious thing to do.”

He ended up going to Harvard Medical School to work with Lewis Cantley, who studies signaling pathways in cells and was receptive to the idea of exploring cancer metabolism. There, Vander Heiden began studying an enzyme called pyruvate kinase M2 (PKM2), which is involved in regulation of glycolysis, a biochemical process that cells use to break down sugar for energy.

In 2008, Vander Heiden, Cantley, and others at Harvard Medical School reported that when cells shift between normal and Warburg (cancer-associated) metabolism, they start using PKM2 instead of PKM1, the enzyme that adult cells normally use for glycolysis. Cantley and Craig Thompson have since founded a company, Agios Pharmaceuticals, that is developing potential drugs that target PKM2, as well as other molecules involved in cancer metabolism.

While at Harvard, Vander Heiden also worked on a paper that contributed to the eventual development of drugs that target cancer cells with a mutation in the IDH gene. These drugs, the first modern FDA-approved cancer drugs that target metabolism, shut off an alternative pathway used by cancer cells with the IDH mutation.

New drug targets

In 2010, Vander Heiden became one of the first new faculty members hired after the creation of netbet sports bettingMIT’s Koch Institute, where he set up a lab focused on metabolism, particularly cancer metabolism.

His research has yielded many insights into the abnormal metabolism of cancer cells. In one study, together with other MIT researchers, he found that tumor cells turn on an alternative pathway that allows them to build lipids from the amino acid glutamine instead of the glucose that healthy cells normally use. He also found that altering the behavior of PKM2 to make it act more like PKM1 could stop tumor cell growth.

Studies such as these can offer insights that may help researchers to develop drugs that starve tumor cells of the nutrients they need, offering a new way to fight cancer, Vander Heiden says.

“If one wants to develop drugs that target metabolism, one really needs to focus on the context in which it’s happening, which is the environment of the cell plus the genetics of the cell,” he says. “That is what defines the sensitivity to drugs.”

Tissue architecture affects chromosome segregation

Biologists discover that the environment surrounding a cell plays an integral role in its ability to accurately segregate its chromosomes.

Ashley Junger | Koch Institute
August 24, 2018

All growth and reproduction relies on a cell’s ability to replicate its chromosomes and produce accurate copies of itself. Every step of this process takes place within that cell.

Based on this observation, scientists have studied the replication and segregation of chromosomes as a phenomenon exclusively internal to the cell. They traditionally rely on warm nutritional cultures that promote growth but bear little resemblance to the cell’s external surroundings while in its natural environment.

New research by a group of MIT biologists reveals that this long-held assumption is incorrect. In a paper published this week, they describe how some types of cells rely on signals from surrounding tissue in order to maintain chromosome stability and segregate accurately.

Kristin Knouse, a fellow at the Whitehead Institute, is the lead author of the paper, which was published online in the journal Cell on Aug. 23. Angelika Amon, the Kathleen and Curtis Marble Professor in Cancer Research in the Department of Biology and a member of the Koch Institute for Integrative Cancer Research, is the senior author.

“The main takeaway from this paper is that we must study cells in their native tissues to really understand their biology,” Amon says. “Results obtained from cell lines that have evolved to divide on plastic dishes do not paint the whole picture.”

When cells replicate, the newly duplicated chromosomes line up within the cell and cellular structures pull one copy to each side. The cell then divides down the middle, separating one copy of each chromosome into each new daughter cell.

At least, that’s how it’s supposed to work. In reality, there are sometimes errors in the process of separating chromosomes into daughter cells, known as chromosome mis-segregation. Some errors simply result in damage to the DNA. Other errors can result in the chromosomes being unevenly divided between daughter cells, a condition called aneuploidy.

These errors are almost always harmful to cell development and can be fatal. In developing embryos, aneuploidy can cause miscarriages or developmental disorders such as Down syndrome. In adults, chromosome instability is seen in a large number of cancers.

To study these errors, scientists have historically removed cells from their surrounding tissue and placed them into easily controlled plastic cultures.

