41
BIOTECH PLAZA Table of contents MISSION STATEMENT 2–3 INSTITUTE OVERVIEW 4–5 INTRODUCTION – TARGET AREAS 6–7 HEALTH AND DISEASE 10–15 NEURODEGENERATION 18–25 SMALL MOLECULE PHARMA 28–39 CELL AND PROTEIN TECHNOLOGIES 42–57 BIG DATA 60–67 PLANTS AND FOOD 70–79

BIOTECH · PDF fileAll of them have arisen directly from BI’s discovery science. ... technology for treating killer diseases caused by defects in the cell’s ... mitochondria, and

Embed Size (px)

Citation preview

BIOTECH PL A Z A

Table of contents

MISSION STATEMENT 2–3

INSTITUTE OVERVIEW 4–5

INTRODUCTION – TARGET AREAS 6–7

HEALTH AND DISEASE 10–15

NEURODEGENERATION 18–25

SMALL MOLECULE PHARMA 28–39

CELL AND PROTEIN TECHNOLOGIES 42–57

BIG DATA 60–67

PLANTS AND FOOD 70–79

32

BI is Finland’s flagship institute for molecular

biosciences, home to more than 250 researchers from

all over the world. Our scientists are at the international

forefront of research in genomics, structural biology,

bioinformatics, molecular neuroscience, cell and

developmental biology and plant sciences. BI’s main

focus is basic science, but we also seek to commercialize

our research findings, for society’s benefit.

This prospectus is the launch document for Biotech

Plaza. It is aimed at potential investors, academic

and industrial partners, funding agencies, business

developers, end-users and any other types of

stakeholder, whom we invite to join with us to turn our

dreams into reality.

Howy JacobsInstitute Director

Here we set out a range of projects in progress, with strong

commercial potential. Some are broad, others narrowly

focused. Some are already close to being realized, whilst

others are long-term and aspirational. Some are aimed at

well-defined niche markets, whereas others seek to establish

technologies to tackle globally important problems.

All of them have arisen directly from BI’s discovery science.

But to develop them will require different kinds of support

and expertise than our scientists can provide on their own.

Biotech Plaza brings together a wide spectrum of advisors,

actors and financing tools to accomplish this task, with a

strong emphasis on the global marketplace. Meanwhile, our

research teams continue to replenish the discovery pipeline.

Troy FaithfullDevelopment Manager, Biotech Plaza

54

GLOBALBI is Finland’s flagship institute for molecular biosciences, home to

more than 250 researchers from all over the world. With a culture

of excellence and a strong track-record of success, it nurtures

discovery, technology, careers and businesses.

CUTTING-EDGEOur scientists are at the international forefront of research

in genomics, structural biology, bioinformatics, molecular

neuroscience, cell and developmental biology and plant sciences.

Our main focus is basic science but, in addition, we vigorously

pursue commercialization of our research findings.

TECHNOLOGYCurrent projects with major potential impact include a revolutionary

technology for treating killer diseases caused by defects in the cell’s

energy system; new drugs to reverse the nerve damage that causes

neurodegenerative diseases, and protein engineering tools based

on the splicing activities of inteins.

INNOVATIONBI’s future innovation potential includes studies of pathogen

resistance in cereal crops, structural analyses of complex viruses

like measles, targeting RNA and RNA metabolism as therapies for

cancer and neurodegeneration, unraveling the genetic circuitry

controlling organ formation, computational tools to interpret the

massive data sets emerging from genome analysis, and screens for

molecules capable of blocking substrate-specific protein secretion.

INVESTMENTBI seeks partnerships from the private sector, in order to develop

and commercialize its inventions. World-class discovery science,

combined with Finland’s transparent business environment and

highly skilled workforce, are ingredients for commercial success.

OPPORTUNITYInvestors and supporters can become stakeholders in BI as a

whole, in specific projects or in startups to be established jointly.

Contact us to explore possibilities.

BI

in numbers

established 1989

7 bioscience

technologies established in

Finland

10startups

>250doctoral theses

25research grants

in excess of $1 million

~300publications in

top-ranked international

journals

145applied patents

bi.helsinki.fiAN INDEPENDENT INSTITUTE OF THE UNIVERSITY OF HELSINKI

5

Contact us to explore

possibilities at

biotechplaza.org

76

HEALTH AND DISEASEMany of the world’s most intractable

diseases arise from disrupted intracellular

communication or homeostasis. Our

scientists are working on novel technologies

to alleviate damage arising from deranged

cell signaling, focusing on key regulatory

pathways affecting proteotoxicity in the

endoplasmic reticulum, oxidative stress in

mitochondria, and interactions between

the cell nucleus and cytoplasm. BI aims to

produce breakthrough treatments for a huge

spectrum of pathological states, ranging

from septic shock, to inherited heart disease,

diabetes and cancer.

NEURODEGENERATIONAge-associated neurodegenerative diseases

are currently incurable, and impose a

massive societal and economic burden, as

well as individual suffering on a colossal scale.

Many labs around the world are working

to understand the underlying pathological

mechanisms as a route to therapy. We have

prioritized ‘out-of-the-box’ approaches,

based on novel strategies to activate

natural, neuroprotective, stress-handling

mechanisms. We believe our new concepts,

presented here, will bring concrete and

dramatic benefits to the treatment of stroke,

movement disorders and dementia within

the next decade.

SMALL MOLECULE PHARMAThe search for new and better drugs for

use in the personalized therapies of the

future is one of the major drivers of today’s

pharmaceutical sector. This involves not just

the identification of more specific targets but

the implementation of ‘designer’ screening

platforms. Biotech Plaza features BI’s ongoing

work to create more specific anticancer

drugs, immune modulators, antibiotics,

and agents directed at new targets in

neurodegenerative and viral diseases. This

builds on our strong profile in structural and

cellular biology.

Biotech Plaza showcases the innovative technologies arising from BI’s scientific work at the frontiers of biological knowledge. We are looking for partnerships and financing to turn our ideas into products and services that will have major societal impact.

CELL AND PROTEIN TECHNOLOGIESFuture medicine will be based upon strategies

for tissue repair and regeneration, and the

delivery of exquisitely targeted therapeutic

drugs and proteins with minimal side-effects.

Biotech Plaza presents a wide spectrum of

new technologies and discovery science to

enable these goals, ranging from new systems

to engineer proteins and drug-conjugates, to

the elucidation of the molecular mechanisms

whereby stem cells stay young and retain

regenerative capacity. We are also developing

services for customized gene expression

and manipulation to serve the research

community, and novel reporter systems to

profile pathological disturbances in cellular

growth status and in subcellular protein

localization.

BIG DATAThe new science of genomics has been made

possible not only by the orders-of-magnitude

advances in DNA sequencing and the equally

astonishing decrease in its costs, but also

by the development of data-handling and

pattern-recognition tools that enable us to

make sense of the vast amounts of data

from biological and clinical specimens that

DNA sequencing generates. BI’s research

teams are at the forefront of developing and

applying computational methods to make

sense of big data emerging from genomic

analyses, and using it to predict the safety

of nanomaterials, the hazards of post-

operative infections or to serve the needs

of conservation biology, environmental

management and agriculture.

PLANTS AND FOODModern genetics opens up enormous

possibilities for altering the properties of

crop plants, to address the major global

challenges of our time: climate change,

resource depletion, pollution, and hunger.

We are focusing on improved pest-resistance

in cereal crops, enhancing the rate of carbon

capture in sustainable forests, modifying

trees so as to produce more suitable raw

materials for the chemical and biofuel

industries, and a more efficient way to

engineer targeted gene modifications in

common crop plants.

Target areas of Research Projects

9

Health & Disease

THE ALTERNATIVE OXIDASE AOX – A REVOLUTIONARY TOOL FOR WIDE-SPECTRUM THERAPY | Howy Jacobs

MANF AS A NOVEL THERAPEUTIC TARGET FOR THE TREATMENT OF DIABETES | Maria Lindahl

MITOCHONDRIAL MEMBRANE PROTEINS – STRUCTURE AND FUNCTION IN HEALTH AND SICKNESS | Vivek Sharma

1110

Howy Jacobs

The Alternative Oxidase AOX – a revolutionary tool for wide-spectrum therapyWe are developing a revolutionary therapy for diseases caused by malfunction of the mitochondria, the energy plants of our

cells. We take advantage of a natural system found in lower organisms for resisting mitochondrial stresses. By deploying

this system in humans, we hope to treat a wide spectrum of currently incurable and fatal diseases, whether caused by

environmental poisons, inherited mutations, metabolic overload or damage due to ‘wear and tear’ during aging.

Mitochondria recover the primary chemical

energy released from the breakdown of

food molecules in forms that the cell can

use. This enables vital processes such as

muscle contraction, electrical conduction

between nerve cells and hormone secretion.

Mitochondrial dysfunction causes many

fatal conditions, including rare, multi-system

diseases of infancy, as well as degenerative

disorders of old age. Major killers such as

heart attack and stroke result from the

inability of mitochondria to shut down and

power up properly, when the oxygen supply

is transiently interrupted. The inflammatory

processes leading to chronic lung diseases

are initiated by specific poisons in toxic

smoke that disable the mitochondria and

prevent the efficient clearance of particulates.

And fatal multiple organ failure in sepsis has

been attributed to mitochondrial shutdown.

Our novel, therapeutic approach employs

the alternative oxidase, AOX. AOX by-passes

the major energy supply pathway when

it is blocked, maintaining cellular functions,

preventing the build-up of toxic oxygen

radicals, minimizing the disruption of normal

metabolism and preventing cell death. We

have shown that transgenic rodents, flies or

mammalian cells endowed with the AOX gene

transplanted from a marine invertebrate are

protected from many pathological insults that

affect mitochondria in humans, including:

• inherited mitochondrial mutations causing

fatal infantile disease

• the toxicity of cyanide or the cocktail of

poisons in vehicle exhaust or cigarette smoke

• multiple organ failure due to bacterial sepsis

• a key indicator of physiological stress

following cardiac infarct, and

• neurodegeneration seen in models of

Parkinson’s and Alzheimer’s diseases

AOX thus has immense and broad potential

in medicine, ranging from a treatment

for rare diseases to the protection of

populations at risk from biohazardous

agents. Our strategy is to supply AOX (using

viral vectors, mRNA-mimetics or as protein

via cell-permeating peptides) to combat

mitochondrial damage.

Our strategy is to supply AOX … to combat mitochondrial damage

10

Our immediate aim is to develop partnerships with others in the field of genetic therapy.

We will develop ways of delivering AOX safely and effectively to affected tissues and organs,

starting with life-threatening conditions where we have solid data from animal trials.

End-users of our technology will initially be in the pharmaceutical sector, but it should also be

applicable to protection of military personnel, civilian first-responders and populations at risk

from terrorism or pollution.

1312

Maria Lindahl

MANF as a novel therapeutic target for the treatment of diabetesWe have identified a new growth factor for pancreatic insulin-producing β-cells. In the future, it could be used to regenerate

the lost β-cell mass in diabetic patients. This would restore normal metabolic control and avoid the severe, life-threatening

complications resulting from diabetes.

