26.07.2019 Views

YSM Issue 92.1

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Yale Scientific<br />

Established in 1894<br />

THE NATION’S OLDEST COLLEGE SCIENCE PUBLICATION<br />

MARCH 2019 VOL. 92 NO. 1 | $6.99<br />

12<br />

PREYING ON<br />

POLLUTANTS<br />

EVADING THE<br />

IMMUNE SYSTEM15<br />

SEQUENCING<br />

HALF A CELL18<br />

LEARNING TO<br />

REMEMBER20<br />

ESSENTIAL<br />

ERRORS22


INDIUM AND<br />

GALLIUM<br />

COMPOUNDS<br />

AND METALS<br />

49<br />

In<br />

114.818<br />

Indium Corporation’s online store allows<br />

you to browse our selection of metals,<br />

compounds, and solder products.<br />

Our ecommerce site:<br />

• Is easy to use<br />

• Accepts credit cards<br />

• Offers a wide variety<br />

of products<br />

Compounds<br />

31<br />

Ga<br />

69.723<br />

EZ-Pour ®<br />

Gallium<br />

Trichloride<br />

Contact our technical engineers today:<br />

techsupport@indium.com<br />

Learn more:<br />

www.indium.com/YALE<br />

From One Engineer<br />

To Another<br />

®<br />

www.indium.com<br />

askus@indium.com<br />

ASIA • CHINA • EUROPE • USA<br />

©2016 Indium Corporation


TABLE OF CONTENTS<br />

VOL. 92 ISSUE NO. 1<br />

More articles available online at www.yalescientific.org.<br />

12<br />

15<br />

18<br />

20<br />

22<br />

PREYING ON POLLUTANTS<br />

COVER ARTICLE: A collaboration between researchers at Yale and Peking University has led to a nanocoagulant that mimics<br />

the sea anemone Actinia. It can remove multiple types of contaminants simultaneously and outperforms conventional<br />

coagulants in multiple classes, thereby offering a cost-effective solution to water purification challenges.<br />

EVADING THE IMMUNE SYSTEM<br />

With the discovery of the immunological role of fibrinogen-like protein 1 (FGL-1) and its potential to aid the growth of cancerous<br />

cells, Yale researchers have solved a mystery that has baffled cancer immunologists for years.<br />

SEQUENCING HALF A CELL<br />

Researchers at Yale have developed a method to sequence both mRNA and microRNA in a single cell, leading to new insights into<br />

non-genetic variability at the single-cell level.<br />

LEARNING TO REMEMBER<br />

When do we start remembering? A new study conducted at the Yale School of Medicine probes this question and reveals<br />

the developmental timeline of memory formation.<br />

ESSENTIAL ERRORS<br />

The accurate translation of genetic material into proteins is essential to life. For parasitic bacteria with mutated genomes,<br />

however, errors in translation are important for their life cycles and could be therapeutic targets, Yale researchers report.<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

3


CAN<br />

WE ENGI-<br />

NEER SPICY<br />

TOMATOES?<br />

By Serena Thaw-Poon<br />

Chili peppers, often conjuring both tears of pain and<br />

pleasure, serve an important role in the culinary exploits of<br />

many cultures. Less well known, however, are the medicinal<br />

properties of the compound responsible for the distinct heat<br />

of chilis. Peppers make a molecule called capsaicin, which has<br />

anti-inflammatory and weight-loss properties. But what if other<br />

plants could be modified to produce capsaicin as well? Could we<br />

make spicy tomatoes? Augustin Zsögön and his team have recently<br />

discovered genes in tomatoes that encode for capsaicin, suggesting<br />

that the “spicy tomato” might become a reality. Not only<br />

would chefs reap the benefits of a culinary marvel, but scientists<br />

would also have an easier time producing capsaicin to research as<br />

tomatoes are easier to breed and cultivate than typical Capsicum<br />

plants. The process of genetically engineering tomatoes, however,<br />

presents certain challenges. Many cellular components combine<br />

forces to make the process possible. “Enzymes that are acting in<br />

the mitochondrion, cytosol, and chloroplast all need to work<br />

together in sequence,” Zsögön said. Consequently, the production<br />

of capsaicin involves the manipulation of multiple<br />

genes and an understanding of how each gene affects the<br />

overall pathway. Zsögön makes it clear that any commercial<br />

opportunities would be unintentional, though<br />

welcome, consequences of his team’s research. “We<br />

don’t really care about the money, we just really<br />

like the ideas and get carried away,” he said,<br />

laughing. Whether for monetary or gastronomical<br />

purposes, spicy tomatoes<br />

are sure to appease everyone’s<br />

appetite.<br />

Q&A<br />

.<br />

CAN TETRIS<br />

HELP TREAT<br />

PTSD?<br />

By Nadean Alnajjar<br />

Tetris is the best-selling videogame of all time, but<br />

what if this simple computer game could relieve symptoms<br />

of an even more prevalent psychiatric disorder? Professor<br />

Henrik Kessler and Aram Kehyayan from Ruhr-Universität<br />

Bochum in Germany studied possible links between<br />

Tetris and post-traumatic stress disorder (PTSD), an anxiety<br />

disorder induced by stressful, recurrent memories called<br />

intrusions. “Our study is the first worldwide study to apply this<br />

Tetris paradigm in real patients with complex PTSD,” Kessler<br />

said. His study involved twenty patients undergoing a six-toeight-week<br />

period of regular inpatient treatment. The treatment<br />

involved playing Tetris after writing about an intrusion. Sixteen<br />

of the twenty patients responded well to the treatment–reporting<br />

a sixty-four percent decrease in intrusions on average. Further<br />

studies with more patients and better controls are now under way<br />

to replicate these results. PTSD intrusions use visuospatial regions<br />

of the brain, which are also activated while playing Tetris.<br />

“You supposedly need the same working memory resources<br />

and capacity that an intrusion would require,” Kessler said.<br />

“Tetris could lock the processing of intrusions.” Another<br />

theory proposes that if you reactivate an old memory, it<br />

becomes vulnerable, and interference through Tetris can<br />

be performed to weaken the memory. While professional<br />

treatment is available for PTSD, few people<br />

utilize it. “The implications of this research are<br />

huge because this would mean that people<br />

who had traumatic experiences could<br />

mend their intrusions by themselves<br />

without needing professional<br />

help,” Kessler said.<br />

4 Yale Scientific Magazine March 2019 www.yalescientific.org


SPIRIT OF CREATIVITY<br />

Great scientific discoveries begin with a pinch of curiosity and a dash of creativity. Our<br />

world faces complex problems which require scientists to work together to compose outof-the-box<br />

solutions. This issue features the work of incredible teams of investigators who<br />

have pushed the limits of human understanding of life in the universe (pg. 28), the immune<br />

system (pg. 15), memory formation (pg. 18), and Earth’s old supercontinents (pg. 34).<br />

Employing unique approaches to solve intricate problems, researchers have grown<br />

artificial brains from stem cells to better understand neurological disorders (pg. 10),<br />

developed a method to sequence two types of RNA in the same cell to learn how they<br />

regulate each other (pg. 22), and designed microscopes mounted on top of the heads<br />

of mice to collect real-time data on hundreds of thousands of neurons (pg. 35).<br />

As the human population explodes, and clean, drinkable water becomes less abundant,<br />

scientists are looking to nature for solutions. In our cover article, researchers<br />

have designed a water purification technology inspired from the feeding process of<br />

a sea anemone known as the “flower of the sea” (pg. 12). Based on the principles<br />

of coagulation, this system could lead to a more<br />

THE<br />

cost-effective approach to making cleaner water.<br />

Researchers have also sequenced the genome<br />

of a giant tortoise named Lonesome George,<br />

whose death at age 100 marked the last of his<br />

kind (pg. 8). Not only has Lonesome George’s<br />

venerable story inspired new conservation efforts<br />

in the Galapagos, but his genome might<br />

EDITORalso<br />

reveal the secrets to living a long life.<br />

All of us want to live long, full lives, but equally<br />

important is ensuring our quality of life doesn’t<br />

decline as we age. One study explains how a<br />

promising new compound restores memories and<br />

neural connections in the brains of mice afflicted<br />

with Alzheimer’s disease (pg. 10).<br />

With a new year comes a new masthead. Each<br />

IN-CHIEF<br />

year we grow as a community of writers, artists,<br />

and designers. We are excited to continue the<br />

work of our predecessors in celebrating compelling,<br />

new research being done at Yale and<br />

around the world. The stories we have chosen<br />

to share this issue have the potential to lead to<br />

a brighter future for our planet, our health, and<br />

SPEAKS<br />

our well-being. One of our goals is to build a<br />

conversation surrounding this research so that,<br />

one day, these visions become reality.<br />

William Burns, Editor-in-Chief<br />

ABOUT THE ART<br />

The cover art for this issue is inspired by the<br />

incredible breakthrough in water purification<br />

technology, modeling the molecular “tentacle”<br />

nano-coagulants of a novel system after<br />

a beautiful organism: the sea anemone. It’s<br />

an elegant and inspiring solution, reminding<br />

us that we need not look farther than natural<br />

forms to develop solutions.<br />

Ivory Fu, Arts Editor<br />

MASTHEAD<br />

MARCH 2019 VOL. 92 NO. 1<br />

EDITORIAL BOARD<br />

Editor-in-Chief<br />

Managing Editors<br />

News Editor<br />

Features Editor<br />

Articles Editor<br />

Online Editors<br />

Copy Editors<br />

Scope Editors<br />

PRODUCTION & DESIGN<br />

Production Manager<br />

Layout Editor<br />

Art Editor<br />

Photography Editor<br />

Webmaster<br />

BUSINESS<br />

Publisher<br />

Operations Manager<br />

Subscriptions Manager<br />

Advertising Managers<br />

OUTREACH<br />

Synapse Presidents<br />

Synapse Vice President<br />

Social Media Coordinator<br />

Outreach Coordinators<br />

STAFF<br />

Sophia Sanchez-Maes<br />

Ameera Billings<br />

Rami Rajjoub<br />

Viola Lee<br />

Miriam Ross<br />

James Han<br />

Ashwin Chetty<br />

Megan He<br />

Isaac Wendler<br />

Sami Elrazky<br />

ADVISORY BOARD<br />

Priyamvada Natarajan<br />

Sandy Chang<br />

Kurt Zilm, Chair<br />

Fred Volkmar<br />

Stanley Eisenstat<br />

James Duncan<br />

Stephen Stearns<br />

Jakub Szefer<br />

Werner Wolf<br />

John Wettlaufer<br />

William Summers<br />

Scott Strobel<br />

Robert Bazell<br />

Craig Crews<br />

Ayaska Fernando<br />

Robert Cordova<br />

Serena Thaw-Poon<br />

Nadean Alnajjar<br />

Katie Schlick<br />

Khue Mai Tran<br />

Tiffany Liao<br />

Grace Chen<br />

Brett Jennings<br />

Alison Ho<br />

Michael Adeyi<br />

Makayla Conley<br />

William Burns<br />

Conor Johnson<br />

Sunnie Liu<br />

Anna Sun<br />

Lukas Corey<br />

Marcus Sak<br />

James Han<br />

Lauren Kim<br />

Isabella Li<br />

Xiaoying Zheng<br />

Kelly Farley<br />

Georgia Woscoboinik<br />

Mafalda Von Alvensleben<br />

Maria Lee<br />

Ivory Fu<br />

Kate Kelly<br />

Matt Tu<br />

Richard Li<br />

Alexandra Brocato<br />

Sebastian Tsai<br />

Tony Leche<br />

Annie Yang<br />

Leslie Sim<br />

Lisa Wu<br />

Hannah Ro<br />

Chelsea Wang<br />

Oscar Garcia<br />

Katherine Dai<br />

Ellie Gabriel<br />

Britt Bistis<br />

Elissa Martin<br />

Anusha Bishop<br />

Antalique Tran<br />

Sandra Li<br />

Adrian Bebenek<br />

Andrea Ouyang<br />

Christie Yu<br />

Carli Roush<br />

Astronomy<br />

Biological and Biomedical Sciences<br />

Chemistry<br />

Child Study Center<br />

Computer Science<br />

Diagnostic Radiology<br />

Ecology & Evolutionary Biology<br />

Electrical Engineering<br />

Emeritus<br />

Geology & Geophysics<br />

History of Science, Medicine, & Public Health<br />

Molecular Biophysics & Biochemistry<br />

Molecular, Cellular, & Developmental Biology<br />

Molecular, Cellular, & Developmental Biology<br />

Undergraduate Admissions<br />

Yale Science & Engineering Association<br />

The Yale Scientific Magazine (<strong>YSM</strong>) is published four times a year by<br />

Yale Scientific Publications, Inc. Third class postage paid in New Haven,<br />

CT 06520. Non-profit postage permit number 01106 paid for May 19,<br />

1927 under the act of August 1912. ISN:0091-287. We reserve the right<br />

to edit any submissions, solicited or unsolicited, for publication. This<br />

magazine is published by Yale College students, and Yale University<br />

is not responsible for its contents. Perspectives expressed by authors<br />

do not necessarily reflect the opinions of <strong>YSM</strong>. We retain the right to<br />

reprint contributions, both text and graphics, in future issues as well as<br />

a non-exclusive right to reproduce these in electronic form. The <strong>YSM</strong><br />

welcomes comments and feedback. Letters to the editor should be under<br />

200 words and should include the author’s name and contact information.<br />

We reserve the right to edit letters before publication. Please send<br />

questions and comments to ysm@yale.edu. Special thanks to Yale STC.