“Chromosome segregation has been studied in a dish for decades,” Knouse says. “I think the assumption was … a cell would segregate chromosomes the same way in a dish as it would in a tissue because everything was happening inside the cell.”

However, in previous work, Knouse had found that reported rates for aneuploidy in cells grown in cultures was much higher than the rates she found in cells that had grown within their native tissue. This prompted her and her colleagues to investigate whether the surroundings of a cell influence the accuracy with which that cell divided.

To answer this question, they compared mis-segregation rates between five different cell types in native and non-native environments.

But not all cells’ native environments are the same. Some cells, like those that form skin, grow in a very structured context, where they always have neighbors and defined directions for growth. Other cells, however, like cells in the blood, have greater independence, with little interaction with the surrounding tissue.

In the new study, the researchers observed that cells that grew in structured environments in their native tissues divided accurately within those tissues. But once they were placed into a dish, the frequency of chromosome mis-segregation drastically increased. The cells that were less tied to structures in their tissue were not affected by the lack of architecture in culture dishes.

The researchers found that maintaining the architectural conditions of the cell’s native environment is essential for chromosome stability. Cells removed from the context of their tissue don’t always faithfully represent natural processes.

The researchers determined that architecture didn’t have an obvious effect on the expression of known genes involved in segregation. The disruption in tissue architecture likely causes mechanical changes that disrupt segregation, in a manner that is independent of mutations or gene expression changes.

“It was surprising to us that for something so intrinsic to the cell — something that’s happening entirely within the cell and so fundamental to the cell’s existence — where that cell is sitting actually matters quite a bit,” Knouse says.

Through the Cancer Genome Project, scientists learned that despite high rates of chromosome mis-segregation, many cancers lack any mutations to the cellular machinery that controls chromosome partitioning. This left scientists searching for the cause of the increase of these division errors. This study suggests that tissue architecture could be the culprit.

Cancer development often involves disruption of tissue architecture, whether during tumor growth or metastasis. This disruption of the extracellular environment could trigger chromosome segregation errors in the cells within the tumor.

“I think [this paper] really could be the explanation for why certain kinds of cancers become chromosomally unstable,” says Iain Cheeseman, a professor of biology at MIT and a member of the Whitehead Institute, who was not involved in the study.

The results point not only to a new understanding of the cellular mechanical triggers and effects of cancers, but also to a new understanding of how cell biology must be studied.

“Clearly a two-dimensional culture system does not faithfully recapitulate even the most fundamental processes, like chromosome segregation,” Knouse says. “As cell biologists we really must start recognizing that context matters.”

This work was supported by the National Institutes of Health, the Kathy and Curt Marble Cancer Research Fund, and the Koch Institute Support (core) Grant from the National Cancer Institute.

The cartographer of cells

Aviv Regev helped pioneer single-cell genomics. Now she’s cochairing a massive effort to map the trillions of cells in the human body. Biology will never be the same.

Sam Apple | MIT Technology Review
August 23, 2018

Last October, Aviv Regev spoke to a gathering of international scientists at Israel’s Weizmann Institute of Science. For Regev, a computational and systems biologist at the Broad Institute of MIT and Harvard, the gathering was also a homecoming of sorts. Regev earned her PhD from nearby Tel Aviv University in 2002. Now, 15 years later, she was back to discuss one of the most ambitious projects in the history of biology.

The project, the Human Cell Atlas, aims to create a reference map that categorizes all the approximately 37 trillion cells that make up a human. The Human Cell Atlas is often compared to the Human Genome Project, the monumental scientific collaboration that gave us a complete readout of human DNA, or what might be considered the unabridged cookbook for human life. In a sense, the atlas is a continuation of that project’s work. But while the same DNA cookbook is found in every cell, each cell type reads only some of the recipes—that is, it expresses only certain genes, following their DNA instructions to produce the proteins that carry out a cell’s activities. The promise of the Human Cell Atlas is to reveal which specific genes are expressed in every cell type, and where the cells expressing those genes can be found.

