23.07.2019 Views

YSM Issue 90.4

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

OCTOBER 2017 VOL. 90 NO. 4 | $6.99


Welcome to Yale!<br />

The Yale Science and Engineering<br />

Association is here for you.<br />

Founded in 1914, the YSEA is one of the oldest university student/alumni<br />

organizations in the world with a focus on STEM.<br />

Whether near or far from New Haven, we help our members realize their<br />

goals and to connect in ways that strengthen the Yale science and<br />

engineering community.<br />

We are excited to be a part of your Yale journey, and we look forward to<br />

supporting you at Yale and beyond!<br />

Join us at: ysea.org


Yale Scientific Magazine<br />

VOL. 90 ISSUE NO. 4<br />

CONTENTS<br />

OCTOBER 2017<br />

NEWS 6<br />

FEATURES 25<br />

ON THE COVER<br />

22<br />

Rsearchers in the Herzon Lab at<br />

Yale have devised a total synthesis<br />

of the antibiotic pleuromutilin,<br />

opening the door to new potential<br />

antibiotics to help bacterial resistance<br />

12<br />

PUTTING THE PATCH<br />

ON RESISTANCE<br />

HOT, DENSE, AND<br />

SPINNING<br />

Just moments after the big<br />

bang, all matter existed in a state<br />

called the quark-gluon plasma.<br />

Yale professor Helen Caines and<br />

her group work with the STAR<br />

collaboration, together aiming<br />

to discover the properties of our<br />

universe this early in its history<br />

15 THE SEARCH<br />

If cancer cells can’t find the highways<br />

of the body, they can’t spread and<br />

become more lethal. A mathematical<br />

model developed by Andre Levchenko<br />

and JinSeok Park of the Yale<br />

Systems Biology Institute provides a<br />

framework to expalin cell migration<br />

behavior that can be implemented<br />

down the line to keep cells searching<br />

longer.<br />

18<br />

IN SEARCH OF LOST<br />

TIME<br />

Yale researchers repair memory<br />

deficits in Alzheimer’s mice using a<br />

drug that targets abnormal protein<br />

interactions in the brain<br />

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

20 NANOPARTICLES<br />

FOR TRANSPLANTS<br />

Yale researchers develop a nanoparticle<br />

delivery system that releases<br />

siRNA capable of protecting ransplanted<br />

organs from rejcetion by the<br />

immune system. the nanoparticles<br />

have the potential to inhibit immune<br />

system’s recognition of transplants<br />

October 2017<br />

Yale Scientific Magazine<br />

3


q a<br />

&<br />

►BY KATHERINE HANDLER<br />

Wheat is an essential part of diets<br />

around the globe. In fact, twenty percent<br />

of the world’s total calorie consumption<br />

is from wheat alone. Thus, scientists are<br />

eager to find out how to produce it faster<br />

and more efficiently, and to do that,<br />

they’re looking back into the past.<br />

Wheat was domesticated ten thousand<br />

years ago in the present-day Middle East,<br />

when humans rapidly modified the crop’s<br />

key traits. Nowadays, we continue to produce<br />

domestic wheat. It differs from wild<br />

wheat in that it has non-shattering spikes,<br />

an adaptation that allows the plant to better<br />

retain its seeds and to be harvested<br />

more easily.<br />

Researchers at Tel Aviv University, led<br />

by Assaf Distelfeld, have been studying<br />

the genetics responsible for non-shattering<br />

spikes. Their work analyses the genome<br />

of wild emmer wheat to better link<br />

►BY SEVERYN KUSHMELIUK<br />

Have you ever noticed yourself<br />

yawning after someone else yawns,<br />

even if you’re not tired? This past<br />

August, researchers at the University<br />

of Nottingham may have figured out<br />

why this phenomenon occurs, discovering<br />

that contagious yawning is<br />

triggered by an area of the brain that<br />

is responsible for motor function: the<br />

left primary motor cortex.<br />

Reflexive yawning is considered<br />

a form of echopraxia, the automatic<br />

imitation of another person’s actions.<br />

While previous studies have<br />

shown that this form of yawning occurs<br />

in humans, chimpanzees, and<br />

dogs, this is one of the first studies<br />

to link contagious yawning and neural<br />

activity.<br />

As part of the study, the researchers<br />

directed four separate groups of<br />

How was wheat domesticated?<br />

IMAGE COURTESY OF PEXELS<br />

►The spike of a wheat plant, with its seeds still<br />

intact.<br />

the grain’s physical traits to the genes responsible<br />

for them. They found two main<br />

genes responsible for shattering spikes in<br />

wild wheat—two genes that are not functional<br />

in domesticated wheat.<br />

“The fact that we find the same mutations<br />

in every domesticated wheat genotype<br />

is amazing because it exemplifies<br />

how strong genetic bottleneck or selection<br />

can be,” Distelfeld said. Human preference<br />

for the non-shattering spike phenotype<br />

was a selective force that drove<br />

the domestication of wheat plant. But<br />

modifications to wheat may not be over,<br />

especially with these new genetic discoveries.<br />

“Now that wheat is in the ‘post-genomic<br />

era,’ many scientists will feel comfortable<br />

working on wheat instead of model<br />

plants, so wheat improvement will be<br />

faster,” Distelfeld said.<br />

Why is yawning contagious?<br />

IMAGE COURTESY OF WIKIMEDIA COMMONS<br />

►Although yawning has long been known to<br />

be contagious, researchers have only recently<br />

discovered the biological basis for why.<br />

people to watch clips of individuals<br />

yawning. Different groups were<br />

instructed to either resist yawning<br />

or let yawning occur naturally, and<br />

participants in booth groups were<br />

hooked up to an electric stimulator—in<br />

true Frankenstein style—<br />

that shocked the brain’s left primary<br />

motor cortex.<br />

The data revealed that attempts to<br />

resist yawning actually increase the<br />

urge to yawn, and that people have<br />

natural tendancy to yawn that is not<br />

affected by instructions. Most importantly,<br />

however, the researchers<br />

found that electrically stimulating<br />

the left primary motor cortex increased<br />

the likelihood of yawning,<br />

revealing that this area of the brain<br />

may play an important role in contagious<br />

yawning.


F R O M T H E E D I T O R<br />

Reaching Out<br />

Millions of Americans traveled this summer to catch a glimpse of our sun turning<br />

black. The Great American Eclipse united the nation in an awe-inspiring display of<br />

nature’s power; at one eclipse viewing, even after organizers ran out of solar glasses,<br />

people simply shared the glasses that were already passed out. They traded eclipse facts<br />

and celebrated the simple joy of witnessing a historic scientific event, one that they<br />

could tell their grandchildren about.<br />

But what if that same excitement carried to all fields of science?<br />

After all, the eclipse is simply the moon passing in front of the sun, and a total eclipse<br />

can be seen somewhere on the Earth every two years or so. How much more amazing<br />

is it when scientists are able to create materials eight hundred million times hotter than<br />

the sun (pg. 12), or when nanoparticles can be used to prevent organ rejection (pg. 20)?<br />

Science is a fundamentally human endeavor, driven by our curiosity and passion to<br />

understand the world. It is why finding diverse, long-dead microbes in ancient rocks<br />

can excite our imagination(pg. 27), and why tiny molecular motors that punch through<br />

diseases in our body can be so surprising (pg. 32). Understanding everything from auditory<br />

hallucinations (pg. 9) to distant galaxy formation (pg. 28) can send tingles down<br />

your spine and set your mind on fire. We are innately drawn to the story of science,<br />

because it is the story of humanity.<br />

Yet we tend to bury these jewels of knowledge beneath a mountain of jargon and<br />

technical language. Research to discover sterile neutrinos (pg. 34) can get bogged<br />

down in dense statistical analyses, while discoveries as elegant as the causes of twisting<br />

flowers (pg. 7) can become lost in abbreviations of genetic markers. The mission of the<br />

Yale Scientific Magazine is to make these beautiful ideas clear and accessible to everyone,<br />

because you shouldn’t need a Ph.D to appreciate the sublime beauty of massive<br />

methane craters in the Arctic sea (pg. 26).<br />

Simultaneously, we seek to train the next generation of science communicators. This<br />

issue, we are especially excited to welcome the writers, artists, photographers, and<br />

designers of the Class of 2021. Already, they have been reporting on diverse topics,<br />

ranging from reclassifying diseases (pg. 25) to biodegradable plastics (pg. 35). We are<br />

eagerly looking forwards to see their continued contributions in the next four years.<br />

As you read this issue, I hope that you will catch our infectious enthusiasm and share<br />

your own wealth of knowledge with the world as well. If we all become better science<br />

communicators, telling the world about the science that excites us, we can truly change<br />

the world.<br />

A B O U T T H E A R T<br />

Chunyang Ding<br />

Editor-in-Chief<br />

What better way to portray the potential of mushrooms in<br />

antibiotics research than to tap into the mushrooms themselves?<br />

The inspiration for this piece came from the unique<br />

shape of the mushrooms described in the cover article — the<br />

Clitopilus passeckerianus. Their pale bodies are fan-like and<br />

upward reaching, furled, and maybe even a little spooky.<br />

While tracing the details of their winding gills was laborious,<br />

it is my hope that the final illustrated product puts these<br />

notable figures of modern antibiotic research on full display.<br />

Editor-in-Chief<br />

Managing Editors<br />

News Editor<br />

Features Editor<br />

Articles Editor<br />

Online Editor<br />

Copy Editors<br />

Special Sections Editor<br />

Yale Scientific<br />

M A G A Z I N E<br />

Established in 1894<br />

OCTOBER 2017 VOL. 90 NO. 4<br />

Production Manager<br />

Layout Editor<br />

Art Editor<br />

Photography Editor<br />

Outreach Designer<br />

Publisher<br />

Operations Manager<br />

Advertising Manager<br />

Subscriptions Manager<br />

Alumni Outreach Coordinator<br />

Synapse President<br />

Synapse Vice President<br />

Outreach Coordinators<br />

Social Media Coordinator<br />

Staff<br />

Yasmin Alamdeen<br />

Sam Berry<br />

Anusha Bishop<br />

Grace Chen<br />

Ashwin Chetty<br />

Serena Cho<br />

Mary Chukwu<br />

Lukas Corey<br />

Keyi Cui<br />

Xander DeVries<br />

Victoria Dombrowik<br />

Ayah Elmansy<br />

Allie Forman<br />

Ivory Fu<br />

Kaija Gahm<br />

Lauren Gatta<br />

Hannah Geller<br />

Vivek Gopalan<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 />

Katie Handler<br />

Jiyoung Kang<br />

Lauren Kim<br />

Theo Kuhn<br />

Severyn Kushmeliuk<br />

Andrew Kuzemczak<br />

Mindy Le<br />

Sunnie Liu<br />

Robert Luo<br />

Tanvi Mehta<br />

Diyu Pearce-Fisher<br />

Jared Peralta<br />

Miriam Ross<br />

Elizabeth Ruddy<br />

Sonia Ruiz<br />

Marcus Sak<br />

Sophia Sanchez-Maes<br />

Isabel Sands<br />

Chunyang Ding<br />

Emma Healy<br />

Sonia Wang<br />

Christine Xu<br />

Diane Rafizadeh<br />

Andrea Ouyang<br />

Kevin Biju<br />

Will Burns<br />

Amy Xiong<br />

Cheryl Mai<br />

Eileen Norris<br />

Richard Hwang<br />

Catherine Yang<br />

Natasha Zaliznyak<br />

Charlie Musoff<br />

Dawn Chen<br />

Kevin Chang<br />

Cheryl Mai<br />

Bryan Ho<br />

Krisstel Gomez<br />

Stephanie Smelyansky<br />

Jessica Trinh<br />

Wai Pan Wong<br />

Archeta Rajagoplaan<br />

Leslie Sim<br />

Anna Sun<br />

Sida Tanag<br />

Vera Villanueva<br />

Eric Wang<br />

Emma Wilson<br />

Alice Wu<br />

Cory Wu<br />

Lisa WU<br />

Jason Yang<br />

Yulan Zhang<br />

Kelly Zhou<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!


NEWS<br />

in brief<br />

Hardware Security “Fingerprints”<br />

By Ayah Elmansy<br />

PHOTOGRAPHY BY XANDER DE VRIES<br />

►DRAM provides each device its<br />

unique “fingerprint,” which can be<br />

used for electronic security.<br />

We all strive to keep our prized possessions<br />

safe. Sometimes those possessions include not<br />

just physical items, but also our digital information.<br />

Inspired to help us keep our data secure, Yale<br />

professor Jakub Szefer is researching security applications<br />

of the tiny differences between devices.<br />

Szefer is an assistant professor of electrical engineering<br />

at the Yale School of Engineering and<br />

Applied Science. Recently, he received the 2017<br />

Faculty Early Career Development Award from<br />

the National Science Foundation, which will provide<br />

funding for his research. Szefer is collaborating<br />

with his research group on this work, which<br />

includes graduate student Wenjie Xiong, as well<br />

as researchers in Germany at Technische Universität<br />

Darmstadt and Ruhr University Bochum.<br />

Szefer’s project focuses on the use of electronic<br />

devices’ unique “fingerprints” to improve computer<br />

security. At the electronic level, every computing<br />

device has distinctive hardware features,<br />

even between identical models. These differences<br />

in the devices’ digital circuits, processors, and<br />

data storage systems are known as dynamic random-access<br />

(DRAM) memory. Small variations<br />

occur when these devices are manufactured,<br />

causing slight deviations in factors such as the<br />

length of the wires of circuits or the capacitors’<br />

thicknesses. These minor deviations accumulate<br />

to give each device its unique “fingerprint,”<br />

which can be used to authenticate and identify<br />

the device.<br />

Szefer wants to explore how these fingerprints<br />

can be applied to the security of our everyday devices<br />

like smartphones, phones, computers, and<br />

embedded devices. “We hope to show that cryptographic<br />

protocols can be designed, which derive<br />

their security from not just difficult mathematical<br />

problems but also from the intrinsic<br />

properties of the hardware,” Szefer said. Through<br />

their continued efforts, Szefer and his collaborators<br />

hope to benefit both the hardware security<br />

community and consumers who use computing<br />

devices every day.<br />

IMAGE CREDIT BY ZUREKS<br />

►The researchers found notothenioids<br />

to be in jeopardy from climate change<br />

and species invasion.<br />

Something’s Fishy<br />

By Namoi D’Arbell Bobadilla<br />

It’s cold in the Antarctic Ocean, but<br />

things are heating up. Yale ecology and<br />

evolutionary biology professor Thomas<br />

Near worked with a research group to study<br />

an ancient Antarctic fish lineage called<br />

notothenioids. These fish are essential<br />

links in Antarctic food webs and are the<br />

basis for several fisheries all over the world.<br />

The researchers used DNA sequences from<br />

89 notothenioid species to reconstruct the<br />

process of how they evolved and to study<br />

their changes in habitat over the years. Their<br />

study, published in June in Nature Ecology<br />

& Evolution, showed that notothenioids are<br />

in danger from climate change and species<br />

invasion.<br />

Historically, notothenioids have survived<br />

natural warming cycles through migration,<br />

such as between the Antarctic continent<br />

and its islands. “Continents tend to produce<br />

biodiversity that emigrates to islands, but<br />

the Antarctic islands are behaving like<br />

a continent,” Near said. In other words,<br />

studies have shown that around Antarctica,<br />

the islands generate a diversity of marine<br />

life. Unfortunately, these island waters are<br />

now warming. This temperature rise allows<br />

fish that usually live in warmer water to<br />

invade and compete with native species<br />

like notothenioids. Notothenioids must<br />

now face both rising temperatures—which<br />

can alarmingly alter Antarctic marine<br />

ecosystems—and increased competition<br />

from invading species.<br />

“This polar ecosystem has probably been<br />

fairly stable for 25 to 30 million years,” Near<br />

said. However, now the climate change<br />

has brought unprecedented instability to<br />

Antarctic ecosystems and has placed its<br />

most dominant lineage, notothenioids, in<br />

jeopardy from habitat change and invasive<br />

species. Near reminds us of our duty to<br />

address such problems: “With our dramatic<br />

utilization of natural resources comes<br />

responsibility for stewardship of the planet<br />

that we’ve perturbed.”<br />

6 Yale Scientific Magazine October 2017 www.yalescientific.org


in brief<br />

NEWS<br />

The Twists and Turns of Flowers<br />

By Ashwin Chetty<br />

Have you ever taken time to enjoy the<br />

beauty of flowers? Professor Vivian Irish and<br />

postdoctoral associate Adam Saffer of Yale’s<br />

Molecular, Cellular and Developmental<br />

Biology department have done so for years,<br />

especially from a scientific perspective.<br />

They study what affects a flower’s shape<br />

and appearance. Recently, they discovered<br />

a molecule that affects the twist of flowers,<br />

which refers to the way flowers turn. In<br />

their Current Biology paper published in<br />

August, they revealed that in a specific type<br />

of flower, Arabidopsis thaliana, a substance<br />

called pectin influences the twisting of plant<br />

cells. Pectin is a common substance in the<br />

kitchen and gives jam its gelatinous quality.<br />

Saffer first looked at a mutation that caused<br />

plant cells to be short and twisted. The<br />

researchers then identified a mutated gene<br />

underlying the helical shape of these cells.<br />

This gene plays a role in the biosynthesis<br />

of a certain kind of pectin called RG-<br />

I. The researchers believe that RG-I may<br />

normally counteract a component of cell<br />

walls that causes cells to twist. However,<br />

when the mutation is present in a cell, less<br />

RG-I is is produced. RG-I inhibits twisting,<br />

so when RG-I levels are low, the unknown<br />

component is free to make cells twist. Thus,<br />

the mutation causes the beautiful helical<br />

shape of plant cells.<br />

The Irish Lab is working to better<br />

understand the role of pectin in providing<br />

cell structure, and Irish is partnering<br />

with researchers at Yale’s department of<br />

mechanical engineering and materials<br />

science to develop models to explain this<br />

left-handed twisting. By identifying a novel<br />

characteristic of pectin, Irish and Saffer<br />

have opened doors for the development of<br />

new biomaterials. While they are excited<br />

for these applications, at the end of the<br />

day, both still like to enjoy the aesthetically<br />

pleasing nature of flowers.<br />

PHOTO BY TANVI MEHTA<br />

►Pectin, the substance that gives jam<br />

its gelatinous quality, can influence the<br />

way that flowers twist.<br />

Raising a child is no easy task—what<br />

would you do if you were put in charge of<br />

raising someone else’s child? In a recent<br />

study, researchers from the Yale Department<br />

of Ecology and Evolutionary Biology<br />

explored whether males of different animal<br />

species would care for offspring that aren’t<br />

their own. The researchers studied how the<br />

energy needed to raise a child could affect<br />

males’ decisions to care for offspring.<br />

Previous theories state that males are most<br />

likely to care for children that are certain to<br />

be their own. Yet in many species, a male<br />

may take care of offspring that were not<br />

conceived by him but rather by a competing<br />

male. Postdoctoral research fellow Gustavo<br />

Requena explained the difference between<br />

his model and those of earlier theories. “In<br />

our study, we used mathematical models<br />

to emulate males’ decisions in different<br />

scenarios and ultimately address the same<br />

question but took into account a more<br />

general biological reality,” he said. This<br />

Game of Sperms<br />

By Lauren Kim<br />

model involves sperm competition games,<br />

which show how males allocate energetic<br />

resources to increase their chances<br />

for success within male-male mating<br />

competition.<br />

Factors that affect the males’ decisions<br />

include female promiscuity, maternal<br />

effort, and the difficulty of providing<br />

care to offspring. Based on these factors,<br />

researchers found that when there is more<br />

energy required, males will provide care<br />

based on relatedness to his offspring. For<br />

example, a male Arowana fish will carry<br />

eggs in his mouth to protect his offspring.<br />

However, in low-cost situations, males will<br />

provide care regardless of relation.<br />

In this way, scientists hope to provide<br />

an answer as to why males continue to<br />

provide energetically-costly care towards<br />

offspring that may not be their own. From<br />

these results, they can develop a greater<br />

understanding of different parenting<br />

patterns in nature.<br />

IMAGE COURTESY OF NATIONAL GEOGRAPHIC<br />

►Male Arowana fish carry the eggs in<br />

their mouths, protecting the offspring<br />

until they are ready to leave permanently.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