Collaborator: Mart Saarma

Diabetes mellitus is a group of metabolic

disorders characterized by hyperglycemia, in

turn caused by the inability of the endocrine

pancreas to maintain sufficient levels of

circulating insulin. Type 1 diabetes (T1D)

results from the autoimmune destruction

of insulin-producing β-cells, leading to total

insulin deficiency, whereas type 2 diabetes

(T2D) develops when the β-cells are no longer

able to respond to an increased insulin

demand due to insulin resistance. Today

more than 380 million people are affected

by the disease, and its prevalence is rapidly

increasing.

Current medications, insulin and anti-diabetic

drugs, are able only to alleviate diabetic

symptoms. Their administration does not

faithfully mirror the physiological response of

β-cells and does not prevent the devastating

micro- and macrovascular complications

of the disease. The reasons for β-cell

destruction both in T1D and T2D are still

unclear, but increasing evidence implicates

endoplasmic reticulum (ER) stress as a key

mechanism, accompanied by prolonged

activation of the signaling pathway linked to

the unfolded protein response (UPR). In turn,

this arrests protein translation and activates

protein refolding and degradation.

Thus, one of the main strategies for

improving diabetes therapy is to define

and validate novel approaches to protect β

-cells from stress, as well as activate their

regeneration. Various nutrients, hormones

and growth factors are known to affect β -cell

proliferation, but their use as therapeutic

factors has remained minimal, because of

lack of specificity. Therefore there is a need

for new and more specific targets for pre-

clinical screening and eventual clinical testing.

Mesencephalic astrocyte-derived

neurotrophic factor (MANF) was originally

identified as a secreted trophic factor for

dopaminergic neurons in vitro. MANF and

its homologue CDNF, discovered by our

group, can protect and repair neurons

and cardiomyocytes in animal disease

models. The protective role of MANF has

been suggested to depend on its ability to

rescue cells from ER stress. To understand

its physiological role in vivo, we generated

MANF knockout (KO) mice that developed

severe insulin-dependent diabetes due to

progressive loss of pancreatic β-cells. One

of the underlying mechanisms was chronic

ER stress. We found that recombinant MANF

protein enhanced β-cell proliferation in

cultured adult mouse islets, whilst pancreatic

MANF overexpression in a mouse T1D model

increased β -cell proliferation and prevented

cell death.

MANF is therefore a promising new

therapeutic candidate for β -cell protection

and regeneration. Our ongoing studies

combine MANF transgenic mouse lines and

animal models for both T1D and T2D, as

well as cell-lines and organ culture. Other

potential approaches will also be investigated

for using MANF as a regenerative protein for

β-cells in diabetes.

12

Background photo: Jari Rossi and Ömer Acar

PhD Student: Tatiana Danilova

1514

Vivek Sharma

Mitochondrial membrane proteins – structure and function in health and sicknessThe main focus of our research is to understand how enzymes involved in ATP generation (respiratory enzymes) function at a

molecular level, and how point mutations cause them to malfunction, giving rise to a range of serious disorders. For this purpose,

we apply state-of-the-art computational methods on the available structural data, and explore spatial and temporal scales that are

not easily reachable within the current experimental framework. Overall, our aim is to shed light on the molecular mechanisms of

enzyme action, and use this knowledge to design possible remedies to combat pathological dysfunction.

Life on earth is powered by ATP, which

is produced primarily in mitochondria

as a result of electron transfer reactions

that are coupled to proton translocation

across the inner-mitochondrial membrane.

These elementary reactions (electron and

proton transfers) involve charged species,

and are efficiently performed by enzymes

embedded in a low-dielectric medium, i.e.

the membrane. Malfunction in one or many

intermediate steps of the catalytic cycle of

these enzymes leads to various metabolic,

neurodegenerative and mitochondrial

disorders. Moreover, under environmental

stress conditions these enzymes produce

excess reactive oxygen species (ROS), which

are harmful to other cellular components, and

lead to various mitochondrial disorders. It is

of the utmost importance to understand how

these ROS are produced, and how that can be

contained to normal physiological levels.

We have performed large-scale atomistic

molecular dynamics simulations and high-

level quantum chemical calculations on

respiratory complex I, which is the first entry

point for electrons and one of the largest

enzymes in the respiratory chain. Our results

show that both electrostatics and long-range

conformational transitions play a key role in

enzyme function, coupling the redox reaction

to proton pumping as far as 200 Å away from

the former. Our work provides one of the first

molecular insights into complex I function,

and moves a step closer to identifying

the underlying causes of mitochondrial

dysfunction.

By performing long timescale fully-atomistic

classical molecular dynamics simulations on

complex IV of the respiratory chain, we have

shown how the enzyme manages to pump

protons with high efficiency (~90%). This is

achieved by microscopic gating elements,

such as conformational dynamics of amino

acid side chains and mobile water molecules.

Here, our work strengthens the view that

evolution has led to optimal nanostructures

that very efficiently convert energy from one

form to another.

We envisage two key areas where our

research will have profound impacts; a)

mitochondrial medicine, and b) bioenergy. By

studying molecular structures of intermediate

states in the catalytic cycle of mitochondrial

enzymes the underlying molecular causes of

diseases can be identified. Second, a deeper

understanding of the molecular mechanism

of respiratory enzymes has enormous

potential in the design and development of

biofuel cells of high efficiency.

A deeper understanding of the molecular mechanism of respiratory enzymes has enormous potential in the design and development of biofuel cells of high efficiency.

15

17

Neurodegeneration

REJUVENATING THE BRAIN: NEW STRATEGIES FOR NEUROREGENERATION | Mikko Airavaara and Andrii Domanskyi

A NOVEL APPROACH TO TREATING PARKINSON’S DISEASE | Jaan-Olle Andressoo

TARGETING RNA TOXICITY IN DEGENERATIVE DISORDERS | Susana Garcia

SYNAPTIC ADHESION MOLECULES - MAKING NEURONAL CONNECTIONS

TO COMBAT COGNITIVE DISORDERS | Tommi Kajander

1918

Mikko Airavaara & Andrii Domanskyi

Rejuvenating the brain: new strategies for neuroregenerationA viable strategy to treat neurodegeneration requires a toolbox for restoring neuronal networks in the damaged or aging brain. Using

a unique set of in vivo and in vitro models and tools, we are developing a broad experimental approach to achieving this goal, focusing

on two of the commonest such disorders, ischemic stroke and Parkinson’s disease. We use cutting-edge in vivo rodent models, combined

with virus- and CRISPR-Cas9-mediated transgenesis, plus surgical and toxicological procedures, to evaluate novel drugs, neurotrophic

factors, and non-coding RNAs with neuroprotective and neurorestorative potential. Using cultured primary cells isolated from embryonic

and adult brain, we also investigate pathways regulating proliferation, migration and differentiation of neural stem cells and astrocytes,

in order to assess the ways these cells could be used for rejuvenation of degenerating neuronal networks.

Aging increases the incidence of

neurodegenerative diseases and stroke,

leading to severe deficits in life quality.

Our ultimate goal is to increase the

quality of life for the elderly, by preventing

neurodegeneration and promoting neuronal

regeneration. Neuronal loss is caused

by various stressors and by the aging

process itself. Current drug therapies

for neurodegeneration are still based on

alleviating symptoms. The major challenge is

to develop safe, curative therapies, based on

enhancing the brain’s endogenous self-repair

mechanisms. Neurotrophic factors such as

Cerebral Dopamine Neurotrophic Factor

(CDNF), Mesencephalic Astrocyte-derived

Neurotrophic Factor (MANF) and Glial cell

line-Derived Neurotrophic Factor (GDNF)

and its family members promote survival of

neurons during development and disease. We

are working with these factors, studying their

interplay, mechanism of action, regulation

by microRNAs and their target genes, in

protecting neurons against stress. These will

lead to novel treatment strategies to combat

neurodegeneration.

We have established novel rodent models

where ischemic injury is restricted to the

cortex, enabling studies of functional recovery,

brain adaptations, neuronal rearrangements

and transdifferentiation of non-neuronal cells.

For Parkinson’s disease, we utilize toxin-based

(6-OHDA, MPTP, lactacystin), α-synuclein

fibril-induced or genetically engineered

rodent models. Applying a broad array of

behavioral tests we have demonstrated the

neuroprotective potential of CDNF and MANF

in these models, and have recently found

that stimulation of the microRNA biogenesis

pathway promotes survival of cultured

dopaminergic neurons. We are currently

testing the efficacy of selected microRNAs and

drugs, used in combination with neurotrophic

factors, in protecting neurons in vivo. Drug therapies for neurodegeneration are still based on alleviating symptoms and the major challenge we have is to develop disease modifying therapies.

WE AIM TO USE OUR

MODELS AND TOOLS:

• to identify new

neuroprotective drugs and

microRNAs, plus their target

genes and pathways, and

evaluate their activity in

promoting neuronal survival.

• to alleviate endoplasmic

reticulum stress as a way

of treating protein-folding

diseases in the brain.

• to mobilize transcription

factors, neurotrophic factors

and microRNAs to stimulate

endogenous self-repair

mechanisms based on

neuroprogenitor cells and

reactive astrocytes.

We are looking for

pharmaceutical, academic,

governmental and non-profit

organizations as partners

to support our mission

in developing efficient

treatments for devastating

neurodegenerative diseases.

19

2120

Our main approach is to target regulatory

areas of genes, called 3´ UTRs. For proof

of concept, we have studied the 3´ UTRs of

two neurotrophic factors, GDNF and BDNF,

that are already known to protect against

Parkinson’s disease (PD). The development

and maintenance of the brain involves

precisely controlled chemoattraction, elicited

by molecules such as neurotrophic factors

(NTFs), that are secreted by the brain’s own

cells. Because NTFs strongly promote the

survival and function of adult neurons, they

have been proposed for the treatment of

neurodegenerative conditions, such as PD.

Intracranial delivery of GDNF has already

been and currently is in clinical trials for the

treatment of Parkinson’s disease. However,

this approach has adverse side-effects and

variable efficacy, hindering its clinical use.

In our new approach, GDNF is not applied

ectopically, avoiding the massive sprouting of

dopaminergic fibers towards the site of GDNF

delivery, with unknown consequences with

respect to side effects, treatment efficacy

and behavior, that has been observed

in previous studies in both experimental

animals and humans. Our results, recently

published in PLoS Genetics (Kumar et al,

2015), imply that measures that promote

elevation of endogenous GDNF levels may

have clinical potential in the treatment of PD.

Our next-generation results, based on this

concept, take us one step closer to defining

a novel, viable treatment strategy for PD [for

reasons of confidentiality, we are able to

disclose further details only under separate

agreement].

We are now looking for investment funding

and/or commercial collaborations for testing

endogenous GDNF/BDNF elevation as a

therapeutic strategy, and for extending the

proof of concept so as to generate better

disease models for validation of novel

treatments.

Jaan-Olle Andressoo

A novel approach to treating Parkinson’s diseaseLittle is known of the therapeutic and scientific potential of elevating normal gene function at native sites of action,

which is hypothetically enormous. Our main objective is to develop novel tools that will enable upregulation of the

expression of specific genes in their native context, aiming to employ these concepts in therapy.

Our main approach is to target regulatory areas of genes, called 3’UTRs.