Speaking to her colleagues at the meeting in Israel, Regev, who is cochairing the Human Cell Atlas Organizing Committee with Sarah Teichmann of the Wellcome Trust Sanger Institute, displayed the no-nonsense demeanor you might expect of someone at the helm of a massive scientific undertaking. The project had been under way for a year, and Regev, an MIT biology professor who is also chair of the faculty of the Broad and director of its Klarman Cell Observatory and Cell Circuits Program, was reviewing a newly published white paper detailing how the Human Cell Atlas is expected to change the way we diagnose, monitor, and treat disease.

As Regev made her way through the white paper, the possibilities began to seem almost endless. At the most basic level, as a reference map detailing the genes expressed by each different type of healthy cell, the Human Cell Atlas will make it easier to identify how gene expression and signaling go awry in the case of disease. The same map could also help drug developers avoid toxic side effects: researchers targeting a gene that’s harmful in one part of the body would know if the same gene is playing a vital role in another. And because the atlas is expected to reveal many new types of cells, it could also add much more sensitivity to a type of standard blood test, which simply counts different subsets of immune cells. Likewise, looking at individual intestinal cells might provide new insights into the specific cells responsible for inflammation and food allergies. And a better understanding of types of neurons could have far-reaching implications for brain science.

The final product, Regev says, will amount to nothing less than a “periodic table of our cells,” a tool that is designed not to answer one specific question but to make countless new discoveries possible. Eric Lander, the founding director and president of the Broad Institute and a member of the Human Cell Atlas Organizing Committee, likens it to genomics. “People thought at the beginning they might use genomics for this application or that application,” he says. “Nothing has failed to be transformed by genomics, and nothing will fail to be transformed by having a cell atlas.”

Cellular circuits

Regev’s interest in cells began at Tel Aviv University, where she was one of just 15 or so entering students in a highly selective program that gave them the freedom to take high-level courses in any subject. “You could go your first day as a freshman and decide to take a graduate class in political science,” she says.

Regev took a genetics class her first semester and got hooked on the computational challenge of finding order in the complex, interconnected networks of proteins and netbet online sports bettinggenes within each cell. She pursued that topic for her doctoral work, characterizing living systems in a mathematical language that had been designed to describe computer processes. As she finished her doctorate in 2002, she was accepted into a program at Harvard’s Bauer Center for Genomics Research that allowed her to start her own lab without first training as a postdoc.

Not long after, Lander, who’d begun his own career as a mathematician after studying algebraic coding theory and combinatorial mathematics at Oxford, was searching for star talent for the newly created Broad Institute, whose mission is to use genomics to study human disease and help advance its treatment. He first met Regev at a lunch at the Bauer Center during which the fellows took turns speaking about their research for five to 10 minutes. “By the time we got all the way around the table I had written down ‘Hire Aviv Regev,’” he recalls.

Convinced by Lander to join the Broad after “many cups of tea” at Cafe Algiers in Harvard Square, Regev continued to apply computational approaches to study the mind-bogglingly complicated machinery of the cell. A single cell is made up of millions of molecules that are in constant conversation as they work together to do all the things cells need to do: divide, grow, repair internal damage, and, in the case of immune cells, signal other cells about threats. Inside the nucleus, the DNA is transcribed into RNA. That in turn gives rise to proteins, the molecules that do the work inside a cell. Meanwhile, proteins on the surface of the cell are constantly receiving molecular messages from outside—glucose is available, an invader has arrived. These must be relayed back to proteins in the nucleus, which will respond by transcribing other DNA, giving rise to new proteins and still more signaling networks.

“It’s like a complex computer that is made of these many, many different parts that are interacting with each other and telling each other what to do,” says Regev. The protein signaling networks are like “circuits”—and you can think about the cell “almost like a wiring diagram,” she says. But using computational approaches to understand their activity first requires gathering an enormous amount of data, which Regev has long done through RNA sequencing. Unlike DNA sequencing, she says, it can tell her which genes are actually being expressed, so it offers a far more dynamic picture of a cell in action. But simply sequencing the RNA of the cells she’s studying can tell her only so much. To understand how the circuits change under different circumstances, Regev subjects cells to different stimuli, such as hormones or pathogens, to see how the resulting protein signals change.

