Reading and writing DNA

Department of Biology kicks off IAP seminar series with a lecture by synthetic-biology visionary George Church.

Raleigh McElvery | Department of Biology
January 31, 2018

Thanks to the invention of genome sequencing technology more than three decades ago, we can now read the genetic blueprint of virtually any organism. After the ability to read came the ability to edit — adding, subtracting, and eventually altering DNA wherever we saw fit. And yet, for George Church, a professor at Harvard Medical School, associate member of the Broad Institute, and founding core faculty and lead for synthetic biology at the Wyss Institute — who co-pioneered direct genome sequencing in 1984 — the ultimate goal is not just to read and edit, but also to write.

What if you could engineer a cell resistant to all viruses, even the ones it hadn’t yet encountered? What if you could grow your own liver in a pig to replace the faulty one you were born with? What if you could grow an entire brain in a dish? In his lecture on Jan. 24 — which opened the Department of Biology’s Independent Activities Period (IAP) seminar series, Biology at Transformative Frontiers — Church promised all this and more.

“We began by dividing the Biology IAP events into two tracks: one related to careers in academia and another equivalent track for industry,” says Jing-Ke Weng, assistant professor and IAP faculty coordinator for the department. “But then it became clear that George Church, Patrick Brown, and other speakers we hoped to invite blurred the boundaries between those two tracks. The Biology at Transformative Frontiers seminar series became about the interface of these trajectories, and how transferring technologies from lab bench to market is altering society as we know it.”

The seminar series is a staple in the Department of Biology’s IAP program, but during the past several years it has been oriented more toward quantitative biology. Weng recalls these talks as being relegated to the academic sphere, and wanted to show students that the lines between academia, industry, and scientific communication are actually quite porous.

“We chose George Church to kick off the series because he’s been in synthetic biology for a long time, and continues to have a successful academic career even while starting so many companies,” says Weng.

Church’s genomic sequencing methods inspired the Human Genome Project in 1984 and resulted in the first commercial genome sequence (the bacterium Helicobacter pylori) 10 years later. He also serves as the director of the Personal Genome Project, the “Wikipedia” of open-access human genomic data. Beyond these ventures, he’s known for his work on barcoding, DNA assembly from chips, genome editing, and stem cell engineering.

He’s also the same George Church who converted the book he co-authored with Ed Regis, “Regenesis: How Synthetic Biology Will Reinvent Nature and Ourselves,” into a four-letter code based on the four DNA nucleotides (A, T, C, and G), subsisted on nutrient broth from a lab vendor for an entire year, and dreams of eventually resurrecting woolly mammoths. He’s being featured in an upcoming Netflix Original documentary, so when he arrived at the Stata Center to give his lecture last week he was trailed by a camera crew.

According to Church, the transformative technologies that initially allowed us to read and edit DNA have grown exponentially in recent years with the invention of molecular multiplexing and CRISPR-Cas9 (think Moore’s Law but even more exaggerated). But there’s always room for improvement.

“There’s been a little obsession with CRISPR-Cas9s and other CRISPRs,” said Church. “Everybody is saying how great it is, but it’s important to say what’s wrong with it as well, because that tells us where we’re going next and how to improve on it.”

He outlined several of his own collaborations, including those aimed at devising more precise methods of genome editing, one resulting in 321 changes to the Escherichia coli genome — the largest change in any genome yet — rendering the bacterium resistant to all viruses, even those it had not yet come into contact with. The next step? Making similarly widespread changes in plants, animals, and eventually perhaps even human tissue. In fact, Church and his team have set their sights on combatting the global transplantation crisis with humanlike organs grown in animals.

“Since the dawn of transplantation as a medical practice, we’ve had to use either identical twins or rare matches that are very compatible immunologically, because we couldn’t engineer the donor or the recipient,” said Church.

Since it’s clearly unethical to engineer human donors, Church reasoned, why not engineer animals with compatible organs instead? Pigs, to be exact, since most of their organs are comparable in size and function to our own.

“This is an old dream; I didn’t originate it,” said Church. “It started about 20 years ago, and the pioneers of this field worked on it for a while, but dropped it largely because the number of changes to the genome were daunting, and there was a concern that the viruses all pigs make — retroviruses — would be released and infect the immunocompromised organ recipient.”

Church and his team successfully disrupted 62 of these retroviruses in pig cells back in 2015, and in 2017 they used these cells to generate living, healthy pigs. Today, the pigs are thriving and rearing piglets of their own. Church is also considering the prospect of growing augmented organs in pigs for human transplantation, perhaps designing pathogen-, cancer-, and age-resistant organs suitable for cryopreservation.