JAAN-OLLE ANDRESSOO, PHD

Principle Investigator

Institute of Biotechnology

P.O. Box 56, Viikinkaari 5D

FI-00014 University of Helsinki, Finland

Tel. +358-2941 59394

GSM +358 503581213

[email protected]

biocenter.helsinki.fi/bi/andressoo

20 Photomicrograph by M.Sc. Kert Mätlik

2322

RNA toxicity results from mutations in genes

where a small region of just a few repeated

nucleotide-pairs is inappropriately amplified

in a repetitive manner. This leads not only

to loss of gene function but also, when

expressed, to toxic transcripts containing

the expanded repeats. Expanded repeat

RNA toxicity is increasingly recognized as

a cause of human degenerative disorders,

from Amyotrophic Lateral Sclerosis (ALS) to

Myotonic Dystrophies (DM), with a wide range

of symptoms that include muscle wasting,

weakness, cataracts, heart conduction

defects and insulin resistance. These

disorders are predicted to share mechanisms

that lead to cellular dysfunction and disease.

Currently they are managed only by early

detection and palliative treatment, costing

several billion dollars annually but with little

return in terms of improved quality of life.

Our research focuses on DM, affecting

at least 1 in 8000 people worldwide, and

constituting a paradigm for RNA toxicity in

disease pathogenesis. We have developed

an animal model of DM, using the worm C.

elegans, providing an opportunity for high-

throughput small-molecule screening to

identify compounds that rescue locomotor

function, and thus hold therapeutic potential

in DM and similar diseases caused by

RNA repeat toxicity. Hits should include

modulators of alternative splicing, RNA

clearance, and other cellular processes.

Pre-clinical, early stage drug discovery with

a whole-animal model offers the advantages

of “fast-fail”, since compounds that are

inherently toxic at the whole-animal level

will be immediately rejected. Compared with

traditional drug-screening platforms, our

approach is faster, more economic, and can

be automated with high-throughput scaling,

key components for the rapid identification of

new therapeutics. We have recently identified

a previously unknown genetic suppressor of

RNA toxicity, the nonsense-mediated mRNA

decay (NMD) pathway, which is conserved in

humans, allowing for rapid translation into

human cell-lines for further validation, with a

high expectation of success.

RNA toxicity has recently been implicated

as an additional pathological mechanism in

a subset of disorders previously believed to

result only from protein repeats, and our C.

elegans model system has detected common

modifiers of DM and one well-studied such

disorder, Huntington’s disease. This widens

the potential of our system in uncovering

modifiers of toxicity, common to a broader

range of neurodegenerative disorders.

The worldwide market for DM-related

therapies is estimated at approximately

$2 billion, but there are currently no

approved drugs. Thus, our approach

has high commercialization potential. A

provisional patent has been filed on our C.

elegans model and we are in the process of

establishing it as a platform to screen small

molecule libraries including FDA approved

compounds. In the second phase, we will

license leads to established pharmaceutical

companies that have expressed interest

in developing DM-targeted therapeutics,

reinvesting licensing revenue into additional

screens and disease-model development.

We are looking for partners interested

in investing in the identification of these

candidate molecules for commercialization.

Susana Garcia

Targeting RNA toxicity in degenerative disorders

Our research focuses on Myotonic Dystrophies, an RNA-based disorder affecting at least 1 in 8000 people worldwide.

Myotonic Dystrophy (DM) worms exhibiting one of the proteins disrupted in DM, tagged with red fluorescence - the splicing factor MBNL-1.

The toxic repeat-bearing RNAs expressed in DM worms can be detected as green fluorescence.

23

2524

Tommi Kajander

Synaptic adhesion molecules - making neuronal connections to combat cognitive disorders

Neurons form a vast network of connections

that underlie all brain functions. Synapses,

where one neuron connects with another,

depend on specific adhesion proteins.

They are required for synaptic assembly,

maturation and stability, and thus play a

central role in the formation and function of

the brain. Moreover, dysfunction of synaptic

adhesion molecules is linked to specific

cognitive impairments such as schizophrenia,

autism and bipolar disorder.

Our broad scientific goal is to dissect the

molecular details of how these proteins

interact, how they are regulated and

expressed, and what are the results of

their inhibition in vitro. This way we hope to

understand the formation and dynamics

of synaptic structures and decipher how

synaptic adhesion proteins are involved in

disease. A longer-term goal is to translate

this knowledge in to clinical use, by creating

tools to treat and eventually cure and prevent

cognitive disorders, which represent a huge

societal burden.

Synaptic adhesion molecules are able to

induce synapse formation by binding to their

receptors on the target cells, where synaptic

differentiation then begins. This involves the

accumulation of different adhesion proteins

and expression of ion channels on the

post-synaptic side, mediated by intracellular

organizer proteins such as PSD-95 or GRIP.

We currently focus on studying the molecular

structures and interactions of the synaptic

leucine rich repeat (LRR) adhesion molecules

and their pre-synaptic ligand-adhesion

receptors, protein tyrosine phosphatase

receptors and neurexins.

We are working to establish several

applications for recombinant synaptic

adhesion molecules in basic research,

together with a wide network of

collaborators. For instance, we are using

these proteins to screen for new ligands

and functions, by affinity purification and

proteomics. Using adhesion protein-

patterned surfaces, we will study their

effects on synapse formation and neuronal

connectivity in cell culture. Starting from cells

derived from patients, where dysfunction

of synaptic adhesion receptors is the

underlying cause of disease, we will use the

technology of induced pluripotent stem-cells

(iPSC) to build disease models that could

be used in future as a platform for drug

screening or for testing other personalized

therapeutic strategies. Finally, we plan to

conduct small-molecule screens to identify

compounds able to enhance or inhibit

synapse formation, specifically targeting the

activity of the LRR adhesion proteins. For all

of these endeavours, we are eager to identify

industrial and academic partners, as well as

funders.

24

27

Small Molecule Pharma

UNDERSTANDING MITOCHONDRIAL PROTEIN SYNTHESIS FOR BETTER TARGETING OF THERAPEUTICS | Brendan Battersby

NOVEL TARGETS TO COMBAT RNA VIRUSES | Sarah Butcher

LEUKOCYTE BETA2-INTEGRINS IN HEALTH AND DISEASE | Susanna Fagerholm

BMH-21, A UNIQUE ANTICANCER MOLECULE TARGETING POL I | Marikki Laiho

TWINFILIN AND CYCLASE-ASSOCIATED PROTEIN INHIBITORS FOR PREVENTING

CANCER INVASION AND CHEMORESISTANCE | Pekka Lappalainen

DISCOVERY OF SUBSTRATE-SELECTIVE SMALL MOLECULE PROTEOSTASIS MODULATORS | Ville Paavilainen

2928

The goal of my research programme is

to understand how biological circuits are

established in the cell for the synthesis of

the distinct subset of 13 proteins required

for aerobic energy metabolism. These

proteins are synthesised in a unique cellular

compartment, the mitochondria, which acts a

metabolic hub and energy generator for the

cell. We focus on discovering the molecular

mechanisms at key regulatory nodes in the

pathways of mitochondrial protein synthesis,

and the response to acute and chronic

disruptions, both of pharmacological and of

genetic origin.

Protein synthesis in mitochondria is

molecularly similar to that in bacteria. Since

various classes of antibiotics target protein

synthesis, a potentially toxic side-effect of

antibiotic usage is disruption of mitochondrial

protein synthesis, which can have profound

effects on human health. Identifying the

mitochondrial targets of antimicrobials is

therefore crucial to enhancing the specificity

of these molecules and reducing their toxicity

to humans.

We have established that the 13

mitochondrial DNA-encoded proteins are

synthesised in excess of demand so that at

least 80% of newly synthesised mitochondrial

proteins are degraded, thus minimizing the

errors that arise naturally during protein

synthesis and folding. The cell requires a

tightly regulated quality-control pathway

to ensure that this labile pool of potentially

harmful proteins is degraded and does not

over accumulate. My current research has

shown how genetic and pharmacological

disruptions to this protein turnover pathway

can be deleterious to the cell and is the

underlying molecular defect in a number of

inherited human diseases. Typically, these

quality control pathways are also upregulated

in cancer cells, where they may become

essential for cell survival.

Fundamental research in my laboratory is

thus generating a molecular understanding

of how acute and chronic disruption to the

quality control of mitochondrial protein

synthesis is linked to cellular function. This

paradigm will provide insight into how

mitochondrial homeostasis is disturbed

by different types of pharmacological and

genetic insults. On the one hand, this will

enable better targeted therapies to limit

such damage, which can be applied, for

example, in neurodegenerative disorders.

On the other, it can also be exploited as a

novel cytotoxic strategy, leading to more

precise treatments for cancer. Finally, it will

inform the development of next-generation

antibiotics, free from damaging side-effects.

Brendan Battersby

Understanding mitochondrial protein synthesis for better targeting of therapeutics

29

Mouse (skin cell) fibroblast stained for mitochondria (red) and alpha-tubulin (green). Mitochondria move throughout the cell on the microtubule cytoskeleton that is composed of alpha and beta tubulin. Image by Paula Marttinen

3130

Sarah Butcher

Novel targets to combat RNA virusesThough non-enveloped icosahedrally-symmetric ssRNA viruses are one of the largest groups of viral pathogens, their assembly

mechanisms are still poorly understood. The control of ssRNA viruses by vaccination is unlikely to ever be possible for all but a small

subset of viruses, so innovative routes to antiviral therapy are urgently required.

Our group has identified sequence- and structure-specific nucleotide motifs within the genome of a human picornavirus that play a

key role in virus assembly. Referred to as “packaging signals”, due to their cooperative interaction with the key structural viral proteins

(capsid proteins), these motifs overlap untranslated and coding regions. Viral assembly is therefore in competition with other functions

of the genome. Disrupting these high affinity packaging signal-protein contacts has deleterious consequences for capsid assembly, and

thus presents a novel antiviral drug target. For a given single-stranded RNA (ssRNA) virus species, the packaging signals share certain

common features that increase the number of target sites per virus per drug, and show a lowered genetic variation between strains.

Drugs targeting such sites will be highly disruptive to ssRNA virus assembly and will not be easily defeated by the development of

resistance. Thus, our strategy offers potential new therapies against clinically relevant viruses such as poliovirus, hepatitis C (significant

chronic infection), and rhinovirus (causing the common cold: economic costs annually in the tens of billions of dollars).

31

We have established a wide range of

experimental and theoretical tools for

the identification and characterization

of packaging signal function(s) in capsid

assembly, revealing a previously unsuspected

principle in the assembly of single stranded

RNA viruses. The implications have the

potential to transform our understanding of

the fundamental biology of these systems,

including the mechanisms of infection and

evolution.

The patent we have applied for relates to

targets for anti-viral agents that either mimic

or bind to packaging signals of RNA viruses

that function in viral capsid formation. It

describes pharmaceutical and plant viral

control compositions for use in the treatment

of viral infections; methods to treat viral

infections; and five methods to screen for

packaging signals in viral RNA genomes.

A second approach we employ aims to identify

the key interactions between pathogenic RNA

viruses and the human host, so as to describe

the molecular pathogenesis of the infections,

and identify novel targets for antiviral agents

and diagnostics. This approach combines a

systematic analysis of all molecular reactions

(interactome), with structure-function studies

on emerging viruses.