7


NEWS<br />

materials science<br />

HARNESSING THE SUN FOR CLEAN WATER<br />

Nanotechnology improves water purification<br />

►BY ALLIE FORMAN<br />

For you and me, obtaining safe drinking water may be as<br />

simple as walking to the nearest sink and getting a cup of tap<br />

water. But 29 percent of people worldwide do not have ready<br />

access to safe drinking water, instead getting their water from<br />

sources contaminated by feces. This leads to over half a million<br />

deaths annually. Climate change and population growth<br />

are expected to exacerbate the issue, making access to potable<br />

water even more challenging.<br />

One solution to this problem is by making salt water drinkable<br />

through desalination. But the most commonly used technique<br />

for desalination, reverse osmosis, requires large inputs<br />

of energy to boil water, making it unsuitable for places without<br />

reliable electricity.<br />

With this in mind, Yale researchers in the School of Engineering<br />

and Applied Science, in collaboration with scientists<br />

at Rice University, Arizona State University, and the University<br />

of Texas-El Paso, are working to improve an alternative<br />

solar-powered desalination method known as membrane distillation.<br />

This process has the potential to be less energy-intensive<br />

than reverse osmosis, making it suitable for underdeveloped<br />

areas and environmentally friendly.<br />

In membrane distillation, salt water and pure water are<br />

placed on opposite sides of a membrane that only allows gases<br />

to pass through. The salty side is heated, while the pure water<br />

is cooled. The difference in partial pressure causes the water on<br />

the salty side to travel through the membrane as a gas, leaving<br />

the salt and other contaminants as solids on the “dirty” side of<br />

the membrane.<br />

Membrane distillation has not been widely used for a number<br />

of reasons. Energy is required to generate and maintain<br />

the temperature difference between the impure salt water and<br />

clean freshwater reservoirs, and the process uses more energy<br />

to produce the same amount of pure water when compared to<br />

reverse osmosis. Along with their collaborators, Yale researchers<br />

Menachem Elimelech and Akshay Deshmukh have optimized<br />

a low-energy membrane distillation technique that uses<br />

solar power coupled with nanoparticle technology.<br />

Their design uses a carbon black-coated membrane that<br />

maximizes uptake of solar energy. This sets up a temperature<br />

gradient in which the salt water closest to the nanoparticle-covered<br />

membrane heats up. The process does not require<br />

an additional power source besides solar energy, and the materials<br />

are widely available.<br />

While the nanoparticle-enabled solar membrane distillation<br />

system is still in development, the scientists have demonstrated<br />

its viability with a paper recently published in the Proceedings<br />

of the National Academy of Sciences. The project is attracting<br />

attention because of its potential applications around the<br />

world, especially in areas lacking access to electricity.<br />

The improved membrane distillation design is ideal for<br />

smaller, decentralized applications, such as a rural village not<br />

connected to the grid. Unlike reverse osmosis—which necessitates<br />

a proper electrical connection for the high-pressure<br />

pumps, the new solar membrane distillation model requires<br />

very little infrastructure. “We wouldn’t use a process like this to<br />

compete with reverse osmosis in a big community,” Deshmukh<br />

said. “But for smaller stuff, which is off-grid, it could be useful<br />

because it’s using sunlight and it’s not a high-pressure process.”<br />

Membrane distillation technology can also be used to remove<br />

other contaminants besides salt from water. “This could<br />

be used in situations where the water is very dirty. For example,<br />

for waste water from industrial sites where the osmotic<br />

pressure is very high, reverse osmosis can’t be used to treat<br />

it, because the membranes can’t deal with the pressure,” said<br />

Deshmukh. He also thinks their design could be implemented<br />

in natural disaster situations, where normal water treatment<br />

infrastructure has been disrupted.<br />

Though the researchers have produced a model of the design,<br />

the technology is not yet ready to hit store shelves. The<br />

scientists now need to refine their design and market approach.<br />

While there is still work to be done, they believe that<br />

the new technology will improve many lives around the globe<br />

by providing much needed access to clean water.<br />

IMAGE COURTESY OF AKSHAY DESHMUKH<br />

►The new design utilizes a nanoparticle layer to heat salt water<br />

with solar energy.<br />

8 Yale Scientific Magazine October 2017 www.yalescientific.org


neuroscience<br />

NEWS<br />

NOW YOU HEAR ME, NOW YOU DON’T<br />

Testing susceptibility of people to hearing voices<br />

►BY SUNNIE LU<br />

PHOTOGRAPHY BY YASMIN ALAMDEEN<br />

►Dr. Powers and Dr. Corlett took MRI scans of people<br />

performing auditory tasks to understand auditory hallucinations.<br />

You hear footsteps coming down the hallway and voices chanting,<br />

“We’re coming.” A shadowy figure suddenly appears in the<br />

corner of your room, while a girl dressed up as a rat looms over<br />

your bed. Although these scenarios sound like they come from<br />

horror movies, they actually are real examples of hypnagogic hallucinations,<br />

which occur during the onset of sleep. Both having<br />

these experiences, Yale psychiatrist and neuroscientist Albert<br />

Powers and Yale neuroscientist Philip Corlett fascinated with<br />

auditory hallucinations. Their research, featured in Science, suggests<br />

that people who hear voices are more likely to experience<br />

induced hallucinations in a lab.<br />

It may seem concerning that both Powers and Corlett have<br />

experienced hallucinations while falling asleep, but these hallucinations<br />

are usually symptomatic of a neurological condition,<br />

not a psychiatric illness. However, people without a psychiatric<br />

condition can hear voices too. The two scientists wanted to figure<br />

out what produces auditory hallucinations and why some<br />

voice-hearing experiences are benign and others require medical<br />

attention.<br />

To explore these questions, they sought out four groups of<br />

test subjects: both psychotic and nonpsychotic voice-hearers<br />

and non-voice-hearers. After identifying potential subjects, the<br />

researchers separated the psychotic and nonpsychotic people<br />

using a questionnaire developed by forensic psychologists to<br />

distinguish between people who were actually experiencing hallucinations<br />

and those who only claimed to do so.<br />

After screening their subjects for hallucinations, Powers and<br />

Corlett induced auditory hallucinations in their subjects to identify<br />

whether psychotic people were more likely than non-psychotic<br />

people to hear conditioned sounds. Using a technique<br />

originally developed at Yale during the 1890s, the subjects were<br />

stimulated with a checkerboard image and a one-second long<br />

sound simultaneously and repeatedly, while getting their brains<br />

imaged by MRIs. This conditioned the subjects to associate the<br />

image with the tone. As the scientists changed the intensity of<br />

tone, sometimes turning it off, the subjects pressed a button when<br />

they thought they heard the tone, while changing the length of<br />

time they pressed the button to show their level of confidence.<br />

Many reported hearing a tone when only the checkerboard<br />

image appeared but no tone played. This inconsistency occurred<br />

more often with the two voice-hearing groups—the people with<br />

schizophrenia and self-identified clairaudient psychics. Both<br />

groups were almost five times more likely to report that they<br />

heard a nonexistent tone than the non-voice-hearing groups.<br />

Furthermore, the two non-voice-hearing groups were 28 percent<br />

more confident that they had heard the tone when no tone<br />

played. These results support a possible explanation for hallucinations.<br />

“The brain makes models for what the outside world is<br />

like,” said Corlett, noting that these models sometimes don’t always<br />

match reality. This study suggests that people hallucinate<br />

when their expectations overweigh what their senses tell them.<br />

Powers and Corlett further understood auditory hallucinations<br />

through analyzing the MRI scans collected: the parts of the<br />

brain that were responsive to the tone were active when people<br />

reported conditioned hallucinations, producing MRI scans of<br />

brains that looked like those of people actually hearing the tone.<br />

The images also revealed that both hallucinating and non-hallucinating<br />

people with psychosis exhibit abnormal brain activity in<br />

regions that monitor internal representations of reality. These results<br />

contribute to the idea that hallucinations stem from internal<br />

representations overruling actual sensory data.<br />

This study was able to distinguish not only between those who<br />

hallucinate and those who don’t, but also between psychotic and<br />

non-psychotic people. “The sooner you catch psychosis and the<br />

sooner you intervene, the better the general outcomes are,” said<br />

Powers. According to Powers, most people with the symptoms<br />

associated with increased chances of psychosis don’t even develop<br />

psychosis. The question then is, who should receive treatment?<br />

This new research may help to answer that key question<br />

by providing the basis for tests to diagnose patients who require<br />

psychiatric treatment early.<br />

“There is no one-size-fits-all anti-psychotic, so there should be<br />

different treatments for different people,” Corlett said. Although<br />

this sort of precision medicine has not existed in psychiatry so<br />

far, Corlett hopes that this study, along with future research, will<br />

lead to more personalized psychiatric treatments, which he believes<br />

would be more effective in helping people suffering with<br />

mental health issues.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

9


NEWS<br />

genetics<br />

MAKING CANCER CELLS NERVOUS<br />

Targeting the genes involved in nerve cells in the brain<br />

►BY DIYU PEARCE-FISHER<br />

Cancer is the second most common cause of death in the<br />

world. Almost one out of every six deaths can be traced back to<br />

this deadly disease. Science has made huge strides towards treating<br />

and curing cancer, but there are many different types of cancer,<br />

and no single cure that works for every case. Understanding<br />

the cause of each type of cancer is crucial to treating each patient,<br />

so much recent cancer research focuses on the genetic factors that<br />

cause normal cells to become cancerous.<br />

Researchers at the Yale Systems Biology Institute led a team of<br />

scientists studying the genes involved in glioblastoma, a type of<br />

brain cancer. This cancer targets star-shaped support cells in the<br />

brain called astrocytes, which serve many purposes in the nervous<br />

system. Glioblastoma is one of the deadliest types of cancer.<br />

While over two-thirds of the people diagnosed with any type of<br />

cancer make it to the five-year survival mark and beyond, the median<br />

survival time for patients diagnosed with glioblastoma is between<br />

1 and 1.5 years. The Yale-led team of researchers developed<br />

a technique enabling them to identify specific gene combinations<br />

that cause glioblastoma. They believe their approach could be applied<br />

to other types of cancer as well, which would revolutionize<br />

how we approach cancer treatment.<br />

Cancer is caused by genetic mutations. However, even if two<br />

patients have the same type of cancer, the patients may or may<br />

not have the same mutations. Prior to the Yale-led study, genome<br />

sequencing identified 223 genes with links to glioblastoma. But<br />

that also means that there are hundreds of thousands of possible<br />

combinations of genes, with certain combinations responding<br />

slightly differently to treatment. Thus, the Yale researchers wanted<br />

to clarify how these different combinations affect glioblastoma<br />

treatment.<br />

To do this, the Yale team employed the use of a technique called<br />

CRISPR. First pioneered in 2012, CRISPR is a gene editing technique<br />

that has revolutionized the biomedical sciences by enabling<br />

researchers to create cells with mutations and test the effects of the<br />

mutations. However, CRISPR does have its limitations. To use this<br />

type of CRISPR, each combination would have to be tested one<br />

by one, which would be hard to compare since the time-intensive<br />

tests would give results two to three years apart.<br />

To address this, the Yale-led team developed a novel CRIS-<br />

PR technique that is less biased, more time-efficient, and more<br />

cost-efficient. Rather than testing each individual mutation and<br />

waiting for the results of each experiment, which would exhaust<br />

both time and resources, the new technique employs a pool of viruses,<br />

each of which targets a specific gene. Infected from a pool<br />

of viruses, each mouse brain cell has the potential to have multiple<br />

mutations. Thus, the team could test for multiple combinations<br />

simultaneously. The researchers found specific combinations of<br />

mutations that could cause glioblastoma, as well as two combinations<br />

that could make the glioblastoma tumor resistant to chemotherapy.<br />

By understanding the results of different combinations of mutations,<br />

doctors will be able to screen for these mutations and decide<br />

the appropriate treatments. For instance, if they find that a patient<br />

has the combination of mutations that cause a tumor to be resistant<br />

to chemotherapy, the doctors will know not to waste time and<br />

resources on an unsuccessful chemotherapy, which could further<br />

harm or discourage the patient. Information like this is especially<br />

important when the average survival time for cancers like glioblastoma<br />

is under two years.<br />

The researchers believe that their approach can be applied to<br />

other types of cancer, which would allow doctors to give treatments<br />

specific to each case, hopefully leading to a higher chance<br />

of success. “In order to find better therapy or treatment, we need<br />

to find the therapeutic responder, or the genes that regulate or<br />

change some response. Now we have the tools—we have the experiment<br />

setting—we can simply do the same experiments but<br />

with gene therapy,” said Sidi Chen, one of the investigators on the<br />

team. By using CRISPR to create the different glioblastoma-causing<br />

gene combinations and applying specific types of therapies to<br />

see how each combination responds, the scientists may be able to<br />

figure out exactly which treatment will work for each case of cancer.<br />

In other words, targeted “cures” for cancer may soon become<br />

an applicable reality.<br />

IMAGE COURTESY OF CHEN LAB<br />

►A gioblastoma induced by new gene-editing and screening<br />

technology.<br />

10<br />

Yale Scientific Magazine October 2017 www.yalescientific.org


public health<br />

NEWS<br />

NOT SO SWEET<br />

Metabolic problems caused by artificial sweeteners<br />

►BY SERENA CHO<br />

PHOTOGRAPHY BY KELLY ZHOU<br />

►Diet drinks contain artificial sweetners that trick our brains<br />

into thinking we are getting a sweet calorie reward.<br />

Diet Coke, Vitamin Water Zero, Minute Maid Light: with<br />

the hype over healthy living and weight loss, the food industry<br />

has introduced many low-calorie drinks to the market.<br />

Most of these beverages use artificial sweeteners, which<br />

provide the same sweet taste with fewer or no calories. But<br />

what effects do the artificial sweeteners have on our body?<br />

Are they safe, after all? The hidden effects of artificial sweeteners<br />

on our body have been long disputed.<br />

According to research conducted by Yale School of Medicine<br />

scientists, the mismatch between the sweet taste and<br />

caloric value in diet beverages could explain the link between<br />

artificial sweetener use and cases of metabolic failure,<br />

such as in diabetes. Because the brain cannot register<br />

how much sugar was consumed, our body cannot properly<br />

digest and process the diet beverages.<br />

A sweet taste helps indicate the amount of energy present<br />

in a food source. “Our bodies evolved to efficiently use<br />

the energy sources available in nature,” said senior author<br />

and Yale psychiatry professor Dana Small. She continued,<br />

“Our modern food environment is characterized by energy<br />

sources our bodies have never seen before.” When a beverage<br />

tastes too sweet or not sweet enough for the number of<br />

calories it contains, the signal communicating its nutritional<br />

value can be disrupted. Our metabolism is confused, and<br />

the beverage cannot be digested properly.<br />

Small and her colleagues discovered the problems of<br />

“diet” beverages when performing an experiment on brain<br />

responses to sugar ingestion. The team observed the brain<br />

activity of fifteen healthy participants after drinking tasteless<br />

sugary drinks. Normally, when sugar is introduced to<br />

the body, the brain releases dopamine, a chemical compound<br />

that induces reward and pleasure. However, when<br />

the participants drank tasteless but sugary drinks, the<br />

amount of dopamine released was not proportional to the<br />

calories in the beverage. “In other words, the assumption<br />

that more calories trigger greater metabolic and brain response<br />

is wrong,” Small said. She was interested in pursuing<br />

this further, commenting, “A strange finding that makes no<br />

sense means that you’re going to learn something new.”<br />

Further research revealed that a sweet taste also determines<br />

how sugar is metabolized and signaled to the brain.<br />

“Calories are only half of the equation; sweet taste perception<br />

is the other half,” Small said. In fact, sweet low-calorie<br />

drinks could produce greater metabolic response and higher<br />

dopamine levels than less sugary higher-calorie drinks.<br />

In nature, when sweetness reflects the number of calories<br />

in the food, the body properly metabolizes the calories and<br />

activates the corresponding brain reward signals. However,<br />

the modern food industry introduces beverages with the<br />

kind of sweetness that our body hasn’t been exposed to before.<br />

“A calorie is not a calorie,” said Small, commenting on<br />

how the impact of these drinks on our bodies goes beyond<br />

just the number of calories. When sweetness and calories<br />

don’t align, like in artificially sweetened drinks, the beverage<br />

fails to trigger metabolism and the brain reward circuits<br />

can’t register the number of calories consumed. The metabolic<br />

failure observed here could help explain the link between<br />

artificial sweeteners and problems like diabetes.<br />

IMAGE COURTESY OF WIKIMEDIA COMMONS<br />

►Artificial sweetners confuse our metabolic systems, possibly<br />

creating mixed signals that lead to health issues like diabetes.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