SELECTING<br />

FOR CANCER<br />

Using evolution to explain the<br />

spread of cancer<br />

BY AMEERA BILLINGS<br />

In 2018, the<br />

International Agency for<br />

Research on Cancer estimated<br />

seventeen million new cancer diagnoses<br />

and 9.5 million cancer-related deaths worldwide.<br />

Though the numbers are grim, researchers have made<br />

great strides in explaining what causes and promotes cancer<br />

through an unexpected lens: evolution.<br />

The Townsend Lab at the Yale School of Public Health has<br />

developed a model to predict the evolutionary progression of<br />

cancerous cells. Now, we can quantify the relative importance<br />

of different gene mutations in stimulating cancer growth–the<br />

so-called ‘cancer effect size’ of a gene. By applying evolutionary<br />

principles, scientists can help analyze what mutations are driven<br />

by natural selection in a cancer lineage. “Selection is what drives<br />

[cancer] once you get a mutation…[causing cells to] replicate<br />

more,” explained Professor Jeffrey Townsend.<br />

Townsend and his team were able to calculate the mutation<br />

rate of cancer cells using tumor DNA sequencing. While some<br />

mutations may actually lead to cancer, others may have just<br />

happened to occur in those tumors. “If we see more [mutations]<br />

than expected by random mutation, then…it was more likely<br />

to lead to a tumor when you had those mutations in that gene,”<br />

Townsend said.<br />

What does this mean for the future of cancer medicine? “The<br />

cure for various cancers will be therapies that manage to corner<br />

all of the evolutionary avenues of an evolving tumor and make<br />

it impossible for it to find a new way to grow,” Townsend said.<br />

Being able to determine the cancer effect size of gene mutations<br />

will provide insight into the development of cancer cells, and<br />

thus, improve treatments for cancer patients in the future.<br />

Paleontology has long been regarded as the domain of dinosaur<br />

bones, teeth, and shells. During fossilization, these hard,<br />

organic scaffolds behave like a rough cast, providing structure<br />

for the assembly of minerals that will endure after the organics<br />

degrade. In this paradigm, soft tissues degrade too quickly<br />

for fossilization timescales, but perplexingly, structures which<br />

morphologically resemble them are common.<br />

A team led by Yale graduate student Jasmina Wiemann investigated<br />

the nature of these structures using a sample of vertebrate<br />

fossils from the Peabody Museum of Natural History<br />

collection. The researchers removed hard material from the<br />

fossils in a process called decalcification to isolate potential<br />

organic matter. Curiously, all the organic samples were brown<br />

in color, suggesting they had a common chemical composition.<br />

To explore this further, the team took fresh tissues rich<br />

in protein and subjected them to accelerated conditions for<br />

fossilization. Remarkably, the transformed tissues exhibited<br />

a brown color and composition reminiscent of the Peabody<br />

specimens. These findings suggest that the organic proteins<br />

in the Peabody specimens survived and metamorphized into<br />

a more stable, discolored polymer, visible to the unaided eye<br />

as the dark tinge on some fossils. The study shows that not<br />

only are organic structures, like cells and even organelles, preserved,<br />

but also that their preservation is surprisingly common<br />

in fossils formed under oxidative conditions.<br />

Scientists often use DNA testing to determine how living<br />

species are related, but DNA, which degrades easily, does not<br />

survive long in fossils. For long-extinct species, preserved<br />

proteins could be the next widely-used bio-informed<br />

molecule to more accurately<br />

reconstruct the tree of life.<br />

DINOSAUR<br />

SECRETS<br />

Preserved proteins reveal a<br />

more accurate tree of life<br />

BY SOPHIA S´ANCHEZ-MAES<br />

6 Yale Scientific Magazine March 2019


We are all familiar with the sensation of pain, but have<br />

you ever wondered why some people experience pain more<br />

vividly than others? For years, perplexed researchers have<br />

documented individual differences in pain tolerance to no<br />

avail. In a paper recently published in the Journal of Neuroscience,<br />

Malgorzata Mis, a postdoctoral associate in the<br />

Department of Neurology at Yale University, working with<br />

a team of researchers from the Yale School of Medicine<br />

and Veterans Affairs Connecticut Healthcare System, has<br />

taken the first steps towards identifying the genes involved<br />

in pain perception.<br />

The researchers analyzed a mother and son with inherited<br />

erythromelalgia, a disease caused by a mutation<br />

within a gene that kindles pain and burning sensations<br />

when expressed in the nervous system. Despite having<br />

the same mutation, the mother and son reported different<br />

levels of pains in a previous study. “The son reported<br />

over one-hundred nightly awakenings due to his mutation<br />

within a fifteen-day period, while the mother reported<br />

only one awakening,” Mis said.<br />

To further study this disparity, the research team grew<br />

sensory neurons derived from the mother’s and son’s<br />

stem cells in a laboratory dish. Using whole exome sequencing<br />

coupled with computer-aided dynamic clamp,<br />

a method that merges the visualization of live neurons<br />

with computational models, the researchers were able to<br />

identify a variation in the mother’s potassium channel<br />

encoded by the KCNQ2 gene, which ultimately led to<br />

her resilience to pain. The team is optimistic about<br />

implementing these new genetic methods<br />

that will guide them toward better<br />

treatments for pain.<br />

RETHINKING<br />

PAIN<br />

Uncovering the genes for<br />

pain resilience<br />

BY RAMI RAJJOUB<br />

THE BRAIN<br />

ON A DISH<br />

Organoids in the Age of<br />

Big Data<br />

BY VIOLA KYOUNG A LEE<br />

Yale neurobiologists<br />

are growing brain organoids to<br />

better understand the causes of neurological<br />

disorders. Brain organoids are artificial<br />

organs which are grown by differentiating pluripotent<br />

stem cells into neuronal tissues.<br />

Researchers in professor Flora Vaccarino’s lab at the Yale School of<br />

Medicine recently verified brain organoids’ ability to closely model fetal<br />

brain development. This project analyzed large-scale gene expression<br />

data from organoids’ RNA transcripts on Yale’s supercomputing clusters.<br />

“An extensive verification of the ability of brain organoids to serve<br />

as a model for the human brain has not been done before,” first author<br />

Anahita Amiri remarks. For years, professor Vaccarino has grown thousands<br />

of organoids to observe the correlation between gene expression<br />

and time-dependent cell differentiation. These brain organoids have<br />

many advantages over traditional neurobiological techniques. “[We<br />

have] the ability to observe the development of human neurons in a living<br />

system, in a time-dependent manner, and in 3D,” Vaccarino said.<br />

With the fidelity of their model system confirmed, the lab can grow<br />

hundreds of organoids and get massive amounts of data, including enhancer<br />

networks–which control which genes will be transcribed at each<br />

specific stage of brain development–and potential interactions with<br />

their target genes.<br />

Vaccarino and her lab are currently using organoids grown from the<br />

cells of patients with Autism Spectrum Disorder (ASD) to gain insights<br />

into how specific enhancers might contribute to ASD. “Once we can<br />

identify ASD biomarkers, the next steps could include developing therapies<br />

to target genes and enhancers involved through gene therapy or<br />

drugs,” Vaccarino said. With her brain organoid research, we are closer<br />

to understanding the mechanisms and causes of some neurodevelopmental<br />

diseases.<br />

March 2019<br />

Yale Scientific Magazine<br />

7


NEWS<br />

genetics<br />

LONESOME<br />

GEORGE’S<br />

LEGACY<br />

Genetic markers<br />

of longevity<br />

in giant tortoises<br />

BY MIRIAM ROSS<br />

IMAGE COURTESY OF FLICKR<br />

“Gone but not forgotten;” an epithet that applies to Lonesome George,<br />

the giant tortoise, and his newly-extinct species. In 2012, Lonesome<br />

George died as the last member of Chelonoidis abingdonii, at the ripe<br />

old age of 100. Galapagos tortoises are among the longest-lived vertebrate<br />

organisms, and researchers have long been curious about which<br />

specific factors contribute to their long lives. In Lonesome George’s<br />

case, the main research to examine tortoise longevity began after his<br />

death. “We were at the point in which it would have been very useful for<br />

us to have a whole genome, [to give] better information of how polymorphisms<br />

of different species are distributed,” said Adalgisa Caccone,<br />

a senior research scientist in Yale’s Ecology and Evolutionary Biology<br />

Department and director of the Center for Genetic Analysis of Biodiversity.<br />

After Lonesome George’s death in 2012, Caccone worked with<br />

colleagues at the Galapagos National Park Service, the Galapagos Conservancy,<br />

and the University of Oviedo to sequence his entire genome<br />

from a blood sample. With the full code of Lonesome George’s genome,<br />

the research team could begin making cross-species comparisons with<br />

smaller DNA sequences coded from several related tortoise species.<br />

In particular, Caccone and her research colleagues wanted to investigate<br />

variants linked to longevity in other species, which affected<br />

functions such as DNA repair, inflammation, and cancer development.<br />

The team had previously identified nine major “hallmarks<br />

of aging” in humans, and found variants affecting six of these “hallmarks”<br />

in the elderly tortoise’s genome–variants which are not found<br />

in the DNA of shorter-lived tortoise species. Of particular interest<br />

were a group of enzyme variants that seemed to offer protection<br />

against the sequence of protein activation which causes Parkinson’s<br />

disease. However, interpreting the meaning of variant changes within<br />

DNA is difficult. “It’s one thing to produce a genome, [and another]<br />

to annotate it, [and to] to know the function of a particular gene,”<br />

Caccone said. Traits are controlled by multiple genes which interact<br />

in multifaceted ways, and isolating the effect of a single variant can<br />

prove especially challenging. Although genome analysis reveals many<br />

variants, there is no easy way to map their functions; what is found<br />

are potential associations. And even in forming these associations,<br />

it is not clear from one genome alone whether a change is associated<br />

with longevity more broadly or effects in an individual organism.<br />

The first step towards identifying a variant’s function comes through<br />

cross-species comparisons. The research team had partial DNA samples<br />

of multiple individuals from related species of long-lived tortoises,<br />

which they compared to the full genome of Lonesome George. “Using<br />

this comparative approach, we can identify the hallmarks among all<br />

these genes, [and] eventually carry out functional studies on other organisms...to<br />

see if the [variants] have any associations with disease [or]<br />

longevity,” Caccone explained. Additionally, cross-species comparisons<br />

can allow the researchers to rule out the importance of changes<br />

which are not shared between species to longevity. The variants most<br />

likely to have functional significance should be those disproportionately<br />

present within the genomes of both Lonesome George and other<br />

Giant Galapagos tortoise species, all of which are long-lived.<br />

In addition to studying longevity, Lonesome George’s genome could<br />

also help with tortoise conservation. An effort is already underway to<br />

bring back extinct Galapagos tortoise species. “The Lonesome George<br />

genome is helping us is to identify molecular markers that are diagnostic<br />

for each species,” Caccone said. In previous research using DNA<br />

analysis, she discovered tortoises living on Galapagos islands that were<br />

actually genetic hybrids between the local species and an extinct one.<br />

The hybrids arose when early mariners brought tortoises with them<br />

for meat, and occasionally dropped them at random islands if the ship’s<br />

hold was too full. Caccone is helping the Galapagos National Park to<br />

start a breeding program that crosses these hybrids with each other to<br />

produce offspring more closely related to the original extinct species,<br />

the first breeding program of this type. The eventual goal is to release<br />

the tortoises’ offspring back onto the islands of their species’ origins.<br />

The sequencing of Lonesome George’s genome has given rise to<br />

many exciting potential associations, but the search for which variants<br />

actively contributed to his long life has just begun. With a group of<br />

potential associations and the sequences of other species, the secret to<br />

tortoises’ longevity will become clearer over time. Lonesome George<br />

may have died as the last member of his species, but discoveries from<br />

his genes can help us both to understand his long life, and perhaps<br />

even bring another extinct species back to their home island.<br />

8 Yale Scientific Magazine March 2019 www.yalescientific.org


cell biology<br />

NEWS<br />

HOPS<br />

INTO<br />

THE CELL<br />

IMAGE COURTESY OF WIKIMEDIA COMMONS<br />

When we take a drug, it flows through our bloodstream before<br />

entering the cells in our bodies–where the action happens. Most<br />

drugs that are currently used are small organic molecules that<br />

perform specific functions according to their chemical composition<br />

and shape. However, there is also substantial interest in using<br />

larger proteins and peptides as therapies for diseases. Unlike<br />

most small organic molecules, which are small enough to cross<br />

cell membranes, larger proteins or enzymes–molecular machines<br />

which have many more functions than small organic molecules–<br />

are tougher to shuttle across membranes inside cells. In a recent<br />

study published in the Proceedings of the National Academy of<br />

Sciences, a team led by Yale professor Alanna Schepartz uncovered<br />

new pathways and mechanisms that help proteins enter cells.<br />

Schepartz focused on two classes of proteins–cell-penetrating peptides<br />

(CPPs) and cell-permeant miniature proteins (CPMPs)–and the<br />

mechanism they use to enter the cell. In the study, Schepartz hoped to<br />

shed light on some of the unknown mechanisms employed by CPPs<br />

and CPMPs to enter cells. Both CPPs and CPMPs use what is called an<br />

endocytic pathway, taking advantage of the built-in cellular machinery<br />

for bringing larger molecules into the cell. During endocytosis, the cellular<br />

membrane surrounds the target molecule, pinches off, and creates<br />

an endosome inside the cell. This process is like the surface of a<br />

bubble pinching off inwards to become a new bubble inside the original<br />

bubble. Though scientists knew CPPs and CPMPs used the endocytic<br />

pathway, they were still unsure how CPPs and CPMPs are able to<br />

escape the endosome and enter the cellular fluid to fulfill their roles.<br />

Schepartz first tested the hypothesis that CPPs and CPMPs<br />

simply rupture the endosome to escape. The researchers used<br />

fluorescent tags to look for signs of endosome damage in the<br />

cell and locate ruptured endosomes. They discovered that the<br />

endosomes in cells that had taken in CPPs and CPMPs were relatively<br />

unharmed. The researchers then tested for signs of leakage<br />

from the endosome, which came back negative. Based on<br />

these results, the researchers were certain that endosomes are<br />

not harmed when the CPPs and CPMPs are released into cells.<br />

The team then decided to look for genes that could be regulating<br />

Uncovering<br />

how proteins<br />

enter cells<br />

BY JAMES HAN<br />

this release of proteins into the cell. They implemented a technique using<br />

small interfering RNAs (siRNAs), which allow researchers to “turn<br />

off” certain genes and observe the effects. After testing genes across<br />

the genome, Schepartz found that if VPS39, a gene that codes for a<br />

piece of the homotypic fusion and protein-sorting (HOPS) complex,<br />

was turned off, CPPs and CPMPs could not leave the endosome and<br />

enter the cell. They compared HOPS to similar complexes, but only<br />

turning off HOPS prevented proteins from leaving the endosome.<br />

Schepartz and her team then decided to look at the effects of<br />

CPPs and CPMPs on the function of HOPS. Using a fluorescence<br />

technique that looks for the products of HOPS’s reactions,<br />

Schepartz found that HOPS remained active even with delivery<br />

of CPPs and CPMPs. Finally, using fluorescent microscopy, the<br />

researchers found that HOPS functions by acting as a guide for<br />

CPPs and CPMPs, directing them to a class of endosome, called<br />

Lamp1, which has factors which facilitate the escape of proteins.<br />

Though the confirmation of HOPS as a key player in cell entry<br />

for proteins is a major breakthrough, further research is needed<br />

to understand the final steps of entry into the cell–particularly,<br />

how CPMPs escape from Lamp1 endosomes. Nevertheless,<br />

Schepartz’s research has great therapeutic potential and furthers<br />

our understanding of movement into and out of cells. As researchers<br />

learn more about these mechanisms, intricate molecular<br />

machines may replace small organic chemicals in medicine.<br />

SCHEPARTZ’S RESEARCH HAS GREAT<br />

THERAPEUTIC POTENTIAL AND<br />

FURTHERS OUR UNDERSTANDING OF<br />

MOVEMENT INTO AND OUT OF CELLS.<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

9


NEWS<br />

neuroscience<br />

PIONEERING<br />

ALZHEIMER’S<br />

TREATMENT<br />

A compound that<br />

can reverse neural<br />

connection loss<br />

BY MIRIAM ROSS<br />

Alzheimer’s disease (AD), which currently affects more that 5.7<br />

million people in the US, is a neurodegenerative disease that greatly<br />

impairs memory. Despite the vast field of research dedicated toward<br />

tackling Alzheimer’s, there is still no cure. Erik Gunther, along with<br />

a team of Yale researchers in Professor Stephen Strittmatter’s lab, has<br />

discovered a promising drug to reverse the progression of AD. After<br />

screening over 60,000 compounds at the Yale Small Molecule<br />

Discovery Center, they developed a new compound that, instead of<br />

simply treating AD symptoms like currently approved drugs, can reverse<br />

neural connection loss and memory defects in mice.<br />

To better unravel the mechanism of the drug, it is important to<br />

understand AD development. AD results from an accumulation of<br />

β-amyloid peptides, which are small proteins present only in low concentrations<br />

in non-Alzheimer’s patients. In AD patients, β-amyloid<br />

peptides clump together to form β-amyloid oligomers, which then<br />

further combine to form β-amyloid plaques that look like “rocks in the<br />

brain,” according to Strittmatter. β-amyloid oligomers are problematic<br />

because they interact with neurons to set off a cascade of molecular<br />

events that damages synapses–the connections between neurons–and<br />

thus lead to memory impairment. Ten years ago, Strittmatter demonstrated<br />

that β-amyloid peptide binds strongly to cellular prion protein<br />

(PrP), a protein on the surface of neurons, so he began to investigate<br />

how to block this damaging interaction from occurring.<br />

Strittmatter’s lab initially screened over 12,000 compounds, hoping<br />

to find one that would inhibit the β-amyloid oligomer interaction<br />

with PrP. They first identified the antibiotic cefixime as a potential<br />

candidate, which, unusually, was only effective after it had degraded–since<br />

most drugs are effective before they start to degrade. If cefixime<br />

had not degraded by the time the research team screened it,<br />

Strittmatter believes, “We might still be searching [for a compound].”<br />

Other antibiotics with analogous structures to cefixime’s yielded similar<br />

results. After further testing, the team switched their focus toward<br />

the degraded product of the antibiotic ceftazidime because it was<br />

more effective than the degraded product of cefixime. Through further<br />

analysis, the research team learned that ceftazidime decomposes<br />

and reacts with itself to create a different polymer, referred to as<br />

IMAGE COURTESY OF FLICKR<br />

Compound Z, that inhibits β-amyloid oligomer interaction with PrP.<br />

Although the researchers knew Compound Z has inhibitory activity,<br />

they were uncertain whether it binds to PrP. Using antibodies constructed<br />

to bind to specific regions of PrP, they found that Compound<br />

Z binds to two regions of PrP that interact with the β-amyloid oligomer,<br />

preventing any interaction between β-amyloid oligomers and PrP.<br />

Once the researchers elucidated this pathway, they tested Compound<br />

Z’s effect on neurons. Even in a petri dish, neurons exposed to β-amyloid<br />

oligomers have damaged synapses, but Strittmatter’s lab found that Compound<br />

Z reduces β-amyloid oligomer binding to neurons by over eighty<br />

percent and reduces the loss of dendritic spines–an essential component<br />

of neuron signaling–by eight-fold. With these promising results, they administered<br />

Compound Z to mice engineered with Alzheimer’s-like disease.<br />

The researchers knew they had discovered a potential treatment for<br />

AD when the mice recovered learning and memory function.<br />

This compound, however, was unable to bypass the brain’s filtration<br />

system, the blood-brain barrier, rendering it difficult to<br />

use as a potential therapeutic. Strittmatter’s lab needed to find a<br />

compound that could cross the blood-brain barrier. They tested<br />

over 56,000 molecules until they landed on a synthetic compound<br />

poly(4-styrenesulfonnic acid-co-maleic acid), abbreviated as PSC-<br />

MA. Similar to Compound Z, PSCMA has inhibitory properties,<br />

but unlike Compound Z it can be administered orally and thus is<br />

considerably more practical as a potential drug.<br />

Though the mice in this study were able to complete memory tasks<br />

with the remaining β-amyloid oligomers in the brain, the results will<br />

not necessarily translate in human subjects. “[The mice models] are<br />

not a complete picture of human AD.” Strittmatter acknowledged.<br />

There are numerous steps before human trials can begin, but the<br />

researchers’ main goal now is to find the ideal balance between maintaining<br />

the drug’s inhibitory activity while optimizing the drug’s access<br />

to the brain. Strittmatter hopes to eventually test whether a drug<br />

from his lab might prove effective in treating AD in humans. “My<br />

optimism is high because I really believe we have a molecular basis<br />

for its effectiveness, and while these animal models are not perfect,<br />

they are the best predictors we have at this time,” he said.<br />

10 Yale Scientific Magazine March 2019 www.yalescientific.org


ecology<br />

NEWS<br />

SOURCES<br />

AND<br />

SINKS<br />

IMAGE COURTESY OF WIKIMEDIA COMMONS<br />

The global carbon cycle is an essential aspect of life on Earth,<br />

and there has been extensive scientific research concerning<br />

how the movement and exchange of carbon compounds can<br />

help manage atmospheric carbon dioxide (CO2) concentrations<br />

and predict climate change. Contemporary carbon cycle<br />

models largely look at carbon that is involved in live plant biomass<br />

and organic matter and the relationship between plants<br />

across landscapes and the atmosphere. Yet, these models ignore<br />

the effects of animals in higher trophic levels of ecosystems.<br />

An interdisciplinary team of researchers led by professor<br />

Oswald Schmitz at Yale sought to dispel the notion that animals<br />

have little or no impact on the carbon cycle. The researchers<br />

argue that wild animal populations are key players<br />

in shaping how ecosystems mitigate climate change and<br />

should be considered when making carbon measurements.<br />

Their paper, published in Science, found that the direct and<br />

indirect effects of animals can control the magnitude of carbon<br />

exchange in various global reservoirs. This can consequently<br />

influence carbon turnover rates, or the time it takes<br />

for carbon fixed in a plant by photosynthesis to be released<br />

back into the atmosphere.<br />

The animals in question are a diverse group spread across<br />

varying locations, including both large and small herbivores,<br />

carnivores, vertebrates, and invertebrates. Herbivores consume<br />

plant biomass and assimilate carbon into their own<br />

animal biomass after digestion, releasing additional carbon<br />

through defecation and respiration. With less plant biomass<br />

comes a decrease in photosynthesis and an increase in respiration,<br />

which reduces the chemical energy created by an ecosystem.<br />

Additionally, the heavy trampling of soil and sediment by<br />

larger animals can change surface temperatures that increase<br />

either carbon retention or release.<br />

These animals can even have conflicting positive and negative<br />

effects on the carbon cycle. For example, in some cases,<br />

grazing and browsing herbivores such as muskox and geese<br />

enhance carbon uptake and storage from the atmosphere into<br />

How animals<br />

shape the<br />

carbon cycle<br />

BY MEGAN HE<br />

organic compounds by twenty to twenty-five percent. In other<br />

cases, these grazing animals cause a fifteen to seventy percent decrease<br />

in CO2 uptake. The predators of these herbivores, on the<br />

other hand, can reverse these effects. In grassland ecosystems,<br />

grasshoppers cause a seventeen percent reduction in CO2 uptake,<br />

but the spiders that forage on these grasshoppers “reverse” the effect<br />

by increasing CO2 uptake by forty-six percent. In freshwater,<br />

stickleback fish that feed on macroinvertebrate stonefly insects,<br />

increase algal CO2 uptake, which in turn decreases concentrations<br />

of dissolved inorganic carbon in the water, enhancing carbon<br />

capture by around ninety percent.<br />

Other underlooked factors in ecosystem models are animal<br />

movement and migration. “Since large animals roam widely<br />

across landscapes, being able to monitor their movements and<br />

quantify the amount of material that is moved along with them<br />

is crucial to understanding their impact,” Schmitz said. Schmitz<br />

outlined a one instrument–named LIDAR–able to provide 3D<br />

portraits of habitat structure, but these methods still are limited<br />

in their ability to quantify the impacts of animals. On-the-ground<br />

sampling is often coupled with remote sensing to produce a more<br />

comprehensive analysis of carbon distribution. For example, the<br />

impact of African elephant populations on densities of woody<br />

vegetation can be monitored through both methods.<br />

An interesting challenge faced by the team was the pushback<br />

from skeptical reviewers, who were not convinced of the<br />

importance of incorporating animals into these models. Ultimately,<br />

however, Schmitz thinks that the data speaks for itself.<br />

“In some cases, you can triple how much carbon you take up if<br />

you protect these animals,” Schmitz said.<br />

What about the role of humans? “We have to protect the<br />

food chains–the animals and plants–that drive carbon dynamics.<br />

Nature provides all these services for our survival benefit<br />

and for other species, so we have to reimagine ourselves as being<br />

part of nature,” Schmitz said. At the end of the day, wildlife<br />

conservation proves to be an essential strategy for creating<br />

carbon storage capacity, and that task is up to the people.<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