With an appropriate pharma partner, our

existing targets plus our capability to produce

additional targets could be harnessed

to deliver a powerful new range of anti-

viral treatments. We thus require further

investment for drug screening, mammalian

animal models and 3D cell culture, to progress

this technology to phase-one drug trials.

Image courtesy of Pasi Laurinmäki

3332

The immune system is of fundamental importance

for human survival as it functions to protect us from

disease. However, it also poses a significant threat to

the individual, as immune cell-mediated diseases such

as autoimmunity and allergy are becoming increasingly

common. We are interested in leukocyte beta2-integrins,

important cell surface receptors in leukocytes which

regulate both adhesion and signaling in immune cells.

Integrin-mediated cell adhesion, migration and signaling

is crucial for proper immune system function. Studying

these processes is therefore fundamental for our

understanding of immunity. Importantly, integrins are

also recognized therapeutic targets. Our group studies

the in vivo roles and regulation of leukocyte beta2-

integrins both in the healthy and the dysfunctional

immune system, using novel animal models. In addition,

we aim to elucidate the role of these receptors

in immune-related diseases such as allergy and

autoimmunity, and investigate their possible targeting

to modify immune responses in diseases such as cancer.

Susanna Fagerholm

Leukocyte beta2-integrins in health and disease

SPECIFIC PROJECTS:

Integrins and SLE. Certain integrin genetic variants are strongly associated

with systemic lupus erythematosus (SLE), an autoimmune disease with

a significant genetic component. By studying these variants and their

signaling capabilities in vitro and in vivo, we aim to reveal novel therapeutic

targets for autoimmune disease. The cell- and animal models we have

already produced, or aim to establish, will also be used in future drug-

development programs aimed at creating more specific therapies for

autoimmune disease, in a personalized medicine approach.

Integrins and cancer. We have recently revealed that integrins work as

a “brake” in certain immune cells, restricting their signaling, activation

and migration. By targeting integrins and/or their signaling pathways, we

therefore aim to modify immune cell behavior to optimize treatment of

disorders with an immune-related component, such as cancer.

32

3534

Marikki Laiho

BMH-21, a unique anticancer molecule targeting Pol ICurrent studies on cancer pathway alterations are focused mostly on cancer genome abnormalities, their impact on the cancer

phenotype, and how they can be targeted therapeutically. However, cancer cells also become dependent on specific cellular metabolic

pathways, which include the need for an increased rate of protein production. This ultimately depends on the synthesis of rRNAs,

governed by RNA polymerase I (Pol I), and on the program of ribosome biogenesis. The Pol I transcriptional machinery is highly

responsive to oncogenic stimuli and commonly deregulated in cancer. Yet, this program has not been viewed as a clinically relevant

target and very few attempts have been made to manipulate it.

Pol I has traditionally been considered to

have a housekeeping function that cannot be

interfered with, and thus to be undruggable.

No markers have been developed to assess

Pol I transcription rates that would support

clinical decision-making, and therapeutic

development towards Pol I is thus still in

its infancy. In response to the critical need

to develop Pol I-targeting drugs, and to

conduct proof-of-principle studies, we have

introduced a novel small-molecule compound

(BMH-21), that targets Pol I activity. This

represents a new therapeutic candidate in

cancer, with huge potential impact.

BMH-21, a unique

pyridoquinazolinecarboxamide, was

discovered in our high-throughput screen

for anticancer small-molecules. We have

demonstrated that it acts by blocking

Pol I transcription and brings about the

destruction of the catalytic subunit of the Pol

I complex. It has broad anticancer activity as

tested in the NCI60 cancer cell panel, with a

potent GI50 of 0.16 µM. BMH-21 is cell and

tissue permeable, orally bioavailable, and

has low toxicity in normal cells compared to

cancer cells. We have conducted structure-

activity relationship analyses of BMH-21

by developing over 40 analogs, and so far

identified several equipotent derivatives. The

physicochemical characteristics of the active

analogs define the activity of the molecule

in a tightly defined chemical space. BMH-21

demonstrates promising activity in preclinical

mouse models, with significant inhibition of

tumor growth in melanoma, prostate and

colon carcinoma models.

Given that Pol I deregulation occurs at very

high frequency in cancers, we predict that a

Pol I targeting approach could have a major

impact in many cancer types. To support this,

we are conducting mechanistic studies on

Pol I complex assembly and stability, aiming

to identify factors that mediate deregulated

Pol I transcription in different cancers. Via

chemogenomic profiling of BMH-21 activity,

we are attempting to identify factors that

sensitize cancer cells to Pol I inhibition. At the

same time, we are proceeding with target-

based chemical biology screens to identify

new Pol I inhibitors.

Our long-term goals are to develop Pol I

targeting, using BMH-21-like molecules,

as cancer therapies. Studying its activity in

combination with other cancer intervention

strategies should reveal further opportunities.

We have patents issued and pending on

BMH-21 and its derivatives in the US and

Europe. The work is a collaboration between

my labs in Helsinki and Johns Hopkins

Universities, with intellectual property jointly

owned, but managed by the Johns Hopkins

University Technology Transfer Office.

35

3736

Pekka Lappalainen

Twinfilin and cyclase-associated protein inhibitors for preventing cancer invasion and chemoresistanceWe use a combination of genetics, cell biology, and biochemistry to study the role of the cytoskeleton in cancer. Our main

aim is to elucidate the structural and biochemical basis of the cancer-associated functions of two key cytoskeletal proteins,

twinfilin-1 and cyclase-associated protein (CAP), which promote cancer cell invasion and chemoresistance.

Coordinated building and break down of the

actin cytoskeleton gives the cell the ability

to undertake many fundamental cellular

processes such as migration, morphogenesis,

adhesion, and cytokinesis. Defects in its

organization and dynamics are therefore

central in cancer. Progression to malignancy

involves the dysregulation of multiple actin-

driven processes. Consequently, several

actin-binding proteins have been directly

linked to cancer invasion and metastasis,

notably twinfilin-1 and CAP.

Twinfilins are highly conserved proteins that

regulate cytoskeletal dynamics in a variety

of cell-types. CAP is a large multifunctional

protein which promotes actin recycling.

Elevated expression levels of CAP and

twinfilin-1 are associated with many

cancers, whilst CAP depletion inhibits the

proliferation and invasion of breast-cancer

cells. These molecules have therefore

emerged as promising new targets for cancer

chemotherapy.

Our studies have revealed the molecular

mechanisms whereby twinfilins control actin

filament assembly. The mammalian non-

muscle twinfilin isoform, twinfilin-1, has been

linked to progression and chemoresistance

of breast cancer and lymphomas. Its

suppression delays lymphoma progression

in mice, and extends animal survival

following chemotherapy. Furthermore, it was

identified as the major functional target of

the regulatory micro-RNA miRNA30c, which is

already used as a prognostic marker in breast

cancer. Twinfilin-1 depletion dramatically

inhibits invasion and spread of breast-cancer

cells and makes them more sensitive to

chemotherapy agents.

These studies allow us to design screens to

identify specific twinfilin-1 and CAP inhibitors

from small compound libraries. These will

serve as useful proof-of-principle compounds

for developing new drugs for use in cancer

chemotherapy.

37

3938

Secreted and membrane proteins constitute approximately 30% of the human proteome and many are important drug targets. Yet, their targeting with small-molecule therapeutics has remained a daunting task.

Ville Paavilainen

Discovery of substrate-selective small molecule proteostasis modulatorsWe are investigating the fundamental question of how protein molecules are correctly targeted to their final cellular locations or

to the extracellular environment. This targeting is achieved by a host of cellular factors that recognize distinct sequence features in

newly-made proteins and deliver them to their ultimate functional destinations. We focus on dissecting and functionally classifying

the chemical information contained in these protein-targeting ‘zip codes’ as well as identifying the cellular machineries responsible

for protein transport to correct locations. Many diseases are associated with dysregulated protein transport and secretion.

Therefore, we are exploring the protein targeting zip code as a novel druggable motif amenable to small-molecule modulation.

About 30% of all proteins are secreted

outside of the cells or integrated into

cell membranes. Secreted and integral

membrane proteins enable cells to exchange

information and respond to their specific

environments. They act as signaling receptors

and enable transport of molecules across

membranes. Most importantly, they represent

the majority of current drug targets.

Our work in this area has two goals. First,

we aim to understand the biological roles

of the protein secretion apparatus and

how its function is altered in disease

states. We aim to functionally identify and

classify information contained in secretory

zip codes and build accurate tools for

predicting secretory protein function from

genomic sequence data. Such tools will

allow prediction of secretory protein loss-

of-function by scanning disease genome

sequences for point mutations. Secondly, we

are exploring the possibility of preventing the

production of specific, mislocalized proteins

associated with disease, by discovering potent

small-molecules with the ability to prevent

their biosynthesis. In this work, we explore

naturally occurring small molecules that have

evolved to target the particular branches

of the protein homeostasis machinery with

highly specific mechanisms of action.

We have thus far demonstrated the ability

of diverse natural product small molecules

to selectively prevent the formation of only

a subset of secreted proteins, and have

evidence that these compounds target

specific recognition elements of the secretion

machinery. We are currently working on

identifying new drug-like natural products and

natural product-inspired small molecules that

can prevent biogenesis of different subsets

of secreted or membrane proteins. We have

established a novel technology platform

for differentiating between functional and

non-functional secretory zip codes and are

using this system to collect vast amounts of

functional sequence data, which will allow

development of different sequence prediction

algorithms. In addition to allowing accurate

prediction of secretory protein function from

genomic data, this method will also act as

a powerful tool for identifying and profiling

mechanistically unique secretory protein

inhibitors with ability to prevent the creation

of different disease-associated proteins.

We envision that new substrate-selective

proteostasis modulators can serve not only

as powerful molecular probes, but also as

lead molecules for the development of future

therapeutics. These compounds would

have much higher selectivity than current

inhibitors of components of the proteostasis

machinery, such as the proteasome or

cellular chaperones.

39

41

Cell & Protein Technologies

CRANIOFACIAL REGENERATION: FROM EVO-DEVO TO THERAPEUTIC APPLICATIONS | Nicolas Di-Poi

“GO IIST” | Hideo Iwai

STEM CELLS AND AGING | Pekka Katajisto

DNA DREAM LAB – EFFORTLESS REALIZATION OF DREAM CONSTRUCTS | Konstantin Kogan

DECLINE OF THE DENTURE: USING STEM CELLS TO REGROW OUR TEETH | Frederic Michon

NOVEL PEPTIDES AND DRUGS TO DIRECT DIFFERENTIATION OF STEM CELLS | Osamu Shimmi

ACTIN-REGULATED TRANSCRIPTION IN CANCER AND LAMINOPATHIES | Maria Vartiainen

PROFILING CELLULAR QUIESCENCE TO DEVELOP NOVEL ANTI-CANCER DRUGS | Norman Zielke

4342

Understanding the key signaling pathways

of craniofacial tissue regeneration

in reptiles will allow us to design

preclinical tests of therapeutic

strategies in mammalian

models and human cell/

organ culture leading

to ground-breaking

clinical applications.