11


WHAT IS HOT, DENSE,<br />

& spins like crazy?<br />

art by Emma Wilson<br />

by Sophia Sánchez-Maes<br />

It all started with a big bang...but then what?<br />

Our universe is 13.8 billion years old—<br />

the most remarkable detail about this fact<br />

is what it implies about its beginning. Space<br />

itself is continuously expanding, and winding<br />

back the clock shows it getting smaller<br />

and smaller, eventually shrinking down to<br />

a single point from whence it all came: life,<br />

the universe, and time itself. Squeezing the<br />

contents of the entire universe into a single<br />

point may seem unimaginably difficult, but<br />

compress matter enough, and that inward<br />

pressure is enough to break molecular bonds<br />

into atoms. Compress further and atoms<br />

themselves break into their components:<br />

protons, neutrons, and electrons. Next, the<br />

stress would break even the bonds that tie<br />

those subatomic particles together, releasing<br />

even smaller particles called quarks and gluons—two<br />

of the 13 elementary particles that<br />

constitute the most basic building blocks of<br />

matter. As carriers of the strong force, gluons<br />

are the glue that acts to bind quarks together<br />

into protons and neutrons. The strong force<br />

governs interactions between all particles<br />

with a color charge, like quarks and gluons,<br />

and is also responsible for binding together<br />

the atomic nucleus.<br />

In order to overcome intermolecular and<br />

interatomic forces and explore the properties<br />

of this primordial cosmic soup, scientists<br />

must replicate the conditions of the early<br />

universe just milliseconds after the Big Bang,<br />

which involved blistering temperatures and<br />

tightly packed matter.<br />

This August, the STAR collaboration, a<br />

group of scientists aiming to deduce the<br />

properties of the quark gluon plasma and<br />

the physics that underlay them, published<br />

their measurement of the vorticity of the<br />

quark-gluon plasma in Nature: a experiment<br />

which elucidates the unexpected fluid properties<br />

of this matter that dominated early in<br />

the universe, and is an important step to bettering<br />

our theory of the strong force.<br />

IMAGE COURTESY OF BROOKHAVEN NATIONAL LABORATORY<br />

►The tracks in this image depict the particles<br />

produced by the collision of gold ions at RHIC,<br />

as captured by the STAR detector’s Time<br />

Projection Chamber.<br />

Exploring the universe, before it was cool<br />

Researchers like Yale’s Helen Caines don’t<br />

need to journey through time to discover<br />

properties of early universe. They have only<br />

to cross the Long Island Sound to Brookhaven<br />

National Laboratory, where the Relativistic<br />

Heavy Ion Collider is cooking up the<br />

same conditions. At Brookhaven, STAR researchers<br />

focus narrow beams of gold ions<br />

(atoms which have lost their outer electrons)<br />

at one another, each traveling close to the<br />

speed of light. Such incredible speeds also<br />

give the ions incredible energy, so any headon<br />

collision between gold ions is able to<br />

dissolve the 79 protons and 118 neutrons in<br />

each gold ion into quarks and gluons, forming,<br />

for a brief instant, the quark gluon plasma,<br />

a soup of quarks and gluons which exists<br />

at extreme temperatures and densities. The<br />

Relativistic Heavy Ion Collider, along with<br />

CERN’s Large Hadron Collider in Geneva<br />

are the only facilities in the world with machines<br />

powerful enough to produce quark<br />

gluon plasma.<br />

Having proven that the produced plasma is<br />

indeed comparable to that of the primordial<br />

cosmos, scientists in the STAR collaboration<br />

moved on to also show that this material is<br />

full of unexpected surprises. For example,<br />

researchers anticipated that such a hot, energetic<br />

state of matter would behave like some<br />

sort of super gas, without the strong collectivity<br />

properties that characterize liquids,<br />

unlike gasses, which diffuse evenly.<br />

In actuality, the quark gluon plasma<br />

(QGP) behaves like a liquid, since its constituents<br />

interact more strongly than those of a<br />

gas. “We know that the gluons themselves<br />

interact, and the quarks interact via gluons,<br />

so it makes sense if they can interact very<br />

strongly with each other,” Caines said.<br />

In this series of experiments, scientists are<br />

aiming to determine the properties of the<br />

quark gluon plasma’s fluid properties. Not<br />

only is the QGP a liquid, but it has the lowest<br />

viscosity of any liquid ever encountered,<br />

meaning that unlike viscous substances like<br />

honey, it has nearly no resistance to flow.<br />

This unique property has prompted scientists<br />

to call it nearly “perfect.”<br />

Since this medium formed under such<br />

extreme temperatures and pressures, it’s<br />

12 Yale Scientific Magazine October 2017 www.yalescientific.org


particle physics<br />

FOCUS<br />

distance, it gets increasingly strong, like a<br />

spring that wishes to retract. Eventually, so<br />

much energy is expended in pulling apart<br />

this particle that another particle antiparticle<br />

pair is pulled from the vacuum to preserve<br />

net colorlessness. This means that it’s<br />

impossible to detect free quarks. This might<br />

be puzzling, since quarks in the QGP aren’t<br />

bound in mesons or nucleons, but zooming<br />

in anywhere in this soup, conditions are “locally”<br />

colorless. Since scientists can’t detect<br />

the quarks individually, the STAR collaboration<br />

scientists instead detect the particles<br />

created from the QGP as it cools, whose motion<br />

is inherited from this fluid that formed<br />

them.<br />

It’s not “just a phase”<br />

not surprising that the medium breaks other<br />

records. These record-setting properties<br />

include a low viscosity, a high temperature,<br />

and a now legendary vorticity, or swirling<br />

properties. These measurements not only<br />

help us understand this unexpected liquid<br />

phase, but also assist in our understanding of<br />

quantum chromodynamics (QCD), a theory<br />

which governs the interactions of the colored<br />

charged particles (quarks and gluons)<br />

which make up the QGP.<br />

The force is strong with this one<br />

Quarks are never observed in alone, but<br />

are always bundled in pairs or groups of<br />

three by gluons, which carry the strong<br />

force. There are six types of quark, with<br />

up and down quarks combining in groups<br />

of three to create protons and neutrons<br />

– the heaviest components of atoms. The<br />

theory governing their interactions is<br />

called quantum chromodynamics (QCD).<br />

Quarks come in three states called ‘colors.’<br />

The strong force, which governs the behavior<br />

of quarks, is incredibly unusual.<br />

Gravity, the force with which we’re most<br />

familiar, only attracts objects toward each<br />

www.yalescientific.org<br />

IMAGE COURTESY OF WIKIPEDIA`<br />

►The color charge acts such all matter must be locally white, binding quarks into combinations<br />

that preserve this colorlessness and preventing the sustained existence of free quarks.<br />

other. Electromagnetism has positive and<br />

negative (anti-positive) charges – but like<br />

gravity, their effect fades with the inverse<br />

square of the distance. Quarks and gluons<br />

have color-charge, which has nothing<br />

to do with color but for convenient<br />

metaphor. There are three colors (red,<br />

green, and blue) and three anti-colors<br />

(anti-red, anti-green, anti-blue) Any stable<br />

state must be colorless – which can<br />

only be achieved by mixing all the colors,<br />

like in protons and neutrons (made up of<br />

three quarks) – or by combining a color<br />

and anti-color, like in particles called mesons<br />

(which consist of a quark and an anti-quark).<br />

Furthermore, mass and charge<br />

are fundamental properties of particles,<br />

whereas color charge can change via gluon<br />

interactions. It is the strong force, and<br />

the color charge that mediates it, that is<br />

responsible for the interactions between<br />

quarks and gluons. By understanding<br />

the behavior of the QGP, scientists are<br />

probing these fundamental forces and<br />

properties.<br />

Try pulling a meson apart and you’ll see<br />

that the strong force operates differently<br />

in another important way: with increasing<br />

The QGP is a fluid, and is defined by superlatives.<br />

At four trillion degrees Celsius,<br />

tens of thousands of degrees hotter than a<br />

supernova, it’s the hottest thing we know<br />

in the universe. It also has the lowest viscosity,<br />

or resistance to flow, of any known<br />

fluid. Now, the STAR collaboration has also<br />

made the first measurement of its vorticity,<br />

or rotation.<br />

When gold ions collide head-on, their<br />

constituent particles combine to create the<br />

QGP; but typically the collision between<br />

ions is more glancing. In that case, region<br />

of contact, like the center of a Venn diagram,<br />

will combine to become QGP, while<br />

the outside regions will continue speeding<br />

away from each other. These glancing collisions<br />

give the resulting soup a net angular<br />

momentum, setting the QGP spinning.<br />

Scientists in the STAR collaboration<br />

aimed to detect this spin by detecting the<br />

particles generated by the plasma as it cools.<br />

October 2017<br />

IMAGE COURTESY OF WIKIPEDIA<br />

►The quark gluon plasma kicked off the start<br />

of the radiation era of the universe’s history<br />

from 10e-12 to 10e-6 seconds after the big<br />

bang, right after cosmic inflation.<br />

Yale Scientific Magazine<br />

13


FOCUS<br />

particle physics<br />

IMAGE COURTESY OF BERKELEY LABS<br />

►The color charge acts such all matter must be<br />

locally white, binding quarks into combinations<br />

that preserve this colorlessness and preventing<br />

the sustained existence of free quarks.<br />

IMAGE COURTESY OF GETTY IMAGES<br />

►This image, taken near RHIC at Brookhaven<br />

Laboratory depicts a subset of the STAR<br />

collaboration.<br />

Any angular momentum in the system will<br />

be passed on to the particles it generates,<br />

whose spins are aligned with the whirling<br />

structures from which they came. On average,<br />

those spins will indicate the net angular<br />

momentum created by the system. As<br />

Yale’s Professor Caines, spokesperson for<br />

the collaboration described, “when you collide<br />

them just a little bit edge on edge you<br />

set it spinning, and that’s basically what we<br />

measured, how fast it’s spinning collectively.<br />

We can detect that angular momentum.”<br />

The experiment used two instruments for<br />

this measurement. The first of these was the<br />

Time Project Chamber, which is filled with<br />

gas that surrounds the collision and allows<br />

scientists to track the particles released<br />

from the collision and therefore generated<br />

by the QGP. This detector measures protons<br />

created by the plasma. The second instrument,<br />

the Beam-Beam Counters, measures<br />

deflections in the paths of particles which<br />

whiz by one another and indicates the magnitude<br />

and direction of angular momentum<br />

in each collision. Correlating this measured<br />

angular momentum with aligned direction<br />

of the detected protons, STAR scientists<br />

searched for preference in direction that<br />

will indicate the vorticity of the system.<br />

This measurement indicates that the<br />

QGP is the most vortical fluid known, a<br />

measured value surprising in its magnitude.<br />

The vorticity of the QGP exceeded<br />

all predicted values – whirling faster than<br />

tornados, Jupiter’s red spot, and previous<br />

vorticity record-holder superfluid helium.<br />

This high vorticity is allowed to perpetuate<br />

by the low viscosity of the fluid – since high<br />

resistance to flow would dampen whirls prior<br />

to measurement. Vorticity is a property of<br />

the flow – it’s not intrinsic to the medium,<br />

but results from the medium and its motion.<br />

But since we know the conditions that<br />

produced the flow inside of the collider, it’s<br />

possible to gain useful information about<br />

the fluid properties. Knowing the specifics<br />

of the collision that generated it allows us to<br />

firm up our theories of how the QGP interacts<br />

and responds to forces.<br />

Changes in temperature and pressure induce<br />

phase transitions. Think briefly of ice:<br />

increasing the temperature will turn it to<br />

water, then to vapor. At 4 trillion degrees<br />

Celcius, the quark gluon plasma is very hot,<br />

which is why the strong mutual interactions<br />

of its constituent particles is puzzling. The<br />

QGP behaves more like a fluid than like a<br />

gas. Instead of expanding out evenly in all<br />

directions, it’s preferential in its flow. Is it a<br />

relic of the strong force, which governs interactions<br />

between its constituent parts? Or<br />

perhaps the extreme pressure of the cosmic<br />

soup? In future experiments, the STAR collaboration<br />

hopes to deduce the equation of<br />

state for the quark gluon plasma to resolve<br />

these questions and test theories about the<br />

strong force.<br />

This vorticity measurement will aid in<br />

improving theories of the plasma and how<br />

it affected the properties of the early universe.<br />

Most importantly, spinning charges<br />

produce magnetic fields, making this measurement<br />

an important first step in making<br />

the first measurements of the magnetic<br />

properties of the QGP, an important step<br />

in better understanding the weird physics<br />

surrounding this hot, flowy, whirling<br />

soup. Although the significance of such<br />

fundamental science isn’t always immediately<br />

clear, Yale’s Li Yi, a scientist with the<br />

collaboration sees this as exciting infinite<br />

possibility. “Maybe it sounds like science<br />

fiction, but in all of the modern world most<br />

of the technology is based on QED (quantum<br />

electro dynamics), our theory of the<br />

electromagnetic force. For example, we<br />

transfer the messages and communications<br />

through electromagnetic waves - this is all<br />

through the QED because we really understand<br />

what we can do. QCD we don’t know<br />

exactly how it works - or what we could use<br />

it for.” The STAR Collaboration’s measurements<br />

of the QGP are among the most important<br />

measurements for formulating and<br />

testing this theory.<br />

ABOUT THE AUTHOR<br />

SOPHIA SANCHEZ-MAES<br />

SOPHIA SANCHEZ-MAES is a junior Physics and Astrophysics double major<br />

in Timothy Dwight college. She works with Professor Jun Korenaga on studying<br />

the origin processes of plate tectonics, and topics in complex systems.<br />

THE AUTHOR WOULD LIKE TO THANK Professor Helen Caines and Dr. Li<br />

Yi for their incredible knowledge and willingness to share it.<br />

FURTHER READING<br />

The Star Collaboration. (2017). Global Λ hyperon polarization in nuclear<br />

collisions. Nature. Retrieved October 1, 2017.<br />

14 Yale Scientific Magazine October 2017 www.yalescientific.org


THE SEARCH<br />

Mathematical model explains<br />

diversity in cancer cell movement<br />

by Charlie Musoff | art by Anusha Bishop


FOCUS<br />

cell biology<br />

You lost your keys again. Without any idea as to where they went, you dart from room to room,<br />

pursuing each lead for just a moment before giving up and trying something else. As you search,<br />

you piece it together and suddenly remember where they are: the refrigerator. Turning away from your<br />

hamper, you make a beeline for the kitchen, and the keys are recovered within moments. The way you<br />

went about finding your keys—the indiscriminate giving way to the direct—is not unique to forgetful<br />

humans. All living things—dogs, bees, cancer cells—devise these strategies as they explore the world<br />

around them. In the last case, however, this probing can do more harm than good. The more cancer cells<br />

move in a persistent beeline, the more efficiently they will spread and the more lethal the cancer will be.<br />

A team at the Yale Systems Biology Institute<br />

led by professor Andre Levchenko<br />

and postdoctorate researcher JinSeok Park<br />

wanted to more comprehensively understand<br />

how cancer cells find their proverbial<br />

keys. To start, it is important to understand<br />

how cells interact with their direct<br />

outside environment, a network of proteins<br />

called the extracellular matrix (ECM) that<br />

cells secrete themselves. Coming into contact<br />

with a neighboring cell’s ECM triggers<br />

two signaling pathways that mediate cell<br />

movement. The balance between the different<br />

behaviors that these two pathways<br />

trigger—the cell’s polarity, so to speak—<br />

influences cells’ migration behavior, their<br />

switch from frenzy to beeline. Previous research<br />

implied that cell contact with the<br />

ECM was responsible for these different<br />

patterns, but how the cells went about this<br />

was poorly understood. Levchenko and<br />

Park derived a mathematical model to predict<br />

a given cell’s behavior based on the activity<br />

of the two principal signaling pathways,<br />

which both cements the link between<br />

cell-ECM contact and migration and, more<br />

importantly, clarifies a new avenue for cancer<br />

treatment.<br />

Migration<br />

The type of cancer called melanoma<br />

rarely kills at its origin, the skin, but rather<br />

takes lives when it attacks other organ<br />

systems, or metastasizes. Therefore, it is a<br />

disease that relies heavily on movement,<br />

a trait that made it a useful model for the<br />

team to study. The critical moment in melanoma<br />

progression happens when the cancer<br />

stops expanding across the skin and begins<br />

tunneling downward. At that point,<br />

it is only a matter of time until melanoma<br />

cells find their way to a blood or lymphatic<br />

vessel, the anatomical highways necessary<br />

for metastasis. But first, these cells have to<br />

navigate the fibers of the dermis, the layer<br />

of tissue right below the skin.<br />

These tissue fibers are like ropes that cells<br />

can pull to move around. There are three<br />

types of cell movements. When the cells<br />

rapidly pull ropes in different directions,<br />

their behavior is random; when cells alternate<br />

pulling forward and backward on the<br />

same rope, their behavior is oscillatory;<br />

and when cells consistently pull one rope in<br />

one direction, their behavior is persistent.<br />

The oscillatory pattern is a cell-specific behavior<br />

and complicates the model—once<br />

a target has been established for the cell,<br />

it continues to travel back and forth in the<br />

vicinity instead of heading straight there.<br />

Levchenko likes to think of oscillation as<br />

a cell overshooting its target and doubling<br />

back repeatedly. Regardless, if a cell is to<br />

reach a destination, persistent behavior is<br />

the most efficient, which, in the context of<br />

cancer, means a faster progression towards<br />

metastasis.<br />

Migration patterns do not happen by<br />

chance. Cells’ direct contact with the fibers<br />

of the ECM sets off two sets of signals<br />

that influence cell movement and so<br />

determine whether cells will display random,<br />

oscillatory, or persistent behavior.<br />

One of these signals is characterized by a<br />

protein called Rac1 and the other by a protein<br />

called RhoA. The two sets of signals<br />

work towards the same goal—to mediate<br />

cell movement—but perform opposite<br />

functions, as Rac1 acts as an accelerator to<br />

RhoA’s brakes. To complicate matters further<br />

RhoA functions to halt cell migration,<br />

its presence is also required for migration<br />

in the first place. This somewhat paradoxical<br />

mechanism only highlights the complexity<br />

of these networks. Research in oncology<br />

typically has focused on how cancer<br />

cells grow and replicate, but these convoluted<br />

and poorly understood processes of<br />

cell migration pushed Levchenko and Park<br />

to establish a coherent model and reconcile<br />

the three migration patterns.<br />

Polarization<br />

The model the researchers derives uses<br />

advanced mathematics to integrate these<br />

three behaviors into the same framework,<br />

an unprecedented feat. The model shows<br />

how cells’ contact with the ECM changes<br />

the balance between the levels of activity<br />

of the Rac1 and RhoA pathways, which<br />

in turn impacts migration. By plotting the<br />

two activation rates on the same axes, the<br />

researchers were able to establish discrete<br />

regions for each migration pattern. For example,<br />

if a cell has a high Rac1 activation<br />

rate and a low RhoA activation rate—a lot<br />

of accelerator and little brakes—it will display<br />

persistent behavior.<br />

The model accounts for not only the balance,<br />

but also the location of this pathway<br />

activation within a given cell. By tracking<br />

sites of high signaling activity, the researchers<br />

were able to visualize the spatial<br />

distribution of these two pathways. In<br />

randomly migrating cells, signaling was<br />

sporadic; in oscillating cells, hotspots of<br />

activity alternated between the front and<br />

the back; and in persistently moving cells,<br />

activity was high on one side and inhibited<br />

on the other. These signals are consistent<br />

with the aforementioned “ropes” of the<br />

ECM and helped Levchenko and Park affirm<br />

that their model accurately represented<br />

cell movement.<br />

This model is unique because Levchenko<br />

and Park were able to map specific populations<br />

of cells onto diagrams that were generated<br />

using the model. Within the same<br />

cancer or even the same tumor, different<br />

cells will display different behaviors, and<br />

the model accounts for these varied responses.<br />

“[Cells] all get the same piece of<br />

information, but they interpret that information<br />

in very different ways,” Levchenko<br />

said. Understanding how real cells move<br />

within and correspond to the rigid math-<br />

16 Yale Scientific Magazine October 2017 www.yalescientific.org


cell biology<br />

FOCUS<br />

ematical model was an especially rigorous<br />

step that confirmed the model’s validity.<br />

Manipulation<br />

IMAGE COUTESY OF JINSEOK PARK<br />

►Professor Andre Levchenko , director of the Yale Systems Biology Institute, worked with<br />

