Biologists answer fundamental question about cell size

The need to produce just the right amount of protein is behind the striking uniformity of sizes.

Anne Trafton | MIT News Office
February 7, 2019

MIT biologists have discovered the answer to a fundamental biological question: Why are cells of a given type all the same size?

In humans, cell size can vary more than 100-fold, ranging from tiny red blood cells to large neurons. However, within each cell type, there is very little deviation from a standard size. In studies of yeast, MIT researchers grew cells to 10 times their normal size and found that their DNA could not keep up with the demands of producing enough protein to maintain normal cell functions.

Furthermore, the researchers found that this protein shortage leads the cells into a nondividing state known as senescence, suggesting a possible explanation for how cells become senescent as they age.

“There are so many hypotheses out there that try to explain why senescence happens, and I think this data provides a beautiful and simple explanation for senescence,” says 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.

Amon is the senior author of the study, which appears in the Feb. 7 online edition of Cell. Gabriel Neurohr, an MIT postdoc, is the lead author of the paper.

netbet online sports betting

To explore why cell size is so tightly controlled, the researchers prevented yeast cells from dividing by modifying a gene critical for cell division, so that it could be turned off at a certain temperature. These cells continued to grow, but they could not divide and they did not replicate their DNA.

The researchers discovered that as the cells expanded, their DNA and their protein-building machinery could not keep pace with the needs of such a large cell. This failure to produce enough protein led to the dilution of the cytoplasm and disruption of cell division. The researchers believe that many other fundamental cell processes that rely on cellular molecules finding and interacting with each other may also be impaired when cells are too big.

“Theoretical models predict that diluting the cytoplasm will decrease reaction rates. Every chemical reaction would occur more slowly, and some threshold concentrations of certain proteins may not be reached, so certain reactions would never happen because the concentrations are lower,” Neurohr says.

The researchers showed that yeast cells with two sets of chromosomes were able to grow to twice the size of yeast cells with just one set of chromosomes before becoming senescent, suggesting that the amount of DNA in the cells is the limiting factor in the cells’ ability to grow.

Experiments with human cells yielded similar results: In a study of human fibroblast cells, the researchers found that forcing the cells to grow to excessive sizes (eight times their normal size) disrupted many functions, including cell division.

“It’s been clear for some time that cells do control their size, but it’s been unclear what the various physiological reasons are for why they do so,” says Jan Skotheim, an associate professor of biology at Stanford University, who was not involved in the research. “What’s nice about this work is it really shows how things go wrong when cells get too big.”

Age-related disease

Amon says excessive growth likely plays a major role in the development of senescence, which occurs in many types of mammalian cells and is thought to contribute to age-related organ dysfunction and chronic age-related diseases.

Senescent cells are often larger than younger cells, and this study, which showed that unchecked cell growth leads to senescence, offers a possible explanation for this observation. Human cells tend to grow slightly larger throughout their lifetimes, because every time a cell divides, it checks for DNA damage, and if any is found, division is halted while repairs are made. During each of these delays, the cell grows slightly larger.

“Over the lifetime of a cell, the more divisions you make, the higher your probability is of having that damage, and over time cells will get larger,” Amon says. “Eventually they get so large that they start diluting critical factors that are important for proliferation.”

A difficult question that remains unanswered is how different types of cells maintain the appropriate size for their cell type, which the researchers now hope to study further.

The research was funded, in part, by the National Institutes of Health, the Howard Hughes Medical Institute, the Paul F. Glenn Center for Biology of Aging Research at MIT, a National Science Foundation graduate research fellowship, the William Bowes Fellows program, and the Vilcek Foundation.

Origin story

Junior Leah McKinney practiced kitchen microbiology on her ranch in Nevada before exploring the intricacies of DNA replication initiation in bacteria at MIT Biology.

Raleigh McElvery
February 6, 2019

Leah McKinney grew up on a 50,000-acre cattle ranch in Nevada — vaccinating sheep, roping calves, digging for fossils, and occasionally hauling home old bovine femurs. She saddled horses, treated sick lambs, and helped ewes struggling to give birth. One Christmas, she even asked Santa for a fetal pig. “He delivered,” McKinney, now a junior in Course 7, recalls with a laugh.

When she was 12 years NetBet sportold, she saved up enough birthday money to purchase a microscope. Even though she permanently dyed the kitchen sink a distinct shade of blue while making slides, her parents (who both hold degrees in animal science) didn’t mind. They even let her grow bacteria in the heater closet and tally them on the kitchen counter — all in the name of the elementary school science fair.