“Hopefully we’ll be doing nonhuman primate trials within a couple of years, and then almost immediately after that human trials,” he said.

Another possibility, rather than cultivating organs in animals for transplant, is to generate them in a dish. A subset of Church’s team is working on growing from scratch what is arguably the most complicated organ of all, the brain.

This requires differentiating multiple types of cells in the same dish so they can interact with each other to form the complex systems of communication characteristic of the human brain.

Early attempts at fashioning brain organoids often lacked capillaries to distribute oxygen and nutrients (roughly one capillary for each of the 86 billion neurons in the human brain). However, thanks to their new human transcription factor library, Church and colleagues have begun to generate the cell types necessary to create such capillaries, plus the scaffolding needed to promote the three-dimensional organization of these and additional brain structures. Church and his team have not only successfully integrated the structures with one another, but have also created an algorithm that spits out the list of molecular ingredients required to generate each cell type.

Church noted these de novo organoids are extremely useful in determining which genetic variants are responsible for certain diseases. For instance, you could sequence a patient’s genome and then create an entire organoid with the mutation in question to test whether it was the root cause of the condition.

“I’m still stunned by the breadth of projects and approaches that he’s running simultaneously,” says Emma Kowal, a second-year graduate student, member of Weng’s planning committee, and a former researcher in Church’s lab. “The seminar series is called Biology at Transformative Frontiers, and George is very much a visionary, so we thought it would be a great way to start things off.”

The four-part series also features Melissa Moore, chief scientific officer of the Moderna Therapeutics mRNA Research Platform, Jay Bradner, president of the Novartis Institutes for BioMedical Research, and Patrick Brown, CEO and founder of Impossible Foods.

How some facial malformations arise

Study explains why mutations that would seemingly affect all cells lead to face-specific birth defects.

Anne Trafton | MIT News Office
January 24, 2018

About 1 in 750 babies born in the United States has some kind of craniofacial malformation, accounting for about one-third of all birth defects.

Many of these craniofacial disorders arise from mutations of “housekeeping” genes, so called because they are required for basic functions such as building proteins or copying DNA. All cells in the body require these housekeeping genes, so scientists have long wondered why these mutations would produce defects specifically in facial tissues.

Researchers at MIT and Stanford University have now discovered how one such mutation leads to the facial malformations seen in Treacher-Collins Syndrome, a disorder that affects between 1 in 25,000 and 1 in 50,000 babies and produces underdeveloped facial bones, especially in the jaw and cheek.

The team found that embryonic cells that form the face are more sensitive to the mutation because they more readily activate a pathway that induces cell death in response to stress. This pathway is mediated by a protein called p53. The new findings mark the first time that scientists have determined how mutations in housekeeping genes can have tissue-specific effects during embryonic development.

“We were able to narrow down, at the molecular level, how issues with general regulators that are used to make ribosomes in all cells lead to defects in specific cell types,” says Eliezer Calo, an MIT assistant professor of biology and the lead author of the study.

Joanna Wysocka, a professor of chemical and systems biology at Stanford University, is the senior author of the study, which appears in the Jan. 24 online edition of Nature.

From mutation to disease

Treacher-Collins Syndrome is caused by mutations in genes that code for proteins required for the assembly and function of polymerases. These proteins, known as TCOF1, POLR1C, and POLR1D, are responsible for transcribing genes that make up cell organelles called ribosomes. Ribosomes are critical to all cells.

“The question we were trying to understand is, how is it that when all cells in the body need ribosomes to function, mutations in components that are required for making the ribosomes lead to craniofacial disorders? In these conditions, you would expect that all the cell types of the body would be equally affected, but that’s not the case,” Calo says.

During embryonic development, these mutations specifically affect a type of embryonic cells known as cranial neural crest cells, which form the face. The researchers already knew that the mutations disrupt the formation of ribosomes, but they didn’t know exactly how this happens. To investigate that process, the researchers engineered larvae of zebrafish and of an aquatic frog known as Xenopus to express proteins harboring those mutations.

Their experiments revealed that the mutations lead to impairment in the function of an enzyme called DDX21. When DDX21 dissociates from DNA, the genes that encode ribosomal proteins do not get transcribed, so ribosomes are missing key components and can’t function normally. However, this DDX21 loss only appears to happen in cells that are highly sensitive to p53 activation, including cranial neural crest cells. These cells then undergo programmed cell death, which leads to the facial malformations seen in Treacher-Collins Syndrome, Calo says.

Other embryonic cells, including other types of neural crest cells, which form nerves and other parts of the body such as connective tissue, are not affected by the loss of DDX21.