professor JinSeok Park on this research.<br />

Once a model had been established, the<br />

next step was to manipulate it. One determinant<br />

of cell migration is the density of<br />

the ECM. To mimic the ECM, the researchers<br />

placed artificial nanoscale posts on a<br />

cell adhesion surface. As more posts were<br />

added, the fraction of persistently migrating<br />

cells decreased. “[The cells] are getting<br />

too much input from the surface, so they<br />

are confused,” Park said. When there were<br />

fewer posts, cells seemed to perceive their<br />

organization into rows and so had stronger<br />

directional cues, but when there were many<br />

posts, the cells were less able to navigate.<br />

The proportions of the migration patterns<br />

as predicted by the model were backed up<br />

with experimental data from real melanoma<br />

cells, and as expected, the more persistent<br />

cell migration was, the farther cells<br />

migrated.<br />

Levchenko and Park went on to alter a<br />

whole host of factors in order to understand<br />

the model’s viability outside their<br />

tightly regulated conditions. When either<br />

the Rac1 or RhoA signaling pathway was<br />

perturbed, the fraction of persistently moving<br />

cells decreased, a finding that is consistent<br />

with the networks’ modulation of cell<br />

movement. Similarly, when a drug that inhibits<br />

the formation of microtubules, elements<br />

of the cytoskeleton that play a role<br />

in migration, was administered, persistent<br />

cells decreased. Lastly, the researchers<br />

tested whether using a different cell line<br />

would yield the same results. All the previous<br />

experiments were done in a cancer<br />

cell known to be particularly invasive. To<br />

determine whether the model would fit a<br />

less aggressive melanoma, it was tested in a<br />

non-invasive cell line for which fewer persistent<br />

cells were expected, as a less invasive<br />

cancer metastasizes less efficiently. Despite<br />

the change in the aggressiveness of the cancer,<br />

the model held, which suggests its application<br />

in a much broader context of cancer<br />

therapy.<br />

Humanization<br />

To expand their discovery, Levchenko<br />

and Park hope to shed further light<br />

on the role of cell-ECM communication<br />

in cancer research. Once a complete map<br />

of the key signaling networks that dictate<br />

a cancer’s behavior is understood,<br />

targeted therapies can infiltrate them. If<br />

the networks can be manipulated to limit<br />

the proportion of persistently migrating<br />

cells, then even a cancer that has begun<br />

to spread can be delayed. Prolonging the<br />

period of time before a cell makes a beeline<br />

for a blood or lymphatic vessel in this<br />

fashion is an untapped therapeutic avenue<br />

towards which this model makes great<br />

strides. On the clinical side, the model allows<br />

for much more accurate predictions<br />

of the interval of time between the onset<br />

of a primary tumor and the cancer’s metastasis,<br />

which can undoubtedly improve<br />

cancer prognosis.<br />

Cancer treatment is ultimately about patients’<br />

well-being. If this model can delay<br />

the cell search and prevent persistent cell<br />

migration, it could prolong life by months,<br />

which can make a significant difference for<br />

a highly aggressive cancer like melanoma,<br />

which has only a 15-20% 5-year survival<br />

rate in its most advanced form. Understanding<br />

how we can target the molecules<br />

that modulate cells’ search period is an<br />

important therapeutic step towards an increase<br />

in these rates. Maybe one day melanoma<br />

cells will never find their keys.<br />

ABOUT THE AUTHOR<br />

CHARLIE MUSOFF<br />

CHARLIE MUSOFF is a sophomore in Davenport College and a molecular,<br />

cellular, and developmental biology major. Besides being Yale Scientific’s<br />

Outreach Designer, Charlie enjoys running, singing with the Baker’s Dozen,<br />

and teaching with Community Health Educators.<br />

THE AUTHOR WOULD LIKE TO THANK Professors Andre Levchenko and<br />

JinSeok Park for their time and insights.<br />

FURTHER READING<br />

Holmes, William R., et al. “A Mathematical Model Coupling Polarity Signaling to<br />

Cell Adhesion Explains Diverse Cell Migration Patterns.” PLOS Computational<br />

Biology, vol. 13, no. 5, 4 May 2017, doi:10.1371/journal.pcbi.1005524.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

17


TURNING back the CLOCK<br />

New drug restores<br />

memory in mice<br />

with fewer side<br />

effects<br />

August would be the first patient diagnosed<br />

with Alzheimer’s Disease. After her<br />

death, Dr. Alzheimer examined her brain<br />

and reported the presence of peculiar alterations<br />

in August’s brain tissue, which would<br />

later be called plaques and tangles. These<br />

changes are now considered hallmarks of<br />

a disease that contributes to the progressive<br />

decline in cognitive function of more<br />

than five million people in the United States<br />

alone.<br />

Researchers in the Strittmatter Lab at the<br />

Yale School of Medicine used a new drug to<br />

restore the memory and learning abilities<br />

of mice with Alzheimer’s. Unlike other Alzheimer’s<br />

drugs that are sometimes linked to<br />

adverse side-effects, this drug, called SAM,<br />

specifically targets the proteins linked to the<br />

disease without affecting essential proteins<br />

in the brain.<br />

A sticky disease<br />

by WILLIAM BURNS<br />

art by SIDA TANG<br />

Over a century has passed since German physician Dr. Alois Alzheimer<br />

documented the case of August Deter, a patient whose<br />

misunderstood and senseless behavior earned her admittance at<br />

a mental institution in Frankfurt in 1901. When Dr. Alzheimer asked<br />

for her name, August responded with the now eminent phrase,<br />

“I HAVE LOST MYSELF.”<br />

Plaques are sticky clusters made up of proteins<br />

called beta-amyloid that accumulate<br />

between nerve cells in the brain. Recent evidence<br />

suggests that small aggregates of beta-amyloid<br />

plaques are more damaging than<br />

the massive plaques themselves. These aggregates,<br />

called oligomers, cluster in synaps-<br />

es—the small gaps separating neurons of the<br />

brain—and block cell-to-cell communication.<br />

When synapses are disrupted, neurons<br />

cannot send electrical signals to each other,<br />

a dysfunction that ultimately contributes to<br />

the loss of memory and brain function characteristic<br />

of Alzheimer’s disease.<br />

On a molecular level, research has shown<br />

that the interaction between two particular<br />

proteins leads to Alzheimer’s disease. These<br />

two proteins are called cell-surface glycoprotein<br />

(PrP) and metabotropic glutamate<br />

receptor 5 (mGluR5). The interaction between<br />

these two proteins relays neurotoxic<br />

signals across the brain, damages synapses,<br />

and drains the brain’s cognitive capacity. Beta-amyloid<br />

peptides play a role in this process<br />

by strengthening the disease-causing<br />

interaction between PrP and mGluR5.<br />

The catch, however, is that these proteins<br />

also serve important roles in the normal<br />

functioning of the body. GluR5, for example,<br />

is a receptor found on membranes of neurons<br />

and interacts with glutamate, a chemical<br />

messenger that relays signals across the<br />

synapses of neurons. Glutamate plays an essential<br />

role in a wide range of neural functions,<br />

including learning, memory, and synaptic<br />

plasticity, or the ability of synapses in<br />

the brain to adapt to new information and<br />

strengthen over time. The more closely neurons<br />

are “wired” together at their synapses,<br />

the more robust the brain’s ability to communicate<br />

between neurons will be.<br />

Introducing SAM<br />

Alzheimer’s drugs that block glutamate interactions<br />

with mGluR5 have been shown to<br />

trigger visual hallucinations, insomnia, and<br />

cognitive dysfunction. As a result, designing<br />

an Alzheimer’s drug that specifically targets<br />

the interaction between beta-amyloid,<br />

PrP, and mGluR5 while leaving glutamate<br />

signaling undamaged has been the goal of<br />

researchers for many years. “A few years<br />

ago, we did an experiment with a drug that<br />

blocks mGluR5, and it ameliorated some<br />

Alzheimer’s conditions, but the drug also<br />

blocks glutamate,” said professor Stephen<br />

Strittmatter. “What we really need, however,<br />

is a drug that stops Alzheimer’s but preserves<br />

the normal physiology of glutamate.”<br />

The researchers contacted the American<br />

pharmaceutical company Bristol Myers<br />

Squibb and requested a schizophrenia drug<br />

called SAM. The researchers noticed that<br />

SAM acted on the same mGluR5 pathway<br />

18 Yale Scientific Magazine October 2017 www.yalescientific.org


neuroscience<br />

FOCUS<br />

they were studying and predicted that SAM<br />

would have an effect on their Alzheimer’s<br />

mice. Encouragingly, the researchers found<br />

that SAM blocks the interaction of a beta-amyloid<br />

with PrP and mGluR5 but leaves glutamate<br />

signaling unaffected in mice. To determine<br />

this, the researchers measured the<br />

amount of calcium ions released inside the<br />

neurons, since glutamate signaling is known<br />

to be involved with the release of that ion. Because<br />

SAM did not alter calcium levels in the<br />

brain, the researchers concluded that SAM<br />

does not disturb glutamate signaling. “We<br />

found that SAM blocks one type of signaling<br />

through GluR5 but maintains another: glutamate<br />

signaling," Strittmatter said. "It stops the<br />

pathology but leaves the physiology."<br />

Like most Alzheimer’s drugs, SAM binds<br />

to PrP and mGluR5 in a way that changes<br />

the overall shape of the group. This prevents<br />

beta-amyloid peptides from sticking to the<br />

group and forming the toxic protein clusters<br />

that damage synapses. Further, the researchers<br />

showed that SAM does not alter<br />

the strength of signals transmitted between<br />

neurons in mice brains. Thus, SAM selectively<br />

targets the disease-causing mechanism of<br />

Alzheimer’s while leaving normal functions<br />

undamaged.<br />

Maze trials<br />

The researchers examined whether SAM<br />

could ameliorate memory deficits in mice<br />

models of Alzheimer’s. The mice’s first task<br />

was a water maze. “When the mice are<br />

young and they don’t yet have accumulated<br />

amyloid in the brain, they can learn tasks<br />

like how to figure out a maze, but as the mice<br />

age, accumulate amyloid, and lose synapses,<br />

they become unable to learn,” Strittmatter<br />

said. The Alzheimer’s mice, the Alzheimer’s<br />

mice treated with SAM, and the healthy<br />

mice were all given the task to navigate the<br />

same maze and find a platform submerged<br />

in a pool.<br />

For the next three days, the mice became<br />

familiarized with the layout of the maze. The<br />

researchers positioned the mice at 24 different<br />

starting points but kept the location of<br />

the platform constant in an effort to enforce<br />

learning. After the third day of training, the<br />

mice were put to the test. While the Alzheimer’s<br />

mice were slow to reach the platform,<br />

Alzheimer’s mice treated with SAM demonstrated<br />

significant improvement, arriving to<br />

the platform only a few seconds behind the<br />

healthy mice.<br />

www.yalescientific.org<br />

Then, the researchers removed the platform<br />

from the pool and observed how the<br />

mice navigated the maze. The healthy mice<br />

spent a significant amount of time swimming<br />

around the zone that the platform used<br />

to be located, showing that they had successfully<br />

learned to correlate the general location<br />

of the platform with safety. In contrast, the<br />

Alzheimer’s mice swam around the pool at<br />

random, indicating that they didn’t “remember”<br />

where the platform should have been.<br />

Mice treated with SAM exhibited restored<br />

memory and spent a comparable amount<br />

of time in the platform zone to the healthy<br />

mice. Accordingly, SAM successfully rescued<br />

memory and learning deficits in Alzheimer’s<br />

mice.<br />

Fighting instinct<br />

"Another innate behavior of mice is that<br />

they scurry around the room and jump into<br />

dark holse in the corner," said Santiago Salazar,<br />

a graduate researcher in the Strittmatter<br />

Lab and second author of the paper. This<br />

natural behavior spurred the researchers to<br />

design another experiment to test the power<br />

of SAM in recovering the learning and<br />

memory aptitudes of mice. The researchers<br />

divided a chamber into a well-lit side and a<br />

dark side. On the dark side, they plated the<br />

floor with a surface that delivered a mild<br />

shock to the mice.<br />

The mice were placed in the light chamber<br />

for 90 seconds before the door separating<br />

the light and dark chambers was opened. As<br />

expected, the mice initially jumped through<br />

the hole to reach the dark chamber only to<br />

be greeted with an electric shock. Eventually,<br />

the healthy mice and the SAM-treated mice<br />

learned to associate the aversive shock with<br />

the dark chamber. In contrast, the Alzheimer’s<br />

mice were not able to make that association<br />

as successfully. “The Alzheimer’s mice<br />

forget that they’re going to get shocked and<br />

jump through the hole much quicker than<br />

the healthy mice type and the mice treated<br />

with SAM do," Salazar said.<br />

Strittmatter, Salzaar, and other lab members<br />

are currently at work to determine the<br />

ideal dose for SAM and the longevity of the<br />

drug’s effects. If all goes well, Strittmatter<br />

hopes to take SAM to clinical trials. Designing<br />

an effective Alzheimer’s drug has been<br />

difficult, partially because there is still a lot<br />

about the disease that we still do not know.<br />

Perhaps the ability of SAM to specifically target<br />

only abnormal protein interactions without<br />

interfering with normal mechanisms in<br />

the brain will yield more promising results.<br />

October 2017<br />

IMAGE COURTESY OF SANTIAGO SALAZAR<br />

►Amyloid beta plaques in the hippocampus of a mouse brain. Blue highlights cell nuclei and<br />

green highlights the plaques. Plaques like these interfere with cell-to-cell communication in<br />

synapses of the brain.<br />

ABOUT THE AUTHOR<br />

WILLIAM BURNS<br />

WILLIAM BURNS is a sophomore Neuroscience major in Morse College.<br />

He is the copy editor for the Yale Scientific Magazine and works in<br />

Professor Forscher’s lab studying the cytoskeletal dynamics underlying<br />

neurodegenerative diseases.<br />

THE AUTHOR WOULD LIKE TO THANK Professor Strittmatter and Santiago<br />

Salazar for their time, and for sharing their passion and dedication to their<br />