“They were always encouraging my weird scientific endeavors,” she says. “I think my love for science, and microbiology specifically, came out of my agricultural upbringing.”

She grew to appreciate basic science because it allowed her to study the fundamental mechanisms behind key biological processes. She arrived at MIT in 2016 determined to major in Biology, and hasn’t wavered in her decision. Although she relishes the subject matter, she initially feared the classes would be tedious and memorization-heavy.

“I was quite happy to learn that’s not the case here,” she says. “MIT Biology values problem-solving over rote memorization, and encourages you to take the information you’ve learned in class and apply it to interesting problems. And that mindset extends from the classroom into the lab.”

One of the things that drew McKinney to MIT was the institute’s Undergraduate Research Opportunities Program (UROP), which allows students to join labs and collaborate with faculty as early as their first year. She recalls that, while other universities touted similar opportunities, MIT placed theirs front and center.

“I’d heard that all you had to do was email a professor and ask to join the lab, but I didn’t believe it — that just seemed way too easy,” McKinney says. “But when I was looking for a UROP, I just emailed my current principal investigator to set up a time to talk, and now I’ve been in his lab for over a year.”

McKinney is part of Department Head Alan Grossman’s lab, which investigates the molecular mechanisms and regulation underlying basic cellular processes in bacteria. The entire group works with the rod-shaped Bacillus subtilis, but some members study horizontal gene transfer while others focus on DNA replication and gene expression. McKinney and her graduate student mentor Mary Anderson are in this second category, examining a protein called DnaA that is required to initiate DNA replication and also modulates the expression of several genes.

In order to successfully grow and reproduce, a bacterium must first replicate its single chromosome before dividing into two identical daughter cells. DnaA is responsible for beginning DNA replication in all bacteria. It binds to the origin of replication on the chromosome, unwinds some of the nearby DNA, and recruits the other proteins needed to copy the chromosome.

This operation is highly regulated to ensure that each daughter cell receives only a single chromosome. B. subtilis controls replication via several proteins, including YabA. When YabA binds to DnaA, it prevents replication from ever getting started.

Since DnaA also serves as a transcription factor — binding to other DNA sequences called promoters to increase or decrease expression of certain genes, including its own gene dnaA — YabA may also impact DnaA’s gene targets. McKinney hopes to eventually determine exactly how.

While McKinney discovers something new about her bacteria each time she conducts a successful experiment, she learns almost as much when her tests go awry. “I’ve had to practice a lot of troubleshooting,” she says, “and that’s not something you can learn in class. But everyone in the lab is incredibly friendly and always willing to answer questions or give advice.”

As a teaching assistant for the lab class 7.02 (Introduction to Experimental Biology and Communication), McKinney had the chance to help other students conduct experiments, answering their questions and grading their lab notebooks. She took 7.02 last spring, but says it’s been enlightening to experience the class through a different lens. She adds: “I definitely understand the material more deeply than I did before.”

In addition to TAing, McKinney teaches an SAT preparatory program run by MIT students. “At first, standing up and talking in front of a 20-person section was rather terrifying, but it’s become so much easier,” she says. “The experience has been really good for me.”

After she graduates, McKinney knows she wants to go to graduate school — likely for microbiology — but beyond that, nothing is concrete. She is sure of one thing, though: joining the Grossman lab was one of the best decisions she’s made at MIT.

She advises all current and prospective students to do a UROP. “Find something you’re really interested in,” she says. “It’s okay not to know a lot coming in; you’re going to learn so much, including topics and techniques you won’t learn in class. And don’t be too disappointed when things don’t work; that’s just part of the process. And when you finally get something to work that you’ve been troubleshooting for a while, the feeling is absolutely amazing.”

Posted 2.5.19
Biologist Adam Martin studies the mechanics of tissue folding

The dynamic process is critical to embryonic development and other cellular phenomena.

Anne Trafton | MIT News Office
February 1, 2019

Embryonic development is tightly regulated by genes that control how body parts form. One of the key responsibilities of these genes is to make sure that tissues fold into the correct shapes, forming structures that will become the spine, brain, and other body parts.

During the 1970s and ’80s, the field of embryonic development focused mainly on identifying the genes that control this process. More recently, many biologists have shifted toward investigating the physics behind the tissue movements that occur during development, and how those movements affect the shape of tissues, says Adam Martin, an MIT associate professor of biology.

Martin, who recently earned tenure, has made key discoveries in how tissue folding is controlled by the movement of cells’ internal scaffolding, known as the cytoskeleton. Such discoveries can not only shed light on how tissues form, including how birth defects such as spina bifida occur, but may also help guide scientists who are working on engineering artificial human tissues.