11


cell biology<br />

FOCUS<br />

The sea anemone Actinia is a deceptive organism, in<br />

both name and appearance. For one, it is known as the<br />

“flower of the sea” for its colorful ring of tentacles. It also<br />

adopts a sessile lifestyle, preferring to lie and wait for food<br />

rather than go looking for it. When an unassuming plankton<br />

or fish passes by and stimulates its tentacles, the marine<br />

predator stuns it, abruptly retracts its tentacles, and<br />

gracefully encapsulates its catch.<br />

In a classic example of biomimetics–the imitation of<br />

natural systems for the purpose of solving human problems–a<br />

team of researchers from Yale and Peking University<br />

has designed a coagulant that can capture contaminants<br />

in water via a shape eversion process reminiscent<br />

of Actinia’s feeding process. Due to its unique structure,<br />

the innovative technology can trap many different types<br />

of contaminants in a single step. This versatile feature can<br />

potentially enable tremendous increases in water purification<br />

efficiency.<br />

Water and Energy: A Modern Compromise<br />

This innovation comes at a crucial time, as it promises<br />

to revolutionize the way we treat water. One in nine people<br />

worldwide do not have access to potable water. Water scarcity<br />

is a multifaceted problem without a clear solution, and cleaning<br />

up water with modern water purification systems has a<br />

high energetic and economic cost. Water and wastewater facilities<br />

are responsible for approximately thirty-five percent<br />

of a typical US municipal energy budget. Today, in order to<br />

supply consumers with clean water, facilities release over forty-five<br />

million tons of greenhouse gases annually–an output<br />

equivalent to that of more than six million cars.<br />

Current water treatment systems feature an extensive<br />

number of steps to ensure complete purification, because<br />

each step cannot remove all contaminants at once. Water<br />

contains many types of contaminants, each with drastically<br />

different chemical properties–think bacteria,<br />

harmful chemicals, and organic matter.<br />

One popular purification method is coagulation,<br />

in which positively charged<br />

coagulants, when added into water,<br />

bind to negatively charged<br />

dissolved organic carbon<br />

(DOC) and carbon-based<br />

remains<br />

of organisms. The<br />

added coagulants<br />

and<br />

organic<br />

mat-<br />

by HANNAH RO<br />

art by ANUSHA BISHOP<br />

PREYING<br />

ON POLLUTANTS<br />

technology imitates life<br />

to generate cleaner water<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

12


FOCUS<br />

cell biology<br />

ter clump together into larger particles, then settle by gravity<br />

to the bottom of the basin so they can be more easily removed<br />

from the water. Another common step, sedimentation, allows<br />

heavier particles to settle, and a further chlorination step kills<br />

remaining microorganisms. Each of these methods requires<br />

specific technologies and tools, and the entire package together<br />

accounts for the high energy costs incurred today.<br />

The newly developed technology developed at Yale and Peking<br />

University, aptly named Actinia-like micellar nanocoagulant<br />

(AMC), streamlines water purification in two ways:<br />

one, it decreases the number of steps; two, it maximizes energy<br />

efficiency. If AMC enters the water purification industry,<br />

it could well eliminate multiple types of contaminants in one<br />

step, an optimal solution to lowering both the time and cost<br />

of cleaning water.<br />

Form Follows Function<br />

Last month, the collaborative project published results<br />

demonstrating that AMC could capture a spectrum of different<br />

contaminants in a single step. Conventional coagulants,<br />

such as aluminum sulfate, can typically only remove bulky<br />

organic matter. AMC inherits its special properties from Actinia.<br />

Like the sea anemone, the coagulant features a flexible<br />

outer shell that can be everted–turned outward/inside-out–<br />

to reveal an inner core, which has contrasting chemical properties.<br />

The positively charged outer shell binds to DOCs and<br />

other organic substances, while the negatively charged inner<br />

core contains organic micelles that capture smaller micropollutants.<br />

Notably, AMC can also extract nitrates, an excess<br />

of which can be harmful to humans.<br />

Both inner and outer surfaces can be accessed<br />

by altering the pH–a scale<br />

from zero to fourteen that<br />

corresponds to a<br />

solution’s acidity–of<br />

the surrounding solution. When stored in acidic conditions<br />

with a pH lower than four, the coagulant maintains its stable<br />

form, with the inorganic shell completely shielding the<br />

organic core. Raising the pH around AMC is akin to stimulating<br />

the tentacles of the Actinia–in wastewater above pH<br />

four, AMC changes its configuration to reveal the organic<br />

core. “Shape eversion of the coagulant adds additional functionality.<br />

Coagulants neutralize the charges on the organic<br />

matter and cause them to clump together so they can settle.<br />

But when the core is exposed, it absorbs additional contaminants<br />

that other coagulants would not be able to,” said Ryan<br />

DuChanois, a graduate student in the Elimelech group, who<br />

worked on the project.<br />

Another unique characteristic of the AMC is its ability<br />

to self-assemble. Much like the phospholipid bilayers that<br />

make up cell membranes, the AMC contains both hydrophobic<br />

and hydrophilic elements that rearrange in an aqueous<br />

solution. During the synthesis of AMC, hydrophobic<br />

carbon chains gather inwards, driven away from water, to<br />

form the organic core, and hydrophilic ionic complexes,<br />

comprising ammonium, silicon, and aluminum, form the<br />

outer shell. The researchers closely regulated the ratios of<br />

the reactants to achieve the product’s desired shape. Thanks<br />

to AMC’s optimized and stable build, it does not aggregate<br />

in solution and instead maintains its size and shape even after<br />

one year of storage.<br />

Having synthesized the particles, the researchers had<br />

to perform characterization to confirm and optimize its<br />

structure. However, one significant challenge of this project<br />

was the nanoscale of the coagulant; specialized equipment<br />

was required to observe the structure and function<br />

of the miniscule AMC particles. Researchers confirmed<br />

the spherical shape of AMC using transmission electron<br />

microscope (TEM) images and approximated its size with<br />

dynamic light scattering (DLS) measurements. For both<br />

types of technology, a beam of electrons, or light, is transmitted<br />

through the molecule to map out its structure. The<br />

caveat is that these methods only work when the molecule<br />

being observed is stationary. This is not the natural<br />

working state of the AMC. As such, the<br />

researchers had to turn to other technologies<br />

to learn about AMC’s mobile<br />

functionality.<br />

To observe how<br />

AMC self-as-<br />

sem-<br />

THE AMC RESEMBLES THE STRUCTURE OF SEA ANEMONE, WITH AN OUTER SHELL AND AN INNER CORE THAT TARGET


cell biology<br />

FOCUS<br />

bles, researchers conducted molecular dynamics<br />

simulations. These computational<br />

simulations, among other visualization<br />

techniques, predict and explain how a coagulant<br />

will behave when exposed to different<br />

types of wastewater. “[Simulations] show<br />

why the shell is removing certain contaminants<br />

and why the core is removing certain<br />

[other] contaminants. You can pinpoint the<br />

exact mechanisms, like electrostatic and hydrophobic<br />

interactions,” DuChanois said.<br />

Comparing Coagulants<br />

Once the structure and function of AMC<br />

were confirmed and better understood, researchers<br />

progressed to the field research<br />

portion of the project. Jar tests–in which<br />

treatment parameters, such as dosage, mixing<br />

rate, and aeration time, are altered to determine<br />

how a coagulant will behave with<br />

specific contaminants–were used to simulate<br />

full-scale water purification processes.<br />

These were performed to compare the<br />

efficiency of AMC and conventional coagulants.<br />

Efficiency was determined by two<br />

factors: the resulting contaminant concentration,<br />

as well as turbidity, a quantitative<br />

measurement of a liquid’s murkiness.<br />

AMC edged out other commercially<br />

used coagulants tested in the study–including<br />

a polymer called polyDADMAC,<br />

aluminum sulfate, and iron(III) chloride–<br />

with an average efficiency of over ninety<br />

percent. While all of the coagulants exhibited<br />

similar efficiency in removing turbidity,<br />

AMC was more successful at lowering<br />

DOC, phosphorous, and nitrate concentrations.<br />

Furthermore, while traditional coagulants<br />

demonstrated negligible removal of<br />

nitrate, AMC’s nitrate removal efficiency<br />

exceeded ninety percent.<br />

Other key contaminants the researchers<br />

considered were organic micropollutants<br />

and certain pharmaceuticals.<br />

Commonly, water is contaminated by<br />

micropollutants from residue left behind<br />

by personal care products, hormones,<br />

and pesticides. Because these particles<br />

are nonbiodegradable, they persist in<br />

wastewater treatment discharges<br />

and enter the environment.<br />

Conventional coagulants<br />

perform<br />

poorly<br />

in this category. Instead, the existing methods<br />

of removing micropollutants are ozonation<br />

and ultrasound. These approaches are energy-expensive,<br />

time-consuming, and work for<br />

only their specific class of contaminants.<br />

In this study, the conventional coagulants<br />

demonstrated removal efficiencies between<br />

zero and sixty percent. AMC outshone all of<br />

them, achieving removal efficiencies of over<br />

ninety percent for all tested micropollutants.<br />

This makes AMC all the more promising as a<br />

future water purification technique because<br />

it offers an entire functional package. “The<br />

real benefit is that if you can remove many<br />

different types of contaminants in one step<br />

instead of using many steps, you can simplify<br />

the process, use less money, and take up<br />

less land. Overall, the process becomes more<br />

efficient,” DuChanois said.<br />

The Big Picture<br />

Ultimately, this project demonstrates<br />

how adaptations which species develop<br />

to face problems in nature can inspire<br />

technological innovations to solve human<br />

problems. Encouraged by the success of<br />

AMC, the researchers hope to continue<br />

applying their novel ideas to other areas<br />

of water treatment and materials science.<br />

“Coagulation has been in use for centuries<br />

and nothing has really changed. You<br />

would think at this point in time, after<br />

thousands of people have thought about<br />

this, that we would have perfected the water<br />

treatment process. But there’s always<br />

opportunity to innovate,” DuChanois<br />

said. Perhaps more innovations, building<br />

on the success of the AMC, will lead to effective<br />

solutions to the abiding challenge<br />

of energy-efficient water purification.<br />

ABOUT THE AUTHOR<br />

IMAGE COURTESY OF RYAN DUCHANOIS<br />

HANNAH RO<br />

HANNAH RO is a first-year in Trumbull from Orange County, California. In addition to writing for the<br />

Yale Scientific, she volunteers with Synapse, researches in the Bergwitz lab, and plans events for Korean<br />

American Students at Yale.<br />

THE AUTHOR WOULD LIKE TO THANK Ryan DuChanois for his time and enthusiasm to share their<br />

research.<br />

FURTHER READING<br />

Jinwei Liu, Shihan Cheng, Na Cao, Chunxiang Geng, Chen He, Quan Shi, Chunming Xu, Jinren Ni, Ryan<br />

M. DuChanois, Menachem Elimelech, Huazhang Zhao. Actinia-like multifunctional nanocoagulant for singlestep<br />

removal of water contaminants. Nature Nanotechnology, 2018; DOI: 10.1038/s41565-018-0307-8<br />

T TARGETS DIFFERENT TYPES OF CONTAMINANTS. IMAGE COURTESY OF RYAN DUCHANOIS<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