research.<br />

FURTHER READING<br />

Haas, Laura et al. “Silent Allosteric Modulation of mGluR5 Maintains<br />

Glutamate Signaling While Rescuing Alzheimer’s Mouse Phenotypes.” Cell<br />

Reports, vol. 20, no. 1, 5 July 2017, pp. 76–88.<br />

Yale Scientific Magazine<br />

19


IMAGE COURTESY OF THE SALTZMAN LAB<br />

►The nanoparticles synthesized in the<br />

Saltzman lab are optimized to be low in<br />

toxicity and are capable of delivering and<br />

releasing siRNA over a few weeks.<br />

Our immune system is the most<br />

useful protection we have from<br />

the world around us—until it<br />

turns on us. While the immune system<br />

is necessary to protect us from the invasion<br />

of harmful substances, it is also<br />

responsible for numerous rejections of<br />

organ transplants every year. One out<br />

of every four kidney recipients and almost<br />

half of all heart recipients experience<br />

an organ rejection within one year<br />

of transplant. When donor organs are<br />

so few and far between—20 people die<br />

each day waiting for a transplant, having<br />

your body attack your newly transplanted<br />

organ would be unfortunate.<br />

However, scientists are working on<br />

various methods of preventing transplant<br />

rejections. One team, led by<br />

Yale professors Mark Saltzman and<br />

Jordan Pober, is attempting to sneak<br />

past the checks of the immune system.<br />

They accomplished this by removing<br />

the tags on transplanted organs<br />

that immune cells recognize<br />

and react violently to. To achieve invisibility,<br />

they deliver small interfering<br />

RNA (siRNA) to the tissue using<br />

nanoparticle vehicles. Astonishingly,<br />

human arteries pretreated with siR-<br />

NA-loaded nanoparticles exhibited<br />

an 80 percent decrease of the tag. The<br />

results are promising for lowering rejection<br />

rates of various types of organ<br />

transplants, including the most common—the<br />

kidney.<br />

Hiding from ourselves<br />

There are currently many approaches<br />

to dampening the damaging response<br />

of the immune system to transplanted organs.<br />

Some groups tackle this challenge by<br />

improving immunosuppressive therapy, a<br />

treatment in which patients take drugs that<br />

reduce the strength of the body’s immune<br />

system. But since immunosuppression cripples<br />

the entire immune system , it also leads<br />

to increased risks of infections and malignancies.<br />

There are alternative approaches,<br />

such as the modification of the graft after<br />

the transplant operation to reduce its ability<br />

to activate the immune system.<br />

Saltzman and Pober are looking at this<br />

problem in a different way. Instead of<br />

tackling the immune system itself, they<br />

want to alter the transplant so that it is<br />

invisible to the immune system. If the<br />

transplant isn’t recognized as alien material,<br />

then the immune system wouldn’t<br />

have any reason to reject it.<br />

Cells of the immune system monitor the<br />

blood for foreign agents, much like guards<br />

at a watchtower looking out for intruders.<br />

All is peaceful—until the immune system<br />

detects a protein called class II major histocompatibility<br />

complex (MHC II), found<br />

on the surface of the transplant’s endothelial<br />

cells. The memory T cell, a type of immune<br />

cell, attaches to MHC II, activating<br />

and alerting the other parts of the immune<br />

system to the foreign tissue and triggering<br />

system-wide inflammation in response.<br />

www.yalescientific.org


molecular biology<br />

FOCUS<br />

However, if researchers can somehow remove<br />

the MHC II proteins that alert the immune<br />

system to false danger, the transplanted<br />

organ may be able to avoid attack. To reduce<br />

the amount of MHC II proteins displayed, researcher<br />

can deliver molecules called small interfering<br />

RNA (siRNA) to the transplant. Normally,<br />

the MHC II proteins are translated, or<br />

produced, using a messenger RNA (mRNA) as<br />

template. mRNA is a direct copy of DNA, our<br />

genetic blueprint, and serves as the instructions<br />

for making any type of protein. siRNA<br />

works by dismantling mRNA and preventing<br />

mRNA from participating in translation and<br />

thus protein production. However, the delivery<br />

of siRNA to the transplant’s endothelial cells, a<br />

cell type lining the interior surface of blood<br />

vessels, poses some complications. Not only is<br />

siRNA unstable, but it also has a hard time permeating<br />

the cell membrane.<br />

Scientists have attempted to overcome these<br />

challenges by engineering a special delivery vehicle<br />

for these siRNA molecules. While these<br />

approaches work in test tube conditions, the<br />

highly charged delivery vehicles are often cytotoxic<br />

inside living organisms. Furthermore,<br />

they only hold and release siRNA for up to three<br />

days, while the transplant is susceptible to rejection<br />

up to several weeks after the operation.<br />

Saltzman and Pober developed a biodegradable<br />

nanoparticle called PACE that is able to get<br />

the job done. Not only does it hold more siRNA,<br />

thereby lengthening the treatment duration, but<br />

the nanoparticle also demonstrates long-lasting<br />

effects on MHC II reduction in test tubes and<br />

in living cells, without damaging the cells. The<br />

siRNA is released over a longer period of time<br />

for increased effectiveness. The resulting reduction<br />

of MHC II on the surface of endothelial<br />

cells prevents the activation of memory T cells.<br />

Without their activation, the rest of the immune<br />

system cannot further damage the endothelial<br />

cells of the transplanted organ.<br />

Furthermore, the timing of PACE delivery<br />

in relation to the transplant operation gives the<br />

approach an advantage. Before a transplant operation,<br />

the donor organ is kept perfused with<br />

blood ex vivo, that is, outside the body, using a<br />

machine. The perfusion system preserves the<br />

quality of the organ. “We recognized that if the<br />

organ is ex vivo, being perfused with blood,<br />

there is an opportunity for treatment during<br />

this period,” Saltzman said.<br />

Thus, instead of injecting the patient<br />

post-operatively with siRNA-loaded nanoparticles,<br />

healthcare providers may be able to first<br />

treat the donor organs with nanoparticles and<br />

then carry out a transplant procedure.<br />

Tuning the nanoparticles<br />

PHOTOGRAPH BY JARED PERALTA<br />

►Endothelial cells are being treated with<br />

drug-loaded nanoparticles.<br />

There are many characteristics and features<br />

of a nanoparticle that can be optimized. “Toxicity,<br />

how readily they are taken up [by endothelial<br />

cells], how slowly they release siRNA,<br />

and how much siRNA they can carry are all<br />

important,” Saltzman said.<br />

To tackle the issue of cytotoxicity, Saltzman<br />

and Pober looked at adjusting the amount of<br />

the molecule 15-pentadecanolide (PDL), a<br />

component of the PACE nanoparticles. PDL<br />

content had been previously shown to decrease<br />

cytotoxicity by neutralizing the surface<br />

charge of the nanoparticles. Nanoparticles<br />

are typically positively charged, and highly<br />

charged particles inside cells can be deadly to<br />

them. To assess these effects, they tested siR-<br />

NA-loaded PACE nanoparticles of different<br />

PDL composition, ranging from 50 to 90 percent,<br />

for cytotoxicity. As expected, very high<br />

levels of PDL in the composition of the PACE<br />

nanoparticles greatly reduced the cytotoxicity.<br />

However, extremely high levels of PDL<br />

in the composition of the PACE nanoparticles<br />

severely hindered the ability of the PACE<br />

nanoparticles to deposit siRNA and suppress<br />

MHC II. As a compromise, Saltzman and Pober<br />

chose a PACE nanoparticle with 70 percent<br />

PDL composition, which greatly reduced<br />

cytotoxicity and still suppressed more than 90<br />

percent of MHC II.<br />

The potential of their nanoparticle delivery<br />

system shines through in several key experiments.<br />

The team pretreated human arteries<br />

with siRNA-loaded nanoparticles for six<br />

hours ex vivo. They then transplanted these<br />

treated arteries into mice without functioning<br />

immune systems, so the arteries would be accepted.<br />

This led to remarkable results, reducing<br />

MHC II expression in the transplanted arteries<br />

for up to six weeks after the transplant.<br />

“We tuned the nanoparticles to slowly release<br />

the siRNA,” Saltzman said. They also demonstrated<br />

that when an ex vivo siRNA-treated<br />

human artery was transferred from a donor<br />

mouse to a recipient mouse, the artery was<br />

protected from T cell infiltration, meaning<br />

that in a mouse model, at least, the arteries<br />

could be transplanted without rejection.<br />

These findings have many future implications<br />

for organ transplant success. Already,<br />

the research team has shown the successful<br />

delivery of nanoparticles without siRNA into<br />

kidneys ex vivo. “We are initially proposing<br />

to test the benefits of reducing expression of<br />

the molecules most strongly targeted by the<br />

recipient's immune system,” said Pober. After<br />

this step, the scientists may move on to<br />

further trials to test the new technology on<br />

organ transplants. With 25,000 organs transplanted<br />

a year, the reduction of organ rejection<br />

would improve the lives of many thousands<br />

for the better.<br />

ABOUT THE AUTHOR<br />

CHERYL MAI<br />

CHERYL MAI is a junior Molecular Biophysics & Biochemistry major in<br />

Davenport College. She is the Special Sections Editor for the Yale Scientific<br />

Magazine and works in Professor Ruslan Medzhitov’s lab studying the function<br />

and regulation of sleep.<br />

THE AUTHOR WOULD LIKE TO THANK Professor Saltzman for his time<br />

and insights.<br />

FURTHER READING<br />

Kseniya Gavrilov and W. Mark Saltzman. 2012. “Therapeutic siRNA:<br />

Principles, Challenges, and Strategies.” Yale J Biol Med. 85(2): 187-200.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

21


FOCUS<br />

organic chemistry<br />

PUTTING A PATCH ON RESISTANCE:<br />

A total synthesis of pleuromutilin opens the door to new long-lasting antibiotics<br />

by SAM BERRY | art by JASON YANG<br />

You’ve heard the news—bacteria have been quickly evolving immunity to the<br />

antibiotics that we’ve long used to fight them off, and diseases that are now<br />

easily treatable could come back in full force. We desperately need new<br />

antibiotics—yet few pharmaceutical companies are willing to invest in developing<br />

a product that natural selection will so quickly render obsolete.<br />

That’s why a team of Yale chemists,<br />

including chemistry professor Seth<br />

Herzon, postdoctoral associate Stephen<br />

Murphy and graduate student Mingshuo<br />

Zeng, have come up with a new way to<br />

synthesize the antibiotic pleuromutilin,<br />

known to slow the process of bacterial<br />

resistance. It isn’t the antibiotic itself<br />

that was significant about the discovery—it’s<br />

been around for decades—but<br />

rather the process of creating it, which<br />

potentially will allow scientists to create<br />

more potent forms of the drug. By devising<br />

a new synthetic scheme for pleuromutilin,<br />

they have opened the doors<br />

for a new world of potential antibiotics<br />

that can provide us new weapons in the<br />

fight against resistance.<br />

Our antibiotic world<br />

We’ve all grown up in an antibiotic<br />

world. Ever since Alexander Fleming’s<br />

accidental discovery of penicillin in<br />

1927, lifespans have grown longer and<br />

many diseases across the developed<br />

world have been all but eradicated. But<br />

even as antibiotics have changed the<br />

way that we live our lives, a new threat<br />

has begun to emerge. Rampant antibiotic<br />

use promotes the survival and<br />

proliferation of bacteria with special<br />

mutations in their genes that<br />

render antibiotics ineffective.<br />

Over time, more and<br />

more strains of antibiotic-resistant<br />

bacteria begin<br />

to appear as only those with<br />

protective mutations survive rounds of<br />

antibiotic treatment.<br />

Though we desperately need<br />

new antibiotics to counter the rising<br />

threat of resistance, this isn’t<br />

happening. There are fewer and<br />

fewer new antibiotics being developed.<br />

“The reason that we<br />

have so much resistance and<br />

no new drugs is this really<br />

weird, twisted conflation<br />

of economics and<br />

evolution,” Herzon said.<br />

Antibiotics aren’t like<br />

other drugs: patients<br />

usually only use them<br />

for short periods of time, and existing<br />

ones are, in Herzon’s words, “dirt cheap.”<br />

And worst of all, antibiotics often last<br />

only a few years before resistance develops<br />

in bacteria, making the actual lifetime<br />

of an antibiotic much shorter than<br />

that of other drugs.<br />

“The result is that there’s a terrible<br />

downward economic pressure not to do<br />

this,” Herzon said of antibiotic development.<br />

And while funding has increased<br />

slightly for small biotech startups in the<br />

past few years, far more new antibiotics<br />

are needed than are currently being<br />

made.<br />

A longer-lasting antibiotic<br />

In 1951, researchers at Columbia first<br />

isolated a powdery white substance from<br />

the edible mushroom Pleurotus mutilis<br />

and showed that it had antibacterial<br />

properties, naming it pleuromutilin. It<br />

was not until 2007, just as the antibiotic-resistant<br />

bacterial strain MRSA was<br />

making a global appearance in a major<br />

outbreak, that the very first antibiotic<br />

derived from pleuromutilin, retapamulin,<br />

was approved for human use. But retapamulin<br />

is different from other antibiotics:<br />

as of 2014, no bacterial resistance<br />

against the drug has emerged during the<br />

seven years that it’s been on the market.<br />

This is a much longer lifespan than the<br />

couple of years most antibiotics last before<br />

emerging resistance makes them<br />

obsolete.<br />

This key property is likely a result of<br />

a special mechanism of action unique<br />

to this compound. Pleuromutilin and<br />

its derivatives function by binding to an<br />

important region of the ribosome, the<br />

www.yalescientific.org


organic chemistry<br />

FOCUS<br />

key player in cellular protein synthesis.<br />

This region is essential to bacterial life<br />

and its sequence is common to almost<br />

all species of bacteria, making it especially<br />

difficult for bacteria to evolve resistance.<br />

Because of this, pleuromutilins<br />

are unique among classes of antibiotics<br />

in in that they treat disease while barely<br />

fostering resistance.<br />

Modifying in the middle<br />

Scientists have been exploiting this<br />

feature of pleuromutilin derivatives by<br />

developing a large number of variants<br />

through a process known as semisynthesis.<br />

Unlike a total synthesis, in which the<br />

product is made from scratch, semisynthesis<br />

involves starting with chemical<br />

compounds isolated from natural sources<br />

and then chemically modifying them.<br />

In this case, the process has involved<br />

taking purified natural pleuromutilin<br />

and typically modifying it at single carbon<br />

atom, called C14 because of its position<br />

in the molecule. C14 is attached to<br />

a chemical group that is easily removed<br />

and replaced with other, customized<br />

groups that afford the chemical new<br />

functionality without resorting to complex<br />

synthetic methods. More than 3,000<br />

derivatives have been made by changing<br />

the chemical groups attached at only<br />

that position.<br />

However, there are many more positions<br />

that could theoretically be modified.<br />

A study showed that changing the<br />

arrangement of chemical groups at a<br />

different carbon atom, C12, can afford<br />

the molecule some degree of efficacy<br />

against a group of harder-to-treat bacteria.<br />

These bacteria are distinguished by<br />

their outer membranes and are harder to<br />

treat because this membrane blocks the<br />

uptake of many traditional antibiotics,<br />

including C14-modified pleuromutilins.<br />

While C12 modifications are theoretically<br />

possible by semisynthesis, they are<br />

much harder to access, and many other<br />

positions that could also potentially afford<br />

a wider range of effects cannot be<br />

accessed at all.<br />

“As a chemist, there’s an obvious hole<br />

here,” Herzon said. “What we don’t have<br />

is a way to modify the other positions<br />

in the molecule.” What they needed was<br />

not pleuromutilin, but a way to make<br />

pleuromutilin, and therefore a way to<br />

vastly broaden the substance’s antibiotic<br />

potential.<br />

Starting from scratch<br />

Expanding the number of possible<br />

pleuromutilin derivatives required the<br />

researchers to start from the very beginning.<br />

“What we’re doing is a total synthesis<br />

of pleuromutilin from scratch,”<br />

said Stephen Murphy, who spearheaded<br />

much of the technical work on the project.<br />

“That gives us total control of the<br />

compound, so we can explore a broader<br />

chemical space.”<br />

The individual steps of the synthesis<br />

can be modified to yield a wide variety<br />

of different products. The full synthesis<br />

process involves a total of seventeen<br />

steps, transforming easily purchased<br />

chemicals into the final product, pleuromutilin.<br />

But synthesizing the product<br />

isn’t the real goal. “What we really<br />

care about is making pleuromutilin-like<br />

structures,” Herzon said. “With this synthesis<br />

in place, we’ve identified the viable<br />

pathways to these structures.” Now<br />

that the researchers have the essential<br />

blueprints to build this molecule, they<br />

can make modifications to any part of it<br />

at any stage of the building process.<br />

They first reported their synthesis in<br />

June and, in the same report, demonstrated<br />

that the final steps of the synthesis<br />

could be easily modified to create<br />

pleuromutilin derivatives with a flipped<br />

arrangement about C12. The total synthesis<br />

allows for many more options to<br />

make antibiotic derivatives that have<br />

never been explored. Because they can<br />

IMAGE COURTSY OF WIKIMEDIA COMMONS<br />

►Antibiotic-resistant bacteria like methicillinresistant<br />

Staphylococcus aureus (MRSA),<br />

shown, provide an immediate threat to public<br />

health.<br />

begin making modifications from earlier<br />

intermediates in the process rather than<br />

working backwards from the finished<br />

product, they can now access synthetic<br />

routes to pleuromutilin-like molecules<br />

that are modified in ways that would<br />

be impossible by semisynthesis, with<br />

functions that have never been studied.<br />

Members of the Herzon Lab are well on<br />

their way to testing these new molecules<br />

for improved antibiotic properties.<br />

“Right now we’re shooting around<br />

in the dark,” Herzon said, describing<br />

the search for the next effective pleuromutilin-based<br />

antibiotic. “Once we<br />

find something more active, we can drill<br />

down and further refine structure with<br />

more precision.”<br />

ABOUT THE AUTHOR<br />

SAM BERRY<br />

SAM BERRY is a junior in Ezra Stiles majoring in Molecular Biophysics &<br />

Biochemistry. He works in the in the lab of Alanna Schepartz, where he<br />

studies protein delivery by cell-permeant miniature proteins.<br />

THE AUTHOR WOULD LIKE TO THANK Professor Herzon and Dr. Stephen<br />

Murphy for their time and enthusiasm when discussing their research.<br />

FURTHER READING<br />

Murphy, Stephen K., Mingshuo Zeng, and Seth B. Herzon. “A Modular and<br />

Enantioselective Synthesis of the Pleuromutilin Antibiotics.” Science 356, no.<br />