“We’d like to understand the molecular mechanisms that tune how forces are generated by cells in a tissue, such that the tissue then gets into a proper shape,” Martin says. “It’s important that we understand fundamental mechanisms that are in play when tissues are getting sculpted in development, so that we can then harness that knowledge to engineer tissues outside of the body.”

Cellular forces

Martin grew up in Rochester, New York, where both of his parents were teachers. As a biology major at nearby Cornell University, he became interested in genetics and development. He went on to graduate school at the University of California at Berkeley, thinking he would study the genes that control embryonic development.

However, while in his PhD program, Martin became interested in a different phenomenon — the role of the cytoskeleton in a process called endocytosis. Cells use endocytosis to absorb many different kinds of molecules, such as hormones or growth factors.

“I was interested in what generates the force to promote this internalization,” Martin says.

NetBet live casinoHe discovered that the force is generated by the assembly of arrays of actin filaments. These filaments tug on a section of the cell membrane, pulling it inward so that the membrane encloses the molecule being absorbed. He also found that myosin, a protein that can act as a motor and controls muscle contractions, helps to control the assembly of actin filaments.

After finishing his PhD, Martin hoped to find a way to combine his study of cytoskeleton mechanics with his interest in developmental biology. As a postdoc at Princeton University, he started to study the phenomenon of tissue folding in fruit fly embryonic development, which is now one of the main research areas of his lab at MIT. Tissue folding is a ubiquitous shape change in tissues to convert a planar sheet of cells into a three-dimensional structure, such as a tube.

In developing fruit fly embryos, tissue folding invaginates cells that will form internal structures in the fly. This folding process is similar to tissue folding events in vertebrates, such as neural tube formation. The neural tube, which is the precursor to the vertebrate spinal cord and brain, begins as a sheet of cells that must fold over and “zip” itself up along a seam to form a tube. Problems with this process can lead to spina bifida, a birth defect that results from an incomplete closing of the backbone.

When Martin began working in this area, scientists had already discovered many of the transcription factors (proteins that turn on networks of specific genes) that control the folding of the neural tube. However, little was known about the mechanics of this folding.

“We didn’t know what types of forces those transcription factors generate, or what the mechanisms were that generated the force,” he says.

NetBet live casinoHe discovered that the accumulation of myosin helps cells lined up in a row to become bottle-shaped, causing the top layer of the tissue to pucker inward and create a fold in the tissue. More recently, he found that myosin is turned on and off in these cells in a dynamic way, by a protein called RhoA.

“What we found is there’s essentially an oscillator running in the cells, and you get a cycle of this signaling protein, RhoA, that’s being switched on and off in a cyclical manner,” Martin says. “When you don’t have the dynamics, the tissue still tries to contract, but it falls apart.”

He also found that the dynamics of this myosin activity can be disrupted by depleting genes that have been linked to spina bifida.

Breaking free

Another important cellular process that relies on tissue folding is the epithelial-mesenchymal transition (EMT). This occurs during embryonic development when cells gain the ability to break free and move to a new location. It is also believed to occur when cancer cells metastasize from tumors to seed new tumors in other parts of the body.

During embryonic development, cells lined up in a row need to orient themselves so that when they divide, both daughter cells remain in the row. Martin has shown that when the mechanism that enables the cells to align correctly is disrupted, one of the daughter cells will be squeezed out of the tissue.

“This has been proposed as one way you can get an epithelial-to-mesenchymal transition, where you have cells dissociate from native tissue,” Martin says.  He now plans to further study what happens to the cells that get squeezed out during the EMT.

In addition to these projects, he is also collaborating with Jörn Dunkel, an MIT associate professor of mathematics, to map the network connections between the myosin proteins that control tissue folding during development. “That project really highlights the benefits of getting people from diverse backgrounds to analyze a problem,” Martin says.

Bacteria promote lung tumor development, study suggests

Antibiotics or anti-inflammatory drugs may help combat lung cancer.

Anne Trafton | MIT News Office
January 31, 2019

MIT cancer biologists have discovered a new mechanism that lung tumors exploit to promote their own survival: These tumors alter bacterial populations within the lung, provoking the immune system to create an inflammatory environment that in turn helps the tumor cells to thrive.

In mice that were genetically programmed to develop lung cancer, those raised in a bacteria-free environment developed much smaller tumors than mice raised under normal conditions, the researchers found. Furthermore, the researchers were able to greatly reduce the number and size of the lung tumors by treating the mice with antibiotics or blocking the immune cells stimulated by the bacteria.