43

Nicolas Di-Poi

Craniofacial regeneration: from Evo-Devo to therapeutic applicationsDevelopmental & Stem Cell Biology has become a crucial field for the understanding of tissue regeneration and the implementation

of regenerative medicine. The ability to repair or regenerate tissue is a fundamental property present in all multicellular organisms,

but there is tremendous diversity in how this process occurs within vertebrates. In particular, adult mammals (including conventional

experimental models such as mice and rats) have limited regenerative capacities compared to other model organisms such as reptiles.

These offer one of the best examples of regeneration in vertebrates. Hence, studying such taxa is a priority for improving our general

understanding of tissue development, patterning and regeneration. Importantly, it should help identify new targets and/or new

therapeutic approaches for regenerative medicine.

Our laboratory is investigating the molecular

genetic basis of the development, evolution

and regeneration of different craniofacial

tissues (tooth, retina, brain) with unique

regenerative capacity in non-classical reptilian

models (lizards and snakes). Surprisingly,

in contrast to the long-lasting interest in

craniofacial development in mammals, and

the numerous studies resulting from it,

relatively little is known about the embryonic

development and adult regeneration of these

tissues in other vertebrate classes. Our focus

on craniofacial research is motivated by the

fact that craniofacial diseases and disorders

account for a considerable and increasing

portion of health problems worldwide. In

addition, craniofacial organs are perfect

targets for evolutionary and ecological

studies, because of their excellent fossil

record, and because they show high levels

of morphological variation and adaptive

innovation. This is of crucial importance in

understanding tissue development from an

evolutionary perspective, which will be needed

to promote regenerative capacity in humans.

Our research projects use a multi-disciplinary

approach that deploys state-of-the-art,

methods combining molecular embryology,

genetics, 3D imaging, phylogenomics,

morphometrics, and theoretical modeling.

The extensive analysis of reptilian models

helps us to understand why and how

regeneration in mammals is absent or

limited. In addition, by revealing the

molecular and cellular principles underlying

regeneration, they identify new targets and

approaches for the treatment of common

human disorders such as neurodegenerative,

retinal, and dental degenerative diseases.

Our systematic approach, combining

Evolutionary Developmental Biology (Evo-

Devo) with Regenerative/Stem Cell Biology is

unique not only in Finland, but worldwide.

Our recent data already indicate that reptile

studies carry enormous potential and high

relevance to common human diseases, since

reptilian tissue structural and functional

circuitry, neuronal and epithelial cell types, as

well as signaling pathways are exceptionally

well conserved (see, e.g., Di-Poi et al., Science

Advances 2016; Milinkovitch, Di-Poi et al.,

Science 2013; Di-Poi et al., Nature 2010).

4544

Hideo Iwai

“GO IIST”

The IIST system, standing for Ion Inducible Self-cleavage tag, is a new technology for protein

purification and ligation, with wide applications.

Every year, the number of peptide-, protein- and antibody-based drugs entering clinical

trials is steadily increasing. There is a pressing demand for more cost-efficient and effective

approaches to the production of peptide-based conjugates for this market.

The IIST system, based on patented technology developed by Dr Hideo Iwai in BI, University

of Helsinki, is a novel and cost-effective way of achieving this goal. Based on natural or

engineered protein-segments with self-cleavage activity, IIST can support many uses both

in basic research and in industry.

In a conventional approach to protein purification and conjugation, a tagged protein of

interest has to be treated with expensive proteases following the affinity-capture step.

These proteases also need to be removed in a final recovery process. IIST eliminates the

need for expensive proteases, by making use of the ion-inducible self-cleavage activity of

the IIST domain, located between the protein of interest and the tag. The final recovery

step needs only a re-use of the affinity column, since the only thing that needs to be

removed from the final product is the tagged IIST domain.

Applications include not only the purification of proteins and peptides, but also the

conjugation of protein and peptide fragments, peptide cyclization and amidation, and

antibody conjugation to drugs or immunotoxins. The conjugation step is enabled by IIST

because of its inherent protein-splicing activity.

The global market for protein therapeutics, including monoclonal antibodies, is expected to

reach $165 billion by 2019. To bring this new technology to market needs the involvement

of one or more commercialization experts or an ‘entrepreneur-in-residence’ to strengthen

the research team, plus the interest of industrial collaborators and potential investors.

The global market for protein therapeutics, including monoclonal antibodies, is expected to reach $165 billion by 2019.

44

4746

Pekka Katajísto

Stem cells and agingYoung stem cells renew tissues constantly,

but old stem cells can no-longer support

functional tissue renewal at the required

rate. The resulting decline manifests as aging.

Stem cells fail at their regenerative task

due to the damage they have accumulated.

We investigate the mechanisms that have

evolved to minimize damage accumulation in

stem cells, and how such mechanisms could

provide points for intervention in aging-related

diseases and even the aging process itself.

One possible way for nature to reduce

damage in a stem-cell lineage would be to

selectively apportion damaged components

away from the new stem cell during

asymmetric cell division. We have discovered

that stem cells do, indeed, harbour distinct

populations of intracellular organelles

of different ages, and that certain old

organelles, for example the mitochondria,

are asymmetrically segregated at stem-

cell division, and become enriched in

the differentiating daughter cell. In other

words, this novel mechanism potentially

ensures that stem cells retain young and ‘fit’

organelles, whilst purging themselves of old

and possibly damaged, cellular components,

which are selectively partitioned into the

differentiating cell lineage.

We have thus established a new scientific

paradigm of age-selective organelle

segregation. At the same time we have

developed a unique set of research tools to

label subcellular components in an age-

specific manner, and then follow their fate

inside live and dividing stem cells. Together,

this concept and toolkit enable us to address

important research questions that are

inaccessible to anyone else. Our current work

aims to determine how stem cells recognize

the age of their organelles, and how age-

selective segregation is regulated.

Once we have identified the molecular

mechanisms underlying this phenomenon,

we will address how to induce or modify

age-selective organelle partition, with the

following aims in view:

• clear away damaged components from

stem cells after harmful events

• rejuvenate old stem cells, so as to promote

renewal of old tissues

• reduce the risk of aging-related diseases

and, by combining these goals

• slow down the aging process itself

Our group is keen to attract funders,

investors and industrial partners to help us

turn our discoveries into viable technologies

to limit, delay and eventually reverse aging.

46

4948

DNA Dream Lab

Konstantin Kogan

DNA Dream Lab – effortless realization of dream constructsDNA Dream Lab (DDL) is a new initiative at BI that aims to serve life-science users, initially in the Helsinki area, but with the

potential to serve the whole of Finland and beyond.

DDL is a one-stop-shop facility for plasmid DNA. It is widely acknowledged that most genetic manipulations revolve

around plasmid DNA, since fast and accurate molecular cloning is at the core of almost any life-science project nowadays.

Our mission is to help both basic and industrial researchers in two major directions: consultation and implementation.

48

Plasmid DNA consultation services include

realization of customer needs by searching

for and designing optimal solutions, with

emphasis on the experiment where these

plasmids are going to be used. Simplicity,

price, and time are the criteria for selecting

the most suitable strategy. Plasmid DNA

experts perform the search using a number

of external service providers (e.g. gene

synthesis companies, plasmid depositories,

local and international cDNA collections)

as well as DDL’s own comprehensive and

constantly growing local depository. Our

deep knowledge and expertise in structural

and molecular biology and protein folding,

coupled with years of experience in

plasmid construction, form a solid basis for

high-quality design of plasmid DNA. Our

awareness of downstream applications and

our ability to obtain any required number

of plasmids of almost any complexity allows

for the specific design of constructs to

increase the success of our client’s further

experiments. This greatly impacts how the

experimental set-up is planned by a lab

researcher using our services.

Plasmid DNA implementation services

complements the plasmid DNA consultation

part. Here we use state of the art methods

in DNA cloning, most of which are sequence

and ligation independent (e.g. SLIC, RF,

Gibson Assembly, LCR, USER). To make the

service complete we also securely store

DNA collections of our customers, having

backups in three different locations, stored

in three different forms. Depending on the

design, we outsource some of our activities

to leading companies in gene synthesis, oligo

manufacturing, and sequencing.

Being a not-for-profit, academic organization

we are completely transparent in our

activities whether it is in design, production

or management, making use of open-source

technologies or outsourcing to licensed

providers at cost. We accept students to

implement their own cloning under our

supervision using our materials, protocols,

and instrumentation. Upon agreement with

researchers, we disseminate the plasmids

as well as all the available information; yet

keep private unpublished plasmids, or

those having patents pending approval.

We implement custom-made information

technology solutions to allow immediate

access, search, and retrieval of any publicly

shared item fast, easily, and cheaply, due to

concise and proper annotation of every single

plasmid, and automated storage systems.

We deliver ready-to-use plasmid DNA for any

experiment in any required amount or purity

to any place in the world.

We are looking for appropriate funding and

partnerships to expand and develop our

services.

5150

Frederic Michon

Decline of the denture: Using stem cells to regrow our teethHistorically, an important goal of developmental biology has been to obtain the knowledge needed to achieve the regeneration of tissues

and organs. Understanding developmental mechanisms, and the underlying networks of regulatory genes, is instrumental for future

tissue and organ bioengineering. We also need to understand the properties of the stem and progenitor cells and the cues that induce

their differentiation. Recent progress in stem-cell research has led to realistic bioengineering of tissues such as bone and cartilage.

However, to regenerate whole organs composed of several cell lineages is far more challenging and needs a deeper understanding of

organogenesis and the roles of different classes of stem cells.

Several organs in vertebrates develop as

appendages of the outmost layer of the

embryo, called the ectoderm. These include

hair, feathers, scales, nails, teeth, cornea,

lachrymal and mammary glands. Their

development shares similar molecular and

morphological mechanisms, leading to the

formation of functional organs. While some

ectodermal organs are heavily studied, less

is known about dental stem cells. The human

tooth is not a renewing or cycling organ.

However, stem cells have been identified in

the dental pulp and periodontal ligament,

which can give rise to hard dental tissues.

These stem cells are not able to generate

whole new teeth: this will undoubtedly

require us to understand their biology in

greater detail. Our current research, started

in Prof. Irma Thesleff’s lab, focuses on the

continuously growing front teeth of mice.

Their renewal is fueled by two specialized

stem-cell niches located at the base of the

tooth, providing an excellent model to study

dental stem-cell biology.

We aim to elucidate the genetic basis of dental

tissue identity, and the origin and lineages of

dental stem cells. As part of a pan-European

research collaboration, our goal is to establish

a clinically applicable tooth bioengineering

protocol. To achieve this, we need to find

out what determines the fate of dental stem

cells, and the genetic networks involved in

their maintenance and differentiation into

specific cell lineages. This will enable us to

design in vitro differentiation protocols that

are the crucial first step towards the dream

of dental bioengineering. Our main focus is

on the specification of the dental epithelial

tissues, responsible for the hard enamel

(tooth crown), and tooth attachment to the

jaw. Our current work is showing the crucial

role of the dental Sox2-expressing cells in the

neo-formation of teeth, ex and in vivo.

The anticipated results will help us

understand how dental stem-cells determine

tissue identity, and may be applicable to

other ectodermal organs. Even though dental

aberrations can nowadays be treated, the

loss of teeth still has a dramatic impact on

human health and self-esteem, and requires

a wide spectrum of expensive medical

interventions over the lifetime of a patient.