14


THE DEFENSES CANCERS RAISE AGAINST<br />

US, AND HOW WE DEFEAT THEM<br />

RESCUING<br />

the<br />

IMMUNE<br />

SYSTEM<br />

BY SAMI ELRAZKY<br />

ART BY ELISSA MARTIN


FOCUS<br />

medicine<br />

The process of drug development is enigmatic<br />

to most of us. Several groups, labs,<br />

and industries contribute considerable<br />

time and resources to producing the medicines<br />

which keep us healthy. The key to a<br />

drug’s success is the interplay between industry,<br />

which ultimately produces the drug,<br />

and academic research, where the “next big<br />

drug” is usually discovered. Research can<br />

be a slow and uncertain process–years pass<br />

seamlessly between major discoveries. In<br />

contrast, the companies that translate these<br />

discoveries into the clinic follow a strict,<br />

fast-moving timeline.<br />

In rapidly expanding fields of science like<br />

cancer immunotherapy, the disconnect between<br />

academic research and industry can<br />

sometimes lead to problems. A recent study<br />

in the Chen group at the Yale School of Medicine<br />

has overturned our traditional understanding<br />

of a certain immunological mechanism–brining<br />

under scrutiny the use of<br />

certain drugs which have been used by pharmaceutical<br />

industry for years. In the study,<br />

the researchers characterized and examined<br />

the interaction between fibrinogen-like protein<br />

1 (FGL-1) and the LAG-3 receptor on<br />

immune T cells. The results of this study<br />

could lead to the development of better treatments<br />

in the field of cancer immunotherapy.<br />

The immune system<br />

The human immune system is an intricate<br />

machine because of the amazing variety<br />

of threats it has to recognize and deal with,<br />

from bacteria, to viruses, to even fungi. Its<br />

operation is centered around innate immunity,<br />

which involves recognizing interactions<br />

between molecular sensors attached<br />

to immune cells and common types of molecules<br />

on foreign cells. But some invaders,<br />

such as viruses, cannot be recognized in this<br />

way. In response, every non-immune human<br />

cell has evolved antibodies and the major<br />

histocompatibility complex ligands class<br />

I or II (MHC I/II), which are attached to the<br />

cell surface and serve as a diagnostic tool for<br />

immune T cells.<br />

The MHC ligand samples protein fragments<br />

from inside the cell and presents it<br />

for inspection by the T cells, which roam<br />

around like policemen. When a virus invades<br />

and forces the cell to produce abnormal<br />

proteins, these proteins will appear on<br />

the MHC-I, and the T cell responsible for<br />

identifying it will be activated. In many cases,<br />

the T cell will kill the infected cell and<br />

stop widespread infection.<br />

In a pinch, this process also allows T cells<br />

to detect and kill cancer cells. Cancer cells<br />

originate from accumulated random mutations<br />

that endow them with an abnormally<br />

high proliferation rate, allowing these cancer<br />

cells to grow unchecked in the body.<br />

These mutations often result in abnormal<br />

protein production, which are sampled and<br />

presented on the MHC-I ligand on the cell’s<br />

surface. T cells can recognize this abnormality<br />

and act as necessary.<br />

I GOT TO THINKING,<br />

MAYBE MHC-II ISN’T<br />

THE MAJOR LIGAND.<br />

MAYBE THE LIGAND<br />

IS SOMEWHERE ELSE.<br />

What is cancer immunology?<br />

This study was supervised by Lieping<br />

Chen, United Technologies Corporation<br />

professor in cancer research and professor<br />

of Immunobiology, Dermatology, and Medicine<br />

at Yale. He had previously revolutionized<br />

cancer treatment with his discovery<br />

and characterization of the PD-L1 ligand, a<br />

cell-death pathway that cancer cells hijack to<br />

prevent an immune response. His research<br />

group works to use the human body’s natural<br />

immune system to fight cancer. To do so,<br />

the team faces a number of challenges. Cancer<br />

cells proliferate rapidly and can thereby<br />

rapidly evolve defenses against T cells. One<br />

such defense is misdirection–when binding<br />

between an abnormal MHC-I ligand and a<br />

T cell receptor sends off an alarm signal, the<br />

cancer cell can induce other interactions that<br />

cancel out that signal, or even kill the T cell.<br />

For example, a cancer cell under attack<br />

by the immune system can evolve the PD-<br />

L1 ligand. The next time a T cell binds to<br />

the MHC-I, the cancer cell will attach its<br />

PD-L1 ligand to the PD-1 receptor on the<br />

T cell. This binding sends a signal to the T<br />

cell to both stop attacking and to self-ter-<br />

16 Yale Scientific Magazine March 2019 www.yalescientific.org


medicine<br />

FOCUS<br />

minate through apoptosis. The PD-L1 ligand<br />

is, in essence, the cancer cell’s “get<br />

out of jail free” card.<br />

To mitigate the cancer cell’s defenses, cancer<br />

immunologists can introduce antibodies<br />

that block the T cell’s PD-1 receptor. The<br />

T cell regains the ability to bind to the MHC<br />

ligand and sound the alarm. This, of course,<br />

prompts the cancer cell line to either find another<br />

way to suppress immune cells or die<br />

out. What results is a game of cat-and-mouse<br />

between the cancer cell and the T cell. Clearly,<br />

an understanding of the various interactions<br />

between cancer cells and immune system receptors,<br />

as well as the cellular responses they<br />

induce, is critical to controlling cancer.<br />

Signals of an issue<br />

IMAGE COURTSEY OF FLICKR<br />

Another receptor that caught the eye of<br />

the Chen group was LAG-3 on T cells. LAG-<br />

3, like PD-1, is an inhibitory receptor on T<br />

cells. That is, if a suitable molecule binds in<br />

a complementary manner to LAG-3, the T<br />

cell loses its ability to attack foreign and cancerous<br />

cells. Discovered before PD-1, the<br />

MHC-II ligand–the version of MHC ligand<br />

on specialized immune cells–was commonly<br />

thought to be the complementary ligand to<br />

the LAG-3 receptor for decades. As a result,<br />

numerous pharmaceutical companies produced<br />

drugs to block LAG-3 from binding<br />

to MHC-II. The rationale was that introducing<br />

molecules that crowd out the MHC-II<br />

ligands will allow LAG-3 will be unbound,<br />

allowing T cells to remain active.<br />

However, evidence had been mounting<br />

that MHC-II is not the major ligand that<br />

pairs with LAG-3. A number of research<br />

studies demonstrated that other kinds of<br />

molecules that prevent LAG-3-mediated inactivation<br />

without blocking MHC-II were<br />

effective at amplifying T-cell responses and<br />

managing tumors, indicating that the MHC-<br />

II/LAG-3 interaction may not be as important<br />

as researchers thought.<br />

Unfortunately, the majority of LAG-3 targeting<br />

drugs pushed to clinical trials are still<br />

MHC-II blockers. Jun Wang, an associate<br />

research scientist in the Chen lab and first<br />

author on the paper, is acutely aware of this<br />

issue. “This phenomenon has been in this<br />

field for more than fifteen years…people<br />

know the issue is there, but they don’t know<br />

the biology and so they continue to produce<br />

MHC-II blockers,” Wang said. It was this<br />

widespread issue that drew Wang to investigate.<br />

“I got to thinking, ‘maybe MHC-II isn’t<br />

the major ligand. Maybe the ligand is somewhere<br />

else,” Wang said.<br />

Dispelling an old misunderstanding<br />

Wang began experimenting by growing T<br />

cells and cancer cells together in a medium<br />

lacking serum, a slurry of the different proteins<br />

and fluids typically used to help cells<br />

remain healthy in laboratory dishes. In this<br />

case, using MHC-II blocking antibodies induced<br />

no therapeutic response whatsoever,<br />

whereas use of the same antibodies on<br />

cells grown on regular serum medium had<br />

ABOUT THE AUTHOR<br />

a small effect on cancer cell growth. This<br />

experiment indicated that the antibodies<br />

weren’t blocking MHC-II, but rather something<br />

in the serum that was responsible for<br />

binding to LAG-3.<br />

To find the molecule responsible in the serum,<br />

the team scanned the human genome<br />

for proteins that would bind to LAG-3. Using<br />

what is termed a GSRA assay, they found<br />

that fibrinogen-like protein 1 (FGL-1), a<br />

protein normally produced in the liver, had<br />

a very high binding affinity for LAG-3. “We<br />

generate antibodies as therapeutic drugs because<br />

they have very high affinity for receptors…but<br />

[the FGL-1/LAG-3 interaction]<br />

has a very high affinity, just like on the antibody<br />

level,” Wang said.<br />

Further experimentation confirmed the<br />

importance of this interaction towards cancer<br />

research. A biophysical study found that<br />

FGL-1 binds to the area of LAG-3 that is<br />

blocked by the non-MHC-II blockers often<br />

used in laboratory studies, finally explaining<br />

the effectiveness of these blockers<br />

in years of studies. Furthermore, knocking<br />

out the FGL-1 gene reduced T cell deactivation<br />

at a comparable level to LAG-3, further<br />

strengthening the connection between<br />

the two. Finally, just like PD-L1, the team<br />

found that FGL-1 was greatly upregulated<br />

in humans with solid tumor cancers, indicating<br />

that the protein acts as a defensive<br />

measure used by cancer cells against T cells<br />

and could serve as a helpful biomarker for<br />

cancer immunotherapy.<br />

This research demonstrates that FGL-<br />

1 is the complementary ligand to LAG-3,<br />

meaning that the numerous drugs produced<br />

to target MHC-II are relying on biological<br />

assumptions proven to be false.<br />

Chen hopes that his team’s research will<br />

lead to the creation of new treatments<br />

which take advantage of this newly discovered<br />

FGL-1/LAG-3 interaction.<br />

SAMI ELRAZKY<br />

SAMI ELRAZKY is a first-year prospective Molecular, Cellular and Developmental Biology major<br />

in Saybrook College. Hailing from Tacoma, Washington, he also writes for the Yale Journal of<br />

Medicine and Law.<br />

THE AUTHOR WOULD LIKE TO THANK Dr. Jun Wang for his extensive explanation of the work, and<br />

Dr. Lieping Chen for his insights into the world of cancer immunology.<br />

FURTHER READING<br />

Wang, Jun, Miguel F. Sanmamed, Ila Datar, Tina Tianjiao Su, Lan Ji, Jingwei Sun, Ling Chen et al.<br />

“Fibrinogen-like Protein 1 Is a Major Immune Inhibitory Ligand of LAG-3.” Cell 176, no. 1-2 (2019):<br />

334-347.<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

17


FOCUS<br />

neuroscience<br />

Can you remember when you started to remember?<br />

At what point in time, and by what<br />

mechanism, does our brain gain the ability to<br />

form memories? A recently published study<br />

conducted by Usman Farooq and George<br />

Dragoi, assistant professor of psychiatry and<br />

neuroscience at the Yale School of Medicine,<br />

has probed the developmental timeline of<br />

memory formation and extracted key information<br />

about this process. This study aimed to uncover<br />

the mechanism through which animals<br />

like us develop the ability to form memories<br />

that link our past experiences with our present<br />

and future selves.<br />

Untangling<br />

the formation<br />

ofmemories<br />

LEARNING<br />

to<br />

REMEMBER<br />

B Y I S A A C W E N D L E R<br />

Conventional views of memory development<br />

The scientific community has long<br />

known that memories are established,<br />

stored, and maintained in the brain. Broadly<br />

speaking, there are three types of memory:<br />

sensory, which lasts only a few seconds;<br />

working, which lasts from a few seconds<br />

to a minute; and long-term, which can last<br />

for years. Different parts of the brain, such<br />

as the hippocampus or amygdala, serve<br />

as mediators for these different types of<br />

memory. These brain regions house the<br />

networks and ‘bundles’ of interconnected<br />

neurons whose interplay forms the physiological<br />

foundation of memory.<br />

Moreover, it is known that newborns<br />

are not able to form long-lasting memories<br />

immediately post-birth; for their first<br />

weeks to months, they are consciously<br />

in the present, after which they gain the<br />

ability to form memories. Although much<br />

is known about the anatomical and neurological<br />

support of new memory formation<br />

in adulthood, scientists know far less<br />

about the exact mechanisms that prompt<br />

the genesis of memory in early animal development.<br />

The present study fills this gap<br />

in knowledge, looking specifically at the<br />

neurological changes during the development<br />

of episodic memory–a type of longterm<br />

memory dealing with first-hand experiences–in<br />

rats.<br />

Stages of memory formation<br />

Farooq and Dragoi wanted to study the<br />

developmental timeline of memory formation<br />

in real time, so they monitored the<br />

electrophysiological activity of different<br />

“neuronal ensembles”–bundles of functionally<br />

connected neurons–in the hippocampi<br />

of rats over the third and fourth<br />

weeks after birth. They chose to monitor<br />

the hippocampi because they are known<br />

to be critical for the formation of new<br />

memories in adulthood. These rats were<br />

placed onto a linear track and allowed to<br />

move freely while their neuronal activity<br />

was recorded and decoded to display<br />

spatial information. “Particular neurons<br />

in the hippocampus ‘code’ or ‘fire’ for discrete<br />

locations; together, a population of<br />

these so-called ‘place cells’ can represent a<br />

whole arena composed of many locations,”<br />

Farooq said. In other words, each instantaneous<br />

location along the track was rep-<br />

18 Yale Scientific Magazine March 2019 www.yalescientific.org


neuroscience<br />

FOCUS<br />

IMAGE COURTESY OF FLIKR<br />

Much of the structure and function of the<br />

animal brain is conserved across different<br />

species; this allows the use of rats as model<br />

organisms for investigating phenomena that<br />

affect even humans<br />

resented by a specific pattern of neuronal<br />

activity. Furthermore, the neural activity<br />

of the rats was recorded during periods of<br />

sleep and “awake rest,” where the rats were<br />

awake but resting on the track.<br />

The goal of the study was to see how<br />

the activity of these neuronal ensembles<br />

changes over time, as the rats gain the ability<br />

to remember their previous experiences<br />

on the track. This experimental design allowed<br />

for direct investigation of the development<br />

of episodic memory.<br />

The researchers observed that episodic<br />

memory development occurs in three separate<br />

stages. In the first stage, around two<br />

weeks after birth, rats displayed neural<br />

activity characterized only by discrete, instantaneous<br />

locations represented by single<br />

neurons, lacking any pattern of activity<br />

characteristic of memory. “These young animals<br />

are completely ‘stuck’ in the present<br />

and cannot form memories,” Farooq said.<br />

In the second stage, around the middle of<br />

week three, the rats began to display signals<br />

that functionally connected different neurons<br />

to one another. Although these signals<br />

were not yet characteristic of full-fledged<br />

memories, the neural patterns of the rats at<br />

this stage gradually started to organize into<br />

sequential motifs that “preplay” the patterns<br />

expressed during memory encoding,<br />

a kind of memory storage practice intrinsic<br />

to the adult animal brain. Interestingly,<br />

these preconfigured patterns were expressed<br />

during sleep, independently of any<br />

external stimuli.<br />

By the onset of stage three–about three<br />

and a half weeks after birth–the rats had<br />

developed an ability to modify their preconfigured<br />

neural network of activity in<br />

response to a new experience. With this<br />

in place, the rats were now able to develop<br />

episodic memories. These memories were<br />

characterized by the strengthening and manipulation<br />

of the preexisting sequential patterns.<br />

Crucially, the rats in this final stage<br />

had developed the ability to use “replay”,<br />

wherein the preconfigured neural connections<br />

are used to relate different neurons–<br />

and thus, different locations on the track–in<br />

a sequential, long-lasting temporal manner.<br />

In other words, these rats had developed<br />

the ability to form memories.<br />

Learning about memory<br />

“Prior to our study, there was essentially a<br />

gap in our understanding of how and when<br />

these sequential patterns develop,” Dragoi<br />

said. Having observed these three stages of<br />

memory formation, the researchers were<br />

able to identify two interdependent prerequisites<br />

for memory development. First, the<br />

preconfigured patterns of sequential neural<br />

activity must form; next, the external<br />

world must impose some changes to these<br />

neural patterns of the brain. These steps<br />

must occur in that order, otherwise the<br />

brain has an insufficient foundation upon<br />

which to build a memory.<br />

These distinct stages of memory development<br />

might have evolutionary roots. In<br />

the same time it takes for a newborn experimental<br />

rat to progress through the developmental<br />

steps in the confines of the linear<br />

track, a wild newborn would leave its nest<br />

to explore the surrounding environment.<br />

The emergence of an episodic memory is<br />

ABOUT THE AUTHOR<br />

almost certainly essential for these animals<br />

to navigate an unforgiving external environment<br />

and stay alive.<br />

The results of this study provide interesting<br />

insights into the animal mind. They bring us<br />

closer to elucidating the system by which the<br />

brain makes and stores memories. “This is an<br />

important question which has attracted the<br />

attention of philosophers for centuries. Now<br />

we’re able to directly test some of their hypotheses,”<br />

Farooq said.<br />

Experimental challenges and future plans<br />

Aside from valuable scientific contributions<br />

made by this study, it is also an archetypal<br />

developmental study. A tremendous<br />

amount of time and preparation was necessary<br />

to lay the groundwork and design the<br />

right approach to the problem. “For this<br />

developmental study, it took close to four<br />

years from the start of the experiments until<br />

its publication,” Dragoi explained.<br />

As for the future, Dragoi and Farooq are<br />

looking to pursue a few different routes.<br />

One possible effort might focus on dissecting<br />

the factors, mechanisms, and neuronal<br />

circuits underlying the developmental processes<br />

investigated in this study. Another<br />

potential direction involves studying the<br />

development of neural patterns in animal<br />

models of neurodevelopmental disorders,<br />

potentially allowing comparisons to the<br />

current findings, to elucidate any deficits in<br />

the developmental system associated with<br />

the disorders.<br />

In any case, this study has managed to<br />

provide some answers to the fundamental<br />

question of memory formation. This understanding<br />

will prove crucial to support<br />

further exploration of the miracle that is the<br />

animal brain.<br />

A R T B Y S U N N I E L I U<br />

Isaac Wendler<br />

Isaac Wendler is a junior in Berkeley College, majoring in Chemistry and MCDB. When he’s not<br />

putting in hours on his chemistry research project, he can be found at Payne Whitney playing<br />

pick-up basketball or representing the Thundercoqs in intramurals.<br />

THE AUTHOR WOULD LIKE TO THANK Dr. George Dragoi and Usman Farooq for their time and<br />

enthusiasm about their research.<br />

FURTHER READING<br />

Farooq, U., and G. Dragoi. "Emergence of preconfigured and plastic time-compressed sequences in<br />

early postnatal development." Science 363, no. 6423 (2019): 168-173.<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