Next comes what she calls “the modeling step”—creating algorithms that try to decipher the most likely sequence of molecular events following a stimulus. And just as someone might study a computer by cutting out circuits and seeing how that changes the machine’s operation, Regev tests her model by seeing if it can predict what will happen when she silences specific genes and then exposes the cells to the same stimulus.

In a 2009 study, Regev and her team examined how exposure to molecular components of pathogens like bacteria, viruses, or fungi affected the circuitry of the immune system’s dendritic cells. She turned to a technique known as RNA interference (she now uses CRISPR), which allowed her to systematically shut genes down. Then she looked at which genes were expressed to determine how the cells’ response changed in each case. Her team singled out 100 different genes that were involved in regulating the response to the pathogens—some of which weren’t previously known to be involved in immune function. The study, published in Science, generated headlines. But according to longtime colleague Dana Pe’er, now chair of computational and systems biology at the Sloan Kettering Institute at the Memorial Sloan Kettering Cancer Center and a member of the Human Cell Atlas Organizing Committee, what really sets Regev apart is the elegance of her work. Regev, says Pe’er, “has a rare, innate ability of seeing complex biology and simplifying it and formalizing it into beautiful, abstract, describable principles.”

From smoothies to fruit salad

There are lots of empty coffee mugs in Regev’s office at the Broad Institute, but very little in the way of decoration. She approaches her science with a businesslike efficiency. “There are many brilliant people,” says Lander. “She’s a brilliant person who can get things done.”

In the fast-changing arena of genomics (“2015 in my field is considered ancient history,” she says), she is known for making the most of the latest innovations—and for helping to spur the next ones. For years, she and others in the field struggled with a dirty secret of RNA sequencing: though its promise has always been precision—the power of knowing the exact code—the techniques produced results that were unspecific. Every cell has only a minuscule amount of RNA. For sequencing purposes, the RNA from millions of cells had to be pooled together. Bulk RNA sequencing left researchers with what she likens to a smoothie. Once it’s blended together, there’s no way to distinguish all the fruits—or in this case, the RNA from individual cells—that went into it. What researchers needed was something more like a fruit salad, a way to separate all the blueberries, raspberries, and blackberries.

In 2011, working with Broad Institute colleague Joshua Levin, PhD ’92, and postdocs Alex Shalek, now at MIT’s Institute for Medical Engineering and Science, and Rahul Satija, now at the New York Genome Center, Regev managed to obtain enough RNA from a single cell to sequence it. To test the method, they sequenced 18 individual dendritic cells from the bone marrow of a mouse. The cells were all obtained in the same way and were expected to be the same type. But to the researchers’ amazement, they were expressing different genes and could be classified into two distinct subtypes. It was like finding out the smoothie you’d been drinking for years had ingredients you’d never known about.

Regev and her colleagues weren’t the only ones figuring out how to sequence a single cell with such sensitivity, nor were they the very first to succeed. Other labs were making similar advances at approximately the same time, each using its own technology and algorithms. And they all faced the same problem: isolating and extracting enough RNA from individual cells was time consuming and expensive. Regev and her colleagues had spent many thousands of dollars to sequence only 18 cells. If the body was full of rare, undiscovered cells, it was going to take an extraordinarily long time to find them.

Skip ahead seven years and the cost of single-cell RNA sequencing is down to only pennies per cell. A critical breakthrough was Drop-Seq, a new technology developed by researchers at Harvard and the Broad Institute, including Regev and members of her lab. The device embeds individual cells into distinct oil droplets with a tiny “bar-coded” bead. When the cell is broken apart for sequencing, some of its RNA attaches to the bead in its droplet. This allows researchers to analyze thousands at once without getting their genetic material mixed up.

Cell theory 2.0

When cell theory was first proposed by German scientists some 180 years ago, it was hard to fathom that our tissues are built from “individual elementary units,” as Theodor Schwann, one of the two scientists credited with the theory, described cells. But it soon became a central tenet of biology, and over the decades and centuries, cells began to give up their secrets. Microscopes improved; new staining and sorting techniques became available. With each advance, new distinctions became possible. Muscle cells could be distinguished from neurons, and then categorized again as smooth or skeletal muscle cells. Cells, it became clear, were all fundamentally similar but came in different forms that had different properties.