Role of DNA damage

The researchers also found that mutations of POLR1C and POLR1D also cause damage to stretches of DNA that encode some of the RNA molecules that make up ribosomes. The amount of DNA damage correlated closely with the severity of malformations seen in individual larvae, and mutations in POLR1C led to far more DNA damage than mutations in POLR1D. The researchers believe these differences in DNA damage may explain why the severity of Treacher-Collins Syndrome can vary widely among individuals.

Calo’s lab is now studying why affected cells experience greater levels of DNA damage in those particular sequences. The researchers are also looking for compounds that could potentially prevent craniofacial defects by making the cranial neural crest cells more resistant to p53-induced cell death. Such interventions could have a big impact but would have to be targeted very early in embryonic development, as the cranial neural crest cells begin forming the tissue layers that will become the face at about three weeks of development in human embryos.

The research was funded by the National Institutes of Health, Howard Hughes Medical Institute, and March of Dimes Foundation.

December 14, 2017

Lab coat meets legislation

Person with red hair in bun stands outside with umbrella.

Undergraduate Courtney Diamond combines biology and policy to tackle real-world challenges

Raleigh McElvery

 

Undergraduate Courtney Diamond arrived at MIT determined to be an oncologist. Five years later, she’s leaving with a broader focus on human health, grappling with real-world, biomedical problems by way of public policy rather than medicine or research.

Although Diamond had completed her requirements for a degree in biology at the beginning of her senior year, she decided then to add a second major: Course 17 (Political Science), and with it a fifth year of study at MIT.

“I came into MIT wanting to be a doctor, but the more I thought about it the less it felt like medical school would be a good fit,” she says. “I spent a long time narrowing my interests within the realm of human health, and recently realized there was another dimension to that interest related to public policy, which was also this common thread among my extracurriculars.”

Diamond grew up in a small town in Massachusetts called Millbury, not too far from MIT, which she describes as special to her but “rather unremarkable” in most other ways — with the exception of one particularly zealous and articulate high school biology teacher. His infectious enthusiasm sparked Diamond’s passion for the life sciences, but over the course of her senior year this interest became far more personal. It was around that time that her mother developed breast cancer, and Diamond resolved to be an oncologist.

“My mom had been diagnosed once before with a different kind of cancer, cervical cancer,” she says. “But I was in sixth grade back then, and assumed she was just at home resting. By the time the breast cancer rolled around, I was old enough to understand that most people are lucky to survive cancer once. But twice?”

Her mother has since entered remission, and the year Diamond began at MIT her interests matured away from a career in medicine and towards biomedical research. In April 2014, she applied to the MIT Undergraduate Research Opportunities Program (UROP). “I wanted to figure out which part of biology excited me — which area I really wanted to drill down on,” she recalls.

She began working with a postdoctoral fellow in Professor Darrell Irvine’s lab at the Koch Institute for Integrative Cancer Research, tackling research questions related to cancer immunology. Diamond’s job was to analyze murine tumors as they developed over time, in order to understand how they were affected by changes to their cellular environments.

After a year, Diamond took a break from research in order to focus on her classes. But she didn’t stay away for long.

“I’ve had a life-long obsession with Australia,” she says, “and in the fall of my sophomore year, I told my advisor, Professor Bob Horvitz, that my dream was to study biochemistry in Melbourne.” One email and two hours later, she received an offer from the Walter and Eliza Hall Institute for Medical Research to spend a summer abroad in Jeff Babon’s lab. “It turns out the director of the Institute did his postdoc at MIT, and liked the UROP system so much he decided to bring it back to Australia,” she explains.

There, Diamond helped to unravel the structure of a protein complex known as JAK-STAT. This complex is involved in many diverse processes — from cell proliferation and programmed cell death to immunity — making it critical to understand how the different molecular components of the complex fit together to influence function.

When she returned to MIT, Diamond decided to maintain her focus on structural biology. She completed her thesis in Professor Thomas Schwartz’s lab, studying the Y complex, a component of the nuclear pore — a channel that allows mRNA and other molecules to pass into the cell’s nucleus. Diamond helped creat a library of fluorescing antibodies that could adhere to the Y complex, allowing her to visualize its position within the nuclear pore. After a year, she opted to broaden her interests by taking classes outside her major.

One of those classes, recommended by a friend, was in political science: 17.309 (Science, Technology, and Public Policy), taught by Professor Kenneth Oye. During one of his lectures, Oye made a quip about a small Massachusetts town called Millbury.

“I came up to him after class to ask him, ‘Did you know I’m actually from there?’ and he thought it was the funniest thing,” she says. “That initial, informal interaction led to more meaningful conversations, and I ended up working with him on a few projects.”