The findings suggest several possible strategies for developing new lung cancer treatments, the researchers say.

“This research directly links bacterial burden in the lung to lung cancer development and opens up multiple potential avenues toward lung cancer interception and treatment,” says Tyler Jacks, director of MIT’s Koch Institute for Integrative Cancer Research and the senior author of the paper.

Chengcheng Jin, a Koch Institute postdoc, is the lead author of the study, which appears in the Jan. 31 online edition of Cell.

Linking bacteria and cancer

Lung cancer, the leading cause of cancer-related deaths, kills more than 1 million people worldwide per year. Up to 70 percent of lung cancer patients also suffer complications from bacterial infections of the lung. In this study, the MIT team wanted to see whether there was any link between the bacterial populations found in the lungs and the development of lung tumors.

To explore this potential link, the researchers studied genetically engineered mice that express the oncogene Kras and lack the tumor suppressor gene p53. These mice usually develop a type of lung cancer called adenocarcinoma within several weeks.

Mice (and humans) typically have many harmless bacteria growing in their lungs. However, the MIT team found that in the mice engineered to develop lung tumors, the bacterial populations in their lungs changed dramatically. The overall population grew significantly, but the number of different bacterial species went down. The researchers are not sure exactly how the lung cancers bring about these changes, but they suspect one possibility is that tumors may obstruct the airway and prevent bacteria from being cleared from the lungs.

This bacterial population expansion induced immune cells called gamma delta T cells to proliferate and begin secreting inflammatory molecules called cytokines. These molecules, especially IL-17 and IL-22, create a progrowth, prosurvival environment for the tumor cells. They also stimulate activation of neutrophils, another kind of immune cell that releases proinflammatory chemicals, further enhancing the favorable environment for the tumors.

“You can think of it as a feed-forward loop that forms a vicious cycle to further promote tumor growth,” Jin says. “The developing tumors hijack existing immune cells in the lungs, using them to their own advantage through a mechanism that’s dependent on local bacteria.”

However, in mice that were born and raised in a germ-free environment, this immune reaction did not occur and the tumors the mice developed were much smaller.

Blocking tumor growth

The researchers found that when they treated the mice with antibiotics either two or seven weeks after the tumors began to grow, the tumors shrank by about 50 percent. The tumors also shrank if the researchers gave the mice drugs that block gamma delta T cells or that block IL-17.

The researchers believe that such drugs may be netbet sports betting appworth testing in humans, because when they analyzed human lung tumors, they found altered bacterial signals similar to those seen in the mice that developed cancer. The human lung tumor samples also had unusually high numbers of gamma delta T cells.

“If we can come up with ways to selectively block the bacteria that are causing all of these effects, or if we can block the cytokines that activate the gamma delta T cells or neutralize their downstream pathogenic factors, these could all be potential new ways to treat lung cancer,” Jin says.

Many such drugs already exist, and the researchers are testing some of them in their mouse model in hopes of eventually testing them in humans. The researchers are also working on determining which strains of bacteria are elevated in lung tumors, so they can try to find antibiotics that would selectively kill those bacteria.

The research was funded, in part, by a Lung Cancer Concept Award from the Department of Defense, a Cancer Center Support (core) grant from the National Cancer Institute, the Howard Hughes Medical Institute, and a Margaret A. Cunningham Immune Mechanisms in Cancer Research Fellowship Award.

Puzzling over Pollen

Graduate student Joe Jacobowitz analyzes new enzymes that could reveal key insights into plant reproduction.

Raleigh McElvery
January 24, 2019

Every morning, fifth-year graduate student Joe Jacobowitz takes the elevator to the seventh floor of the Whitehead Institute, passes the soil bins, “false winter” fridges, and toasty growing chambers, and enters his favorite workspace: the greenhouse. There, among the myriad of tall, stout, grass-like, and blooming plants, he attends to his organism of choice, Arabidopsis thaliana. With four simple, white petals interrupted by protruding, yellow stamens, “it looks like something that would grow in the cracks of a sidewalk,” Jacobowitz says. While you or I might pass by it and not think twice, Jacobowitz and the Weng lab hold that Arabidopsis could reveal key insights into pollen development, in particular which enzymes are critical for plant reproduction.

Jacobowitz became fascinated by enzymes as a biochemistry major at Brandeis University, studying the evolution of a single enzyme found in the deadliest form of malaria. After arriving at MIT Biology for graduate school and joining Jing-Ke Weng’s team at the Whitehead, Jacobowitz shifted his focus from biochemistry and biophysics to plant development. His work investigating the pollen-bearing chamber known as the anther represents just one facet of the Weng lab — which probes the origin and evolution of plant metabolism, as well as the small molecules plants produce to interact with their environments.