19


FOCUS<br />

cell biology<br />

E S S E<br />

In the average adult human, half a pound<br />

of proteins is turned over daily–destroyed<br />

and rebuilt from scratch. Among the many<br />

biological processes that occupy an organism<br />

throughout its lifetime, protein production<br />

ranks among the most intensive. Most forms of<br />

life therefore depend on accurate protein production.<br />

For instance, in humans, single base<br />

pair mutations in DNA blueprints, leading to<br />

a change in one amino acid building block, are<br />

known to cause diseases ranging from sickle<br />

cell anemia to cancer. However, a recent study<br />

at Yale has found an exception. Researchers in<br />

professor Dieter Soll’s group, working in collaboration<br />

with Antonia van den Elzen from<br />

professor Carson Thoreen’s group, examined<br />

the genomes–the complete set of genetic material<br />

in an organism–of thousands of bacterial<br />

species and found that parasitic bacteria with<br />

reduced genomes lack protein proofreading<br />

organisms do not face the same evolutionary<br />

pressures as free-living organisms. They do<br />

not have to be the best in their habitat to survive<br />

and reproduce. They merely need to be<br />

good enough.<br />

As a result, parasites can tolerate mutations<br />

and genomic deletions that weaken their survival<br />

ability. As free-living organisms transfer<br />

to parasitic lifestyles, they lose small pieces of<br />

their DNA over time due to the imperfect nature<br />

of DNA replication. Unlike in free-living<br />

organisms, these small losses of functionality<br />

are not important in the relatively noncompetitive<br />

host environment, so the bacteria survive<br />

and continue replicating, eventually losing up<br />

to ninety-five percent of their DNA. This process,<br />

known as “erosive evolution” or “Muller’s<br />

Ratchet,” explains why the genomes of parasitic<br />

organisms are much smaller than those of the<br />

free-living organisms from which they evolved.<br />

This erosion of the genome has consequences<br />

for the cellular machinery. During<br />

this reduction of genetic code, certain features<br />

typically thought to be essential for<br />

life can be lost, such as the ability of the<br />

protein production machinery to proofread<br />

the amino acid sequence being generated.<br />

After studying 10,423 genomes of 2,277<br />

bacterial species, Melnikov and his colleagues<br />

determined that parasitic bacteria<br />

with reduced genomes lose mechanisms responsible<br />

for protein quality control. “Parasitic<br />

life eradicates one important property<br />

of cells: accurate processing of genetic information,”<br />

Melkinov said.<br />

According to their research, an unusually<br />

large percentage of the parasitic bacteria<br />

studied had mutations in their protein<br />

synthesis quality control system. One<br />

such system identified was the aaRSs editing<br />

site of tRNA, where messenger RNAs<br />

coming from the organism's DNA are<br />

converted to protein. Seventeen percent<br />

of the bacteria had multiple mutations in<br />

one synthetase, an enzyme that links two<br />

amino acids together, and five percent had<br />

multiple mutations in several synthetases.<br />

Mutations were more common in species<br />

with reduced genomes. For instance, only<br />

6.3 percent of species with genome sizes<br />

greater than three million base pairs had<br />

editing-site mutations, while 83.8 percent<br />

of species with genome sizes less than one<br />

million base pairs had percent editing-site<br />

mutations. These mutations indicate that<br />

these species do not correct erroneous amino<br />

acids incorporated into the produced<br />

protein.<br />

ADVANTAGEOUS ERROR-<br />

mechanisms. How is it that these bacteria survive–and<br />

even thrive–with chunks of faulty<br />

protein production machinery?<br />

Parasites only have to be “good enough”<br />

The parasitic lifestyle may provide an answer.<br />

Organisms can be divided into two major<br />

lifestyles: free-living or parasitic. Free-living<br />

organisms live in open environments–air,<br />

soil, water, or ground–while parasitic organisms<br />

live inside other organisms. “These<br />

lifestyles are fundamentally different because<br />

of the difference in competition,” said Sergey<br />

Melnikov, a postdoctoral associate who led<br />

the project. While a free-living organism<br />

competes with others for environmental resources,<br />

a parasitic organism does not; the<br />

resources available inside its host are plentiful.<br />

Without intense competition, parasitic<br />

Loss of quality control<br />

Surviving and thriving with mutations<br />

Although their amino acid sequences may<br />

be incorrect, these parasites somehow still<br />

survive. To explain this, Melnikov uses an<br />

analogy between headphone cords and proteins.<br />

“Think of headphones. It is very easy for<br />

them to form knots and fold incorrectly, but<br />

only one form of the untangled headphones<br />

cord is useful to you,” Melnikov said. Just like<br />

headphone cords, proteins can misfold into<br />

incorrect structures that prevent them from<br />

executing their intended function.<br />

To offset the increased possibility of protein<br />

misfolding, parasites have elevated amounts<br />

of chaperone proteins. These chaperone proteins<br />

help guide the folding of the protein into<br />

its biologically active form. Consequently,<br />

parasitic proteins are still functional despite<br />

defects in their sequence. Moreover, protein<br />

E R R<br />

20 Yale Scientific Magazine March 2019 www.yalescientific.org


cell biology<br />

FOCUS<br />

N T I A L<br />

IMAGE COURTESY OF FLICKR<br />

Helicobacter pylori, a common bacterium that can<br />

lead to stomach ulcers and eventually stomach<br />

cancer. Its protein production mechanism is<br />

characteristic of a parasite, making it a potential<br />

target for amino acid therapy.<br />

clumping, a common and catastrophic consequence<br />

of misfolding, is less likely.<br />

All of the errors in protein synthesis proofreading<br />

actually help the bacteria survive the<br />

host’s immune defenses. This is because the<br />

errors result in incredible protein diversity.<br />

Leveraging the errors<br />

of the parasite could be suppressed without<br />

affecting the growth of the host.<br />

The reliance of parasites on chaperone proteins<br />

can also be targeted with small molecules<br />

that inactivate chaperones. Although<br />

this therapeutic approach could impact the<br />

chaperone proteins of the host as well, it likely<br />

will affect the parasites much more than it<br />

will the host, meaning that even a low dose<br />

could disrupt the growth of parasites without<br />

significantly impacting host growth and survival.<br />

Potential targets for these approaches<br />

include the parasites responsible for ulcers<br />

(Helicobacteri pylori), sexually transmitted<br />

diseases (Ureaplasma parvum), and Lyme disease<br />

(Borrelia afzelii).<br />

Studying genomes of over 10,000 bacteria<br />

has shown that a parasitic lifestyle leads to<br />

the degeneration of the genome, severely diminishing<br />

quality control systems in protein<br />

PRONE DNA TRANSLATION<br />

While typical proteins are constructed from<br />

twenty stock amino acids, these bacterial<br />

proteins were found to have over a hundred<br />

additional amino acid variants. The erroneous<br />

use of these unnatural amino acids<br />

would typically be prevented by the tRNA<br />

proofreading system, which is mutated in<br />

these bacteria. The result is that every “copy”<br />

of a protein produced is slightly different<br />

from the next.<br />

The constant flux in protein structure allows<br />

the parasite to evade the host immune<br />

system. As soon as the host develops immunity–that<br />

is, learns to recognize and destroy<br />

certain parasite proteins–the parasite changes<br />

its identity. These organisms have evolved<br />

to perfect their role as parasites.<br />

Researchers are devising novel approaches<br />

to fight parasite infection that exploit precisely<br />

this property. “What we are doing here<br />

is developing a treatment not based on what<br />

the parasite has, but based on what the parasite<br />

does not have: the ability to proofread<br />

amino acids when building proteins,” Melnikov<br />

said.<br />

Traditionally, therapeutics target a feature<br />

that is present in a specific parasite, but not<br />

in the host, in order to selectively destroy the<br />

specific parasite. But this knowledge of faulty<br />

protein synthesis can now be exploited to create<br />

a therapeutic that targets parasites in general.<br />

By increasing the availability of unnatural<br />

amino acids, the protein synthesis of the parasite<br />

could be disrupted without disrupting<br />

the protein synthesis of the host, whose proofreading<br />

mechanisms would prevent the use of<br />

these erroneous amino acids. If the amino acid<br />

sequence is sufficiently disrupted so that the<br />

proteins can no longer properly fold and perform<br />

functions necessary for life, the growth<br />

ABOUT THE AUTHOR<br />

synthesis. These errors are a parasite’s greatest<br />

strength, as its ever-changing composition<br />

ensures it avoids the host’s immune response,<br />

but they also present a key weakness to be exploited<br />

by future therapeutic approaches.<br />

KELLY FARLEY<br />

KELLY FARLEY is a first-year in Morse College interested in science communication. She is the Yale<br />

Scientific Magazine's Special Sections Editor, teaches a bilingual 3rd grade science class through<br />

Demos, and leads dance classes for children at Yale-New Haven Hospital through Peristalsis.<br />

THE AUTHOR WOULD LIKE TO THANK Dr. Sergey Melnikov for his time, knowledge, and enthusiasm<br />

while explaining his work.<br />

FURTHER READING<br />

Melnikov, Sergey et al. 2018. “Loss of protein synthesis quality control in host-restricted organisms.”<br />

Proc Natl Acad Sci USA.<br />

A R T B Y S U N N I E L I U<br />

O R S21<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine


FOCUS<br />

molecular biology<br />

CONQUER<br />

DIVIDE &<br />

Sequencing Two<br />

Types of RNA in<br />

a Single Cell<br />

BY MATT TU<br />

ART BY SUNNIE LIU


molecular biology<br />

FOCUS<br />

Imagine a police officer chasing two suspects<br />

down a narrow alleyway. He almost<br />

catches up to them, when suddenly, the<br />

alleyway splits into two paths going in opposite<br />

directions. One of the suspects takes<br />

the left path while the accomplice takes<br />

the right path. No matter which choice he<br />

makes, one convict will go behind bars,<br />

and the other will remain at large.<br />

This might seem a far-fetched scenario<br />

for scientists, but they in fact face a similar<br />

dilemma when genetically sequencing<br />

a single cell. Conventionally, much like the<br />

one policeman, researchers can sequence<br />

either the messenger RNA (mRNA) or<br />

micro RNA (microRNA) of a single cell,<br />

but never both simultaneously. Therefore,<br />

it remains poorly understood how both<br />

types of RNA affect and regulate each<br />

other within a single cell. Recently, a collaborative<br />

study involving the Lu and Fan<br />

labs at Yale has led to a method that allows<br />

both types of RNA from the same cell to<br />

be sequenced. This method could prove<br />

useful in a variety of fields, ranging from<br />

epigenetics to cancer research.<br />

A look inside the protein factory<br />

RNA is generally known to serve as a<br />

template and regulator in protein synthesis,<br />

but the true complexity of the specific<br />

roles played by various types of RNAs<br />

and how they interact with each other is<br />

less well-known. Each RNA type is like a<br />

worker with a unique job on an assembly<br />

line, and each of these workers coordinates<br />

with every other component in the overall<br />

production pipeline. The three main types<br />

are large messenger RNAs (mRNA), the<br />

intermediate between DNA and protein,<br />

transfer RNAs (tRNA), and ribosomal<br />

RNAs (rRNA), both of which are involved<br />

in the translation of mRNA into functional<br />

proteins. Less commonly known is microRNA,<br />

a small RNA composed of only<br />

twenty-two nucleotides that inhibits gene<br />

expression by attaching itself to specific<br />

strands of mRNA, physically blocking<br />

translation. MicroRNA, alongside a myriad<br />

of other RNA types, is crucial in gene<br />

expression regulation. For instance, abnormal<br />

activities of some microRNAs have<br />

been linked to the development of cancer<br />

and the onset of Alzheimer’s disease.<br />

Divide and conquer<br />

As a result, many researchers are interested<br />

in better understanding microRNA<br />

and other types of small RNA, especially<br />

at the single-cell level. For Jun Lu, associate<br />

professor of Genetics and co-principal<br />

investigator of the study, studying microRNA’s<br />

role in gene expression can help<br />

explain slight variations in functionally<br />

similar cells. “Looking at many genes in a<br />

single cell at a time has allowed us to detect<br />

variations of gene expression in cells<br />

of the same tissue…We are interested in<br />

understanding microRNA in single cells<br />

and their relationship to this variability,”<br />

Lu said.<br />

The only way to exactly determine the<br />

effects of microRNA on a cell’s genetic<br />

variability would be to also simultaneously<br />

sequence its messenger RNA, since the two<br />

regulate each other. Although it has long<br />

been possible to sequence both large RNA<br />

and small RNA in a group of cells, no previous<br />

methodology has proven successful<br />

at sequencing both large and small RNA<br />

types in a single cell. Directly combining<br />

current approaches does not work. “Just<br />

like how certain cooking ingredients cannot<br />

be added together, any technique to sequence<br />

large RNA, such as mRNA, cannot<br />

be combined with techniques to sequence<br />

small RNA like microRNA,” Lu explained.<br />

Therefore, in the first part of their<br />

study, Lu and his collaborator Rong Fan,<br />

associate professor of Biomedical Engineering,<br />

worked together to develop their<br />

own methodology that could co-sequence<br />

both types of RNA. “If we could somehow<br />

split the single cell in half, then we could<br />

sequence the microRNA and mRNA separately,”<br />

Lu said. In other words, what if<br />

the police officer in the beginning of this<br />

story had a partner to catch the other suspect?<br />

Initially, the team thought about<br />

engineering an extremely precise tool to<br />

cut the cell in half like a microscopic cake<br />

cutter. However, they eventually realized<br />

that this approach was nearly impossible<br />

to implement in practice, due to physical<br />

constraints and the inevitability of damaging<br />

the genetic material inside the cell.<br />

Instead, they decided to divide the cell using<br />

an approach called half-cell genomics.<br />

In this method, the membranes of single<br />

PHOTOGRAPHY BY KATE KELLY<br />

Professor Jun Lu holds a component of the<br />

laboratory robotics system the team used in<br />

their methodology.<br />

cells were first broken. The resulting lysate<br />

was split into two “half-cell lysates” where<br />

one was sequenced for its microRNA while<br />

the other was sequenced for its mRNA. To<br />

account for the smaller amount of genetic<br />

material in a half cell compared to a group<br />

of cells, the researchers chose sensitive sequencing<br />

techniques.<br />

Perfecting the “cell-splitting” methodology<br />

was difficult. For instance, when the<br />

researchers realized that each “half-cell”<br />

didn’t have equal amounts of microRNA,<br />

they had to tinker with freezing and heating<br />

processes in the cell lysis protocol to<br />

produce lysates that had equal amounts of<br />

mRNA and microRNA. With the kinks sorted<br />

out, the technique proved to be remarkably<br />

successful–even to the surprise of the<br />

researchers. Out of twenty trials with single<br />

cells, nineteen of them passed the quality<br />

check for both microRNA and mRNAs.<br />

Insights from the new method<br />

In addition to verifying their methodology<br />

on single cell RNA samples, the researchers<br />

also analyzed the resulting sequencing<br />

data to determine the degree to<br />

which variations in microRNA expression<br />

related to mRNA expression in a single<br />

cell. As predicted, the variation of abundantly<br />

expressed microRNAs was significantly<br />

anti-correlated with the mRNAs<br />

that those microRNAs targeted, suggesting<br />

that differences in cell-specific microRNA<br />

expression alone can lead to non-genetic<br />

variation between cells. Furthermore, the<br />

researchers found evidence suggesting that<br />

some large RNAs could also regulate small<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

23


FOCUS<br />

molecular biology<br />

LU AND FAN BELIEVE<br />

THAT THEIR "HALF-<br />

CELL GENOMICS"<br />

METHOD CAN SERVE<br />

AS A POWERFUL<br />

MOLECULAR TOOL<br />

TO UNDERSTAND<br />

GENOMICS ON A<br />

CELL-BY-CELL BASIS.<br />

of thousands and even millions. To further<br />

advance this technology and broaden its<br />

impact, designs of higher throughput is the<br />

direction to go.” Chen said. Another foreseeable<br />

direction is to automate and enhance<br />

the microRNA sequencing process<br />

using tools like microfluidics.<br />

A large part of this project’s success was<br />

attributed to the close collaboration between<br />

the Lu and Fan labs, and a shared<br />

interest in developing single-cell techniques<br />

to study microRNA. “It was natural<br />

for us. We had expertise on the molecular<br />

aspects of small RNA research, while Dr.<br />

Fan had expertise in developing single-cell<br />

technologies,” Lu said. Zhuo Chen, a graduate<br />

student in the Fan lab who was part of<br />

the project team, spoke to the contrasting<br />

strengths of the collaborators. “Engineers<br />

are more results-driven and are always trying<br />

to build things that work. Biologists<br />

are more detail-oriented and curious about<br />

why things happen,” he said.<br />

The collaboration between the Lu and<br />

Fan labs has led to the creation of a new<br />

and important method in single-cell gene<br />

sequencing and will likely continue to reap<br />

benefits given the multiple promising directions<br />

in sight. As in the case of co-sequencing,<br />

it often takes two collaborators<br />

to divide and conquer.<br />

RNAs like microRNAs, suggesting an interdependent<br />

relationship rather than conventional<br />

one-way regulation of microR-<br />

NAs inhibiting mRNAs.<br />

Lu and Fan believe that their “half-cell<br />

genomics” method can serve as a powerful<br />

molecular tool to understand genomics on<br />

a cell-by-cell basis. Specifically, this tool<br />

could help researchers study the relationship<br />

between the expression of cancer-initiating<br />

microRNAs and differences in individual<br />

cells in cancer tissue. “If we can<br />

continue improving this methodology, we<br />

could look at cancer tissue in high definition…and<br />

extensively study one cell’s<br />

gene expression patterns in a manner unlike<br />

other techniques,” Lu said. Moreover,<br />

this methodology opens the door to other<br />

methods that can further examine the underlying<br />

molecular “wiring” between small<br />

RNAs and large RNAs. “Since we can now<br />

sequence both small and large RNAs in a<br />

single cell, we can start to use computational<br />

tools to figure out the logic behind<br />

how they regulate each other,” Lu said. Ultimately,<br />

however, the researchers hope to<br />

have their new method adopted by others,<br />

who can apply it to solve a wide variety of<br />

specific problems.<br />

Collaboration and continuation<br />

Despite this recent success, both labs<br />

acknowledge the limitations of their current<br />

implementation of “half-cell genomics”<br />

and seek to improve the speed and<br />

scalability of this technique. “If you take<br />

a look at other single-cell measurements,<br />

you will be amazed at how many single<br />

cells they can measure in a single run–tens<br />

ABOUT THE AUTHOR<br />

PHOTOGRAPHY BY KATE KELLY<br />

Professor Jun Lu demonstrates operation of the<br />

automatic pipetting machine in the Lu and Fan<br />

labs at Yale.<br />

MATT TU<br />

MATT TU is a first-year and a prospective Statistics and Data Science major. He is the Webmaster of<br />

Yale Scientific and is planning to work in Ty Cannon’s psychology lab over the summer.<br />

THE AUTHOR WOULD LIKE TO THANK Professor Jun Lu and Zhuo Chen for time taken to explain<br />

their research.<br />

FURTHER READING<br />

Wang, Nayi, Ji Zheng, et al. "Single-cell microRNA-mRNA co-sequencing reveals non-genetic heterogeneity<br />

and mechanisms of microRNA regulation." Nature Communications 10, no. 1 (2019): 95.<br />