By the 21st century, 200 to 300 major cell types had been identified. And while biologists have long recognized that the true number of cell types must be higher, the extent of their diversity is only now coming into full focus, thanks in large part to single-cell RNA sequencing. Regev says that the immune system alone can now be divided into more than 200 cell types and that even our retinas have 100 or more distinct types of neurons. She and her colleagues have discovered several of them.

The idea that knowing so much more about our cells could lead to medical breakthroughs is no longer hypothetical. By sequencing the RNA of individual cancer cells in recent years—“Every cell is an experiment now,” she says—she has found remarkable differences between the cells of a single tumor, even when they have the same mutations. (Last year that work led to Memorial Sloan Kettering’s Paul Marks Prize for Cancer Research.) She found that while some cancers are thought to develop resistance to therapy, a subset of melanoma cells were resistant from the start. And she discovered that two types of brain cancer, oligodendroglioma and astrocytoma, harbor the same cancer stem cells, which could have important implications for how they’re treated.

The excitement in the field has become tangible as more new cell types have been found. And yet Regev realized that if the aim was comprehensive knowledge, the approach needed to be coordinated. If each lab were to rely on its own techniques, it would be hard to standardize the computational tools and the resulting data. The new studies were producing “very nice glimmers of light,” Regev says—“a thing here, a thing there.” But she wanted to make sure those findings could be connected.Regev has also been busily mapping cells from the immune system, brain, gut, and elsewhere. She is not alone. Other labs have started their own mapping projects, each tackling a different part of the body. Last year researchers at the University of Washington attempted to classify every cell type in the microscopic worm C. elegans. “Every single field in biology is saying, ‘Of course we have to look at single-cell resolution,’” says Lander. “How did we ever imagine we were going to solve a problem without single-cell resolution?”

Regev began to advocate creating something more unified: a map that would allow researchers to chart gene expression and cell types across the entire body. Sarah Teichmann had been thinking along the same lines. When she reached out to Regev in late 2015 about the possibility of joining forces, Regev immediately said yes.

A Google Maps for our cells

The Human Cell Atlas is a collaboration among hundreds of biologists, technologists, and software engineers across the globe. Results from single-cell RNA sequencing will be combined with other data points to provide a comprehensive catalogue of all human cells.

But the many researchers involved won’t simply be compiling spreadsheets listing different cell types. The atlas will also reveal where the cells are located in the body, how many there are, what forms they can take, even the developmental history of different cell types as they differentiated from stem cells. And all netbet online sports bettingof this will be made accessible through a data coordination platform and a rich visual interface that Regev compares to Google Maps. It will allow users to zoom in to the molecular level of our cells, but zooming out to the level of tissues and organs will be important too. As a 2017 overview of the Human Cell Atlas by the project’s organizing committee noted, an atlas “is a map that aims to show the relationships among its elements.” Just as corresponding coastlines seen in an atlas of Earth offer visual evidence of continental drift, compiling all the data about our cells in one place could reveal relationships among cells, tissues, and organs, including some that are entirely unexpected. And just as the periodic table made it possible to predict the existence of elements yet to be observed, the Human Cell Atlas, Regev says, could help us predict the existence of cells that haven’t been found.

The plan is not to sequence all 37 trillion cells but to sample from every part of the body. As Regev talks about the project, her enthusiasm evident, she digs up a slide to demonstrate how effective sampling can be. The slide, first only an empty frame of white, begins to fill in, pixel by pixel, with specks of blue and yellow. Soon, even though many of the pixels haven’t yet been filled, the image on the screen is unmistakable: it is Van Gogh’s Starry Night. Likewise, Regev explains, the Human Cell Atlas can give a complete picture even if not every single cell has been sequenced.