Above his lab desk, next to hand-drawn sketches and photos of friends, Jacobowitz has taped intricate microscopy images detailing the many complex stages of anther development. The pollen grains inside this structure contain the plant’s male gametes, which are transferred via wind and passersby to the female part, the pistil, of another flower. In the case of Arabidopsis, a single flower can self-pollinate and reproduce on its own, generating seeds and engendering the next generation. As the pollen grains mature, they become coated in a tough outer layer made of the material sporopollenin. This polymer, Jacobowitz explains, has helped sculpt the terrestrial ecosystem we know today.

Nearly 500 million years ago, the first plants migrated from sea to land, and eventually developed this durable coating to protect their delicate pollen grains from the stresses of living above water, such as UV radiation and desiccation. Today, researchers understand the basic sequence of events required for pollen development, but it’s been historically difficult to identify the genes involved — or even break down the resilient sporopollenin to determine its composition. In December of 2018, Weng lab postdoc Fu-Shuang Li and his team became the first to report the successful degradation of this virtually indestructible material and determine its chemical structure.

“Now that we have a better grasp of what this pollen coating looks like at a molecular level,” says co-author Jacobowitz, “we can improve our understanding of the genes that are already known to produce the pollen wall, and make predictions about new enzymes that also likely contribute.”

Jacobowitz aims to pinpoint which enzymes add certain chemical groups to sporopollenin, as well as the molecular players required for anther development. As he puts it, the general premise of his current project is to “examine genes that no one has looked at before.”

Jacobowitz spent almost a year sifting through online databases to compile a list of enzymes that could potentially play a critical role in anther development. He ordered knockout lines that eliminated each enzyme one at a time, and watched as the plants matured.

At first, nothing happened. Jacobowitz was simply rearing a bunch of normal plants. But then it occurred to him that perhaps nature had built in some redundancy, allowing plants to survive these genetic errors. If one enzyme was incapacitated, another might compensate for the loss and assume its function so development could proceed as usual.

“Even though my screens were pretty unsuccessful at first, I still enjoyed the entire process,” he recalls. “That’s when I started to realize that I really like genetics. There’s always this possibility that you’ll stumble upon a new gene, or a new function of a known gene, that no one ever suspected. That was the opposite of my undergraduate experience in biochemistry, where we drilled down into the intimate details of a single, well-studied enzyme.”With this in mind, Jacobowitz crossed two knockouts together and created a double mutant, simultaneously erasing what he suspected were two relatively similar enzymes. This time, he saw an effect — the walls of the anther began to swell, invading the space containing the pollen and preventing the grains from developing properly. He’d made a sterile plant, indicating that these two enzymes (encoded by the PRX9 and PRX40 genes, respectively) were critical for pollen development

Post-MIT, Jacobowitz is considering pursing a postdoc in genetics. He’s open to studying any organism, so plants aren’t off the table just yet.

“As humans, we rely heavily on plant-based medicines and agricultural products,” he says. “In today’s changing climate, it’s especially important recognize our dependence on plants, and put necessary resources into understanding the basic principles governing their reproductive cycle.” In fact, our own lives could depend on it.

Posted 1.24.19
Sallie “Penny” Chisholm awarded the 2019 Crafoord Prize

Institute Professor honored for discovering <i>Prochlorococcus,</i> the most abundant photosynthesizing organism on Earth.

Allison Dougherty | Department of Civil and Environmental Engineering
January 22, 2019

MIT Institute Professor Sallie “Penny” Chisholm of the departments of Civil and Environmental Engineering and Biology is the recipient of the 2019 Crafoord Prize.

Announced on Jan. 17, Chisholm was awarded the prize “for the discovery and pioneering studies of the most abundant photosynthesizing organism on Earth, Prochlorococcus.”

Prochlorococcus is a NetBet live casinotype of phytoplankton found in the ocean that is able to photosynthesize like plants on land.  The process of photosynthesis is responsible for the oxygen humans breathe, which makes it critical to life on Earth. Prochlorococcus accounts for approximately 10 percent of all ocean photosynthesis, which draws carbon dioxide out of the atmosphere, provides it with oxygen, and forms the base of the food chain.