6341 (June 6, 2017).<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

23


alumni<br />

FEATURE<br />

ESSAY<br />

CONTEST<br />

BREAKING THROUGH OCEAN ACIDIFICATION<br />

►BY CLARA BENADON from poolesvile high school<br />

As a Marylander, one of my favorite things to do is to make the<br />

trek up to the Chesapeake Bay. Its sparkling waters and abundant<br />

wildlife make it a prime jewel of the East Coast. Nothing can compare<br />

to the experience of paddling down the Potomac River on a<br />

sunny day, the boughs of a sycamore arching overhead.<br />

Apart from being a stunner, the Bay provides major cultural and<br />

economic benefits. Its unique way of life is perfectly encapsulated<br />

in the small towns of Smith Island, where watermen make a living<br />

from the estuary’s riches. On a recent visit, one local said, “We<br />

truly build our lives around the water.” From the individual fisherman<br />

to larger commercial operations, the Chesapeake provides<br />

$3.39 billion annually in seafood sales alone—part of a total economic<br />

value topping $1 trillion.<br />

The stability of these waters is endangered by growing ocean<br />

acidification due to absorption of carbon dioxide from the atmosphere.<br />

Acidification disintegrates the protective carbonate coverings<br />

of shellfish, killing off large amounts of oysters, mussels, and<br />

scallops. Without a thriving population of oysters, which filter the<br />

Bay, harmful pollutants run rampant. Acidity also causes low oxygen<br />

levels, hindering fish respiration. Even with survivable oxygen<br />

levels, low pH can be fatal for fish.<br />

The plummeting numbers of these Chesapeake staples make a<br />

dent on the economy. According to the Chesapeake Bay Foundation,<br />

Maryland and Virginia have suffered losses exceeding $4<br />

billion over the last three decades stemming from the decline of<br />

oyster health and distribution. High acidity stunts oyster growth,<br />

and shellfish fisheries cannot profit from the smaller shells.<br />

The losses aren’t economic alone. An estimated 2,700 species<br />

call the Bay their home, and the loss of even one species causes<br />

a ripple effect through the entire food web. According to a 2004<br />

study in Science, the survival of threatened and nonthreatened<br />

species is closely intertwined. Moreover, biodiversity keeps in<br />

check the amount of carbon dioxide in any body of water. Now,<br />

zooming out from the Chesapeake Bay, skyrocketing acidity is<br />

present in almost every aquatic biome on our planet. When pH<br />

is low, coral reefs cannot absorb the calcium carbonate that makes<br />

up their skeleton. Corals—along with snails, clams, and urchins—<br />

disintegrate. A particularly disturbing image of ocean acidification<br />

is its effect on the neurology of fish. Their decision-making skills<br />

are significantly delayed to the level where they sometimes swim<br />

directly into the jaws of predators.<br />

Economically, the UN estimates that ocean acidification will<br />

take a $1 trillion bite out of the world economy by the year 2100.<br />

This massive cost has direct human implications, including its<br />

harm on health, job security, and cultural heritage. In addition,<br />

the economies of many countries are wholly dependent upon reef<br />

-based tourism and other activities built around the water.<br />

We need a solution to our world’s rapidly acidifying oceans;<br />

solving this problem would be beneficial on an unprecedented<br />

scale. Methods that at first appeared brilliant have either been limited<br />

by their feasibility or have been rejected due to their negative<br />

side effects, ultimately prolonging the search for a solution.<br />

The method of dumping significant iron sulphate into the water<br />

is based on the principle that iron fertilizes phytoplankton, or<br />

microscopic organisms found in every body of water. The energy<br />

phytoplankton gain from the iron allows them to bloom, absorbing<br />

CO 2<br />

from the atmosphere and the ocean. When the phytoplankton<br />

die, they sink to the bottom of the ocean, locking the<br />

CO 2<br />

there for centuries. In 1988, the late oceanographer John<br />

Martin proclaimed, “Give me a half tanker of iron, and I will give<br />

you an ice age.” It is theorized that fertilizing two percent of the<br />

Southern Ocean could set back global warming by ten years.<br />

Why not implement this magic fix? A 2016 study in Nature determined<br />

that the planktonic blooms would deplete the waters of<br />

necessary nutrients. Additionally, when the large bloom dies, it<br />

would create large “dead zones,” areas devoid of oxygen and life.<br />

Side effects aside, this technique may be entirely ineffective. Carbon<br />

dioxide may simply move up the food chain when the phytoplankton<br />

are eaten and be respired back into the water. This was<br />

observed when the 2009 Lohafex expedition unloaded six tons of<br />

iron off the Southern Atlantic.<br />

Alternatively, planting kelp is less drastic. Revitalizing expansive<br />

forests of algae has proven to be effective in sucking up underwater<br />

CO 2<br />

. Kelp grows as quickly as 18 inches a day, provides a habitat<br />

for marine species, and removes nutrient pollution. Researchers<br />

from the Puget Sound Restoration Fund, who have been monitoring<br />

the capability of this process, have found that kelp forests are<br />

effective at diminishing acidification on a local scale. While planting<br />

carbonsucking species across the ocean would not be a feasible<br />

global solution, kelp forests could help solve the acidification crises<br />

found in less expansive areas.<br />

To date, there is no straightforward fix to combat ocean acidification.<br />

If a scientific breakthrough were to occur, it would perhaps<br />

be comprised of a combination of methods. However, as science<br />

continuously evolves, the key to deacidifying our oceans may well<br />

turn out to be something beyond our wildest dreams.<br />

24 Yale Scientific Magazine October 2017 www.yalescientific.org


genetics<br />

FEATURE<br />

NATURE AND NURTURE<br />

Examining genetic and environmental factors<br />

►BY ANNA SUN<br />

IMAGE COURTESY OF PIXABAY<br />

►This study’s enormous computational power unlocks new<br />

genetic and environmental connections between diseases.<br />

Imagine you have a factory that produces flaky concrete and<br />

that this concrete is shipped to construction sites across the city.<br />

You discover that one building has a crack in the roof, another<br />

has a crack in the walls, and a third has a crack in the basement.<br />

All of these incidents can be traced back to the brittle concrete<br />

produced by this one factory. Now imagine that, another day,<br />

an earthquake causes similar cracks in buildings made from<br />

smooth, unblemished concrete. What’s the difference? In this<br />

analogy, the first scenario represents the genetic contributions<br />

to diseases, while the second represents the environmental factors<br />

that affect diseases.<br />

This is how Andrey Rzhetsky, a researcher at the University<br />

of Chicago, explains the different contributions of genetics<br />

and the environment to diseases. In the past, disease classifications<br />

were largely arbitrary, based mostly on vaguely similar<br />

features or even cultural similarities. However, in a study<br />

published in Nature Genetics in August 2017, researchers from<br />

the University of Chicago, Microsoft Research, and Vanderbilt<br />

University developed a largely computational method of reclassifying<br />

diseases based on their shared genetic and environmental<br />

correlations. By analyzing data from over one-third of the<br />

U.S. population, they discovered that complex diseases might<br />

be unexpectedly more or less interconnected than we previously<br />

thought.<br />

Diseases that can be traced back to a variation in a single gene<br />

are classified as Mendelian disorders, whereas those affected by<br />

both genetic and environmental factors are complex disorders.<br />

The researchers examined vast amounts of health insurance<br />

data, ultimately deriving genetic and environmental correlations<br />

for a wide range of 29 complex diseases—including bipolar<br />

disorder, type I diabetes mellitus, asthma, migraine, irritable<br />

bowel syndrome, and cystitis/urethritis. Individual data were<br />

then grouped by families to better determine how diseases can<br />

occur non-randomly. The researchers used statistical models to<br />

estimate genetic correlations, which suggested that even seemingly<br />

unrelated complex diseases share some similarities.<br />

According to Rzhetsky, if the genetic correlation for a certain<br />

disease is low, the contribution must be mostly environmental.<br />

“This is good because we can prevent the diseases by putting<br />

patients away from the responsible environmental factors,”<br />

Rzhetsky said. For better visualization of their findings, the researchers<br />

transformed their correlations into relative distances<br />

in a refined tree model to portray the inherent similarities and<br />

differences between the selected diseases. Diseases with shorter<br />

branches in the model were more correlated and thus more<br />

similar than those separated by longer branches.<br />

As a result of the reclassification, the researchers were able to<br />

easily identify new, unexpected relationships between the complex<br />

diseases. For instance, a migraine, previously assumed to<br />

be similar to neuropsychiatric conditions, was found to have a<br />

closer distance to general immune system diseases, such as irritable<br />

bowel syndrome and cystitis/urethritis.<br />

Hallie Gaitsch, an undergraduate researcher from Yale University<br />

who also worked on this study, best portrays the significance<br />

of the research with the following scenario: no matter<br />

how many patients from different families with different genetic<br />

diseases visit a one doctor, that doctor would still not be able<br />

to grasp the links between the familial genetic and environmental<br />

contributions of these diseases. The possible onset of a<br />

complex disease could significantly increase with another related<br />

one, but comorbidity in patients could be masked for years<br />

since some diseases are assumed to have such vastly different<br />

causes and origins. Only with the enormous computational<br />

power utilized in this study would these unforeseen similarities<br />

between apparently different diseases come to light.<br />

An important implication is the potential use of pre-existing<br />

tools and treatments to treat similar diseases or perhaps even<br />

the discovery that a drug might have adverse effects on another<br />

disease. This research establishes a strong foundation in its<br />

interpretation of the intertwined relationship between genetics<br />

and the environment on complex diseases.<br />

“Our work is a hypothesis-generator for other scientists to<br />

use what we’ve shown statistically in going forth with their own<br />

experiments and seeing these new relationships,” Gaitsch said.<br />

She believes that future research could not only more deeply<br />

analyze the interconnectedness of certain diseases, but also test<br />

the efficacy of treatments on these closely related diseases.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

25


FEATURE<br />

geology and geophysics<br />

ARCTIC OCEAN CRATERS<br />

Sudden methane release linked to ice sheet retreat<br />

►BY THEO KUHN<br />

In the depths of the arctic Barents Sea, north of the northernmost<br />

point of Norway, the seafloor is pocked with large craters and<br />

mounds. These craters don’t have their origins in fire, but in ice. Researchers<br />

believe that they formed due to the sudden breakdown<br />

of gas hydrate—an ice-like substance found both in seafloor sediments<br />

and on land, which can rapidly transition from a solid to<br />

a gas. New research led by Karin Andreassen at the University of<br />

Norway suggests that these craters released great expulsions of gas<br />

at the end of the last Ice Age. Furthermore, the study concludes that<br />

receding ice sheets were a primary factor in drawing these gases—<br />

which mostly consist of methane, a powerful greenhouse gas—out<br />

of the seafloor and potentially releasing them into the atmosphere.<br />

Gas hydrates are substances that form when hydrocarbon gases<br />

interact with water and sediment under the appropriate conditions<br />

and crystallize. Though they are relatively simple compounds,<br />

the factors that control their stability—determining whether they<br />

remain solid or decompose into gas and liquid water—are surprisingly<br />

complex. The most important factors are pressure and temperature;<br />

the pressure must be high and the temperature must be<br />

low for the hydrate to remain solid.<br />

To develop a model for the origin of the craters, Andreassen’s<br />

team combined their observations of the seafloor with a chemical<br />

analysis of the hydrates and a model of the physical conditions that<br />

would have existed in their study area over the past 30,000 years.<br />

Their findings paint a picture in which crater formation was the direct<br />

result of deglaciation.<br />

Near the end of the last Ice Age, which occurred 17,000 years<br />

ago, more than a kilometer of solid ice lay on the seafloor at the<br />

study location. Underneath it, there was a large zone with the optimal<br />

conditions for methane hydrate formation. But within two<br />

thousand years, that ice sheet receded from the area due to the<br />

warming climate. “It happened quite fast,” Andreassen said.<br />

The loss of such a large mass of ice caused a sudden change in<br />

pressure, and, to a lesser extent, temperature, that shrank the zone<br />

in which methane hydrates were stable. As methane hydrates became<br />

unstable and decomposed, the newly-released gas collected<br />

underneath a shrinking cap of solid hydrate. Eventually, the high<br />

concentration of gas and hydrate in a dwindling area caused the<br />

seafloor to distend and fracture, further lowering the pressure and<br />

allowing the remaining hydrates to suddenly decompose. The<br />

scars of this runaway process are the craters that remain to this<br />

day.<br />

Andreassen’s model is especially intriguing because it doesn’t<br />

just connect the sudden decay of gas hydrates to ice sheets; it also<br />

suggests that glaciation helps to form large quantities of gas hydrates<br />

in the first place. Methane typically leaks towards the surface<br />

from hydrocarbon deposits that lie in the bedrock far beneath<br />

the surface—the types of formations that can be drilled into for natural<br />

gas extraction. As an ice sheet advances over these area, its immense<br />

weight forces gas upwards and towards the surface where it<br />

can form gas hydrates, and as ice sheets recede, much of this hydrate<br />

then decomposes. “It’s like a big bulldozer going back and forth,”<br />

Andreassen said. “The ice will pump the gas up from the deeper<br />

reservoirs and into the shallow surface, where it can form gas hydrates.”<br />

Methane is 25 times more potent than carbon dioxide as a greenhouse<br />

gas. Methane is currently seeping from the seafloor, but most<br />

of it doesn’t reach the atmosphere—its slow release rate allows seawater<br />

to absorb it before it breaches the surface. In contrast, the<br />

events that formed the craters are believed to have occurred abruptly.<br />

Thus, these large quantities of gas released may have been able to<br />

bubble to the surface and enter into the atmosphere.<br />

Researchers hope to find traces of these events in atmospheric<br />

records that will help them determine if the events contributed to<br />

the change in climate that occurred at the end of the last Ice Age. In<br />

the short term, Andreassen’s team hopes to understand the extent<br />

of gas hydrates and craters across the Arctic Ocean, since large areas<br />

have experienced a similar glacial history and may have undergone<br />

the same process of hydrate production. By developing a clearer<br />

picture of the history of gas hydrates in Arctic areas, researchers<br />

may be able to predict how a similar process might unfold in the<br />

future—to find ice sheets that are quickly receding, one may need<br />

not look back fifteen millennia.<br />

IMAGE COURTESY OF WIKIMEDIA COMMONS<br />

► Methane hydrate from the seafloor near Oregon. Methane<br />

hydrate can be found within sediments and sedimentary rocks,<br />

primarily within the continental shelves of the world’s oceans.<br />

26 Yale Scientific Magazine October 2017 www.yalescientific.org


geology and geophysics<br />

FEATURE<br />

ΜICROBIAL DIVERSITY<br />

How environmental niches affect biological diversification<br />

►BY JIYOUNG KANG<br />

IMAGE COURTESY OF NIAID/RML<br />

►Researchers examined the diversification of microbes by<br />

studying rock formations that are millions of years old.<br />

Have you ever marveled at the diversity of life on Earth?<br />

Over the past 500 million years, the development of new<br />

environmental niches has played a huge role in the diversification<br />

of plants and animals. These macro-organisms<br />

have colonized new continents and expanded to various<br />

habitats, developing novel evolutionary adaptations that<br />

have led to radiation of species. But what about the microbes?<br />

Even though we cannot see them with our naked<br />

eye, they have evolved for a much longer period of time:<br />

the past four billion years. A team of researchers sought<br />

out a possible explanation for their diversification in ancient<br />

rocks.<br />

Led by Eva Stüeken at University of St. Andrews, the<br />

team investigated whether the diversification of microbial<br />

populations was also prompted by the expansion of<br />

environmental niches. They studied five formations in<br />

the Neoarchean lower Fortescue Group, located in Western<br />

Australia. The depositional settings possess distinct<br />

hydrogeologic properties, meaning that they differ in<br />

what elements they are made out of and whether water<br />

constantly flows in and out of the lakes. But they were all<br />

formed about three million years ago, which assures that<br />

any diversity in microbes observed by the researchers is<br />

due to the difference in habitats and not time. To assess<br />

the biological and geological properties of the formations,<br />

they gathered samples from drill cores and measured<br />

traces of different elements.<br />

This allowed the researchers to identify the type of<br />

bacteria that lived in the region. When some bacteria<br />

eat, they change the type of carbon left in the environment,<br />

while other bacteria change the type of sulfur. If<br />

researchers found more of a certain type of carbon or<br />

sulfur, then they would be fairly certain that the corresponding<br />

bacteria was once in the area.<br />

After taking these measurements, the researchers concluded<br />

that geographic and hydrologic parameters indeed<br />

impact the diversification of microbes. For example,<br />

Mt. Roe Formation is made out of basaltic rocks that<br />

can react with water to generate methane. The researchers<br />

examined the site’s carbon isotope values and inferred<br />

the presence of methanotrophs—bacteria that use<br />

methane as their main source of energy. Similarly, they<br />

found evidence of different metabolisms present in other<br />

formations with various environmental properties. This<br />

is because distinct minerals release different types of elements,<br />

which shape the biological pattern of microbial<br />

populations as the organisms evolve to utilize different<br />

nutrients.<br />

“It is a pretty new concept that environmental diversification<br />

may have triggered microbiological diversification”,<br />

Stüeken said. In fact, the implications of the research<br />

extend even to astrobiology, in the effort to find<br />

extraterrestrial life. Based on the data, it may be important<br />

to focus on planets with diverse environments. “If<br />

you have a planet that only has a big ocean and no land<br />

masses, then it would perhaps be much more difficult to<br />

develop a diverse biosphere,” Stüeken said. Since there<br />

are fewer niche spaces, there would also be fewer types<br />

of bacteria.<br />

Naturally, there were challenges that followed the study<br />

of ancient rocks. The scientists had to work with the possibility<br />

that the rocks could have been metamorphosed<br />

from the exposure to great heat and pressure over the<br />

years, potentially skewing the data. Gathering samples<br />

was also difficult, as they are quite weathered and scarce.<br />

Still, the researchers hope to further the study by analyzing<br />

older and younger rocks with a similar method<br />

by comparing marine rocks to non-marine rocks. This<br />

work opens a new window into the field and may be an<br />

important step towards acknowledging the effect of environmental<br />

diversity on biological diversity.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

27


FEATURE<br />

astronomy<br />

S T A R R Y<br />

FUEL TANKS<br />

What fuels the<br />

starburst phases<br />

of galaxies?<br />

A long time ago, in galaxies far, far away, stars were<br />

churned out at unprecedented rates; over 100 solar masses<br />

were produced annually. These luminous, dusty starburst<br />

galaxies were 1000 times more common in the very early<br />

universe than they are today. But the light from these stars<br />

is only now reaching the earth, ending its multi-billion year<br />

journey through the cosmos as it reaches our telescopes.<br />

By looking out into the universe and back into the past, we<br />

gain a better understanding of how these starburst galaxies<br />

formed and how they sustained such rapid rates of star<br />

production.<br />

In the early universe, galaxies differed in their abilities to<br />

produce stars. Many of the galaxies at this time were unable<br />

to support such rapid star formation, as the process blasted<br />

away the hydrogen gas, preventing the creation of future<br />

stars. Yet others continued to produce hundreds of stars per<br />

year for periods up to 100 million years, long after other<br />

galaxies had subsided. Why did some galaxies remain fertile<br />

while others died down? Recent research suggests that we<br />

can find the answer with the help of CH+, a rare but useful<br />

cation (a positively-charged, electron-deficient molecule).<br />

Using the Atacama Large Millimeter Array (ALMA)<br />

radio telescopes in Northern Chile, astronomers studied six<br />

early starburst galaxies. They found that CH+ is abundantly<br />

present in all of them, providing insight into what set these<br />

galaxies apart from the others. CH+ is very rare and was<br />

only discovered in 1941, as it can only form in extremely cold<br />

temperatures of about 20 Kelvin and with extremely high<br />

energy inputs equivalent to 4000 Kelvin. With such a high<br />

by ELIZABETH RUDDY || art by SONIA RUIZ<br />

energy requirement, CH+ must be formed in the presence<br />

of strong ultraviolet radiation or mechanical energy. It also<br />

has an extremely short lifespan, so it cannot be transported<br />

far. Therefore, its presence in these galaxies suggests that they<br />

must have recently undergone violent energy shocks.<br />

“CH+ traces how energy flows in a galaxy. Think of plankton<br />

fluorescence which is excited by little shocks that generate<br />

turbulence in the water. When you throw a stone in, you<br />

light up trails of fluorescent plankton,” said Edith Falgarone<br />

of the École Normale Supérieure and Observatoire, where<br />

the research was conducted.<br />

Falgarone and her team of researchers studied the emission<br />

and absorption spectral lines of this CH+ cation in samples<br />

from the six starburst galaxies. Each spectral line emitted<br />

by the interstellar gas corresponds to a different compound<br />

present in the galaxy, so the CH+ spectral lines can tell us a<br />

lot about how the CH+ was formed. The lines revealed the<br />

presence of extremely turbulent hydrogen gas surrounding<br />

the galaxies, extending far outwards from the cores where<br />

stars form.<br />

The discovery of this turbulent gas elucidates how galaxies<br />

grow and how these star-forming engines are fueled.<br />

The width of the CH+ lines are broader than 1000 kilometers<br />

per second, suggesting that it was born in enormous<br />

shock waves. The researchers suggest that these motions<br />

are powered by energetic outflows originating in the core<br />

of the galaxy. These outflows exit the galaxy in such a way<br />

that leaves matter trapped within the galaxy’s gravitational<br />

pull. This culminates in vast, turbulent reservoirs of cool,<br />

28 Yale Scientific Magazine October 2017 www.yalescientific.org


low-density gas circling the galaxy, up to 30,000 light-years<br />

from the galaxy core.<br />

While it may seem that these high-powered inner-galactic<br />

winds would quench the starburst phase of the galaxy,<br />

slowing the rate of star production by blowing out much of<br />

the hydrogen gas needed to create new stars, the researchers<br />

suggest that the opposite is actually true. Transforming<br />

previous models for galaxy formation, Falgarone and her<br />

team propose that the winds actually extend the star-formation<br />

phase by feeding these vast reservoirs of “fuel” for future<br />

stars.<br />

“What we have found with CH+ is that this stellar feedback<br />

generates turbulence in the galactic environment, so energy<br />

is lost and the outward momentum of the gas is lost too.<br />

Indeed, most of the gas expelled from the galaxy eventually<br />

astronomy<br />

FEATURE<br />

gas reservoir surrounding the galaxy but would like to see<br />

some more data before making the conclusion that this<br />

reservoir is what prolongs the starburst phase. “I don’t see<br />

the jump personally between the data and the conclusions,”<br />

Arce said. “This is not to say that the results are invalid in<br />

any way —just that the beauty of the data could have perhaps<br />

been more fully presented in a longer piece.”<br />

Larson and Arce also both expressed excitement about<br />

what Falgarone’s work means for future research into star<br />

formation. We are currently seeing very exciting results<br />

coming from the ALMA telescopes. They observe in the<br />

millimeter and sub-millimeter wavelengths, so they’re useful<br />

for observing dust and molecular-level matter such as CH+.<br />

“This paper is an example of the great work people are doing<br />

with the ALMA telescopes,” Arce said.<br />

IMAGE COURTESY OF ESO<br />

►An artist’s impression of how cold hydrogen gas fuels star production in distant starburst galaxies. The turbulent gases that surround the<br />

galaxy extend far beyond outwards of the starbust core where stars are formed.<br />

falls back on it, feeding further star formation instead of<br />

quenching it,” Falgarone explains.<br />

The study has, however, raised a few questions among relevant<br />

academic circles. “The authors suggest that turbulence<br />

in the outlying molecular gas slows down its infall and prolongs<br />

star formation. To me, this seems plausible but unproven.<br />

I don’t see how the generation of strong turbulence would<br />

contribute to the fueling of a starburst; if anything, I would<br />

expect it to inhibit gas infall,” said Richard Larson of the Yale<br />

Astronomy Department.<br />

Yale professor Héctor Arce, who is currently researching<br />

star formation in the Milky Way galaxy, had similar questions<br />

about the data. According to Arce, the CH+ indicates<br />

massive outflows of gas from the center of the galaxies. He<br />

agrees that this likely means that these outflows feed a cool<br />

The work acknowledges that the mass outflow rates<br />

caused by the winds alone do not completely account for<br />

the extreme rates of star production. Something else, still<br />

unknown, is nourishing these reservoirs. Falgarone and her<br />

team suggest that perhaps this extra mass is produced by<br />

galactic mergers or possibly accretion from streams of gas<br />

that are sucked into the gravity of the galaxy.<br />

Falgarone’s work sheds light on questions that have been<br />

puzzling scientists for years—they have wondered, how did<br />

starburst galaxies come by their extra fuel? We may now<br />

have some answers, but the result also raises new questions.<br />

What causes the hot, violent winds at the centers of certain<br />

galaxies, powering the cool gas reservoirs? Why do some galaxies<br />

have them and others do not? Astronomers continue to<br />

scour the cosmos for answers.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