To do the sequencing, Regev and Teichmann have welcomed and recruited experts in each different tissue type. Though expected to take years, the project is moving ahead rapidly with such backers as NIH, the EU, the Wellcome Trust, the Manton Foundation, and the Chan Zuckerberg Initiative, which pledged to spend $3 billion to battle disease over the next decade; this year alone it will fund 85 Human Cell Atlas grants. Early results are already pouring in. In March, Swedish researchers working on cells related to human development announced they had sequenced 250,000 individual cells. In May, a team at the Broad made a data set of more than 500,000 immune cells available on a preview site. The goal, Regev says, is for researchers everywhere to be able to use the open-source platform of the Human Cell Atlas to perform joint analyses.

Plenty of challenges remain before the atlas can become a reality. New visualization software must be developed. Sequencing and computational approaches will need to be standardized across a huge number of labs. Conceptual issues, such as what distinguishes one cell type from another, have to be worked through. But the community behind the Human Cell Atlas—including more than 800 individuals as of June—has no shortage of motivation.

One of Regev’s own recent studies, published in August in Nature, is perhaps the best example of how the project could change biology. In mapping cells of the lungs, Regev and Jay Rajagopal’s lab at Massachusetts General Hospital found a new, very rare cell type that primarily expresses a gene linked to cystic fibrosis. Regev now thinks that these rare cells probably play a key role in the disease. More surprising yet, researchers had previously thought that a different cell type was expressing the gene.

“Imagine if somebody wanted to do gene therapy,” Regev says. “You have to fix the gene, but you have to fix it in the right cell.” The Human Cell Atlas could help researchers identify the right cell and understand how the gene in question is regulated by that cell’s extraordinarily complicated molecular networks.

For Regev, the importance of the Human Cell Atlas goes beyond its promise to revolutionize biology and medicine. As she once put it, without an atlas of our cells, “we don’t really know what we’re made of.”

Antidepressant restores youthful flexibility to aging inhibitory neurons

Neural plasticity and arbor growth decline with age, study in mice shows.

David Orenstein | Picower Institute for Learning and Memory
August 20, 2018

A new study provides fresh evidence that the decline in the capacity of brain cells to change (called “plasticity”), rather than a decline in total cell number, may underlie some of the sensory and cognitive declines associated with normal brain aging. Scientists at MIT’s Picower Institute for Learning and memory show that inhibitory interneurons in the visual cortex of mice remain just as abundant during aging, but their arbors become simplified and they become much less structurally dynamic and flexible.

In their experiments published online in the Journal of Neuroscience they also show that they could restore a significant degree of lost plasticity to the cells by treating mice with the commonly used antidepressant medication fluoxetine, also known as Prozac.

“Despite common belief, loss of neurons due to cell death is quite limited during normal aging and unlikely to account for age-related functional impairments,” write the scientists, including lead author Ronen Eavri, a postdoc at the Picower Institute, and corresponding author Elly Nedivi, a professor of biology and brain and cognitive sciences. “Rather it seems that structural alterations in neuronal morphology and synaptic connections are features most consistently correlated with brain age, and may be considered as the potential physical basis for the age-related decline.”

Nedivi and co-author Mark Bear, the Picower Professor of Neuroscience, are affiliated with MIT’s Aging Brain Initiative, a multidisciplinary effort to understand how aging affects the brain and sometimes makes the brain vulnerable to disease and decline.

In the study the researchers focused on the aging of inhibitory interneurons which is less well-understood than that of excitatory neurons, but potentially more crucial to plasticity. Plasticity, in turn, is key to enabling learning and memory and in maintaining sensory acuity. In this study, while they focused on the visual cortex, the plasticity they measured is believed to be important elsewhere in the brain as well.

The team counted and chronically tracked the structure of inhibitory interneurons in dozens of mice aged to 3, 6, 9, 12 and 18 months. (Mice are mature by 3 months and live for about 2 years, and 18-month-old mice are already considered quite old.) In previous work, Nedivi’s lab has shown that inhibitory interneurons retain the ability to dynamically remodel into adulthood. But in the new paper, the team shows that new growth and plasticity reaches a limit and progressively declines starting at about 6 months.

But the study also shows that as mice age there is no significant change in the number or variety of inhibitory cells in the brain.