While the organism is the most abundant photosynthesizer on the planet (the total amount of Prochlorococcus on Earth has been estimated to be 3*1027, or 3,000,000,000,000,000,000,000,000,000), it wasn’t until the mid-1980’s that Prochlorococcus was discovered by Chisholm and colleagues at the Woods Hole Oceanographic Institution. The reason the organism remained unknown for so long can be attributed to its small size. The tiny bacteria is half of a micrometer in size, 1/100 the width of a human hair, making it the smallest photosynthesizing organism.

Since its discovery, Chisholm and her team have found that although each cell has only 2,000 genes, the species as a whole has more than 80,000 different genes in its gene pool, which is four times more than the genetic makeup of humans. This vast diversity of genes distributed among the global population contributes to why Prochlorococcus is able to exist prominently in various environments containing different levels of light, heat, and nutrients.

Chisholm, who has been at MIT since 1976, now studies how Prochlorococcus interacts with various components of seawater and other microorganisms found in the ocean; its role in shaping the ocean ecosystem over evolutionary time; and how its populations may shift in response to climate change.

In April, Chisholm delivered a TED Talk that dove deeper into the properties of Prochlorococcus, comparing the organism’s genetic diversity to iPhone apps, and expanded on the the beauty of this microorganism as the smallest living thing that can convert solar energy and carbon dioxide into fuel through photosynthesis. Understanding its simple design could aid in efforts to engineer artificial photosynthesis machines — reducing our dependency on fossil fuels.

Prochlorococcus has even inspired Chisholm to educate future generations of scientists through a series of children’s books called the “Sunlight Series,” with co-author and illustrator Molly Bang. The series describes the Earth’s natural processes in layman’s terms and through imagery. While none of Chisholm’s books mention Prochlorococcus by name, Chisholm says the simplicity of Prochlorococcus compelled her to create the series.

Chisholm will present her prize lecture in Sweden at Lund University on May 13, and will receive her prize at the Royal Swedish Academy of Sciences prize award ceremony on May 15, in the presence of H. M. King Carl XVI Gustaf and H. M. Queen Silvia of Sweden.

The Crafoord Prize is awarded in partnership between the Royal Swedish Academy of Sciences and the Crafoord Foundation, with the academy responsible for selecting the Crafoord Laureates. Awards are presented in one of four disciplines each year: mathematics and astronomy, geosciences, biosciences, or polyarthritis (such as rheumatoid arthritis).

From microfluidics to metastasis

New platform enables longitudinal studies of circulating tumor cells in mouse models of cancer.

Bendta Schroeder | Koch Institute
January 23, 2019

Circulating tumor cells (CTCs) — an intermediate form of cancer cell between a primary and metastatic tumor cell — carry a treasure trove of information that is critical to treating cancer. Numerous engineering advancements over the years have made it possible to extract cells via liquid biopsy and analyze them to monitor an individual patient’s response to treatment and predict relapse.

Thanks to significant progress toward creating genetically engineered mouse models, liquid biopsies hold great promise for the lab as well. These mouse models mimic many aspects of human tumor development and have enabled informative studies that cannot be performed in patients. For example, these models can be used to trace the evolution of cells from initial mutation to eventual metastasis, a process in which CTCs play a critical role. But since it has not been possible to monitor CTCs over time in mice, scientists’ ability to study important features of metastasis has been limited.

The challenge lies in capturing enough cells to conduct such longitudinal studies. Although primary tumors shed CTCs constantly, the density of CTCs in blood is very low — fewer than 100 CTCs per milliliter. For human patients undergoing liquid biopsy, this does not present a problem. Clinicians can withdraw enough blood to guarantee a sufficient sample of CTCs, just a few milliliters out of five or so liters on average, with minimal impact to the patient.

A mouse, on the other hand, only has about 1.5 milliliters of blood in total. If researchers want to study CTCs over time, they may safely withdraw no more than a few microliters of blood from a mouse each day — nowhere near enough to ensure that many (or any) CTCs are collected.

But with a new approach developed by researchers at the Koch Institute for Integrative Cancer Research, it is now possible to collect CTCs from mice over days and even weeks, and analyze them as the disease progresses. The system, described in the Proceedings of the National Academy of Sciences the week of Jan. 21, diverts blood to a microfluidic cell-sorting chip that extracts individual CTCs before returning the blood back to an awake mouse.

A menu of sorts

The inspiration for the project was cooked up, not in the lab, but during a chance encounter in the Koch Café between Tyler Jacks, director of the Koch Institute and the David H. Koch Professor of Biology, and Scott Manalis, the Andrew and Erna Viterbi Professor in the departments of Biological Engineering and Mechanical Engineering and a member of the Koch Institute.