Today, she is pursuing a final UROP with Oye, looking at technologies and policies related to synthetic biology. At Oye’s weekly working group of graduate students and postdocs, she debates the possible repercussions of using gene editing techniques like CRISPR-Cas9 to control the transmission of certain traits throughout a given population. For example, what would happen if mosquitos in the regions where malaria is most prevalent carried a gene encoding malaria resistance — would that eradicate the illness? But might there be unintended, negative consequences?

As part of a separate project, Diamond is researching U.S. consent and privacy policies in the realm of health information technology. She’s also hard at work on her political science thesis, focusing on ways to incentivize companies and researchers to develop new and more effective antibiotics to combat antimicrobial resistance.

Diamond is now applying for public health consulting jobs, and she plans to pursue graduate training in epidemiology, followed by a master’s in public health. Long-term, she sees herself at the Centers for Disease Control and Prevention or the World Health Organization.

“I mean, that’s the current plan,” she says. “Check back in with me in two years.”

Photo credit: Raleigh McElvery
From DNA forensics to cancer metabolism

Carolyn Lanzkron discovered bench science while attending community college with her son, and followed her newfound passion to MIT

Raleigh McElvery
December 3, 2017

From DNA forensics to cancer metabolism

Person in black hat and purple shirt sitting in front of lab building.

Carolyn Lanzkron discovered bench science while attending community college with her son, and followed her newfound passion to MIT

Raleigh McElvery

 

Carolyn Lanzkron spent 20 years as a stay-at-home mother raising five children before starting at MIT. Life has taught her patience, which she, in turn, has tried to pass on to her kids: “A successful person falls down many times and gets up — just pick a direction and move forward.”

Those were the same words she told her teenage son back in 2011 when she encouraged him to attend community college.

“I figured I would just take a few courses with him,” she says. “He enjoyed his chemistry classes, so I was looking at the chemistry offerings, and on the wall there was a poster for Dr. Bruce Jackson’s unique Forensic DNA Science program.” Lanzkron was intrigued, and decided to enroll.

The students aided Jackson with real cases, and were given dedicated lab space and materials to follow their curiosities, as well as design their own inquiries. The program was based on a peer-mentoring model, and Lanzkron was appointed chief of peer mentors and forensic case manager. Under Jackson’s tutelage, she worked on lineage cases tracing ancestry and criminal cases for defense and prosecution.

“I was hoping my son would join me in a chemistry class, but he wasn’t so interested in having his mom as a lab partner — go figure,” she says. “But we carpooled to school together for a year, and by that time I’d developed a love for bench science.”

After two years, Lanzkron had completed her degree, but it wasn’t enough. So she applied to several institutions within her carpool radius, including MIT. Like all transfers here, she began as a sophomore.

“I love bench science because I really appreciate the combination of being part of a team and solving a big, important question, but at the same time having tasks in my day that allow me to focus on small details — like keeping track of the labels on my tubes,” she says. “That balance works really well for me; it satisfies my need for a quest while still having control over a small environment.”

She’s turned her attention from DNA forensics to cancer metabolism, an interest which has become far more personal over the past year. Last spring, Lanzkron’s mother was diagnosed with lung cancer, and Lanzkron took a leave of absence to care for her.

“Right now, my mother is doing really well, and we are enjoying a window of stability,” Lanzkron says, “which has allowed me to come back to MIT and finish my degree.”

Although Lanzkron is not currently in a lab, lest that period of stability suddenly end, she’s worked in several over the course of her three years at MIT. She began in Jean Francois Hamel’s chemical engineering lab, adapting an adherent cell line to grow in a suspension-like culture in various bioreactors using microcarriers.

Later, Lanzkron joined David Sabatini’s lab in the Whitehead Institute for Biomedical Research, aiding two separate projects: one spearheaded by then-postdoc Yoav Shaul, and the other led by MD-PhD student Walter Chen.

Chen was hard at work developing a new method for profiling undamaged mitochondria, while Shaul had discovered a unique set of 44 metabolic genes that were upregulated in certain cancers that expressed mesenchymal markers (which he called the “Mesenchymal Metabolic Signature,” or “MMS”), indicating that those cells were acquiring cancerous characteristics. Lanzkron collaborated with Shaul as he worked to further characterize the metabolic requirements and behavior of the MMS. She also helped him refine his web-based gene analysis tool, Metabolic gEne RApid Visualizer (MERAV), which queries a database comprising ∼4,400 microarrays, representing human gene expression in normal tissues, cancer cell lines, and primary tumors.

The summer after Shaul completed his postdoctoral training, Lanzkron interned in his lab in at the Hebrew University of Jerusalem at Hadassah Ein Kerem through the MISTI/Israel program, to continue working with him on these projects.