Our research will lead to improved clinical

treatment and prevention of dental defects,

in particular loss of teeth.

51

Photo courtesy of Jukka Jernvall

5352

Osamu Shimmi

Novel peptides and drugs to direct differentiation of stem cellsMembers of the Bone Morphogenetic Protein (BMP) family have been associated with many pathologies, including vascular diseases,

obesity, diabetes and cancer. Several BMPs have been shown to be clinically useful, and BMP2 and BMP7 have been approved for

treatment of spinal fusion, fracture healing and dental tissue engineering. The BMP2:BMP6 heterodimer has also been proposed for

use in vitro to promote the differentiation of embryonic stem cells (ESCs).

Implantation of BMPs together with

autologous stem cells has emerged as a

promising technique in tissue engineering,

aiming at the regeneration of various body

parts. However, large-scale production of

BMP ligands remains costly due to multiple

preparation and purification processes,

and clinically effective doses of such

recombinant BMPs are much higher than

their physiological levels. To overcome these

issues, development of a novel, “superBMP”

agonist is desired. One such way is to develop

ligands with improved bioactive properties

through increased solubility, stability and

receptor affinity.

Due to the fact that BMPs play crucial

roles in many different contexts in animal

development, their protein sequences in

mammals are highly conserved. We study

Drosophila BMPs to understand the ways

in which they have become functionally

optimized during evolution. The Drosophila

BMP5-8-like protein Scw is in a unique

position since it is exclusively expressed in

the early embryo and is evolutionarily highly

derived. Scw is required for generating peak

levels of the BMP gradient in Drosophila

embryos. Thus, its protein sequence appears

to have evolved to provide maximal biological

activity. We have also shown that post-

translational modifications of Scw modulate

BMP signaling activity both in vitro and in vivo.

We postulate that tolerance for structural

changes of BMPs is dependent on a range of

developmental and evolutionary constraints.

By employing optimized sequences through

natural selection of novel peptides, the

biological properties of BMP ligands can be

improved. Combined with our knowledge

of how evolution has shaped Scw, we aim

to develop a superBMP, as an efficient

and specific differentiation factor for stem

cells. Such a product should be applicable

for differentiation of stem cells into

chondrocytes, osteoblasts or cardiomyocytes.

This will involve not only optimization of the

peptide sequence, but also post-translational

modifications that can have a profound

effect on its potency, specificity, stability and

solubility.

We are looking for partner organizantions

to help us implement the various technical

steps in our project, enabling us to produce

and evaluate BMP variants based on our

predictions. These approaches will create and

develop novel products for commercialization

in the regenerative medicine sector.

53

5554

Maria Vartiainen

Actin-regulated transcription in cancer and laminopathiesActin operates in the cell’s cytoplasm as a key part of the cytoskeleton, which provides the cell its shape and enables movement. The

actin cytoskeleton is therefore critical for cell migration, which is a pre-requisite for cancer metastasis. Thus, it is not surprising that it is

deregulated not only in many cancers, but also other diseases, which show disturbed tissue architecture. In recent years, we discovered

that actin plays an important second role in cellular processes, inside the cell nucleus as well, where it influences gene expression. This

provides a mechanism whereby actin could contribute to disease development and progression independently of its direct effects on cell

structure and motility. Our data also supports the involvement of nuclear actin in laminopathies. These are a group of genetic disorders

caused by mutations in genes encoding proteins of the nuclear lamina, the fibrillar structure responsible for the internal organization of

the nucleus. Laminopathies display a large variety of clinical symptoms, such as skeletal and cardiac muscular dystrophy, lipodystrophy,

diabetes, and premature aging, progeria. One conceptual puzzle with laminopathies has always been the fact that, whist they mainly

affect specific tissues, the underlying genes are expressed everywhere in the body.

NUCLEAR ACTIN AND CANCER

The hallmark of cancer cells is unregulated

growth. However, most cancer deaths are

not due to the primary tumor, but rather

the spread of cancer cells, metastasis, to

other organs. In normal cells, cell growth

and migration are tightly regulated by many

signaling pathways and transcriptional

programs. Nuclear actin has been shown

specifically to regulate two transcriptional

programs critical for cancer progression:

the growth-regulating Hippo-pathway and

metastasis-regulating MKL1-SRF pathway. Our

preliminary data reveals extensive connections

between these two programs, and we are

examining how this crosstalk influences

cancer progression. By high-throughput

approaches, we aim to identify common

players and small molecules targeting both

pathways. This will not only shed light on the

basic mechanisms of cancer progression. It

will identify novel targets and molecules for

diagnosis and, importantly, a new generation

of more effective anti-cancer drugs.

NUCLEAR ACTIN AND LAMINOPATHIES

In collaboration with researchers from the

US, we demonstrated a novel mechanism

that could provide insights into the

cause of the cardiac phenotype in many

laminopathies. We showed that two

components of the nuclear lamina, lamin

A/C and emerin, modulate nuclear actin

and thus regulate gene expression through

the MKL1-SRF pathway. This regulation is

disrupted in laminopathies. Subsequently we

discovered a physical association between

the transcription coactivator MKL1 and

components of the nuclear lamina, and we

are currently analyzing their relevance for

MKL1-SRF-regulated gene expression. Here

too, high-throughput screening will help us

identify novel regulators and small molecule

targeting the pathway, and test their ability

to correct impaired MKL1–SRF signaling in

laminopathic cells. These approaches should

enable us to ameliorate the drastic cardiac

pathology associated with laminopathies.

PERSPECTIVE: We seek major funding to

support our high-throughput screening

pipeline, aiming to identify compounds to test

further in cell and animal models of cancer

progression and laminopathy, ahead of

clinical trials.

55

5756

Norman Zielke

Profiling cellular quiescence to develop novel anti-cancer drugsThe tissues and organs of multi-cellular organisms are comprised of specialized cell types, which originate from stem cells.

Stem-cell pools are typically heterogeneous in regard to many cellular processes, including transcription, cell signaling,

proliferation status, and susceptibility to apoptosis. Superficially minor differences between cells can have a profound impact

on cell fate. Heterogeneity is also found in most tumors, resulting in differential sensitivities to cytotoxic or anti-proliferative

drugs. Our work aims to develop and exploit tools that can discriminate between superficially similar cells that differ in key

properties such as proliferation status and drug susceptibility.

Understanding the characteristics of

quiescent stem cells has been hampered

by the rarity of these cells in many tissues.

To bypass this limitation, we are currently

developing a biosensor that facilitates

detection and purification of quiescent cells.

The isolated cells can be further analyzed by

various omics methods, thereby providing

novel insights into the transcriptome,

and the epigenetic status of quiescent

cells. In addition, this reporter system can

aid screening for novel, more effective

anti-cancer drugs. Many conventional

chemotherapeutics target key regulators of

cell proliferation. However, a large fraction

of the targeted cell-cycle genes are down-

regulated in quiescent cells, which may

explain why certain cells in a tumor are

resistant to drug treatment. Therefore, we

plan to use our biosensor-based system to

screen for compounds specifically targeting

quiescent cells, or aimed at releasing them

from quiescence to make them susceptible to

conventional treatments.

FUCCI (fluorescent ubiquitination-based cell

cycle indicator) biosensors are a promising

approach for studying cell proliferation at

single-cell resolution. By assigning specific

fluorescent labels to different cell cycle

stages, it provides a reliable readout for cell

proliferation that can be applied to live-cell

imaging or high-content screening. Combined

with flow cytometry it enables detailed

molecular characterization of isolated single

cells. We have recently extended this concept

to the fruit fly Drosophila melanogaster,

where it can be applied in conjunction with

the powerful genetic toolkit available in this

model organism. Since its introduction in

2014 Fly-FUCCI has proven to be a reliable

method for monitoring the proliferation

status of stem cells e.g. in the posterior

signaling center (PSC) of the Drosophila lymph

gland or the intestinal stem cells (ISCs) in

the regenerating midgut. We are currently

developing a next-generation FUCCI sensor

that allows robust detection of quiescent cells

in flies and mammalian cell culture.

We aim to develop this technology further

in two directions: first, we plan to use it to

assess how variability of the proliferation

status affects cell-fate decisions. This will

help us to understand how abnormalities in

cell-fate programming in stem cells can lead

to cancer or degenerative disease. Second,

it will support high-content screening in 2D

and 3D cell culture, as well as small model

organisms such as Drosophila, for drugs or

druggable targets that switch cells into or out

of quiescence, or target them specifically for

destruction.

Micrograph of an eye imaginal disc derived from the fruit fly Drosophila melanogaster, in which their characteristic

cell cycle pattern was visualized with the Fly-FUCCI (fluorescent ubiquitination-based cell cycle indicator) system

(Zielke et al. 2014). Cells residing in G1 phase were marked in green; cells in S phase were labeled red and cells in

G2 phase were labeled yellow. Image courtesy of N. Zielke and H. Lorenz (ZMBH – Imaging Facility).

59

Big Data

TOOLS AND SERVICES FOR ‘BESPOKE’ GENOME ANALYSIS | Petri Auvinen

PREDICTING SAFE AND NOVEL USES OF NANOMATERIALS | Dario Greco

BIOINFORMATICS SOLUTIONS FOR PRECISION MEDICINE | Liisa Holm

WASABI – WEB-BASED VISUALIZATION AND ANALYSIS OF COMPARATIVE SEQUENCE DATA | Ari Löytynoja

6160

Petri Auvinen

Tools and services for ‘bespoke’ genome analysis

Genome analysis requires a lot more than

just DNA sequencing, even though efficient

and accurate high-throughput sequencing

is a necessary starting point. Our group

combines this ‘bedrock’ service with

comprehensive bioinformatic analyses, using

tools also developed in-house for key steps

in the process, such as sequence alignment,

annotation and protein-function prediction.

Throughout the work, we establish and

maintain close interactions with the teams of

biologists behind the various projects that we

undertake.

We have created our facility primarily to

implement the assembly, annotation, and

functional and evolutionary analysis of

large eukaryotic genomes, building on early

experience as part of a wide collaboration to

determine the genome sequence of barley.

The first two examples of species whose

genomes have been fully analysed by our

group are the Glanville fritillary butterfly

(Melitaea cinxia; Nymphalidae) and the

silver birch (Betula pendula). Based on these

successes we are now involved in a number

of other genome projects, notably the ringed

seal, where our focus is the endangered

population unique to Lake Saimaa, and the

strawberry. In addition, we have developed

extensive expertise in metagenomics.

This has been applied to the gut and skin

microbiome in relation to disease, to food

spoilage, and to the analysis of environmental

samples, such as the microbial communities

in deep crystalline rocks.

This varied portfolio has enabled us to

build up not only a state-of-the-art toolkit

for genome analysis, but also the expert

knowledge required to interface and team-

up with a broad spectrum of biologists, so

as to extract information from genomes in

the ways most relevant to specific scientific

questions and real-world applications. This

has led to biologically significant findings. In

the case of the Glanville fritillary butterfly,

comparative genome analysis revealed a

thus-far unique evolutionary feature in the

Lepidotera, not seen in other groups of

animals such as mammals, whereby the

order of genes along the chromosomes

seems to be stable extending back at least

140 million years. The silver birch genome, on

the other hand, has been studied because of

its economic importance in boreal forestry.