24 Yale Scientific Magazine March 2019 www.yalescientific.org


ROBOT<br />

biotechnology<br />

SELF-ORGANIZING ABILITY OF ROBOTS WITH-<br />

OUT CENTRAL CONTROL<br />

BY ANTALIQUE TRAN<br />

IMAGE COURTESY OF WIKIMEDIA<br />

A swarm of kilobots, cheap robots that the research team used<br />

for modeling, demonstrate the morphogenetic capability of a<br />

true robot swarm.<br />

Can technology develop a mind of its own? The robot<br />

apocalypse is not yet upon us, but the Sharpe group from<br />

European Molecular Biology Laboratory (EMBL) in Barcelona<br />

has recently demonstrated the capability of a group of<br />

robots to self-organize into a swarm without a central control.<br />

Modeled after morphogenesis, the spatial organization<br />

of biological systems during embryonic development, the<br />

spatial behavior of the robot swarm works through feedback<br />

loops rather than through top-down control. Since<br />

self-organizing systems have the advantage of adaptability<br />

in size and shapes and can function despite the unreliability<br />

of an individual, replicating this biological process in human<br />

technology could pave the way to innovations in machinery,<br />

architecture, and pharmaceuticals.<br />

Previous morphogenetic engineering studies had only<br />

addressed robotic swarms in small scale simulations or<br />

through top-down control, but could a true robot swarm<br />

self-organize on a larger scale? The group addressed this<br />

question by programming kilobots–cheap robots that allowed<br />

for large-scale testing of the functioning of a whole<br />

despite the imperfections of the individual–to follow Turing<br />

patterns, or patterns derived from feedback mechanisms<br />

rather than from positional information.<br />

Combined with robot migration, a Turing pattern determined<br />

by virtual molecules allowed the swarm to generate<br />

a pattern without relying on individuals’ precise distances<br />

and angles from one another. Robots moved only if they<br />

were on the edge of the swarm and if they were in a region<br />

of low virtual molecular concentration; they were to stop<br />

once they reached an area of high molecular concentration<br />

called a “Turing spot.” LED lights on the robots represented<br />

molecular gradients that allowed the team to observe the<br />

spatial patterns formed.<br />

After the team decided that spots, as opposed to stripes<br />

or inverted spots, were the best pattern for exploring morphological<br />

development, a larger-scale study tested the<br />

swarm’s ability to produce more complex morphological<br />

shapes. This showed that the swarm could create organic<br />

shapes just as biological cells are able to. Test trials without<br />

the programmed Turing patterns did not organize to make<br />

the same shapes, confirming that the results were indeed a<br />

result of patterning rather than simple robot movement. In<br />

line with the adaptability of local self-organized systems,<br />

the robotic swarm could also reproduce Turing spots no<br />

matter what the shape or size of the original swarm was,<br />

and if the swarm was damaged; the swarm could still recover<br />

the general pattern even if it was split if half.<br />

This robot swarm thus provides all the advantages of an<br />

organic, self-organizing system–as the patterns that formed<br />

were dynamically adaptable. The morphological patterns<br />

persisted throughout the trials despite varying the size of<br />

the swarm–ranging from 110 to 300 robots–or varying the<br />

initial shape of the swarm–circular or rectangular. “Tissue<br />

healing” also occurred with any inflicted damage. These<br />

characteristics could be especially useful for applications in<br />

nanoengineering, such as drug targeting via a nanoparticle<br />

swarm, or in dynamic machinery and structures, such as a<br />

bridge that adapts to its river.<br />

There is a still a long way to go before applications like<br />

that. Although this proof-of-concept has been achieved<br />

here using Turing patterns and feedback mechanisms, the<br />

team hopes for a more sophisticated control system in the<br />

future able to achieve more complicated shapes.<br />

THIS ROBOT SWARM THUS PROVIDES ALL<br />

THE ADVANTAGES OF AN ORGANIC, SELF-<br />

ORGANIZING SYSTEM—AS THE PATTERNS THAT<br />

FORMED WERE DYNAMICALLY ADAPTABLE.<br />

FEATURE<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

25


GO WITH<br />

MODELING FLEXIBLE MICROSCOPIC SHEETS THAT<br />

FEATURE materials science<br />

SCULPT THEMSELVES<br />

The Swiss Army Knife is a necessity for survivalists and<br />

the outdoorsy, providing tools for any situation you might<br />

encounter. Researchers at the University of Pittsburgh’s Department<br />

of Chemical Engineering have created a computational<br />

model for just that—on a microscopic scale. Researchers<br />

Abhrajit Laskar, Oleg Shklyaev, and Anna Balazs<br />

have developed a model for flexible, chemically-active<br />

sheets capable of interacting with a fluid environment to<br />

create fluid flow, which in turn moves and alters the shape<br />

of the sheet. Each sheet is composed of a network of flexible<br />

chemical bonds connecting nodes coated with catalysts,<br />

chemical compounds that speed up chemical reactions.<br />

This potential for autonomous deformation and locomotion<br />

opens up new opportunities for mechanical and biomedical<br />

engineering on the microscopic scale.<br />

A key of the model is a chemical reaction that has reactants<br />

and products of different volume. “Think of the reactants as<br />

being little balloons and the products as being big balloons,”<br />

Balazs said. “The big balloon and little balloon displace different<br />

amounts of fluid in the container, and so they make<br />

these local density gradients in the fluid.” This phenomenon,<br />

known as solutal buoyancy, is the crux of the model.<br />

The reaction occurs through catalysts which break down<br />

reactants introduced into the fluid. An example reaction is<br />

the decomposition of hydrogen peroxide (H2O2) into water<br />

(H2O) and oxygen (O2). This reaction is sped up with an enzyme,<br />

called catalase, coated onto a sheet in the shape of a petaled<br />

flower. The less-dense products rise upward in the fluid,<br />

changing the direction of fluid flow and causing the petals to<br />

rise. As the amount of H2O2 remaining in solution decreases,<br />

the sheet flattens again due to the drop in products made.<br />

By intermittently introducing H2O2 into the system, the catalyst-coated<br />

petals can be made to open and close cyclically.<br />

Coating the petals with different catalysts and setting up a<br />

chain of catalytic reactions allow for further control over the<br />

sheet’s shape. However, differing reactions’ rates sometimes<br />

pose a challenge if the reaction rate of one is significantly slower<br />

than another’s. This can be compensated for by coating the<br />

walls of the container with an appropriate catalyst to increase<br />

the surface area catalyzing the reaction, or by changing the areal<br />

density of catalyst on the surface of the sheet.<br />

The sheets can be made to move through the chamber<br />

by two primary methods. One is modeled by small bumps<br />

representing unevenness along the bottom surface of the<br />

container. A catalase-coated sheet would tumble over each<br />

bump because of its flexibility and chemical activity generating<br />

upward local fluid flow. In this way, an active sheet<br />

can be made to move through the channel over obstacles at<br />

BY MICHAEL ADEYI<br />

IMAGE COURTESY OF FLICKR<br />

If a chemical reaction involving reactants and products of different<br />

volumes are symbolized as small balloons and large balloons, the two<br />

balloons would displace different amounts of fluid in the container.<br />

the bottom until there is no more reactant to be consumed.<br />

The second method is to have a catalyst-coated sheet with<br />

edges heavier than the interior of the sheet, allowing the sheet<br />

to move like an inchworm—the products flow upward and create<br />

a bulge in the lighter center. After the reactant is consumed,<br />

the sheet flattens out, displaced to a new position. Addition of<br />

more H2O2 can repeat the process and propel the sheet further.<br />

Another method to induce inchworm locomotion would be to<br />

coat the sheet with an uneven concentration of catalyst. “You’d<br />

have more of the catalyst in one region than the other, and you<br />

could potentially achieve the same thing,” Balazs said.<br />

The researchers think the sheets could be used to identify<br />

diseased cells and, acting as claws, remove them from<br />

the bloodstream for examination. “The gripper itself has<br />

to be on the same size scale as the cell and also has to be<br />

very flexible and gentle,” Balazs said. Another use of the<br />

sheets would be for construction. “You can have these little<br />

hands that can pick up things in the microfluidic device<br />

and then do construction.” This could pave the way<br />

for what could be thought of as microscopic foundries.<br />

“It’s still challenging to make the sheet sufficiently flexible<br />

and compliant that it will fold and bend nicely without<br />

crumpling,” Balazs said. Coating flexible sheets with<br />

catalyst is not a technology most are familiar with, so<br />

bringing the model to life in a wet lab may be difficult.<br />

Balazs and her collaborators have certainly put us on the<br />

right track, and, regardless of the challenges, this technology<br />

is sure to do more than open cans and bottles.<br />

THE FLOW<br />

26 Yale Scientific Magazine March 2019 www.yalescientific.org


PESTS OR<br />

ecology<br />

HOW TERMITES HELP MITIGATE THE EFFECTS<br />

OF RAINFOREST DROUGHT<br />

BY MAKAYLA CONLEY<br />

IMAGE COURTESY OF FLICKR<br />

Termites move soil from deep down towards the surface, contributing<br />

to increased soil moisture and nutrient heterogeneity in rainforests<br />

during droughts.<br />

When termites come up in everyday conversation, horror<br />

stories of tented houses and damaged house foundations<br />

usually soon follow. However, of all the termite species in<br />

the world, only four percent are actually pests. Rather, most<br />

termites help maintain healthy ecosystems and soil integrity.<br />

This is what led researcher Louise Ashton of the University<br />

of Hong Kong and a group of scientists from around<br />

the world to fly to Borneo and study the effects of termite<br />

populations on tropical rainforest ecosystems.<br />

The researchers set about creating a large-scale experiment<br />

to test how native termite species interact with the ecosystem.<br />

In order to see the difference between environments with<br />

and without termite populations, the scientists killed off termite<br />

populations in several controlled plots of land. First, they<br />

set up four eighty-by-eighty-meter plots in the rainforest and<br />

physically removed all termite mounds within those areas.<br />

They then placed toilet paper dipped in low concentrations<br />

of insecticide across the plots. Since toilet paper is an appealing<br />

meal for termites, the insects ate the paper with insecticide<br />

and brought it back to their nests, thus effectively suppressing<br />

most of the termite population in the experimental zones.<br />

Ready to watch for an effect on the ecosystem, the timing<br />

of their experiment proved serendipitous. “We had<br />

originally intended to understand termites and ecological<br />

processes, but inadvertently we had data on termites<br />

during a major drought,” Aston said. The researchers soon<br />

realized that they had set up an experiment to collect data<br />

right in time for the 2015 El Niño drought, one of the<br />

strongest and most damaging droughts in recent history.<br />

The initial results shocked the researchers. During the<br />

drought, termite numbers doubled. “Everything else in<br />

the forest slows down in the drought, but the termites<br />

thrived,” Ashton explained. According to Aston, the termites<br />

acted as soil engineers. “They bring clay from underneath<br />

the soil to the surface to make the sheeting that<br />

they move around,” Ashton said. This results in greater<br />

soil heterogeneity and more soil moisture compared to the<br />

plots where termites were suppressed.<br />

Soil moisture and heterogeneity are key features of rainforest<br />

ecosystems intricately tied to biodiversity. Previous<br />

research shows that seedlings in the rainforest usually start<br />

growing around five centimeters under the soil surface,<br />

where researchers in this study noted that termites had a direct<br />

impact. To study the effect of termites on seedling survival,<br />

they conducted a transplantation experiment using<br />

liana seedlings in both the control and termite suppression<br />

plots. They quantified the number of seedlings that survived<br />

in soil with and without termite suppression and found that<br />

the control plots with termites had a significantly higher<br />

seedling survival rate during the drought compared to termite<br />

suppression plots. In this way, termites help the rainforest<br />

maintain a healthy ecosystem even during dry spells.<br />

In order to confirm that the differences in the initial experiment<br />

were due to the drought and not some other potential<br />

factor, the scientists conducted a second experiment the following<br />

year when the Borneo rainforests experienced normal<br />

rainfall. The findings of this second study confirmed the results<br />

of the drought experiment. There was no significant difference<br />

in the seedling survival rate of the control plots versus<br />

termite suppression plots during non-drought conditions,<br />

suggesting it was the doubled termite population that benefitted<br />

seedling growth during the drought. While it is not yet<br />

known exactly why termites thrive in drought conditions, tunneling<br />

and foraging ability in drier ground might play a role.<br />

This was the first large-scale termite suppression experiment<br />

ever to be conducted, serving as a model for future<br />

work in discovering the profound impact little critters<br />

have on entire ecosystems. While this study focused on the<br />

rainforests of Borneo, there is further research to be done.<br />

“There’s significant remaining work on other parts of the<br />

landscape, like agricultural systems or other disturbed areas,<br />

to see if they have lost this insurance policy that the termites<br />

confer,” Ashton said. Understanding how organisms contribute<br />

to an ecosystem’s response to extreme weather conditions<br />

has the potential to affect environmental policy and shape<br />

the way we interact with rainforests in the years to come.<br />

FEATURE<br />

www.yalescientific.org<br />

ENGINEERS?<br />

27<br />

March 2019<br />

Yale Scientific Magazine


STARING<br />

by || ELLIE GABRIEL<br />

art by || ELISSA MARTIN<br />

AT THE<br />

SKY<br />

NASA satellite finds new exoplanets<br />

just months after launch<br />

Many of us wonder what lies beyond<br />

Earth and our solar system. Star Wars, The<br />

Martian, and other science fiction works<br />

have helped to quench our imaginations,<br />

but perhaps it need not all be fiction. NA-<br />

SA’s recently retired Kepler satellite lasted<br />

approximately one decade on a mission to<br />

discover Earth-sized planets in habitable regions<br />

of stars. The Kepler satellite looked at<br />

one part of the Milky Way for years, whereas<br />

a promising new satellite is currently<br />

exploring the entire sky utilizing similar<br />

technology. NASA launched the Transiting<br />

Exoplanet Survey Satellite (TESS) in April<br />

2018 for a two-year mission. In its first four<br />

months of surveying, it has already successfully<br />

identified eight exoplanets, planets<br />

that lie beyond our solar system, and<br />

observed more than three hundred others<br />

for follow-ups. These are of great interest to<br />

scientists, as the data collected by satellites<br />

like Kepler and TESS can help answer questions<br />

about life in the universe and the suitability<br />

of other planets to sustain life.<br />

Transiting—the science behind satellites<br />

Kepler and TESS are able to determine<br />

the presence of exoplanets by monitoring<br />

the brightness of a particular star over<br />

time and detecting dips in brightness due<br />

to transiting, the movement of a planet in<br />

front of the star. A planetary transit reduces<br />

the brightness of a star as it passes across.<br />

The planet’s orbit can be deduced from the<br />

time between dips in brightness.<br />

TESS is expected to be a more effective<br />

planet hunter than Kepler, because TESS<br />

surveys a new patch of sky each month. It<br />

will take just two years for TESS to scan all<br />

360 degrees of sky seen from Earth. Since<br />

TESS spends a short amount of time scanning<br />

each sky segment, scientists do not expect<br />

it to find many planets with periods,<br />

the time it takes for a planet to make one<br />

full orbit, longer than one Earth month.<br />

Longer-period planets transit less frequently;<br />

therefore, TESS is expected to primarily<br />

detect planets with periods less than ten<br />

Earth days.<br />

Nevertheless, TESS has already proudly<br />

confirmed the existence of HD 21749b, an<br />

exoplanet with a period of 35.61 Earth days.<br />

The density of HD 21749b suggests a rather<br />

gaseous atmosphere. “The density is really<br />

valuable. It gives a sense of the composition<br />

of the planet,” said Diana Dragomir, postdoctoral<br />

researcher at the MIT Kavli Institute<br />

for Astrophysics and Space Research<br />

and member of the TESS team. Calculations<br />

like density, mass, temperature, and planet<br />

size are made possible by TESS technology,<br />

specifically through measuring how big of a<br />

dip in brightness occurs during transit. Furthermore,<br />

the magnitude of the dimming is<br />

related to the planet’s mass and diameter<br />

28 Yale Scientific Magazine March 2019 www.yalescientific.org


astronomy<br />

FOCUS<br />

relative to the star’s size. The star’s size can<br />

be determined using asteroseismological<br />

data from TESS. Additionally, the orbital<br />

size of the planet and the temperature of the<br />

star can be used to calculate the temperature<br />

of the planet. Evidently, planets cannot<br />

be considered isolated bodies, but part of a<br />

larger celestial system, particularly dependent<br />

on the stars they orbit.<br />

TESS and the life question<br />

IMAGE COURTESY OF WIKIMEDIA<br />

Researchers are able to explore the totality of the<br />

milky way galaxy with TESS, which they previously<br />

could not do.<br />

TESS is about the size of a big fridge and<br />

has four telescopes, each about ten centimeters<br />

in diameter, rather small compared to<br />

the Hubble Telescope. “It has a pretty funky<br />

orbital; it approaches Earth once per orbit<br />

and then swings out to avoid light reflected<br />

from Earth on the camera,” Dragomir said.<br />

Such light from Earth would be considered<br />

as contaminating measurements of light<br />

from distant stars. “We are looking in the<br />

inhabitable zone of stars for planets able to<br />

store water,” Dragomir explained. Earth is<br />

the perfect distance from the sun, making it<br />

able to hold water and sustain the immense<br />

diversity of life that it does. TESS attempts<br />

to find exoplanets that, like Earth, are located<br />

in what is known as the Goldilocks zone.<br />

“We think the results will answer the life<br />

question,” Dragomir said.<br />

Outlandish new worlds<br />

Of the over three hundred exoplanets<br />

that TESS has observed in its first four<br />

segments of the galaxy, many are already<br />

wondering what remains to be found. One<br />

strange finding was Pi Mensae c, reported<br />

in September 2018. Pi Mensae c has a 6.27-<br />

day orbit and is 2.14 times the diameter of<br />

Earth, with 4.8 times Earth’s mass, such that<br />

its density is close to that of water. Another<br />

planet, Pi Mensae b, orbits the same star<br />

but much more slowly, with a period of 5.7<br />

years, and has 3170 times Earth’s mass. It<br />

is rather unusual that Pi Mensae c is able<br />

to survive the eccentric orbital swinging<br />

caused by Pi Mensae b.<br />

TESS has also discovered a planet covered<br />

in lava known as LHS 3844b. It is 1.3 times<br />

the size of Earth but maintains an orbit of<br />

eleven Earth days. Its surface temperature<br />

of 540 degrees Celsius makes it interesting–<br />

but unfortunately, not a viable alternative to<br />

Earth for human life.<br />

The TESS mission goes beyond simply<br />

finding exoplanets. The information collected<br />

by TESS is continuing the space<br />

revolution with more rigor than any satellite<br />

before. “We will have<br />

the means to detect things<br />

like oxygen on other planets,”<br />

Dragomir said. Every<br />

detail discovered about<br />

other planets will help scientists<br />

understand Earth<br />

more thoroughly, as well<br />

as help improve TESS to<br />

plan future missions. Processes<br />

like planet formation<br />

can be more clearly<br />

understood by collecting a<br />

large amount of data from<br />

TESS’s search. For instance,<br />

planetary composition<br />

is key information<br />

that can help researchers<br />

discern the chemical reactions<br />

able to take place on<br />

individual planets–some<br />

of which are essential for<br />

sustaining life.<br />

Infinite promise<br />

“We’re working to extend the mission<br />

from two years,” Dragomir said. She believes<br />

that TESS will be in space for a while. In an<br />

effort to make full use of the TESS mission,<br />

NASA plans to launch the James Webb Space<br />

Telescope in 2020 to follow up on some of<br />

TESS’s most prominent discoveries. Besides<br />

directly hunting for planets and analyzing<br />

their physical characteristics, TESS is able<br />

to indirectly learn about planets through<br />

asteroseismology, a technique where stellar<br />

sound wave vibrations in stars are used<br />

to determine the composition of stars, their<br />

ages, and their sizes. Such sound waves result<br />

from temperature changes inside the star’s<br />

convection zone, an area between the core<br />

and visible surface where hot plasma rises,<br />

cools, and falls on repeat. When the plasma<br />

falls toward the core, it releases an energy<br />

wave that makes the star expand and contract,<br />

essentially making the star comparable<br />

to a bell. This bell rings through space. TESS<br />

is capable of sensing the star sounds while in<br />

space by seeking changes in star brightness,<br />

which is indicative of star “ringing.” Knowing<br />

more about stars can do much to shed<br />

light on the planets that orbit them.<br />

TESS has shown incredible promise in a<br />

rather short time frame. The conclusions of<br />

the mission are impossible to predict but exciting<br />

to imagine. TESS has proven that it is<br />

capable of turning science fiction into reality.<br />

Visual of Venus transiting across the sun.<br />

IMAGE COURTESY OF GBPHOTODIDACTICAL<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

29


FOCUS<br />

applied physics<br />

LIGHT<br />

READINGBY XIAOYING<br />

Lights on, lights off<br />

ral magnetization of the signal. In the past<br />

decade, since its discovery in gadolinium/<br />

As technology continues to evolve, data iron/cobalt alloys, AOS technology has<br />

writing is growing as an area of interest. seen an influx of attention and a boom in<br />

Simultaneously, the emerging field of spintronics<br />

studies how the spin and magneting<br />

the speed and energy efficiency of data<br />

research, as it shows potential in increasic<br />

properties of an electron can be used writing in spintronic memory devices.<br />

for information processing. All Optical This type of data memory relies on setting<br />