“When I went to Israel, my husband stayed in Boston and took care of the kids,” she recalls. “Without family responsibilities, I could work in lab around the clock, and that was great. I was actually able to finish things up, prepare them for the next day, and cover for other people and really focus; I look forward to being able to do that again as the kids get older.”

Lanzkron admits these aren’t the only aspects of the MIT undergraduate experience she’s missed — not just because she lives off campus and can’t meet at odd hours of the night to collaborate on problem sets — but also because she’s a generation and a half older than her classmates.

But in some ways she considers this an advantage. For instance, she now has the tools to guide her own children through today’s college process.

“I no longer have this outdated view of what it’s like to apply to schools and navigate the SAT,” she says. “Granted, MIT is not your average school. It’s been quite the ride to be at the community college where I had to bring my own masking tape to complete the gel trays because we didn’t have any sealing rings — I didn’t even know there was such a thing as a seal back then. And to go from that to the MIT Department of Biology and the Whitehead Institute where the resources are phenomenal, it’s just mind blowing. I have learned so much from both situations — having to make do, and having an abundance of resources.”

While Lanzkron intends to graduate this spring, her future plans depend on her mother’s health.

“I picked my classes this semester so that I could take her to her cancer treatment,” Lanzkron says, “so, though I’m ultimately planning to go to graduate school, right now things are still in flux.”

While maintaining this school-family balance would be inconceivable for most, Lanzkron takes her personal and academic responsibilities in stride.

“Honestly I’m so happy here at MIT,” she says. “I tell my kids, ‘Don’t get too worked up about the college process. You’ll get where you need to go — the starting point almost doesn’t matter; what matters is what you do when you get there.’”

Photo credit: Raleigh McElvery
Sizing up cancer

Graduate student Zhaoqi Li investigates how cancer cells grow by harnessing exceptional chemical reactions

Justin Chen
January 11, 2018

Cancer cells use extreme measures to fuel their growth. In fact, researchers like Zhaoqi Li, a third-year graduate student, witness chemical reactions in these cells that would be impossible in the context of normal cells. In a petri dish, normal cells stop dividing once they cover the bottom of the dish and fit neatly together like mosaic tiles. In contrast, cancer cells continue to proliferate and pile haphazardly into small mounds. Within the human body, this abnormal growth — when combined with the spread of cancer cells throughout the body — interferes with organ function and causes death.

Li, a member of Professor Matthew Vander Heiden’s lab located in the Koch Institute, studies cancer metabolism. His work describes the chemical reactions cancer cells use to create energy and materials to make new cells such as membranes, proteins, and DNA. By tracking the flow of nutrients through cancer cells, Li and his labmates are learning how such cells change their metabolism to stimulate growth. These insights will help scientists develop new ways to treat the disease.

Cell metabolism comprises all the chemical reactions occurring in the cell, but researchers are particularly interested in a few reactions that aren’t required by normal cells but are critical for cancer growth. Stopping these reactions with drugs would disrupt the metabolism of cancer cells and hinder tumor development.

“Even though many people may not think of metabolism as a treatment target for cancer, this strategy has been used unwittingly for a long time,” Li says. “Many chemotherapies, such as antifolates, were originally used by doctors without knowing exactly how they worked. Since then, we’ve discovered that those treatments target metabolic pathways. By understanding the details of cancer metabolism we are hoping to design drugs in a more rational way.”

– –

Li might never have joined the Vander Heiden lab or studied cancer metabolism were it not for the unique structure of graduate training at MIT.

During their first year at MIT, graduate students are required to take four classes. Unlike their counterparts at many other PhD programs, they do not work in laboratories until their second semester. This allows students to focus initially on coursework — covering biochemistry, genetics, and research methodology — designed to build a foundation of knowledge. As a result, students discover new interests and develop the confidence to move out of their comfort zones. When it comes time to select a lab, they can choose from 56 spread across six locations, spanning a wide breadth of biological research.

Li could study how the brain forms memories, interpret X-rays to deduce protein structure, or even build miniature organs for drug testing. Before making his decision, he rotated in three laboratories. During each month-long rotation, he performed a small project allowing him to experience the culture of the lab and learn more about its research.

“The first two labs I visited were studying topics I was familiar with and thought were interesting,” he says. “But when I visited the Vander Heiden lab it was so different and caught me off guard. That’s why I eventually joined, even though I had never imagined myself working in a metabolism lab before.”

Diagram showing a metabolism pathway
Cellular metabolism is comprised of a network of interconnected biochemical reactions resembling a subway system. Zhaoqi Li compares normal and diseased cells to determine the differences in the way nutrients travel through this network. Credit: Justin Chen

– –

Although he is new to the community of researchers specializing in metabolism, Li has long known that he wanted to interact with the world through science. As an immigrant who moved from China to southern Tennessee at the age of six, Li struggled to learn English and began to view science as a universal language that transcended culture.