Combined with appropriate field work, the

genome sequence opens up the possibility

of identifying the molecular basis of growth

rate regulation, wood quality and pest-

resistance. In the case of the Saimaa seal, the

emphasis is on conservation biology, and on

the bottleneck effect on speciation in a large

mammal. By correlating patterns of genomic

variation with specific biological traits we

hope to understand the molecular basis of

adaptation to locally changing environments

in a species under extreme threat.

Metagenomics is becoming an increasingly

important tool for detecting environmental

changes before they become irreversible.

It will also have an increasingly important

predictive role in medicine, as we learn more

about the interaction between microbial

communities and ‘host’ metabolism and

disease processes.

In the future, the expertise we have acquired

enables us to contemplate extending beyond

purely academic collaborations and launching

‘bespoke’ genome analysis as a commercial

service directly offered to animal or plant

breeders, wildlife conservation agencies,

healthcare providers and diverse other

organizations around the world. Thus, we are

in the early phase of a search for investors

and developers to identify the most relevant

markets and create a viable business out

of what hitherto has been curiosity-driven

research.

60

6362

Dario Greco

Predicting safe and novel uses of nanomaterialsNanoparticles are molecules with at least one dimension smaller than 100 nm. Nanotechnology is categorized as one of the ‘Key

Enabling Technologies’ in the EU’s current Research and Innovation program, Horizon 2020. The industry already employs more than

300,000 people in Europe, and the global market value of nanotechnology-enabled products is estimated at $2 trillion in 2015. These

products are already present in our everyday life and new Engineered NanoMaterials (ENM) are continuously produced. However, the

same physical, chemical, magnetic and electrical properties that make ENM extremely valuable, namely their high reactivity, are also

regarded as a potential threat to human health and the environment. The possible risks associated with ENM are a major impediment

to their adoption worldwide. Moreover, current safety testing of ENM is much too slow and laborious. This even applies to novel,

controlled and safe applications of existing ENM in the field of nanomedicine. Thus, there is a tremendous need to accelerate

hazard assessment and to predict the effects of ENM. On the other hand, some ENM effects could provide a great opportunity, for

instance, in the contexts of innovative therapeutic protocols. Hence, it is essential to understand nano-bio interactions in depth and at

multiple levels, in order to maximize ENM usability as well as safety.

The focus of our research group is

predictive and systems nanotoxicology. We

use cutting-edge genomic technologies,

artificial intelligence methods and systems

biology approaches to formulate predictive

computational models of ENM toxicity and

biomedical usability. OMICs technologies

allow us to thoroughly characterize the

effects on human cells of exposure to ENM.

Novel bioinformatics methods are then

developed to integrate the experimentally

derived big data-sets, with the aim of

characterizing the mode of action of ENM

and revealing relevant biomarkers for use in

future regulatory applications. The respiratory

system is the main exposure route for

humans, especially in work environments,

and is thus the context for one of our most

important current research projects.

Our group is currently completing the

development of the first such computational

tool, INSIdE nano (Integrated Network of

Systems bIology Effects of NANOmaterials),

allowing systematic contextualization of

the mode of ENM action with the respect

to interactions with other chemicals,

ability to cause diseases, and prospective

combinatorial use with drugs. Thus, we

aim not only to predict hazards, but also to

enhance existing therapeutic strategies.

Our long-term goals are (1) to develop

procedures for rapid, large-scale hazard

assessment of ENM; (2) to formulate

predictive algorithms capable of indicating

the potential adverse effects of ENM, based

just on the knowledge of their intrinsic

chemico-physical properties; (3) to produce

computational tools for the systematic

prediction of ENM uses in broader biomedical

contexts.

Screenshot from our tool INSIdE nano tool depicting the network of interactions that Parkinson’s disease (PD, purple) have with chemicals (eg. the known dopaminergic neuron toxin MPTP, green), drugs (eg. Levodopa the preferred drug for the treatment of PD, red) and nanomaterials (blue). INSIdE nano is helping scientists and regulators to formulate hypotheses concerning the involvement of nanomaterials in pathogenesis of specific types of diseases (as in this example), or it could otherwise suggest a possible use of nanomaterials as a treatment for diseases.

6564

Liisa Holm

Bioinformatics solutions for precision medicine

Post-operative complications caused

by microbial pathogens in the wound

occur in approximately 2% of all surgical

interventions. In the US, they claim about

8,200 lives annually, and their societal cost is

estimated at $1.6 billion.

One of the most essential steps in the

diagnosis and management of surgical site

infections and other healthcare-associated

infections is the prompt, accurate, and

reliable identification of the etiological

agents. To date, however, pathogen

detection from clinical samples has relied

heavily on culture-based approaches and

phylogenetic marker gene identification that

are narrow in scope and unreliable for the

characterization of many pathogens.

These limitations in conventional diagnostics

techniques may compromise patient health

and delay the initiation of the correct

antimicrobial therapy. The consequences

are a matter of life and death for individual

patients, including the danger of long-term

disability and decreased life quality. At the

societal level, the deficiencies in pathogen

recognition have contributed markedly to

the favoring of untargeted, broad-spectrum

antibiotics in infection control, an extremely

detrimental approach in the era of increasing

antimicrobial resistance. To facilitate more

appropriate treatment and to minimize

the unnecessary use of broad-spectrum

antibiotics, it is imperative to develop novel

techniques for identification of infectious

pathogens and their virulence and antibiotic

resistance determinants.

Our project aims to advance our

understanding of the etiology of surgical

infections and develop cost-effective tools

for early recognition and customized

treatment. As a pilot project in personalized

medicine, it combines top expertise in

microbiology, computational biology, surgery

and clinical wound management. We employ

cutting-edge DNA sequencing, healthcare

informatics, and bioinformatics algorithms

to define the relationship between infections

and the microbiota residing in surgical sites.

Our group coordinates and implements the

computational aspects of the project. Our

expertise is in developing bioinformatics

analysis pipelines that can annotate big

data sets, as generated by metagenomics

(typically hundreds of gigabytes) in a clinically

relevant time frame. This is accomplished

using in-house algorithms deployed in

superfast sequence database search servers.

Our approach belongs to a new generation

of algorithms which is orders of magnitude

faster than BLAST, the industry standard.

Importantly, these algorithms scale better

because search time is independent of

database size, enabling the processing

of larger quantities of data, against

exponentially growing databases, with the

same computer power.

We have tested our computational pipeline

on test samples from patients and spiked

controls, and shown that it identifies the

potentially infectious agents.

The project is a wide collaboration between

BI, clinical and non-clinical departments, and

hospital services. As a spin-off it will establish

novel molecular tools and a tailored pipeline

for analysing microbial community genomics

data. It thus has substantial potential for

improving the quality and cost-effectiveness

of healthcare, and should be of interest to a

wide and global market.

The Overview

SURGICAL SITE INFECTION INFECTION INFORMATION

• Wound biopsies

• Swab Samples

• Patient information

METATRANSCRIPTOMICS SAMPLE PROCESSING

• Homogenization

• Extraction of total RNA

• DNAse I digestion

• Bead-based enrichment of bacterial RNA

RNA-SEQ LIBRARY PREPARATION

• Ribosome depletion

• Fragmentation

• Adapter ligation

HIGH-THROUGHPUT SEQUENCING DATA GENERATION

DATA AND SPECIMEN COLLECTION FUNCTIONAL ANALYSIS

• Species/strains

• Antibiotic resistance

• Virulence factors

SECONDARY ANALYSIS

• Feature counting

• ORF mapping

• ORF calling

PRIMARY ANALYSIS

• rRNA removal

• Human mRNA removal

• Quality trimming

• Adapter removal

Infected surgical siteUninfected surgical siteBurn wound

6766

Ari Löytynoja

Wasabi – Web-based visualization and analysis of comparative sequence data

Wasabi, developed by our team in BI, is a web-based

environment for analysis and visualization of comparative

sequence data. The Wasabi analysis environment is versatile

and adapts to diverse tasks in molecular genetics: for

example, the phylogenetic profiling of human mutations,

comparative analyses of microbial protein fragments,

amplicon analyses of marker genes or epidemiological studies

of pathogens. Wasabi runs inside a web browser and does

not require any installation: it can be immediately accessed

and implemented, via wasabiapp.org. The Wasabi visualization

library is a self-contained package for web-based display of

sequence data. Wasabi windows can be embedded in web-

pages (see figure) and in web-applications, and allow live,

browsable views of underlying data and fast data manipulation

(translation, filtering, text search), performed locally within the

browser. A prototype of embedded Wasabi visualization for

scientific publishing is shown at wasabiapp.org/journal.

Wasabi represents a specific output from our work, serving

the academic community. As such, the Wasabi environment

is based on open-source software and wasabiapp.org is

available to all as a free service. However, for users requiring

strict confidentiality or having special needs, custom services

can also be provided.

67

These can include:

• in-house installation of Wasabi software

with user training and support

• extension of the Wasabi environment

with custom analysis tools

• construction of reference data sets

(e.g. microbial protein families,

pathogen marker genes)

• embedded data browser,

e.g. for scientific journals or

sequence databases

Wasabi data visualization is currently

integrated in the Ensembl genome

browser (ensembl.org). Our aspiration

is to develop these extended services

on a commercial basis, serving diverse

business communities, such as publishers,

pharmaceutical companies, healthcare

providers or public agencies.

For further details, see the videos and

our publication describing Wasabi at

wasabiapp.org/about.

Live, browsable Wasabi window embedded on a web-page, showing cetacean-specific changes in the haptoglobin gene, potentially preventing hypoxia-induced cell damage as an adaptation to deep diving.

69

Plants & Food

MOLECULAR ENGINEERING THE FOREST: RATE AND QUALITY OF WOOD FORMATION | Yrjö Helariutta

WARMING TREES TO COMBAT GLOBAL WARMING | Howy Jacobs

REGULATING GENE EXPRESSION AND HORMONAL SIGNALING TO BOOST

BIOMASS PRODUCTION IN TREES | Ari-Pekka Mähönen

A TOOL FOR IMPROVING THE EFFICIENCY AND TARGETING OF GENE MODIFICATIONS IN PLANTS | Alan Schulman

A ROBUST RESISTANCE GENE FOR YELLOW (STRIPE) RUST | Alan Schulman

7170

Yrjö Helariutta

Molecular engineering the forest: rate and quality of wood formationThe aim of our research is to understand molecular mechanisms controlling plant vascular development, and use this knowledge to

develop ways of enhancing economically useful traits in forest trees. The plant vasculature, which comprises the conducting tissues of

phloem and xylem, provides structural support to the plant and enables the transport of water and sugars between shoot and root.

Wood, the secondary xylem of plant stems, represents most of the carbohydrate biomass of the plant, and its composition is under

genetic control. Our research thus has immense potential for the forest industry: the better we understand the molecular mechanisms

controlling wood formation and composition, the easier it is to apply this knowledge. The molecular regulators that we have identified

represent optimal target genes for tree breeding and forest biotechnology.

In our labs, we explore the molecular basis of

vascular development in two complementary

model systems: herbaceous Arabidopsis

thaliana and woody forest trees. Despite

its small size, Arabidopsis produces wood

and has emerged as a versatile model for

plant vascular development. Based on our

Arabidopsis research, we have identified a

wealth of novel regulators of wood production.