Switching (AOS) is a related technique the spin of electrons. Each electron can assume<br />

two states: up and down. These can<br />

that reverses magnetization of a magnetic<br />

material with short laser pulses. It allows<br />

one optical signal to<br />

patterns of spin can represent bits of infor-<br />

be utilized to represent data, as different<br />

control another optical<br />

mation. Because AOS allows information<br />

signal through interfering<br />

with the natu-<br />

appeal in its efficiency if possible.<br />

to travel at near light speed, it has wide<br />

The AOS mechanism still has<br />

ZHENG<br />

ART BY<br />

ANTALIQUE TRAN<br />

flaws, causing it to lose some speed and energy<br />

efficiency, but researchers have begun<br />

to solve some of the issues.<br />

In the past decade, researchers discovered<br />

that AOS in Rare Earth-Transition<br />

Metals (RE-TM) alloys can be performed<br />

in a single pulse process driven by heat.<br />

AOS mechanisms in rare earth transition<br />

metal alloys, as well as their existence in<br />

ferromagnetic thin films and multilayers,<br />

were shown. Additionally, AOS has been<br />

connected to two promising spintronics<br />

research areas, namely racetrack memory<br />

and next-generation magnetic random-access<br />

memory. Racetrack stores magnetic<br />

signals in regions of nanowires that are oppositely<br />

oriented, called racetracks. Electric<br />

pulses are then applied to the nanowires,<br />

which create so-called domain walls<br />

between them. The magnetic orientation<br />

of each region is then used to store bits of<br />

data. If AOS is integrated successfully in<br />

racetrack memory, it has the potential to<br />

store a high volume of information compared<br />

to existing storage devices such as<br />

flash drives. However, even though AOS<br />

was found to be possible in these methods,<br />

it requires many pulses to switch memory<br />

states, which bars this type of AOS from<br />

being used in fast spintronic devices. Only<br />

a thermal single-pulse AOS mechanism<br />

can be successfully integrated into spintronic<br />

devices.<br />

Searching for new AOS materials<br />

In recent work done by a team at Eindhoven<br />

University of Technology, it was<br />

found that the thermal single-pulse AOS<br />

mechanism needed for spintronic integration<br />

existed in multilayers made of a platinum/cobalt/gadolinium<br />

(Pt/Co/Gd) synthetic-ferrimagnetic<br />

stack, stacks of alloys<br />

that display weak permanent magnetism.<br />

www.yalescientific.org


www.yalescientific.org<br />

IMAGE COURTESY OF V3 NEWSDESK<br />

With continued application of AOS to spintronic<br />

memory devices, methods for memory storage<br />

are becoming more efficient.<br />

A member of the team, professor M. L. M.<br />

Lalieu, has led research demonstrating that<br />

this Pt/Co/Gd stack is ideal for integrating<br />

AOS with spintronics and racetrack memory.<br />

In this study, Lalieu shows that clear<br />

single-pulse AOS in Pt/Co/Gd racetracks<br />

is possible, suggesting that the domain<br />

walls (which separate magnetic domains)<br />

of the optically written domains are chiral<br />

Neél walls, meaning they can be moved<br />

along the racetrack with the Spin Hall Effect<br />

(SHE), a means of transport that relies<br />

on the accumulation of homogeneity<br />

in spin direction. Additionally, the SHE<br />

efficiency in this Pt/Co/Gd racetrack and<br />

domain wall velocities are both predicted<br />

to be high. Domain wall velocity can be<br />

used to predict speed of memory devices,<br />

making its value integral to gaining insight<br />

regarding the mechanism of AOS in spintronics.<br />

In order to affirm that single-pulse AOS<br />

can be found in Pt/Co/Gd wires, tantalum/<br />

platinum/cobalt/gadolinium/platinum<br />

stacks were deposited on silicon substrates<br />

coated with silicon dioxide (SiO2) at room<br />

temperature and formed into wires. Each<br />

wire had a structure called a Hall Cross.<br />

The magnetization in a Hall Cross is measured<br />

in order to investigate AOS in these<br />

wires. The Anomalous Hall Effect (AHE),<br />

a phenomenon that occurs in ferromagnetic<br />

solids that measures magnetization, was<br />

used in order to do this. To identify single<br />

pulses in the AHE measurement, the team<br />

utilized slow, repeated laser-pulses. It was<br />

shown that the magnetization in the Hall<br />

Cross region oscillates between positive<br />

and negative states of spin. When measurements<br />

with AOS were repeated for a longer<br />

time, a one-hundred percent success rate of<br />

the AOS was shown, which means that single-pulse<br />

AOS of magnetization is, indeed,<br />

present in the Pt/Co/Gd wires.<br />

It was expected that the domain walls in<br />

the Pt/Co/Gd were chiral Neél walls, which<br />

means that the two domain walls were<br />

symmetrical with respect to spin organization.<br />

These domain walls could move<br />

through the wire by means of electrical<br />

current through SHE and the accumulation<br />

of a common spin. The direction of motion<br />

experienced by these domain walls could<br />

be determined by the sign of magnetization<br />

and chirality of domain walls. In the top<br />

layer of the wire, the Pt layer, this motion<br />

was reported to go against the direction of<br />

electron flow.<br />

Spintronics on the fly<br />

On-the-fly data writing, which is data<br />

writing that is changed as the process of<br />

writing data is being carried out, has been<br />

established through a combination of<br />

SHE-driven transport of optically written<br />

domains, or domains that are coded with<br />

optical signals, and single pulse AOS in<br />

racetrack memory. In this type of measurement,<br />

AOS is used for writing a domain in a<br />

Pt/Co/Gd wire with two Hall Crosses while<br />

a current is also flowing through the wire.<br />

Because both the domain walls that have<br />

the written domains share the same chirality,<br />

they move in the same direction, along<br />

the direction of the current, immediately as<br />

they are written. In this, the full domain is<br />

transported through the wire. It is then recorded<br />

using AHE.<br />

SHE efficiency and domain wall chirality<br />

in optically written domains were also<br />

analyzed quantitatively through performing<br />

field-driven SHE-assisted domain wall<br />

velocity measurements. In this method, an<br />

out-of-plane field is applied while a current<br />

is sent through the wire. Depending on the<br />

polarity of the current, domain wall motion<br />

is affected by SHE (either assisted or hindered),<br />

so an increase or decrease in velocity<br />

is seen. The wires used for these measurements<br />

have two Hall Crosses that are<br />

exposed to gallium ion irradiation. When<br />

the measurement is started, the applied<br />

March 2019<br />

applied physics<br />

FOCUS<br />

field causes the domain to expand through<br />

the wire and pass the two Hall Crosses. This<br />

is recorded by a switch in the AHE signal.<br />

It has been demonstrated that thermal single-pulse<br />

AOS- and SHE-induced domain<br />

wall motion can be combined in a racetrack<br />

to utilize the chiral Neél nature of domain<br />

walls for efficient and smooth motion of optically<br />

written domains. Because of this, the<br />

Pt/Co/Gd racetrack is an ideal mechanism<br />

for integrating AOS into spintronics. The<br />

integration of AOS with racetrack memory<br />

could potentially pave the way for integrated<br />

photonic memory devices.<br />

IMAGE COURTESY OF CNRS NEWS<br />

Depiction of electron spin, one of the foundational<br />

principles of spintronics.<br />

This discovery holds much power in the<br />

world of spintronics, as it seems that a potential<br />

mechanism for creating efficient<br />

photonic memory devices is finally plausible.<br />

However, many professionals in the<br />

field are still skeptical about the application<br />

of AOS to widely accessible memory devices.<br />

“I don’t think any of these ideas are reality<br />

yet,” stated a Yale electrical engineering<br />

professor who asked not to be named.<br />

“They have remained in the laboratory or<br />

on paper for now.” However, if perfected,<br />

AOS could alter the field of data writing<br />

and memory, offering society more efficient<br />

memory devices.<br />

Researchers use electron<br />

science to write data in<br />

photonic memory<br />

Yale Scientific Magazine<br />

31


FOCUS<br />

medicine<br />

Take a Deep Breath<br />

of Fresh mRNA<br />

||| by Britt Bistis ||| art by Ellie Gabriel |||<br />

converting cells into drug synthesizers<br />

A recently published study from the Koch<br />

Institute for Integrative Cancer Research at<br />

MIT gets glowing results, literally. Using a<br />

messenger RNA (mRNA) transcript that<br />

encodes for the bioluminescent, light-emitting<br />

protein, called luciferase, MIT researchers<br />

were able to track the expression of the<br />

aerosolized polymer-bound mRNA that had<br />

been delivered to mice epithelial cells via a<br />

nebulizer. The findings hold hope for more<br />

effective treatments of lung diseases.<br />

Tackling drug delivery<br />

Many believe that RNA drugs will be the<br />

next major achievement in drug development<br />

technology. These types of drugs either<br />

target RNA or use RNA directly as a therapy.<br />

Other types of commonly used drugs are<br />

small chemical treatments and protein drugs.<br />

Chemical drugs are generally non-specific<br />

and diffuse throughout the body. They are often<br />

not naturally degraded and can accrue to<br />

toxic levels–sometimes producing considerable<br />

side effects. The development of protein<br />

drugs addressed many of these limitations.<br />

Protein drugs are well-known, well-established,<br />

and well-funded, with the 2016 global<br />

market for protein drugs estimated at 172.5<br />

billion USD. However, protein drugs also<br />

have drawbacks—the largest arguably being<br />

that proteins are sometimes immunogenic,<br />

meaning that they are prone to induce an immune<br />

response from the body.<br />

However, DNA has received recent attention<br />

as a potential therapy. An inhalable<br />

lipid-DNA complex that contains the gene<br />

Cystic Fibrosis Transmembrane Conductance<br />

Regulator (CFTR), which can cause cystic<br />

The development of protein drugs addressed<br />

many of these limitations. Protein<br />

drugs are well-known, well-established,<br />

and well-funded, with the 2016<br />

global market for protein drugs estimated<br />

at 172.5 billion USD. However, protein<br />

drugs also have drawbacks–the largest<br />

arguably being that proteins are sometimes<br />

immunogenic, meaning that they<br />

are prone to induce an immune response<br />

from the body.<br />

fibrosis when mutated, is currently going<br />

through clinical trials. By delivering the correct<br />

copy of the CFTR gene, researchers had<br />

noted some improvement in patients' conditions.<br />

While researchers are working to op-<br />

32 Yale Scientific Magazine March 2019


medicine<br />

FOCUS<br />

timize the delivery system and enhance the<br />

delivery system of the DNA treatment, using<br />

DNA poses specific challenges. "One issue<br />

with DNA delivery is that a lot of the DNA<br />

vectors, or molecules used to carry DNA, are<br />

viral," reports Padmini Pillai, a current postdoctoral<br />

fellow doing immunoengineering research<br />

at MIT after receiving her PhD in immunobiology<br />

from Yale. “This is less favorable<br />

since it can induce an immune response. Secondly,<br />

DNA needs to enter the nucleus, which<br />

THE NEW DESIGN, NAMED<br />

THE S-HINGE, MAKES<br />

MATERIALS BETTER ABLE<br />

TO OVERCOME THE STRESS<br />

FROM APPLIED FORCES.<br />

is a big ask," Pillai said. These challenges have<br />

led MIT researchers and others to pioneer the<br />

investigation of using RNA instead of DNA in<br />

aerosolized treatments.<br />

The promise of RNA drugs<br />

Despite a deep understanding of how RNA<br />

can regulate gene expression, researchers<br />

discounted RNA as a viable therapeutic primarily<br />

due to how readily it is degraded. The<br />

experiments conducted in the MIT study employ<br />

the natural protein-building machinery<br />

of the cell. By introducing messenger RNA<br />

into the cell, the cell will use its own synthetic<br />

pathways to translate this mRNA into<br />

a functioning protein. Previous studies have<br />

examined the possibility of using nebulized<br />

nucleic acids to treat diseases and disorders in<br />

the lungs, while others have investigated the<br />

use of systemic delivery of mRNA to combat<br />

tumors. Both lines of study have yielded positive<br />

results. Injections of mRNA packaged<br />

in lipid nanoparticles, fat blobs designed to<br />

protect RNA from degradation, have shown<br />

potential in treating cancer in a mouse model<br />

of melanoma. An mRNA-based vaccine that<br />

would target tumor cells is currently in clinical<br />

trial.<br />

The mRNA transcript does not require delivery<br />

inside a viral vector like DNA. Furthermore,<br />

mRNA is more easily transported and<br />

only temporarily active as it is quickly degraded<br />

after it has been translated into protein, enabling<br />

researchers to control mRNA expression<br />

with precision through readministration.<br />

Additionally, mRNA only has to enter the cell<br />

membrane, rather than both the cell membrane<br />

and the nucleus. One limitation of most<br />

current forms of RNA delivery is that they are<br />

non-specific. For example, even if only certain<br />

cell-types in the lung are diseased or cancerous,<br />

an injection of mRNA transcript, and by<br />

extension the protein that it will encode, will<br />

result in mRNA expression systemically.<br />

Breathe easy<br />

The research team investigated two questions:<br />

Could aerosolized mRNA be administered<br />

effectively through a nebulizer, and<br />

what type of nanoparticle would better facilitate<br />

mRNA delivery? The inhalation of<br />

aerosolized nanoparticles is a noninvasive<br />

treatment that has enabled researchers to target<br />

the epithelial cells in the lungs with precision.<br />

Systemic drugs, which are injected or<br />

absorbed into the bloodstream, are expressed<br />

throughout the entire body. "The benefit of<br />

the nebulized delivery system of treatment<br />

is that it facilitates targeted delivery that is<br />

isolated to the lung. Further, the nebulizer is<br />

completely noninvasive; no needles or sharps<br />

[sharp objects] are needed," Pillai said. This<br />

technique initially showed that RNA was able<br />

to enter one-quarter of the target epithelial<br />

cells in mice, evenly distributed throughout<br />

all chambers of the lungs.<br />

The MIT team further engineered a<br />

polymer that more efficiently delivered<br />

nebulized mRNA to the epithelial cells of<br />

the lung. To deliver mRNA via inhalation<br />

through a nebulizer, researchers needed to<br />

first bind the mRNA to nanoparticles that<br />

would help facilitate delivery and protect<br />

the mRNA from degradation before they<br />

enter cells. The polymer the MIT research<br />

team used as their nanoparticle is hPbAE,<br />

which is positively charged, biodegradable,<br />

hyperbranched polymer. The positive<br />

charge on the polymer facilitates stronger<br />

binding between the nanoparticle and negatively<br />

charged mRNA. Additionally, the<br />

positively charged nanoparticle will more<br />

easily penetrate the negatively charged cell<br />

membrane. Studies cited by the MIT team<br />

indicate that a hyperbranched polymer is<br />

better suited to aerosolization than a linear<br />

polymer. Pillai explains that the branched<br />

polymers terminated with polar groups are<br />

more efficient due to their increased solubility.<br />

The increased solubility results in a<br />

nanoparticle that can be easily encased by<br />

water molecules and therefore nebulized<br />

into water breathable gaseous water particles<br />

more easily. Further, the nanoparticle<br />

needs to be biodegradable, as previous studies<br />

have indicated that non-biodegradable<br />

polymers can accumulate in toxic levels.<br />

An RNA future<br />

“While previous studies have used another<br />

biomaterial, PEI, to deliver nucleic acids, it is<br />

not biodegradable, and its accumulation in the<br />

body can lead to side effects,” Pillai said. Pillai<br />

is optimistic of the aerosolized-mRNA delivery<br />

method due to its ability to facilitate targeted<br />

delivery of mRNA to a specific cell type,<br />

the lung epithelial cells. She further projects<br />

that the aerosolized delivery of mRNA could<br />

have a variety of therapeutic applications, not<br />

only in cystic fibrosis, but also to a wide range<br />

of diseases affecting the epithelium of the<br />

lungs, perhaps even viral infections. The field<br />

of RNA research is developing therapeutics<br />

for a broad range of medical conditions, including<br />

cancer. Whatever the condition, this<br />

new potential treatment, breaking through a<br />

sea of criticism and doubt, is sure to provide a<br />

breath of fresh RNA.<br />

March 2019<br />

Yale Scientific Magazine<br />

33


T R A C I N G A N C I E N T<br />

M O T I O N I N R O C K S<br />

Pangea is Earth’s most well-known and recent theory rests on geological evidence of a “ring of<br />

supercontinent. It began to break apart only fire” surrounding the Pacific, where plates in the<br />

around two hundred million years ago, but the deep Earth come together and drop downwards<br />

history of Earth’s continental movement starts like a carpet being pulled into a slit in the ground.<br />

many more hundreds of millions of years ago. As a result, continents, like the furniture on the<br />

David Evans, director of the Yale Paleomagnetic carpet, collide and can continue to move neither<br />

Laboratory, professor of geology and geophysics, forwards nor backwards.<br />

and head of Berkeley College, studies this ancient More recently, Evans has turned his attention<br />

movement. Recent developments in his research to Rodinia, a supercontinent that existed eight<br />

have uncovered a new hypothesis.<br />

hundred to nine hundred million years ago. His<br />

Unlike Pangea, whose geological record of lab is currently investigating a relatively novel<br />

breakup is still intact in the ocean floor, the older hypothesis of this supercontinent’s structure,<br />

supercontinents that Evans studies require a refuting the idea that California was once<br />

different arsenal of methods, as the oceanic records connected directly to Australia or South China<br />

of their motion have since descended back into as many other experts have suggested, and<br />

Earth’s interior. His lab uses paleomagnetism, in looking for other candidates. “People started<br />

which the magnetic signatures of certain minerals out looking at the big intact pieces of the Earth’s<br />

in ancient rock are analyzed to draw new insights. crust—Australia is one of those big pieces…For<br />

By associating the orientations of their magnetic all the pieces that have been suggested, we’ve<br />

fields with their ages, Evans can hypothesize the been able to refute them through a combination<br />

path of their movement.<br />

of magnetic studies or comparisons of geology,”<br />

Studying continental drift is complicated by the Evans said. Researchers realized that relying on<br />

fact that bodies of land tend to become deformed these “big pieces” might not work in places like<br />

as they move. Evans compares the gradual wear Central Asia, where its relatively recent collision<br />