“My parents were also non-native speakers and the English as a Second Language classes in my elementary school were geared towards Spanish speakers, so I had a really hard time,” Li says. “I joke that the only reason I passed the first grade was because I was good at math.”

Li’s contrasting relationship with science and English continued as an undergraduate at Columbia University. There he majored in biochemistry and also studied literature of the Western Canon to fulfill his general degree requirements.

“I took four semesters worth of classes that started with Plato and ended with Virginia Woolf,” he says, “It was an eye-opening experience, but I never really loved it. I found biology more intuitive because it doesn’t rely on being familiar with a specific cultural lens. Most every society in the world values the scientific method to some extent.”

Li began working in a lab during his sophomore year at Columbia. To his surprise, he was mentored by a professor who valued his input and encouraged creative thinking. Li’s supervisor also introduced him to basic science — a type of research driven not by the desire to find a specific answer or cure, but by curiosity and the need to better understand the natural world.

– –

During his second semester rotation at MIT, Li searched for similarly open-minded environments, and was attracted to cancer metabolism because the field was relatively young.

“In other more established areas of biology, if you have a question someone has probably answered it in some capacity,” Li says. “The Vander Heiden lab was using new techniques so there was a lot of space to explore. Many questions I asked — even during my initial rotation —  didn’t have an answer, which was exciting.”

The great challenge confronting the metabolism field is translating decades’ worth of research on enzymes — proteins that manage chemical reactions — from the test tube to the cell and human body. By studying enzymes individually in the controlled setting of test tubes, researchers have documented almost all the chemical reactions that occur in the cell. When combined, these reactions look like a giant subway map where each stop, indicated by a dot, is a different molecule, and the line between stops represents a chemical reaction where atoms are added or subtracted. Some pathways are a straight line but others have nodes or intersections where a molecule can take part in several different reactions. Other pathways are circular where the molecule that starts the pathway is remade at the end so that the line circles back on itself.

Despite the ability to study chemical reactions in a test tube, scientists have struggled to understand what is actually happening in the complex environment of cells, which coordinate millions of reactions that not only affect each other, but are also influenced by outside stresses like nutrient deprivation.

To Li, using the metabolism map to figure out what chemical reactions are occurring and how atoms are moving through the cell is like using a subway map to track how people are traveling through a city.

“The map describes all the possible routes people could take,” Li says, “but you have to track the passengers to figure out where they are actually going. You could imagine people commuting into the city during the week and going to entirely different places on the weekend. There are a lot of different patterns of movement that you can’t infer just from looking at a map.”

To analyze what chemical reactions are occurring in the cell, Li utilizes cutting edge technology to track carbon atoms — an essential element that is required to build all components of the cell. By tagging carbon with an extra neutron, Li makes the experimentally altered atom heavier and distinguishable from naturally occurring carbon in the cell. Feeding cells nutrients like glucose made with heavy carbons allows Li to compare how molecules are broken down and used by normal and cancerous cells.

Person at lab instrument with sample“Returning to the subway map analogy, this labeling technique is similar to not only being inside the subway, but also giving everyone in Downtown Boston a red shirt,” Li says. “After 12 hours, we can look at the rest of the city. If we see a lot of red shirts in Allston, we would know that this particular route is really popular.”

In the case of glucose, Li and his labmates observed that normal cells break down the sugar to release energy and heavy carbons in the form of carbon dioxide. In contrast, cancer cells alter their metabolism so that the heavy carbons originally found in glucose are used to build new parts of the cells that are required for cancer cells to grow, such as membranes, DNA, and proteins.

Li’s observations demonstrate how cancer cells sustain abnormal growth by accumulating carbon. For his thesis project, Li has chosen to investigate one of the main tricks cancer cells use to hoard carbon atoms: a process known as carbon fixation. This type of chemical reaction, originally studied in plants performing photosynthesis, attaches carbon dioxide to other molecules. Li’s initial findings suggest that a protein, Malic Enzyme 1, helps cancer cells use carbon dioxide to build components required for growing and dividing.

“This is surprising,” he says, “because the textbook version of this enzyme actually catalyzes the reverse reaction in normal cells where carbon dioxide is removed from molecules.  Malic Enzyme 1 is an example of how cancer performs remarkable chemical reactions — who would have thought that cancer cells use carbon like plants do?”

Li is at the beginning stages of his research, and can’t predict where his project will take him. His current goal is to determine how cancer cells react when they are missing Malic Enzyme 1. Such loss could slow growth, but Li will have to perform experiments to be sure, since cancer is a resourceful and elusive target.