As luck and lunch lines would have it, the pair would discuss thesis work being done by then-graduate student Shawn Davidson, who was using a dialysis-like system to track metabolites in the bloodstream of mice in the laboratory of Matthew Vander Heiden, an associate professor of biology. Jacks and Manalis were inspired: Could a similar approach could be used to study rare CTCs in real time?

Along with their Koch Institute colleague Alex K. Shalek, the Pfizer-Laubach Career Development Assistant Professor of Chemistry and a core member of the Institute for Medical Engineering and Science (IMES) at MIT, it would take Jacks and Manalis more than five years to put all the pieces of the system together, drawing from different areas of expertise around the Koch Institute. The Jacks lab supplied its fluorescent small cell lung cancer model, the Manalis lab developed the real-time CTC isolation platform, and the Shalek lab provided genomic profiles of the collected CTCs using single-cell RNA sequencing.

“This is a project that could not have succeeded without a sustained effort from several labs with very different sets of expertise. For my lab, which primarily consists of engineers, the opportunity to participate in this type of research has been incredibly exciting and is the reason why we are in the Koch Institute,” Manalis says.

The CTC sorter uses laser NetBet live casinoexcitation to identify tumor cells expressing a fluorescent marker that is incorporated in the mouse model. The system draws blood from the mouse and passes it through a microfluidic chip to detect and extract the fluorescing CTCs before returning the blood back to the mouse. A minute amount of blood — approximately 100 nanoliters — is diverted with every detected CTC into a collection tube, which then is purified further to extract individual CTCs from the thousands of other blood cells.

“The real-time detection of CTCs happens at a flow rate of approximately 2 milliliters per hour which allows us to scan nearly the entire blood volume of an awake and moving mouse within an hour,” says Bashar Hamza, a graduate student in the Manalis lab and one of the lead authors on the paper.

Biology in their blood

With the development of a real-time cell sorter, the researchers could now, for the first time, longitudinally collect CTCs from the same mouse.

Previously, the low blood volume of mice and the rarity of CTCs meant that groups of mice had to be sacrificed at successive times so that their CTCs could be pooled. However, CTCs from different mice often have significantly different gene expression profiles that can obscure subtle changes that occur from the evolution of the tumor or a perturbation such as a drug.

To demonstrate that their cell-sorter could capture these differences, the researchers treated mice with a compound called JQ1, which is known to inhibit the proliferation of cancer cells and perturb gene expression. CTCs were collected and profiled with single-cell RNA sequencing for two hours prior to the treatment, and then every 24 hours after the initial treatment for four days.

When the researchers pooled data for all mice that had been treated with JQ1, they found that the data clustered based on individual mice, offering no confirmation that the drug affects CTC gene expression over time. However, when the researchers analyzed single-mouse data, they observed gene expression shift with time.

“What’s so exciting about this platform and our approach is that we finally have the opportunity to comprehensively study longitudinal CTC responses without worrying about the potentially confounding effects of mouse-to-mouse variability. I, for one, can’t wait to see what we will be able to learn as we profile more CTCs, and their matched primary and metastatic tumors,” says Shalek.

Researchers believe their approach, which they intend to use in additional cancer types including non-small cell lung, pancreatic, and breast cancer, could open new avenues of inquiry in the study of CTCs, such as studying long-term drug responses, characterizing their relationship to metastatic tumors, and measuring their rate of production in short timeframes — and the entire metastatic cascade. In future work, researchers also plan to use their approach for profiling rare immune cells and monitoring cells in dynamic contexts such as wound healing and tumor formation.

“The ability to study CTCs as well other rare cells in the blood longitudinally gives us a powerful view into cancer development. This sorter represents a real breakthrough for the field and it is a great example of the Koch Institute in action,” says Jacks.

The paper’s other co-lead authors are graduate students Sheng Rong Ng from the Jacks lab and Sanjay Prakadan from the Shalek lab. The research is supported, in part, by the Ludwig Center at MIT, the National Cancer Institute, the National Institutes of Health and the Searle Scholars Program.

Study shows how specific gene variants may raise bipolar disorder risk

Findings could help inform new therapies, improve diagnosis.

David Orenstein | Picower Institute for Learning and Memory
January 18, 2019

A new study by researchers at the Picower Institute for Learning and Memory at MIT finds that the protein CPG2 is significantly less abundant in the brains of people with bipolar disorder (BD) and shows how specific mutations in the SYNE1 gene that encodes the protein undermine its expression and its function in neurons.