Using both transgenic and non-transgenic

gene editing methods, we can translate

this knowledge to modify the analogous

developmental processes in a tree trunk.

Besides boosting biomass production in

forest trees, we aim to optimize its properties

for various industrial purposes. Our current

focus is to modify lignocellulosic plant

biomass by adding callose to cellulose-rich

cell walls. Both callose and cellulose are

glucose polysaccharides, but with strikingly

different structures: in contrast to cellulose,

callose does not form recalcitrant microfibril

structures. By modifying the function

of a callose synthase enzyme, we have

already engineered transgenic plants for

overproduction of callose. Callose-enriched

wood should require less energy to liberate

simple sugars from cell walls and thus

significantly reduce the costs of industrial

processes that use the released sugars, for

example, to yield biofuels.

PERSPECTIVE: Our ultimate aim is to

engineer forest trees, so as to optimize

biomass production to yield high-value

products in the biorefineries of the future.

70

7372

Howy Jacobs

Warming trees to combat global warmingThe Boreal Forest is one of the world’s major carbon sinks. Accelerating its growth or extending its range would contribute

significantly to decreasing net CO2 emissions. When combined with sustainable forestry, it could result in increased

production of renewable fuels and many other products that today are sourced primarily from fossil materials.

We are working on a strategy to address

the challenge of accelerating tree growth,

by taking advantage of a natural system

present in all plants, the alternative oxidase,

AOX. AOX buffers stress and overload in the

mitochondrial energy system, by serving as

a by-pass that conducts electrons released

by biological oxidations directly to oxygen.

In so doing, it releases energy as heat,

rather than as ATP. This pathway is used

naturally by some plants to generate heat,

for example, by arum lilies to volatilize insect

attractants. AOX activity is induced under

extreme cold in many plants, and may play

a role in resistance to freezing. However,

in the model plant species Arabidopsis,

engineering AOX over-expression has already

been shown to promote growth of the plant

even at the more temperate condition of 15

°C. In effect, some of the energy captured

from sunlight by photosynthesis is being

‘wasted’ to produce heat. But the net effect

on biomass production is strongly positive,

because of the temperature-dependence of

the chemistry of biosynthesis.

Extending this concept to tree species

means that we can realistically contemplate

engineering trees to over-express AOX,

thus generating heat that will promote their

growth at sub-optimal temperatures, such

as those encountered globally across the

far north, where the majority of the world’s

remaining forests are located.

Previous studies have shown that the growth

of the major tree species in the Boreal Forest,

notably Scots Pine, Norway Spruce and Silver

Birch, is strongly influenced by temperature

during the growth season, and this effect

is most pronounced at environmental

temperatures encountered near the northern

limits of the range. Ironically, global warming

itself has already had a demonstrable effect

on tree growth in Finland. The advent of

gene-editing technology now makes it

theoretically feasible to engineer AOX over-

expression in commercial trees, promoting

greatly accelerated growth, increased carbon

capture and, in the longer term, increased

production of renewable raw materials.

Our research group is now looking for

investors and commercial partners

to accelerate this project, developing

and licensing this technology for use in

commercial forestry.

73

7574

Ari-Pekka Mähönen

Regulating gene expression and hormonal signaling to boost biomass production in trees

Wood (or xylem) constitutes a large fraction

of global biomass. It is produced in plants

by a tissue known as the vascular cambium,

the cylindrical secondary meristem located

in stems and roots, that serves as a stem-cell

compartment for producing the adjacent

cell layers. Cambium activity is particularly

important in tree trunks. Understanding the

molecular regulatory mechanisms directing

cambial development brings us powerful tools

to enhance the biomass production of trees,

formed as the lignocellulosic carbohydrates

that we recognize as wood. Work by us

and others has revealed a central role for

the plant hormone cytokinin in promoting

cambial development in the model plant

species Arabidopsis thaliana and in the

poplar (Populus tremula x tremuloides).

Following up our earlier studies on cytokinin

and the cambium, my group has identified

downstream factors of cytokinin signaling

whose function is specifically to regulate

cambial development. Overexpression of

these factors in Arabidopsis resulted in

increased secondary growth. Our aim is to

translate this knowledge to tree species,

where it could have profound economic and

environmental benefits.

Our next step in this project is to overexpress

the Arabidopsis cambial factors in the

cambium of poplar, and select lines with

increased secondary growth and production

of lignocellulosic biomass. This approach

will serve as a proof-of-concept step to

show the potential of these factors to breed

commercially important tree species.

In Finland, silver birch (Betula pendula) has

emerged as an intensively studied tree

species because of its suitability for genetic

breeding programs, plus its economic

importance in Finland and for the entire

boreal forest. Using available genome editing

techniques (CRISPR/Cas9) we will next target

birch homologues of the Arabidopsis cambial

genes, to generate both gain-of-function

and loss-of-function mutations predicted to

enhance the response to cytokinin signaling.

Genome editing techniques, combined with

our specific knowledge of the mechanisms

promoting cambial development, will enable

us to breed birch varieties with increased

biomass production or altered wood quality

characteristics much more rapidly than any

classical selection-based techniques.

We plan also to initiate discussion with

forestry companies, in order to collaborate

on the use of poplar lines already engineered

for overexpression of cambial factors, or to

generate new overexpression lines in other

economically important species cultivated

globally, such as Eucalyptus.

Our long-term goal is to use the information

provided by our studies in model species

to generate tree varieties with enhanced

production of lignocellulosic biomass, or

improved wood quality characteristics. This

has the potential to revitalize forestry in

Finland and elsewhere, helping to address the

challenges of climate change and the need to

switch to renewable, biologically sourced raw

materials.

75

7776

Alan Schulman

A tool for improving the efficiency and targeting of gene modifications in plants

Production of transgenic plants for

agriculture is increasingly important

worldwide; 182 million hectares of GM

crops were planted in 2014, with an annual

increase of 3-4 %. Soybean (79% GM), maize

(32%), cotton (70%) and rapeseed (24%)

have the greatest proportion of total area

in GM varieties. However, whilst wheat and

barley are both in the world’s top four cereal

crops, no GM barley or wheat is grown. One

limitation is that only a few varieties are

transformable; frequencies above 25% are

seldom achieved in barley, and are far lower

in wheat. Another, more general problem is

the lack of a targeting mechanism to direct

the transgene to a particular site in the

genome. With current methods, a transgene

may be integrated anywhere. Given the

high proportion of heterochromatin in

barley, wheat and other large genomes,

most insertions occur in areas with poor

expression. As a result, many transgenic lines

must be produced in order to recover just

one with suitable expression levels. There

is also the issue of silencing due to position

effects, which can occur even if transgene

expression is initially satisfactory.

A solution to this problem is provided by

retrotransposons and retroviruses, nature’s

genetic engineers, which insert their own

genomes into the DNA of the cell as part of

their natural life cycle. Retrotransposons are

abundant, non-infectious components of all

plant genomes, carrying out their transposition

using an enzyme called integrase. We have

studied the retrotransposon BARE, found in

both barley and wheat, as well as in related

cereals, for over 25 years, accruing a large

body of knowledge of its life-cycle and

integrase function.

We propose to develop BARE integrase as a

commercially viable transformation vector for

the targeting of transgenes to specific locations

in cereal genomes. Although the integrase

normally inserts its substrate semi-randomly

throughout the genome, specificity can be

achieved by tethering the integrase to another

protein with DNA or protein-binding capacity.

For example, linking integrase to a particular

transcription factor will lead to integration

wherever in the genome that transcription

factor binds. Proof-of-concept has been

achieved using retroviral (e.g. HIV-1) integrases

to create targeted integrations for gene

therapy. Investment is now needed to develop

tethered integrases and transformation

complexes for cereal genome modification,

extending this proof-of-concept to BARE.

The resulting technology would offer many

benefits. It could be used as a proprietary

system for in-house transformation, offered

as a service for plant breeding companies,

or licensed further. It could in principle be

tailored for targeted integration of transgenes

in any crop species where BARE integrase

will function. Because integrases are highly

conserved, this is likely to cover a wide

species spectrum, certainly including the

Triticeae (wheat, barley, rye, timothy) and

Pooideae (e.g. oats).

Competing technologies are problematic.

Homologous recombination for specific

targeting is possible, but is extremely inefficient

and needs selection. Specific removal of

genes has been reported with the piggyBac

insect transposon. The CRISPR/Cas9 system

is becoming widely used for site-specific

mutagenesis, but still depends on homologous

recombination for targeted insertion. Thus,

tethered BARE integrase should have a secure

place in a potentially vast market.

76

7978

Alan Schulman

A robust resistance gene for yellow (stripe) rust

Wheat is the world’s second most important

grain crop, planted on 219 million Ha

and yielding 716 million tons worldwide

(FAOSTAT, 2013). Wheat supply and demand

has seesawed over recent years, with eight

seasons of surpluses and 12 of deficits

over the last two decades. As witnessed

by the North African food riots of 2008,

political stability is closely linked to wheat

prices, which are in turn affected by wheat

production. The yellow (or stripe) rust disease

is today one of the most signficant threats to

wheat production. It is caused by the fungus

Puccinia striiformis f. sp. tritici, the spores

of which can be wind-dispersed over long

distances, even thousands of kilometers.

While the best strategy to overcome

the disease is to breed resistant wheat,

the pathogen has repeatedly developed

immunity against available resistance genes

by evolving increased virulence.

Wild emmer wheat, Triticum dicoccoides,

is an important source for novel yellow-

rust resistance (Yr) genes, which provide a

potential solution. Yr15, a major gene located

on chromosome 1BS of T. dicoccoides, was

previously reported to confer resistance to a

broad spectrum of stripe-rust isolates, at both

seedling and adult plant stages. Introgressions

of Yr15 into cultivated T. aestivum (bread

wheat) and T. durum (pasta wheat) have been

widely used since the 1980s, but remain

problematic, due largely to associated “linkage

drag” of undesirable flanking genes.

We have been working on a long-term project

to clone Yr15, using a variety of genetic and

genomic methods. We currently have a

candidate gene identified and under testing

for verification. The candidate gene is not a

typical NBS-LRR type resistance gene, which

explains why the resistance conferred by Yr15

has, unusually, not been broken in the 30

years since it was first used. Final confirmation

of the Yr15 gene identity will require a

combination of knockout and gain-of-function

strategies. Knowledge of the molecular details

of the resistance gene (sequence, control

of expression, natural variation) will enable:

1) direct transfer of the gene without the

problem of linkage drag; 2) editing of the gene

for introducing new variants to overcome

any emerging resistance; 3) potential design

of new resistance genes for other crops and

fungal diseases or direct use of Yr15 in barley

and related cereals.

The confirmed Yr15 gene can be made

available on a commercial basis for in-house

or licensed use by plant breeding companies.

Its benefits will include the development not

only of improved disease-resistant bread-

and durum-wheat varieties, but of fungal

pathogen-resistant strains of other crops.

The Yr15 gene has no current competition,

as it is the only gene that confers sustained

resistance to all known yellow-rust isolates.

Other resistance genes tend to lose

effectiveness over time, as the fungal strains

evolve. Thus, Yr15 can provide a unique and

robust answer to a major scourge of world

agriculture.

79