Retraction and inflexibility with age

Instead the changes the team observed were in the growth and performance of the interneurons. For example, under the two-photon microscope the team tracked the growth of dendrites, which are the tree-like structures on which a neuron receives input from other neurons. At 3 months of age mice showed a balance of growth and retraction, consistent with dynamic remodeling. But between 3 and 18 months they saw that dendrites progressively simplified, exhibiting fewer branches, suggesting that new growth was rare while retraction was common.

In addition, they saw a precipitous drop in an index of dynamism. At 3 months virtually all interneurons were above a crucial index value of 0.35, but by 6 months only half were, by 9 months barely any were, and by 18 months none were.

Bear’s lab tested a specific form of plasticity that underlies visual recognition memory in the visual cortex, where neurons respond more potently to stimuli they were exposed to previously. Their measurements showed that in 3-month-old mice “stimulus-selective response potentiation” (SRP) was indeed robust, but its decline went hand in hand with the decline in structural plasticity, so that it was was significantly lessened by 6 months and barely evident by 9 months.

Fountain of fluoxetine

While the decline of dynamic remodeling and plasticity appeared to be natural consequences of aging, they were not immutable, the researchers showed. In prior work Nedivi’s lab had shown that fluoxetine promotes interneuron branch remodeling in young mice, so they decided to see whether it could do so for older mice and restore plasticity as well.

To test this, they put the drug in the drinking water of mice at various ages for various amounts of time. Three-month-old mice treated for three months showed little change in dendrite growth compared to untreated controls, but 25 percent of cells in 6-month-old mice treated for three months showed significant new growth (at the age of 9 months). But among 3-month-old mice treated for six months, 67 percent of cells showed new growth by the age of 9 months, showing that treatment starting early and lasting for six months had the strongest effect.

The researchers also saw similar effects on SRP. Here, too, the effects ran parallel to the structural plasticity decline. Treating mice for just three months did not restore SRP, but treating mice for six months did so significantly.

“Here we show that fluoxetine can also ameliorate the age-related decline in structural and functional plasticity of visual cortex neurons,” the researchers write. The study, they noted, adds to prior research in humans showing a potential cognitive benefit for the drug.

“Our finding that fluoxetine treatment in aging mice can attenuate the concurrent age-related declines in interneuron structural and visual cortex functional plasticity suggests it could provide an important therapeutic approach towards mitigation of sensory and cognitive deficits associated with aging, provided it is initiated before severe network deterioration,” they continued.

In addition to Eavri, Nedivi and Bear, the paper’s other authors are Jason Shepherd, Christina Welsh, and Genevieve Flanders.

The National Institutes of Health, the American Federation for Aging Research, the Ellison Medical Fondation, and the Machiah Foundation supported the research.

Altering Neuronal Activity at Will

Graduate student Nicole Aponte Santiago uses fruit flies to probe the relationship between the many nerve cells passing signals from the brain to the muscles.

Raleigh McElvery
August 16, 2018

As fifth year graduate student Nicole Aponte Santiago puts it, she has the power to alter nerve cell activity in fruit flies at will. She’ll also tell you point-blank that flies are her favorite model organism, although that wasn’t always the case. They have many well-cited advantages — they procreate often, have short lifespans, and many of their genes overlap with humans while being easy to manipulate. But Aponte suggests an additional reason for her change of heart. She suspects “looking them straight in the eye” so frequently ultimately convinced her, using their physical features to deduce their genetic makeup, and from there investigate the relationship between the many nerve cells (neurons) passing signals from the brain to the muscles.

Aponte credits her initial interest in science to her father, a dentist who used to take her for long walks on the beaches of Puerto Rico where she grew up. Together the two would examine the minute details of the coastal world around them, discussing various phenomena. Where does sand come from? How do the tides form? What types of shelled critters reside at the sea’s edge?

She first set foot on netbet sports betting appMIT’s campus in January of 2011 while a sophomore at the University of Puerto Rico, Río Piedras, attending the weeklong Quantitative Methods Workshop to learn various quantitative tools and programming languages. In June of that same year she returned, this time for 10 weeks as an MIT Summer Research Program (MSRP) student. Aponte joined Hazel Sive’s lab at the Whitehead Institute for Biomedical Research, gathering preliminary data for a paper that was eventually published in Fluids and Barriers of the CNS.