29


FEATURE<br />

molecular biology<br />

MOLECULAR SPEAK:<br />

How gut bacteria communicate with human cells<br />

by STEPHANIE SMELYANSKY |<br />

art by SUNNIE LIU<br />

More than 10,000 different bacterial species occupy the human<br />

body, outnumbering human cells ten to one. At first glance, those<br />

numbers may seem alarming, but fear not, we need these bacteria<br />

to live normal, healthy lives. These bacteria—from the ones in<br />

our stomachs to those on our skin—have a symbiotic relationship<br />

with us: we provide them with the nutrients they need to survive,<br />

and they help us digest food and defend our body against disease-causing<br />

agents. Our lives are so intertwined with these helpful<br />

bacterial species that the bacteria have perhaps learned how<br />

to speak the language of human cells. Researchers at Rockefeller<br />

University have identified gut bacteria that produce signaling<br />

molecules that can interact with human cells, opening the door<br />

for a wide variety of medical treatments for various gastrointestinal<br />

disorders.<br />

“The majority of FDA-approved drugs are either copies of, or<br />

inspired by, naturally-occurring compounds,” said Sean Brady,<br />

30 Yale Scientific Magazine October 2017 www.yalescientific.org


molecular biology<br />

FEATURE<br />

a professor at Rockefeller University whose lab conducted the<br />

study. According to Brady, bacteria are an incredibly common<br />

source for those compounds. “Most of our antibiotics and immunosuppressants<br />

come from looking at products produced by<br />

bacteria,” Brady said. Many of these drugs were found by searching<br />

soil bacteria to see if they produced molecules with antibiotic<br />

properties, which is one of the main focuses of Brady’s lab.<br />

However, in this project, the lab decided to screen the human microbiome—the<br />

set of bacteria that live within the human body—<br />

for molecules that might be able to interact directly with human<br />

cells. The crux of the idea is simple: these bacteria already live in<br />

our bodies, so researchers wanted to find out whether we can use<br />

these microbes to manipulate human physiology.<br />

To get there, the scientists had a challenge to overcome: searching<br />

through the thousands of bacterial species living in the human<br />

body to find just one or two that can “talk” to human cells.<br />

A few decades ago, researchers would have had to do so by hand,<br />

culturing every bacterial strain individually and testing it for active<br />

molecules. In the twenty-first century, however, this task is<br />

much more feasible. The researchers had already identified one<br />

bacterial molecule, commendamide, that interacted with a class<br />

of human cell receptors called G-protein coupled receptors (GP-<br />

CRs). Commendamide is an N-acyl amide, a type of biologically<br />

active organic molecule, that is often produced by bacteria. Taking<br />

advantage of this fact, the researchers used the Human Microbiome<br />

Project database to search for all of the bacterial genes<br />

in the human microbiome encoding proteins that can produce<br />

N-acyl amides like commendamide. They identified a total of 143<br />

human microbial genes that code for such proteins, of which 44<br />

were unique enough to merit testing in the lab for biological activity.<br />

The researchers determined that these 44 genes comprised<br />

six distinct classes of N-acyl amides that are naturally produced<br />

by bacteria, four of which are produced by gut bacteria.<br />

The bacterial molecules identified by the researchers are quite<br />

similar to signaling molecules already produced by the human<br />

body; their structures mimic human signaling molecules incredibly<br />

closely. Furthermore, the bacterial molecules bind to the<br />

GPCR receptors just as well as the human signaling molecules<br />

do, such that the physiological result of GPCR activation is indiscernible<br />

between the human and bacterial molecules. Brady<br />

hypothesizes that as we learn more about the chemistry of the<br />

human microbiome, we’re going to find more cases of bacterial<br />

mimicry in the future. “More and more often you’re going to find<br />

molecules that maybe aren’t identical to, but resemble, the molecules<br />

that we as humans already make to target our own receptors,”<br />

Brady said.<br />

Since the molecules these bacteria produce have such a strong<br />

effect on human physiology, the researchers decided to see if they<br />

could harness that ability for medical purposes. These commensal<br />

bacteria can interact with GPCRs, which is a lucky coincidence<br />

for researchers because GPCRs are implicated in a wide variety of<br />

metabolic disorders. These receptors are the largest and most diverse<br />

group of human cell receptors, and GPCRs make up about<br />

one third to one half of all drug targets are. Many GPCRs are<br />

located in the human gut as well, which is where the N-acyl-amide-producing<br />

bacteria were isolated from. In fact, GPCRs in the<br />

gut have been implicated in hunger, glucose absorption, and diabetes,<br />

which is exactly what the researchers decided to study.<br />

IMAGE COURTSY OF WIKIMEDIA COMMONS<br />

►An electron microscopy image of E. coli cells isolated from the<br />

human small intestine—just one of the many bacterial species living<br />

in the human gastrointestinal tract.<br />

The different N-acyl amide ligands were surveyed to see if they<br />

would bind to 240 different GPCRs. Of these, one GPCR in particular—denoted<br />

GPR119—bound the bacterial molecules particularly<br />

well. GPR119 is implicated in the regulation of glucose<br />

homeostasis, and has historically been a drug target for Type 2<br />

Diabetes. Specifically, activation of GPR119 receptors in the gut<br />

can prevent the rapid change of blood sugar levels in hyperglycemic<br />

patients. Brady and his team wanted to see that activation<br />

of GPR119 with the bacterial N-acyl amides could produce the<br />

same effects as human signaling molecules.<br />

To see if the bacterial molecules were capable of affecting<br />

GPR119 receptors strongly enough to regulate glucose levels, the<br />

researchers infected mice with E. coli engineered to produce the<br />

molecules of interest. They then measured the blood sugars of the<br />

mice. As predicted, the mice that were infected with N-acyl-amide-producing<br />

bacteria had lower blood sugar levels than mice<br />

that weren’t infected with the genetically engineered bacteria.<br />

This shows that some bacteria produce biologically active molecules<br />

that not only can interact with GPR119 in the gut, but can<br />

also regulate blood sugar in ways similar to many blood sugar<br />

medications.<br />

Like any recently published study, these findings still have a<br />

long way to go from the lab to becoming viable medical treatments<br />

for diseases like diabetes. According to Brady, human<br />

physiology is incredibly complicated, and people are still working<br />

on how to apply these findings in mice and cell culture models to<br />

the human body. But that doesn’t stop Brady and his team from<br />

thinking about ways that these bacterial molecules could be harnessed<br />

as a drug. He suggests that perhaps people can ingest the<br />

pure molecule just like any other drug. “You could also imagine<br />

introducing the organism regularly, like in a yogurt,” Brady said.<br />

This is perhaps more enjoyable than taking a pill! Whatever the<br />

mode of action, these commensal bacteria provide us with ways<br />

to manipulate the human body in new, inventive, and potentially<br />

less invasive ways in the hopes of providing new treatments for<br />

common diseases.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

31


FEATURE<br />

ecology<br />

Researchers led by James Tour of Rice University, Robert Pal of<br />

Durham University, and Gufeng Wang of North Carolina State<br />

University recently developed and tested nano-sized molecular<br />

machines that may hold the key to treating diseases through<br />

mechanical attacks that leave healthy cells unharmed. The original<br />

design for these molecular motors was based on work by Nobel laureate<br />

Bernard Feringa.<br />

Collaboration between these three groups of scientists lent these<br />

nanomachines biomedical applications, opening the door to an exciting<br />

new frontier in medical treatment. The researchers at Rice synthesized<br />

ten molecular motors with novel functions, the most significant<br />

being the ability to drill through the lipid bilayer membranes that<br />

encapsulate cells. The North Carolina State researchers then tested<br />

these new motors on artificial lipid vesicles, which are fluid-filled<br />

cavities surrounded by a fatty membrane, and finally, the Durham<br />

researchers conducted experiments on live cells.<br />

The key difference between the molecular motors and traditional<br />

pharmaceuticals is their method of action. “It’s a whole new mechanism<br />

of treatment. We have a mechanical effect at the molecular<br />

level,” Tour said. The molecular motors, in effect, punch holes in the<br />

membranes of cells through pure force, an attack that is less vulnerable<br />

to cellular resistance than chemical attacks that interrupt cell<br />

processes like DNA replication. Unlike chemotherapy, which uses<br />

toxic chemicals to kill tumor cells, these nanomachines can target<br />

diseased cells more directly.<br />

These molecular motors, except for the controls, each consist of<br />

a UV light-activated rotor and a larger portion of the motor that<br />

remains fixed during rotation and provides a magnetic pole around<br />

which the rotor turns. When hit with the correct wavelength of<br />

light, the motor will rotate at a frequency of up to two or three<br />

million rotations per second. If the motors have attached themselves<br />

to a cell, when UV light causes rapid spinning of the motor,<br />

it effectively drills through the cell’s lipid bilayer, causing the cell to<br />

spill its contents and die.<br />

The sheer variety of motors developed provides a testament not<br />

only to the wide array of biomedical applications that these nanomachines<br />

can have, but also to the complex molecular interactions that<br />

govern the success of molecular motors. While the main components<br />

of each molecular motor are the same, each motor can vary<br />

and size and can be modified with different chemical groups to give<br />

it new capabilities. For example, the motors can be equipped with<br />

fluorophores, which are fluorescent chemicals that allow the motor<br />

to be tracked easily inside of cells. Several of the motors were also<br />

32 Yale Scientific Magazine October 2017 www.yalescientific.org


designed with attachments that bind specifically to protein groups<br />

found on cancer cells, thus testing the ability of the nanomachines to<br />

target specific cells.<br />

To confirm whether the motors could open bilayers, the<br />

researchers tracked the release of dye from vesicles in the presence<br />

of activated motors. In the first set of experiments, the Wang<br />

group encapsulated both dye and molecular motors modified with<br />

fluorophores inside of synthetic vesicles. Application of UV light<br />

significantly decreased the fluorescence intensity within these<br />

vesicles, as the dye and molecular motors diffused out. In contrast,<br />

in the absence of motors, the vesicles released negligible amounts<br />

of dye when exposed to UV light—proving that the vesicles burst<br />

because of the motors themselves, not the UV light.<br />

Next, the Pal group performed several experiments exposing live<br />

cells to the motors. The researchers first introduced the motors to cells<br />

without UV activation. Although cells internalize some of the motors<br />

through endocytosis—a normal cellular process by which cells take<br />

up materials from their extracellular environments—the experiments<br />

showed that cells remained healthy in the presence of the inactive<br />

motors, an important consideration for potential treatments.<br />

Armed with the knowledge that the inactive nanomotors are<br />

non-toxic, the researchers then activated the motors with UV<br />

light to observe how quickly the motors would accelerate cell<br />

death. The results were remarkable: the most effective motors<br />

killed both mouse fibroblast cells (a common cell-type found in<br />

connective tissue) and prostate cancer cells 50 percent faster than<br />

UV light did alone. In absolute terms, cells died within a matter<br />

of minutes after UV-activation.<br />

Perhaps most interestingly, the researchers were able to demonstrate<br />

that molecular motors could selectively target which cells they<br />

want to kill. In the experiments—performed on mammalian cells in<br />

petri dishes—the motors were modified with protein attachments<br />

physical chemistry<br />

FEATURE<br />

and were able to target cancer cells that were overexpressing certain<br />

protein groups. The motors were able to kill only these desired targets,<br />

and cell death was completed in just two to three minutes. This finding<br />

could mark the beginning of a new kind of photodynamic, or<br />

light-based, therapy research that could offer new sources of hope for<br />

patient treatment.<br />

Still, the molecular motors are not without their challenges when<br />

it comes to translating their functions into potential treatments. For<br />

example, UV light has poor penetration of human tissue, so the labs<br />

are actively investigating alternative sources of activation that have<br />

much deeper penetration. In the meantime, the labs expect the<br />

molecular motors to become viable options for diseases that occur at<br />

or near the surface of the skin, such as eczema and melanoma. Such<br />

treatments can have a strong impact: there are about 31.6 million<br />

people in the U.S. who live with eczema, and the American Cancer<br />

Society estimates that more than 87,000 new cases of melanoma will<br />

be diagnosed in 2017 alone.<br />

The molecular motors may also have a flexible range of functions<br />

beyond killing cells, such as in drug delivery. “We are looking first at<br />

cancers because we have a good understanding of cancer cells, but any<br />

cell that you need to kill can be targeted in this way,” Tour said. “You<br />

can also use molecular motors as a treatment source. In other words,<br />

turn on the molecular motors momentarily and then drugs can enter<br />

the cell.” With these options, molecular motors have the potential to<br />

enhance everything from antibiotic delivery to gene therapy.<br />

In the future, the labs will focus on applying the molecular motors<br />

to the issues of pancreatic cancer, carcinomas and eczema, and on<br />

developing different methods of activation that reach deep tissues<br />

within the body. Indeed, these motors provide hope for targeting<br />

nearly incurable diseases like pancreatic cancer in a way that minimizes<br />

side effects. As research progresses, a safe, quick, and effective<br />

photodynamic therapy could become a reality.<br />

IMAGE COURTESY OF WIKIMEDIA COMMONS<br />

► White blood cells defend the body from disease partly by<br />

distinguishing the body’s own healthy cells from pathogens and<br />

cancerous cells. Molecular motors can similarly target diseased cells<br />

within the body and selectively destroy them.<br />

IMAGE COURTESY OF WIKIMEDIA COMMONS<br />

► Prescription drugs come in many shapes and sizes. Molecular<br />

motors may in the future work independently of or alongside such<br />

drugs to enhance treatment, while causing less damage to patient<br />

cells and fewer side effects.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