Like a detour rerouting travelers around a closed metro stop, cancer cells may further contort their metabolism, taking advantage of little-used or still unidentified chemical reactions to maintain growth. In the face of such adaptability, Li and his labmates believe the best course of action is to be as curious as possible to understand as much as they can about how cancer works. Working together, they discuss confounding results, adjust hypotheses, and design new experiments.

“It’s really encouraging to be part of Matt’s lab and the Koch Institute in general where researchers take a basic science approach,” Li says. “We try to keep an open mind because there’s probably no single thing that cancer cells depend on. Everyone’s work builds together to form a cumulative understanding.”

Photo credit: Raleigh McElvery
Chris A. Kaiser

Education

  • PhD, 1987, MIT

Research Summary

The Kaiser lab studied protein folding and intracellular trafficking in the yeast S. cerevisiae. Their work focused on the protein folding in the endoplasmic reticulum (ER), quality control mechanisms in the ER, and membrane protein sorting in Golgi compartments. They combined genetic, biochemical, and cell biological methods to gain an understanding of the molecular mechanisms underlying each of these processes. Chris Kaiser is no longer accepting students.

Douglas Lauffenburger

Education

  • PhD, 1979, University of Minnesota
  • BS, 1975, Chemical Engineering, University of Illinois, Urbana-Champaign

Research Summary

The Lauffenburger laboratory emphasizes integration of experimental and mathematical/computational analysis approaches, toward development and validation of predictive models for physiologically-relevant behavior in terms of underlying molecular and molecular network properties. Our work has been recognized as providing contributions fostering the interface of bioengineering, quantitative cell biology, and systems biology. Our main focus has been on fundamental aspects of cell dysregulation, complemented by translational efforts in identifying and testing new therapeutic ideas. Applications addressed have chiefly resided in various types of cancer (including breast, colon, lung, and pancreatic cancers along with leukemias and lymphomas), inflammatory pathologies (such as endometriosis, Crohn’s disease, colitis, rheumatoid arthritis, and Alzheimer’s disease), and the immune system (mainly for vaccines against pathogens such as HIV, malaria, and tuberculosis). We have increasingly emphasized complex tissue contexts, including mouse models, human subjects, and tissue-engineered micro-physiological systems platforms in association with outstanding collaborators. From our laboratory have come more than 100 doctoral and postdoctoral trainees. Many hold faculty positions at academic institutions in the USA, Canada, and Europe; others have gone on to research positions in biotechnology and pharmaceutical companies; and others yet have moved into policy and government agency careers.

Awards

  • Bernard M. Gordon Prize for Innovation in Engineering and Technology Education, National Academy of Engineering, 2021
  • American Association for the Advancement of Science, Member, 2019
  • American Academy of Arts and Sciences, Fellow, 2001
  • John Simon Guggenheim Memorial Foundation, Guggenheim Fellowship, 1989
Matthew Vander Heiden

Education

  • PhD, 2000, University of Chicago; MD, 2002, University of Chicago
  • SB, 1994, Biological Chemistry, University of Chicago

Research Summary

We study the biochemical pathways cells use and how they are regulated to meet the metabolic requirements of cells in different physiological situations. We focus on the role of metabolism in cancer, particularly how metabolic pathways support cell proliferation. We aim to translate our understanding of cancer cell metabolism into novel cancer therapies.

Awards

  • Howard Hughes Medical Institute Faculty Scholar, 2016
  • SU2C Innovative Research Grant Recipient, 2016
Rethinking transcription factors and gene expression

Study shows that, like proteins, genomes must fold appropriately to function properly and that some transcription factors provide the structural support.

Nicole Giese Rura | Whitehead Institute
December 7, 2017

Transcription — the reading of a segment of DNA into an RNA template for protein synthesis — is fundamental for nearly all cellular processes, including growth, responding to stimuli, and reproduction. Now, Whitehead Institute researchers have upended our understanding of how transcription is controlled and the role of transcription factors in the process.

The paradigm shift, described in an article online on Dec. 7 in the journal Cell, hinges on a small protein that plays a key role in genome structure and gives us new insights into how changes in the control of transcription and gene expression can lead to disease.

Transcription has several important players that must all be in the right place at the right time: the transcription machinery, transcription factors, promoters, and enhancers.  According to the existing model, transcription factors are proteins that bind to enhancer regions of the genome and recruit the transcription machinery to the promoter DNA regions, which then initiate the genes’ transcription.