“I was introduced to basic science through both programs,” Aponte says. “Discovery-based research lays the foundation for translational research later on, and it allows you to address questions that no one has answered before. If you’re lucky enough to witness those discoveries, for a few moments you’re the only person in the world with that knowledge, and I find that prospect incredibly exciting.”

As a first-year graduate student in 2013, she tested out several different labs and ultimately chose Troy Littleton’s lab located in the Picower Institute for Learning and Memory. She was drawn to the friendly and collaborative atmosphere, as well as the breadth of potential projects — from those related to the central nervous system and Huntington’s disease to the connections between nerve cells.

Aponte investigates how nerve cells interface with one another and neighboring muscles, focusing on the tiny gap between neurons and muscles, known as neuromuscular junctions. Multiple neurons can form connections with the same neuron or muscle cell, but which connections remain depends on competition between the nerve cells and how often those connections are used. New ones are constantly being shaped as we learn new things and acquire new experiences, while less frequently used connections get trimmed away. Aponte wants to understand how neurons that talk to the same muscle cell interact with each other, determining which lines of communication will be strengthened and which will be pruned away.

When Aponte first began in the Littleton lab five years ago, she screened scores of unique drivers — segments of DNA that “drive” gene expression in specific cells during a specific time in development. She hoped to find drivers that could trigger the expression of a green fluorescent protein (GFP) at the neuromuscular junction, and ultimately identified two different drivers which caused GFP expression in two separate neurons targeting the same muscle near the back of the fly. These drivers would allow her to control the expression of not just GFP, but also other genes that she connected to the drivers, including genes that can increase or decrease the activity of the neurons, or even kill them.

“It’s really from there that my entire project was born,” Aponte recalls.

Each morning, she heads to the fly room to collect virgin flies and determine which ones to breed in order to generate progeny with her desired genetic makeup. She can infer something about the flies’ chromosomes based on their physical characteristics, which allow her to keep track of their genes and create offspring with the GFP marker in her neurons of interest. As is true for most things these days, there’s an app for that. As we chat, she takes out her phone and begins to scroll through images that illustrate which genetic markers engender which physical characteristics. The Tubby mutation makes the flies rather fat, whereas Bar makes their eyes noticeably skinnier.

“In the same way that I can express GFP in just one of the two motor neurons to distinguish between them and monitor their shape,” she says, “I can also express genes that increase or decrease activity of one of the neurons, or even kill one of the neurons. In this way, I create an input imbalance by increasing or decreasing the activity in either neuron to see how that affects the activity of its partner neuron.”

In the afternoons, sometimes she uses molecular fluorophores to highlight the changes in structure of these neurons. She also assesses the activity in the muscles by poking them with electrodes and observing what happens when the muscle receives more or less input from one of the motor neurons. Additionally, she explores what happens to surrounding muscles when these inputs are modified

Recently, she’s stumbled upon a rather baffling phenomenon. When she kills one of the two neurons (let’s call it “Neuron A”) the other one, Neuron B, remains relatively unchanged. Yet, when she kills Neuron B, Neuron A shrinks. This suggests Neuron B may be sending a “permissive” signal to Neuron A, telling it to contact the muscle. She is monitoring the neuromuscular junction over time to determine if Neuron A will ever reach the muscle, and if the muscle will change over time in response.

“This has never been seen before, so right now I’m trying to identify the smaller neuron to see if it is functional,” she says. “I’m going to look at it during different stages of development to see if it starts and stops communicating with the muscle at some point.”

This endeavor will constitute her main focus over the next few months. After she graduates, Aponte intends to pursue an academic postdoc studying flies, despite developing an allergy to her subjects over the past year. (She’s since acquired a face mask that keeps dust and fly particles away.) “I’m just excited to see where the research will take me,” she says. Now that’s dedication.

Photo credit: Raleigh McElvery