33


COUNTERPOINT<br />

GOOD PROSPECTS FOR NEUTRINO PHYSICS<br />

►BY ISABEL SANDS<br />

In the time it takes you to read this sentence, more than 100 billion<br />

neutrinos from the sun will pass through your fingertip. You’re not<br />

likely to notice—the neutrino is a tiny, subatomic particle with virtually<br />

no mass. It interacts with matter through two of the four fundamental<br />

forces of nature: gravity and the weak nuclear force, which causes<br />

radioactivity. However, even this gravitational interaction is feeble due<br />

to the neutrino’s infinitesimal mass. And yet, despite its imperceptible<br />

nature, this elusive particle may be the key to unlocking some of the<br />

deepest secrets of the physical universe.<br />

Neutrinos have no electric charge and belong to a class of subatomic<br />

particles called leptons. Leptons possess very little mass, do not<br />

undergo strong interactions, and are characterized as the building<br />

blocks of matter. According to our current understanding of particle<br />

physics, there are three “flavors” of neutrino: the electron, muon,<br />

and tau neutrino. Each of these uncharged leptons associates with<br />

a specific type of charged leptin—the electron neutrino matches<br />

with the electron, while the muon and tau neutrinos pair with the<br />

electron’s heavier, less stable siblings: the muon and tau particles.<br />

Some physicists, however, have hypothesized the existence of a fourth<br />

flavor: a “sterile” neutrino that only interacts with gravity, making it<br />

difficult to detect.<br />

Recently, measurements from the Daya Bay power plant in China<br />

generated excitement over the possibility of sterile neutrinos. Nuclear<br />

reactors produce a flow of electron antineutrinos, which are the<br />

antiparticles of electron neutrinos. Physicists can model this flow by<br />

understanding the reactions that occur within the nuclear reactors.<br />

But at the Daya Bay power plant, these models predicted a higher<br />

number of flowing antineutrinos than were experimentally within the<br />

IMAGE COURTESY OF YALE WRIGHT LABORATORY<br />

►The result of a US-China-Russia collaboration, the Daya Bay reactor<br />

neutrino experiment promises to shed light on the mysterious nature<br />

of neutrinos.<br />

reactor. This phenomenon, called the “reactor antineutrino anomaly,”<br />

has also been measured at other sites. Neutrinos can spontaneously<br />

change flavors in a phenomenon called “oscillation,” so is it possible<br />

that some percentage of neutrinos are oscillating into undetectable,<br />

sterile neutrinos?<br />

Careful analysis of the data from Daya Bay initially seemed to<br />

rule out sterile neutrinos. As nuclear reactors consume fuel, the<br />

elemental composition of the fuel changes. Scientists found that the<br />

degree of antineutrino deficit varies with the fuel’s composition. Thus,<br />

they suspect that these elemental changes are responsible for the<br />

antineutrino anomaly. Specifically, they hypothesize that isotopes of<br />

uranium—forms of the atom with a different atomic mass—are the<br />

primary culprits. They believe they are overestimating the number<br />

of antineutrinos produced by uranium due to the variation between<br />

isotopes. However, further analysis has revealed that uranium isotopes<br />

cannot entirely explain the antineutrino anomaly.<br />

Karsten Heeger, a Yale physics professor and the director of Yale’s<br />

Wright Laboratory, is currently working on the Daya Bay experiment.<br />

“The immediate conclusion people jumped to was, ‘Oh, this might<br />

explain everything about the reactor antineutrino anomaly!’” Heeger<br />

said. “But it does not. People have now taken a closer look and realized<br />

that two effects could be taking place, wrong nuclear physics and<br />

sterile neutrinos.” The combined effect of the two theories fits the<br />

data better than that of either hypothesis alone, which leads Heeger to<br />

believe that multiple causes contribute to the anomaly.<br />

Heeger is now leading a new, Yale-led neutrino experiment called<br />

PROSPECT at the Oak Ridge National Laboratory’s research reactor.<br />

By using only one isotope in the fuel for the reactor at Oak Ridge,<br />

PROSPECT will be able to test nuclear reactor models more precisely.<br />

“We can see if over the distance of several meters, these neutrinos<br />

oscillate into sterile neutrinos,” he said.<br />

The implications of this research are certain to be profound. “Instead<br />

of neutrinos being a byproduct, we’re now starting to see them as a<br />

way to probe into nuclear reactions,” Heeger remarked. PROSPECT<br />

will hopefully provide data that allows scientists to refine their nuclear<br />

reaction models.<br />

PROSPECT also has the potential to transform modern physics:<br />

sterile neutrinos are not included in the Standard Model, the theory<br />

that describes all known elementary particles. If PROSPECT finds<br />

evidence for sterile neutrinos, “It would revolutionize particle<br />

physics,” Heeger said. “It would require us to come up with something<br />

completely new and different.”<br />

Researchers and students at the Wright Laboratory will complete<br />

construction on PROSPECT this fall, and installation at Oak Ridge<br />

National Laboratory will take place by the end of the year. Heeger says<br />

there may be a definitive answer to the antineutrino anomaly by 2018,<br />

but for now, he remains cautious. “I have no predictions,” he said. “We<br />

need to get the data.”<br />

34 Yale Scientific Magazine October 2017 www.yalescientific.org


INN VATI N<br />

STATION<br />

Decluttering our Landfills<br />

►BY LESLIE SIM<br />

Half of the plastics we use today are used once and then<br />

left to accumulate in landfills. This statistic is symbolic of<br />

a greater issue: given our ever-increasing rate of plastic<br />

consumption, overstuffed landfills will create dire problems<br />

for future generations. And while humans eventually will<br />

have to face the consequences of plastic pollution on land—<br />

if we aren’t doing so already—plastic pieces are currently<br />

floating in our oceans, being ingested by marine animals and<br />

contaminating natural habitats.<br />

Reducing our plastic consumption is certainly one way to<br />

address this issue, but researcher Yiqi Yang and his team at the<br />

University of Nebraska-Lincoln have an even better solution:<br />

biodegradable plastics. Yang believes that his lab may have<br />

found a way to create a cost-effective biodegradable plastic<br />

that targets the textile industry, where polyester plastics are<br />

a big source of plastic consumption. Production of these<br />

plastics requires a lot of petroleum, a limited resource.<br />

Other researchers have designed biodegradable plastics in<br />

the past. For example, polylactic acid (PLA) was the first and<br />

largest-scale biopolymer produced from annually renewable<br />

resources such as corn starch and sugarcane. Biopolymers<br />

like PLA are very long molecules consisting of a biologicallyrelevant<br />

repeating subunit. However, PLA’s limitations make<br />

it ineffective for use in the textile industry: “Others have<br />

been able to find biodegradable products such as PLA, but<br />

those products cannot be used widely in the textile industry<br />

because they are easily hydrolyzed at high temperature,”<br />

Yang said. Hydrolysis refers to the degradation of a plastic<br />

by breakdown into its subunits. PLA-based plastics risk<br />

being too soft and too easily broken-down, and are thus<br />

not suitable for textile materials. While engineers have<br />

made some progress, the main problem today isn’t finding<br />

a biodegradable product, but rather one that is both costeffective<br />

for manufacturers and has the properties necessary<br />

for a quality plastic.<br />

The plastics we use daily have the opposite problem. Most<br />

plastics are too stable and stick around in landfills or natural<br />

habitats for a long time, unable to biodegrade. After some<br />

experiments, however, Yang and his team have developed<br />

a biodegradable plastic without those shortcomings. In<br />

their experiments, the team obtained two biopolymers,<br />

poly-L-lysine (PLL) and poly-D-lysine (PDL). With these<br />

biopolymers, they formed plastic fibers and employed a<br />

thermal treatment to form tighter structures with strong<br />

interactions between the polymers. This key process<br />

ultimately decreased the plastic’s sensitivity to hydrolysis,<br />

especially at high temperatures.<br />

Several experiments testing the new plastic’s properties<br />

show that Yang’s team may indeed have the key to tackling<br />

the two main problems in plastic development (hydrolysis<br />

and softness at high temperatures). However, until their<br />

materials are examined in large-scale production, they can’t<br />

say for sure whether their plastic will work on the industrial<br />

scale. The next step for Yang and his team is to test the plastic<br />

on a small scale in the textile industry. From there, they can<br />

target other industries. Nonetheless, Yang and his team have<br />

gotten a step closer to making a product that will hopefully<br />

prove useful in decreasing, if not eliminating, plastic waste.<br />

The impact of a biodegradable plastic in reducing plastic<br />

pollution is far-reaching. Currently, animals on land and in<br />

oceans are easily harmed by ingesting plastic microparticles<br />

or by getting caught in large pieces of plastic. For humans,<br />

plastic remains in our landfills for far too long, and we’re<br />

running out of space. Yang also cautions that tap water from<br />

all around the world contains microscopic pieces of plastic,<br />

and without our knowing, we could be accumulating plastic<br />

in our bodies.<br />

Since demand for plastic is growing exponentially as the<br />

human population grows, it is especially critical for us to<br />

find a sustainable resource that meets our needs without<br />

damaging our environment. As we move in the direction<br />

of finding cost-effective and useful forms of biodegradable<br />

plastic, we can rest assured that researchers like Yang and his<br />

team are on the case.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

35


PETER WANG (TD ‘18)<br />

A MODERN DAY RENAISSANCE-MAN<br />

►BY ERIN WANG<br />

UNDERGRADUATE PROFILE<br />

IMAGE COURTESY OF ERIN WANG<br />

►Peter has a wide variety of interests, from molecular biology to<br />

hiking through Europe.<br />

Scientists are often stereotyped as people buried deep in research papers<br />

and focused singularly on the pursuit of science—this is hardly the<br />

case for current Yale College senior Peter Wang (TD ’18). Although he<br />

is majoring in Molecular, Cellular and Developmental Biology, his interests<br />

range from photography and poetry to violin-playing and graphic<br />

design. Science first caught his attention in middle school, but he wanted<br />

to be an architect when he was younger. “I got interested in appreciating<br />

structures, appreciating how things connect and form,” Wang said.<br />

This interest steered him towards science—specifically, to biomolecules.<br />

“The complexity and sheer scale of molecules are incredible,” he said.<br />

Wang’s scientific endeavors took off at Yale. Here, he has been immersed<br />

in classes that give him more nuanced understandings of subjects.<br />

Moreover, he has been able to engage in cutting-edge RNA research:<br />

the summer after his first year at Yale, Wang began working<br />

on methods to decipher the interactions and conformations of long<br />

non-coding RNAs in Matthew Simon’s lab, where he has been ever<br />

since. These RNAs play various important roles in cells, such as regulating<br />

gene expression, so studying their structures and functions is a<br />

worthwhile endeavor. He has thrown himself into research with a fervent<br />

passion—self-evident in his current juggling of multiple independent<br />

research projects within his lab and his recent second-author publication<br />

in ACS Biochemistry.<br />

Not only is Wang passionate about studying science himself, but he<br />

also wants to make science more accessible to others. In fact, his interest<br />

in communicating biology drove him towards graphic design. “Biology,<br />

because of its complexity, relies on a lot of concepts and processes that<br />

you need very clear visual tools to help people understand,” Wang said.<br />

He believes that in many cases, art is one of the most effective ways to<br />

improve science communication to the general public.<br />

Similarly, Wang has tried to improve the accessibility of research to<br />

students at Yale. He, along with his teammates at the Yale Undergraduate<br />

Research Association (YURA), felt that students were having difficulty<br />

searching through faculty to find suitable research mentors due to different<br />

department website layouts and the fact that key information, like<br />

faculty research interests, was often difficult to find. To this end, he led<br />

a five-person team in a yearlong endeavor to build the YURA Research<br />

Database, the first of its kind at Yale. This database gives students an easy<br />

way to sort through professors and their research interests. Under Wang’s<br />

guidance, the YURA Research Database is now an invaluable tool for students<br />

looking for research at Yale, and another improved version is in the<br />

works.<br />

Wang is also an avid environmentalist. As president of the Yale Student<br />

Environmental Coalition, he helped send four Yale student observers<br />

to the 2015 United Nations Climate Change Conference in Paris.<br />

He participated in The GREEN program, through which students learn<br />

more about the environment by global exploration. Through the program,<br />

he traveled to Iceland and Hawaii—places that have adopted virtually<br />

fully renewable energy standards. From these trips, Wang learned<br />

more about how their communities maintain sustainable lifestyles.<br />

Along the way, he built friendships with locals, learned more about their<br />

cultures, and experienced the natural beauty these places provided.<br />

Outside of exploring science, Wang also enjoys exploring the world.<br />

Recently, he went on a month-long solo trek in Europe. “I had never<br />

been to Europe. I wanted to travel. I wanted to explore somewhere I’d<br />

never been,” Wang said. He plotted a ten-city path through Europe, using<br />

Airbnb to find places to stay. “That was probably the most adventurous,<br />

exciting, packed, and happy month of my life,” Wang said. The<br />

most memorable moments of his journey took place at a Zürich train<br />

station. “The sunset reflected gold on the rail tracks, and the late evening<br />

trains brought back tired home-bound people from their work.<br />

It kind of encapsulated a lot of what that trip was—catching beautiful<br />

sights, and feeling what home felt like for different people around Europe,”<br />

Wang said.<br />

Wang is currently working on applications to graduate school. In ten<br />

years, he hopes to be working in academia on exciting projects in a lab<br />

with people he enjoys being around, all the while making meaningful<br />

contributions to science. He is not certain if he will be working on<br />

non-coding RNA in the future, but he would not hesitate to investigate<br />

it if the opportunity arises.<br />

36 Yale Scientific Magazine October 2017 www.yalescientific.org


ALUMNI PROFILE<br />

SANDY CHANG (YC ‘88)<br />

BUILDINGTHENEXTCREATIVETHINKERSINSCIENCE<br />

►BY JESSICA TRINH<br />

Sandy Chang wears a lot of hats. As a professor at the Yale School of<br />

Medicine, he signs out clinical cases and runs an NIH-funded research<br />

lab, studying telomeres and their relationship to cancer and aging. He<br />

holds professorships in the departments of Laboratory Medicine, Pathology,<br />

and Molecular Biophysics and Biochemistry. Last spring, he<br />

was appointed to the role of Associate Dean for Science and Quantitative<br />

Reasoning Education at Yale College, a role that involves overseeing<br />

the quality of STEM education and administration of undergraduate research<br />

fellowships. If that wasn’t enough, he also teaches a popular freshman<br />

seminar called “Topics in Cancer Biology.” For Chang, the time he<br />

invests is worth it, as his top priority is nurturing future scientists.<br />

Born in Taiwan, Chang moved to Sarasota, Florida at the age of seven,<br />

where his love of science blossomed. namored by the vast open skies, he<br />

became interested in astronomy. Despite doing astronomy-related research<br />

in high school as part of his dream of becoming an astronomer,<br />

Chang later found himself drawn toward molecular biology. So, in 1984,<br />

he attended a college well-known for its Molecular Biophysics and Biochemistry<br />

major: Yale University.<br />

While at Yale, Chang pursued a diverse array of interests, from writing<br />

about his undergraduate research for our very own Yale Scientific<br />

Magazine to joining the fencing team. He started conducting laboratory<br />

research during the summer after his first year at Yale. Immediately, he<br />

noticed the lack of funding resources available for undergraduate students,<br />

a problem he would always remember.<br />

Mentorship was key to Chang’s success. During his time in college,<br />

Chang found no shortage of mentors, from undergraduate seniors who<br />

gave him advice on MD-PhD programs to the principal investigators<br />

of research labs who nurtured his love of scientific inquiry. Outside of<br />

class, Chang volunteered in the pediatric oncology department at the<br />

Yale New Haven Hospital, where he engaged with patients. The combination<br />

of volunteer work and scientific research led Chang to realize he<br />

couldn’t just pursue an MD or a PhD degree—he wanted both.<br />

Chang graduated from Yale in 1988 and went on to the prestigious<br />

tri-institutional Weill Cornell/Rockefeller/Sloan-Kettering MD-PhD<br />

program. Afterwards, he did his residency in clinical pathology at the<br />

Brigham & Women’s Hospital and a postdoctoral fellowship at the Dana<br />

Farber Cancer Institute, Harvard Medical School. Chang then moved to<br />

Houston, Texas to start his first real job as an assistant professor at the<br />

MD Anderson Cancer Center. In 2010, Chang received an opportunity<br />

to return to Yale as a tenured associate professor at Yale Medical School.<br />

When it comes to scientific research opportunities for students, Chang<br />

believes in leveling the playing field. He is a firm believer that anyone<br />

who desires to do research should be able to do so, regardless of their<br />

financial obligations. In particular, the First-Year Summer Research Fel-<br />

PHOTOGRAPHY BY SUNNIE LIU<br />

►Between research and serving as Dean of Science and QR, Sandy<br />

Chang still finds plenty of time to mentor undergrads.<br />

lowship provides financial support that enables students to conduct research<br />

under the supervision of a Yale faculty member. The fellowship<br />

includes a stipend that is more than enough to cover Yale’s summer income<br />

contribution, allowing students to focus on their research.<br />

A recent survey of students who completed the fellowship show<br />

promising results: over 91 percent of rising sophomores stated they<br />

would continue research in the future. Chang strongly believes that early<br />

exposure to quality research opportunities is crucial for fostering a<br />

love of science. “The second you find yourself in a research laboratory<br />

with an inspirational mentor who guides you, it’s like being bitten by the<br />

research bug—you fall immediately in love with the research you are<br />

doing,” Chang said. Funding and support from fellowships are just one<br />

important component to building foundations for those interested in<br />

pursuing research—equally important is mentorship.<br />

For instance, Chang was the first-year advisor to Cayla Broton (ES<br />

’16). Following graduation, Cayla spent a year working with Chang,<br />

making important contributions to telomere biology. She is now getting<br />

interviews to top MD/PhD programs. In addition, in his “Topics<br />

in Cancer Biology” course, Chang guides first years through reading<br />

primary research articles and crafting their own research grant proposals.<br />

These are crucial tools that he believes students will utilize long after<br />

they finish the course.<br />

Today, Chang continues to make time for his undergraduates. You will<br />

often find him having meals with students. He loves to teach, to mentor,<br />

and to foster opportunities for undergraduates to be bitten by that research<br />

bug. “It’s a privilege to help students and to watch them blossom<br />

into scientists,” Chang said.<br />

www.yalescientific.org<br />

October 2017<br />

Yale Scientific Magazine<br />

37


FEATURE<br />

documentary and book review<br />

SCIENCE IN THE SPOTLIGHT<br />

DOCUMENTARY REVIEW : CHASING CORAL<br />

►BY CORY WU<br />

A pristine bed of corals shimmers pure white, surrounded by silence.<br />

Water calmly brushes over these stagnant structures, with no other life<br />

in sight. This seemingly beautiful scene is quite deceiving, as it depicts a<br />

coral graveyard after a mass bleaching event. The vivid, vibrant colors<br />

of ordinary corals have been usurped by a sickening white, and entire<br />

schools of fish that once relied on the coral have disappeared. Over just<br />

a two-month period, an entire coral reef ecosystem has been destroyed.<br />

The culprit? An increase in the average ocean temperature by two<br />

degrees Celsius. While two degrees seems minimal, even slight increases<br />

in ocean temperature can wipe out entire reefs. Heat waves cause coral<br />

to eject their main food source, symbiotic algae, and lose their vivid<br />

colors. Thus, begins the first stage of death for the coral: coral bleaching.<br />

Jeff Orlowski, director of the award-winning documentary “Chasing<br />

Ice,” recently released a follow-up documentary called “Chasing Coral,”<br />

which focuses on a lesser-known danger of climate change: coral<br />

bleaching. The film follows a team of divers who endeavor to bring the<br />

realities of climate change and coral bleaching to the public. Viewers<br />

follow the divers as they continue the daily struggle of watching beautiful<br />

fields of coral slowly wither away.<br />

The film truly excels at the art of “show, don’t tell.” Shocking facts<br />

are coupled immediately with haunting imagery. Salient side-by-side<br />

comparisons of coral before and after bleaching in the documentary<br />

illustrate how the bustling, lively nature of coral reefs is destroyed by<br />

BOOK REVIEW: THE TELOMERASE REVOLUTION<br />

►BY ROBERT LUO<br />

Death is an inevitable consequence of the passage of time.<br />

This notion underlies much of our literature and films, and even<br />

affects the choices we make in everyday life. Death and poor<br />

health plague our futures. But what if there were an elixir that<br />

might allow us to live even longer than today’s centenarians—a<br />

life devoid of age-related illnesses, like Alzheimer’s? It might<br />

sound like something out of a science fiction novel, but some<br />

scientists and physicians argue that sometime in the future, we<br />

could turn science fiction into reality.<br />

One of these physicians is Michael Fossel, MD-PhD and a<br />

professor of clinical medicine at Michigan State University,<br />

who studies the science of aging. In his book, The Telomerase<br />

Revolution, Fossel introduces telomerase as a component in<br />

treating illnesses like heart disease and dementia—and even<br />

in reversing aging itself. Telomerase is an enzyme that repairs<br />

telomeres, the regions at the ends of chromosomes. Within the<br />

last two decades, scientists have found that the shortening of<br />

telomeres causes cellular aging, thus giving rise to the telomere<br />

theory of aging.<br />

In the book, Fossel excellently explains the elegance of this<br />

theory. He contextualizes the theory with respect to other<br />

theories of aging—vitalist, hormonal, and genetic theories,<br />

amongst others—and proposes a strong unifying argument<br />

for telomeres as a cause of aging. Poignantly, Fossel illustrates<br />

the powerful implications of the telomere theory of aging; by<br />

activating telomerase in lab mice, signs of aging can not only<br />

bleaching, leaving only foreign algae<br />

to drape the skeletal remains of the<br />

coral. “Chasing Coral” masterfully<br />

illustrates the emotional impact coral<br />

bleaching can have on people, which is<br />

particularly apparent when the divers<br />

are visibly shaken by the destruction they<br />

photograph daily.<br />

In its middle, the film’s pacing<br />

becomes somewhat sluggish. It often<br />

focuses too deeply on the methodology<br />

of capturing images of coral, instead of<br />

analyzing the process of bleaching or the<br />

methods through which corals support<br />

ecosystems. The documentary also<br />

jumps between the perspectives of multiple people too quickly at times,<br />

which can detract from the significant experiences of each person.<br />

Still, the climax and ending masterfully cinch the documentary<br />

together by reassuring viewers of the many ways we can contribute in<br />

the fight against climate change. The film’s seamless combination of<br />

informative commentary and engaging storytelling make “Chasing<br />

Coral” not only an entertaining watch, but a necessary one to understand<br />

the full threat climate change already poses to our environment.<br />

be stopped, but also reversed.<br />

Through telomerase therapy,<br />

Fossel argues, humans have the<br />

potential to not only live longer,<br />

but also live more healthily.<br />

Although Fossel explains the<br />

effects of telomere shortening on<br />

the human body, he only does so<br />

on a surface level. Those curious<br />

about mechanistic explanations<br />

will be scratching their heads<br />

searching for deeper answers.<br />

Further, Fossel goes beyond the<br />

biology of telomerase to explain<br />

the highs and lows of telomerase<br />

therapy, often muddled in politics or stalled by lack of financial<br />

support. Finally, Fossel discusses the societal implications of<br />

telomerase therapy. How will we value life when we can all live<br />

into old age? To those curious about telomerase therapy and its<br />

potentially earth-shaking implications, this book is definitely<br />

worth a read.<br />

While the full implications of the purported “telomerase<br />

revolution” are still speculative, the book ends with a<br />

commentary about what we can currently do to fight the<br />

negative effects of aging. According to Fossel, the best answers<br />

are often the most simple: good diet, exercise, and meditation.<br />

38 Yale Scientific Magazine October 2017 www.yalescientific.org


cartoon<br />

FEATURE<br />

BEFORE/AFTER<br />

►BY LISA WU<br />

Interested in writing for<br />

Contact us at<br />

ysm@yale.edu!

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!