“We’ve always assumed that the role of transcription factors was to recruit the transcription machinery to genes to turn them on or turn them off,” says Richard Young, a Whitehead Insistute member and professor of biology at MIT. “But we never imagined that the transcription factors we’ve studied for three decades actually contribute to the genome’s structure. And as a consequence, they regulate genes. So we now look at genomes like proteins: They have to fold up appropriately in order to control genes.”

Scientists have known that the genome’s structure — how it bends and folds — is essential for efficiently compressing two meters of DNA into each human cell, which is the equivalent of packing a strand ten football fields long into a space the size of a marble. Yet until recently, researchers have not had the tools necessary to appreciate this architecture’s importance in fine control of gene expression or study the genome’s structure at sites ready for transcription.

In 2014, Young and his lab determined that portions of the genome reside in loop-based structures, creating insulated neighborhoods that bring enhancers, promoters, and genes into close proximity. Each loop is tied at the top by a pair of molecules, called CTCF, that are bound together. This structure is essential for proper gene control: If the loop structure is broken, gene expression is altered, and cells can become diseased or die.

In the current research, Young along with co-first authors Abraham Weintraub and Charles Li took a closer look at a protein that is well known but not well understood: Yin Yang 1 (YY1). Hundreds of scientific papers have linked YY1 dysfunction to diseases such as viral infections, cancer, and arthritis, and yet the studies produced seemingly contradictory observations of YY1’s function.

According to Young and colleagues, YY1 is a unique transcription factor that occupies both enhancers and promoters, is essential for cell survival, and is found in almost every cell type in humans and mice. Like CTCF, YY1 can also pair with itself and bind to DNA to form loops that enhance DNA transcription.

“YY1 is expressed broadly, and it is necessary for establishing enhancer-promoter loops in multiple cell types,” says Weintraub. “That’s its job, not recruiting the transcription apparatus. When the structure created by YY1 is removed, the genome is no longer folded properly, gene control is lost and transcription of the affected genes is significantly diminished, which can cause dysfunction.”

This model of YY1’s function could account for its association with a number of disparate diseases. Earlier this year, scientists reported YY1 syndrome — a genetic syndrome causing cognitive disabilities in people with mutations in their YY1 gene.

According to Young, YY1 is probably not the only transcription factor with this loop-forming role, and his lab will be searching for additional factors with similar functions.

“YY1 is most likely just the first one, and there are probably a bunch of collaborators that have similar roles,” says Young. “Instead of the classic function that we thought these transcription factors had — interacting with the transcription apparatus and giving instructions on how much or how little of a gene’s transcript to produce — they are bringing together regulatory elements with the gene. The whole job of these transcription factors is just making structure. We are realizing that the things that form physical structures are much more important than we had appreciated.”

The researchers’ work was supported by the National Institutes of Health, the Ludwig Graduate Fellowship funds, the National Science Foundation, the American Cancer Society, a Margaret and Herman Sokol Postdoctoral Award, the Damon Runyon Cancer Research Foundation, and the Cancer Research Institute. The Whitehead Institute has filed a patent application based on this study.

Richard O. Hynes

Education

  • PhD, 1971, MIT
  • MA, 1970, Biochemistry, Cambridge University
  • BA, 1966, Biochemistry, Cambridge University

Research Summary

We study the mechanisms underlying the spread of tumor cells throughout the body, known as metastasis. We are particularly interested in the role of the extracellular matrix — a fibrillar meshwork of proteins that surrounds both normal and tumor cells, which plays many important roles in tumor progression. We also investigate changes in the metastatic cells themselves and in the contributions of normal cells, both in terms of metastasis and other bodily functions.

Awards

  • Paget-Ewing Award, Metastasis Research Society, 2018
  • Inaugural American Society for Cell Biology (ASCB) Fellow, 2016
  • American Association for Cancer Research (AACR) Academy, Fellow, 2014
  • Distinguished Investigator Award, International Society for Matrix Biology, 2012
  • Earl Benditt Award, North American Vascular Biology Organization, 2010
  • Robert and Claire Pasarow Medical Research Award – Cardiovascular, 2008
  • E.B. Wilson Medal, American Society for Cell Biology, 2007
  • President, American Society for Cell Biology, 2000
  • Gairdner Foundation International Award, 1997
  • National Academy of Sciences, Member, 1996
  • National Academy of Medicine, Member, 1995
  • Royal Society of London, Fellow, 1989
  • Howard Hughes Medical Institute, HHMI Investigator, 1988
  • American Association for the Advancement of Science, Fellow, 1987
  • American Academy of Arts and Sciences, Fellow, 1987
  • John Simon Guggenheim Memorial Foundation, Guggenheim Fellowship, 1982

Media Inquiries

For media inquiries, please email rhynes-admin@mit.edu.