and tear of continents to having multiple fenderbenders<br />

on a bumper car and trying to figure<br />

out what it looked like originally. “Or, using a<br />

puzzle piece analogy, someone snipping off all the<br />

interlocking bits and somebody else sanding away<br />

a bit of the picture of each piece. Of course, many<br />

pieces also get lost and you’re trying to figure this<br />

out without a picture on a box,” Evans said.<br />

Despite these obstacles, deducing a complete<br />

history of continental movement is important<br />

for finding patterns and predicting future<br />

supercontinents. There are three models for the<br />

assembly of new supercontinents: introversion,<br />

extroversion, and orthoversion. Introversion<br />

COU<br />

NTER<br />

PO<br />

BY<br />

INT<br />

GRACE<br />

CHEN<br />

hypothesizes that new supercontinents form in the<br />

same location as previous ones; a supercontinent<br />

breaks up, temporarily opens up an ocean, and<br />

drifts back together in the same place, perhaps<br />

in a different orientation. The extroversion<br />

model predicts exactly the opposite, that broken<br />

supercontinents rejoin on the opposite side of<br />

the Earth. The last theory of orthoversion is an<br />

intermediate theory which Evans’s lab coined<br />

several years ago. Orthoversion states that<br />

continents are instead confined to no more than<br />

ninety degrees from their point of origin. This<br />

with India has broken up the land into much<br />

smaller pieces. “What we’re recognizing now is<br />

there was a big piece there, but you can’t recognize<br />

it anymore,” Evans added.<br />

From analysis of field samples collected from<br />

the area, Evans’s data showed that part of China<br />

could not have been where people initially<br />

thought it was, on the side of Rodinia. Instead,<br />

his data directs it to a spot right in between<br />

California and Australia. “One of the things that<br />

really impressed me about the possibilities now is<br />

that we might need to look at some of these areas<br />

that have been chopped up by younger events<br />

and consider those as maybe once-larger intact<br />

blocks,” Evans said.<br />

“The ultimate goal is to finish the entire puzzle,<br />

which is actually a series of puzzles strung<br />

together across the vast reaches of time,” Evans<br />

started. Moving forward, Evans hopes to fill in<br />

another piece of the puzzle by leading his lab<br />

to Morocco, which used to be part of the West<br />

African Craton. “When you can play an entire<br />

movie back and forth of seeing three or four<br />

supercontinents assemble and break up and<br />

reassemble again, that is when we can start to find<br />

patterns,” Evans added.<br />

34 Yale Scientific Magazine March 2019 www.yalescientific.org


CURIOUS<br />

HEAD MOUNTED<br />

MICROSCOPES<br />

BY BRETT JENNINGS<br />

To study an animal’s brain in real-time as it<br />

navigates its surroundings, researchers typically<br />

implant electrodes into the animal’s brain. These<br />

electrodes track electrical changes in neurons,<br />

detecting when the neurons are activated. However,<br />

this method is limited in how many neurons can be<br />

analyzed simultaneously. “When you are looking at<br />

activity with electrodes, you are unsure if it is coming<br />

from one neuron or another area,” said Catherine<br />

Dulac, professor of molecular and cellular biology<br />

and researcher at Harvard University.<br />

To procure a broader view of the brain, Dulac<br />

and her team have begun to used microscopes<br />

mounted on top of the heads of mice. “Typically,<br />

a microscope is something on your desk and you<br />

put a slide under the lens to look at tissue or fats,”<br />

Dulac said. “[But] this microscope is a miniature<br />

version–light enough to sit on the head of a<br />

mouse.” Small and portable, these miniaturized<br />

versions of fluorescent microscopes can sit<br />

on the head of mice, collecting live data while<br />

the mice perform various activities. These<br />

microscopes are connected to the neurons in the<br />

brain through an endoscopic tube, or a camera<br />

that peers inside an organism. Instead of tracking<br />

the change in electrical charge between neurons,<br />

researchers can now view the fluorescence of<br />

calcium binding in neurons, for example, to<br />

show signaling between neurons in the brain. To<br />

pass on a message, one neuron releases calcium<br />

into the synapse to activate signal receptors<br />

on the next neuron. The movement of calcium<br />

produces a change in fluorescent intensity that<br />

can be observed by researchers. This method<br />

allows researchers to visualize the activity of<br />

hundreds of thousands of neurons all at once,<br />

rather than focusing on just a few.<br />

www.yalescientific.org<br />

Dulac’s team has begun to use these portable<br />

microscopes to investigate the brain processes<br />

underlying the perception of social cues. The<br />

portability of these microscopes enables the research<br />

team to track the activity of the medial amygdala–<br />

the brain region that responds to social stimuli–of a<br />

mouse as it interacts with other mice. The scientists<br />

can then see how a mouse’s brain interprets the cues<br />

which determine its response. For example, the<br />

researchers compared the brain activity of a male<br />

mouse attempting to mate with a female mouse<br />

with an onlooking male mouse who decided to<br />

combat the mating mouse. These microscopes<br />

can also collect data to make predictions for other<br />

neurons. “You can take half of the neurons that<br />

you have recorded and model the form of activity<br />

mathematically to predict the activity in the other<br />

half,” Dulac said. “To some extent, you can simply<br />

look at neural activity and make a guess of what the<br />

animal is thinking and doing.”<br />

In the future, researchers hope to apply these<br />

microscopes to image multiple areas at once.<br />

This function would give researchers the ability<br />

to see the dialogue between multiple areas of the<br />

brain, investigating how brain signals travel and<br />

pass between different brain regions.<br />

Additionally, researchers are looking into a<br />

number of improvements, such as sharpening the<br />

spatial resolution of the microscope’s images to<br />

detect more fluorescent colors. Dulac’s team is also<br />

working on a smaller, potentially wireless version of<br />

the device as well as a multifunctional head-mounted<br />

microscope with an electrical probe to stimulate<br />

or inhibit neural activity. “For a neuroscientist, it’s<br />

a dream come true to have a window to what the<br />

brain does when an animal is looking around and<br />

making decisions,” Dulac added.<br />

March 2019<br />

METHODS<br />

Yale Scientific Magazine<br />

35


UNDERGRADUATE PROFILE<br />

NICOLE ESKOW (PC ’19)<br />

LOOKING FORWARD IN CANCER BIOLOGY<br />

BY TONY LECHE<br />

PHOTOGRAPHY BY MEHANA DAFTARY<br />

Nicole Eskow (PC ’19) has always been asking questions.<br />

“After losing both of my grandmothers to cancer at a young<br />

age, I began to inquire about cancers and where these diseases<br />

originate,” Eskow said. Determined to find answers,<br />

she started to think critically about diseases such as HIV<br />

and cardiovascular disease. Eskow was not satisfied with the<br />

information she found on the internet. At fifteen, she began<br />

conducting research on leukemia treatments in her high<br />

school’s cell biology laboratory. Her passion for research has<br />

only blossomed since. Soon after, Eskow started shadowing a<br />

pediatric hematologist-oncologist at Hackensack University<br />

Medical Center. After that summer, Eskow knew she wanted<br />

to pursue both research and clinical medicine.<br />

Now a Molecular, Cellular, and Developmental Biology (MCDB)<br />

major, Eskow spends much of her time working in the Krause Lab,<br />

which is dedicated to researching the mechanism that regulates<br />

hematopoiesis, the formation of new blood cells, and how that<br />

process can go awry. Eskow has focused her time on understanding<br />

acute megakaryoblastic leukemia, a type of pediatric cancer<br />

that primarily affects newborns. This kind of cancer disrupts the<br />

development of megakaryocytes, cells that produce platelets,<br />

which are important in blood clotting. “Interestingly, previous research<br />

has suggested that this cancer begins to develop in the fetus,<br />

before a baby is born,” Eskow said. Her project investigates the<br />

role of a protein known to be mutated in many patients with acute<br />

megakaryoblastic leukemia. For her accomplishments and dedication<br />

to research, Eskow was selected for the Rosenfeld Science<br />

Scholar Program, which supports promising students conducting<br />

summer research under the supervision of a Yale faculty member.<br />

Eskow has also played an integral role in the Yale Undergraduate<br />

Research Association (YURA) since joining her freshman<br />

year. As the former co-president of YURA, she has helped give<br />

students easier access to research opportunities on campus.<br />

During her sophomore year, Eskow compiled a list of all the research<br />

labs at Yale, featuring descriptions of what the lab studied<br />

as well as the contact information and general interests of<br />

the head researcher. This program was released to the Yale community<br />

in 2016 and is now known as the YURA Research Database<br />

(RDB). “I find it incredible whenever I hear that first-years<br />

are finding research labs through the YURA RDB,” Eskow said.<br />

After graduating, Eskow hopes to pursue an MD-PhD to<br />

dedicate her time to both research and patient care. “While<br />

I haven’t completely decided what area of medicine I’d like to<br />

pursue, I know that I’m very passionate about oncology and<br />

cancer research,” Eskow said. “I’ve really enjoyed researching<br />

“I KNOW THAT 20 OR 30 YEARS<br />

DOWN THE ROAD I’M NOT GOING TO<br />

REMEMBER THE EXACT NUMBER OF<br />

PAPERS I PUBLISHED OR MY GPA.”<br />

hematologic malignancies like leukemia and lymphomas, and<br />

it’s definitely an area I can see myself pursuing in the future.”<br />

Looking back, Eskow has gained a better understanding of the<br />

relationship between her research and her life. “I know that 20<br />

or 30 years down the road I’m not going to remember the exact<br />

number of papers I published or my GPA,” Eskow said. “Over the<br />

past four years, I’ve learned that investing time in the people who<br />

love and support you is equally as important as building a career. I<br />

couldn’t imagine my life without the friends and family members<br />

who have stood by me and helped me get to where I am today.”<br />

36 Yale Scientific Magazine March 2019 www.yalescientific.org


ALUMNI PROFILE<br />

CHRISTINA AGAPAKIS (YC ’06)<br />

Being different has always been the norm for Christina<br />

Agapakis (YC ’06). After receiving her PhD in biological and<br />

biomedical sciences from Harvard University in 2011, she became<br />

the creative director in 2015 for Ginkgo Bioworks, an<br />

organism design company looking to apply biological methods<br />

to engineering challenges. As a member of the 2012 Forbes 30<br />

Under 30 list, Agapakis managed to combine her passions in<br />

art and biology to bring biotechnology to the greater public.<br />

Growing up, Agapakis has always loved art. “I was interested<br />

in art in high school and really enjoyed art classes and art making,<br />

especially drawing and making jewelry. I love the feeling<br />

of being able to translate something from inside my head into<br />

something real,” Agapakis said. However, she originally thought<br />

that she could not meld her interests in art and science together.<br />

“I used to think that liking art and science were sort of incompatible<br />

and totally separate. It was not until later in my life that I<br />

realized the power of the combination,” Agapakis said.<br />

At home, her parents, who both studied in STEM fields, have<br />

constantly supported her love of science, encouraging her to join<br />

programs like Science Olympiad and Odyssey of the Mind. In fact,<br />

Science Olympiad cemented her interest in biology. “I remember<br />

an event called ‘Science of Fitness’ where I learned about the Krebs<br />

cycle, and the biochemistry of it just blew my mind,” she said.<br />

However, an artist at heart, Agapakis craved a way to combine<br />

art and biology, but she did not believe that art could contribute<br />

to her education and career as a scientist until her senior year at<br />

Yale, when she took Intro to Architecture and Sculptures as Object.<br />

“These two courses really helped me rewire my brain and allowed<br />

me to approach problems differently,” Agapakis explained.<br />

While pursuing her PhD, she met Pam Silver, an RNA expert<br />

pioneering the field of synthetic biology, who became Agapakis’s<br />

mentor. During this time, Agapakis finally realized how art could<br />

be part of her scientific practice: artists ask questions, solve problems,<br />

and find different ways to see the world–missions also essential<br />

to scientists. “Art [has] helped me be a better scientist and totally<br />

shifted my perspective on what a scientist should be,” she said.<br />

Many of the questions she hoped to answer in synthetic biology<br />

required her to think beyond her laboratory work–towards<br />

more general scientific principles. Agapakis began blogging at<br />

Harvard, exploring the impact and implications of biotechnology.<br />

Her pieces range from discussing the social and political<br />

“ecologies” of producing cheese to the irony of design evolution.<br />

“I believe that the intersection of society and biotechnology is<br />

so powerful. By making scientists more engaged and aware of<br />

social issues, it makes us better scientists,” Agapakis explained.<br />

BRIDGING BIOLOGY AND ART<br />

BY TIFFANY LIAO<br />

IMAGE COURTESY OF ERIN CHIA<br />

More importantly, blogging enabled her to finally combine her<br />

interests in biology and art. “By writing, I was able to put out<br />

my thoughts, even if they were unfinished. Then, people would<br />

reach out, and that is how I started connecting with artists and<br />

designers working on synthetic biology,” she said.<br />

After finishing her postdoctoral studies at UCLA, Agapakis<br />

joined her interests in art, biology, and writing together at Ginkgo<br />

Bioworks, which shares her vision to make the world of biotechnology<br />

more approachable. She now leads a Ginkgo venture nicknamed<br />

Project Cretaceous, which works with multidisciplinary<br />

artist Daisy Ginsberg and scent researcher Sissel Tolaas to bring<br />

back the smell of extinct flowers. The resulting artwork will be presented<br />

at numerous galleries and exhibitions as an interactive piece<br />

in which individuals can smell the scents of ancient flowers such as<br />

the Hawaiian mountain hibiscus (Hibiscadelphus wilderianus) and<br />

the Falls of the Ohio scurfpea (Orbexilum stipulatum). “This project<br />

is the result after years of doubt, wondering how in the world<br />

would I combine these two interests of mine,” Agapakis explained.<br />

“The most important thing is to keep pushing and changing<br />

what you think of when you think ‘scientist,’” she said. “We<br />

need more political, artist, teacher, feminist scientists–individuals<br />

who have multiple interests and are willing to combine<br />

them to better the world.”<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

37


SEX ON THE KITCHEN TABLE<br />

THE ROMANCE BETWEEN PLANTS AND YOUR FOOD<br />

B Y K H U E T R A N<br />

Vulgar, dirty, and exposed–these descriptors may come to mind upon reading the title of<br />

Norman Ellstrand’s new book. However, in Sex on the Kitchen Table: The Romance of Plants<br />

and Your Food, Ellstrand actually illustrates a much more demure topic: the importance of<br />

understanding the relationship we have with the food we consume.<br />

Each chapter highlights a specific crop, seamlessly melding its history, anatomy, and special<br />

reproduction system. Using humorous metaphors and a distinctly easygoing voice, Ellstrand<br />

discusses the reproduction, economics, and politics surrounding the future of each<br />

plant, culminating in a delectable homemade recipe. “I can’t pick just one to be called my<br />

favorite,” Ellstrand said. Ellstrand gives human-like sexuality and mechanisms to each plant:<br />

the bisexual tomato plant, for example, self-pollinates using its flowers. Though playful, each<br />

recipe demonstrates our exploitation of plant sexuality.<br />

Ellstrand explores the complex, twisted history of–as well as humanity’s influence behind–each<br />

plant’s reproduction system, revealing that the foods we eat evolved from very<br />

different looking plants thousands of years ago. Currently a professor at the University of<br />

California, Riverside, Ellstrand teaches a class called “California Cornucopia,” where he<br />

utilizes fruits mentioned in his book, such as the sterile banana, to teach plant biology.<br />

IMAGE COURTESY OF FLICKR<br />

scie<br />

in<br />

spot<br />

“You can’t understand why sex is important without examining something without sex,”<br />

Ellstrand said.<br />

Additionally, Ellstrand illuminates the potential problems that may arise in these vegetable<br />

sexual reproduction systems. For instance, when a plant possesses only “female” or “male”<br />

reproductive parts, it runs into self-incompatibility issues and thus, cannot reproduce.<br />

While Ellstrand wrote Sex on the Kitchen Table, his second book, to be humorous and casual<br />

pleasure reading, his first book, Dangerous Liaisons, was a scholarly science book directed<br />

at policy makers, selling less than nine hundred copies in fifteen years. Although he feared<br />

that his colleagues would criticize Sex on the Kitchen Table for being unscientific, the general<br />

public loved the simple, beautiful analogies he employed to describe biological functions<br />

of plants in this book. For example, Ellstrand said, “Tomatoes are self-fertile individuals<br />

[that] have the potential to serve as both mother and father to one or more of their kids.”<br />

This book will not only change your perspective on tomatoes, bananas, avocados, beets,<br />

and squash, but it will also morph your view of vegetables into the erotic plants that they<br />

truly are, opening your eyes to observe the nature around you as perhaps a bit spicier.<br />

38 Yale Scientific Magazine March 2019 www.yalescientific.org


nce<br />

the<br />

light<br />

IMAGE COURTESY OF FLICKR<br />

Ian Cheney’s (YC ’02) 2018 film The Most Unknown takes a different approach than most science documentaries.<br />

Although his film explores how people study organisms, space, quantum physics, geobiology,<br />

and the human mind, Cheney, a graduate of Yale College and the Yale School of Forestry and Environmental<br />

Studies, ultimately tells a story about the field of science itself–one about what it means to think, question,<br />

experiment, and live as a scientist.<br />

The Most Unknown’s opening credits acknowledge that the documentary itself is an experiment. The film<br />

takes viewers around the globe, from ocean floors to mountain peaks, to meet nine accomplished scientists.<br />

As the quirky stories of those researchers intertwine and each travels to learn about the work of another,<br />

Cheney showcases the excruciatingly detailed and thoughtful processes behind scientific discoveries. The<br />

film highlights the passion that these scientists throw into their work and the joy that comes with contributing<br />

even a bit more to both the “answered” and “yet-to-be-understood” piles within their respective fields.<br />

A microbiologist obsessed with slime visits a physicist who enthusiastically explains how his “simple” dark<br />

matter-detecting machine works. Sardonically, the physicist refuses to speak on the whereabouts of dark<br />

matter because he believes that scientists should not speculate without substantial reasoning and experimentation.<br />

Later, we travel to the peaks of Hawaii with an astrophysicist and an astrobiologist: macro- and<br />

micro-scales of science align when the astrobiologist, who spends most of his time looking through microscopes,<br />

saw how similar the speckled night sky through a telescope looks to the cosmos-like arrangement of<br />

archaea under a microscope. Finally, we meet a cognitive psychologist, Yale professor and Silliman Head of<br />

College Laurie Santos, on an island near Puerto Rico inhabited by monkeys. As some of the last images taken<br />

before Hurricane Maria hit just three weeks later, the beautiful footage from the island resonates especially<br />

deeply with audiences.<br />

Regardless of the various scientific fields and special environments in which they work, these nine scientists<br />

all have stories that beautifully stitch together to shed light on universal unknowns. Santos, whose work<br />

has centered on applying scientific answers in ways that directly help people, hypothesized that most scientists<br />

do not have what psychologists refer to as a need for certainty. Scientists are comfortable admitting the<br />

extents of their knowledge. Thus, The Most Unknown illuminates the human face of science–a particularly<br />

important feat given how the field often seems so distant, sterile, and daunting. “How I think about science<br />

in just the last few years has expanded–not just answering these questions in ivory tower pursuits but finding<br />

ways to bring the science back to people,” Santos said.<br />

The Most Unknown humbly reminds us that even scientists lack all of the answers. After all, questions are<br />

what perpetuate the grand scientific experiment of uncovering the unknowns.<br />

CHENEY’S REVOLUTIONARY SCIENCE DOCUMENTARY<br />

B Y K A T I E S H L I C K<br />

THE MOST UNKNOWN<br />

www.yalescientific.org<br />

March 2019<br />

Yale Scientific Magazine<br />

39


Interested in writing for<br />

Contact us at<br />

yalescientific@yale.edu<br />

AD_Yale_SC_ENG_half Winter <strong>Issue</strong>_2019.qxp_8 2/11/19 1:05 PM Page 1<br />

Kudos to the YSEA Award Winners!<br />

Your achievements inspire all of us in the Yale STEM community.<br />

Advancement of Basic and Applied<br />

Science Award:<br />

Daniel Prober, Ph.D.<br />

Professor of Applied Physics, Physics and<br />

Electrical Engineering<br />

Experimental solid state physics and<br />

superconductivity<br />

Distinguished Service to Industry,<br />

Commerce or Education:<br />

Michael Gold ’60 BS, M.Eng<br />

Founder & President, Gold<br />

Metallurgical Services, LLC<br />

Former Principle Engineer, Babcock &<br />

Wilcox Power Generation<br />

Meritorious Service to Yale:<br />

Brian Scassellati, Ph.D.<br />

Professor of Computer Science, Cognitive<br />

Science & Mechanical Engineering<br />

Director of the NSF Expedition on Socially<br />

Assistive Robotics<br />

Thank you for continuing Yale's rich tradition in STEM.<br />

Awards shall be presented at the YSEA Annual Dinner, April 12, 2019.<br />

For more information visit: ysea.org

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!