Led by Elly Nedivi, professor in MIT’s departments of Biology and Brain and Cognitive Sciences, and former postdoc Mette Rathje, the study goes beyond merely reporting associations between genetic variations and psychiatric disease. Instead, the team’s analysis and experiments show how a set of genetic differences in patients with bipolar disorder can lead to specific physiological dysfunction for neural circuit connections, or synapses, in the brain.

The mechanistic detail and specificity of the findings provide new and potentially important information for developing novel treatment strategies and for improving diagnostics, Nedivi says.

“It’s a rare situation where people have been able to link mutations genetically associated with increased risk of a mental health disorder to the underlying cellular dysfunction,” says Nedivi, senior author of the study online in Molecular Psychiatry. “For bipolar disorder this might be the one and only.”

The researchers are not suggesting that the CPG2-related variations in SYNE1 are “the cause” of bipolar disorder, but rather that they likely contribute significantly to susceptibility to the disease. Notably, they found that sometimes combinations of the variants, rather than single genetic differences, were required for significant dysfunction to become apparent in laboratory models.

“Our data fit a genetic architecture of BD, likely involving clusters of both regulatory and protein-coding variants, whose combined contribution to phenotype is an important piece of a puzzle containing other risk and protective factors influencing BD susceptibility,” the authors wrote.

CPG2 in the bipolar brain

During years of fundamental studies of synapses, Nedivi discovered CPG2, a protein expressed in response to neural activity, that helps regulate the number of receptors for the neurotransmitter glutamate at excitatory synapses. Regulation of glutamate receptor numbers is a key mechanism for modulating the strength of connections in brain circuits. When genetic studies identified SYNE1 as a risk gene specific to bipolar disorder, Nedivi’s team recognized the opportunity to shed light into the cellular mechanisms of this devastating neuropsychiatric disorder typified by recurring episodes of mania and depression.

For the new study, Rathje led the charge to investigate how CPG2 may be different in people with the disease. To do that, she collected samples of postmortem brain tissue from six brain banks. The samples included tissue from people who had been diagnosed with bipolar disorder, people who had neuropsychiatric disorders with comorbid symptoms such as depression or schizophrenia, and people who did not have any of those illnesses. Only in samples from people with bipolar disorder was CPG2 significantly lower. Other key synaptic proteins were not uniquely lower in bipolar patients.

“Our findings show a specific correlation between low CPG2 levels and incidence of BD that is not shared with schizophrenia or major depression patients,” the authors wrote.

From there they used deep-sequencing techniques on the same brain samples to look for genetic variations in the SYNE1 regions of BD patients with reduced CPG2 levels. They specifically looked at ones located in regions of the gene that could regulate expression netbet sports betting appof CPG2 and therefore its abundance.

Meanwhile, they also combed through genomic databases to identify genetic variants in regions of the gene that code CPG2. Those mutations could adversely affect how the protein is built and functions.

Examining effects

The researchers then conducted a series of experiments to test the physiological consequences of both the regulatory and protein coding variants found in BD patients.

To test effects of non-coding variants on CPG2 expression, they cloned the CPG2 promoter regions from the human SYNE1 gene and attached them to a “reporter” that would measure how effective they were in directing protein expression in cultured neurons. They then compared these to the same regions cloned from BD patients that contained specific variants individually or in combination. Some did not affect the neurons’ ability to express CPG2 but some did profoundly. In two cases, pairs of variants (but neither of them individually), also reduced CPG2 expression.

Previously Nedivi’s lab showed that human CPG2 can be used to replace rat CPG2 in culture neurons, and that it works the same way to regulate glutamate receptor levels. Using this assay they tested which of the coding variants might cause problems with CPG2’s cellular function. They found specific culprits that either reduced the ability of CPG2 to locate in the “spines” that house excitatory synapses or that decreased the proper cycling of glutamate receptors within synapses.

The findings show how genetic variations associated with BD disrupt the levels and function of a protein crucial to synaptic activity and therefore the health of neural connections. It remains to be shown how these cellular deficits manifest as biopolar disorder.

Nedivi’s lab plans further studies including assessing behavioral implications of difference-making variants in lab animals. Another is to take a deeper look at how variants affect glutamate receptor cycling and whether there are ways to fix it. Finally, she said, she wants to continue investigating human samples to gain a more comprehensive view of how specific combinations of CPG2-affecting variants relate to disease risk and manifestation.

In addition to Rathje and Nedivi, the paper’s other authors are Hannah Waxman, Marc Benoit, Prasad Tammineni, Costin Leu, and Sven Loebrich.

The JPB Foundation, the Gail Steel Fund, the Carlsberg Foundation, the Lundbeck Foundation and the Danish Council for Independent Research funded the study.