the human microbiome: time for effective...

26
The Human Microbiome: Time for Effective Translation March 29-30, 2018 California NanoSystems Institute, UCLA, Los Angeles

Upload: vuonganh

Post on 21-Apr-2018

254 views

Category:

Documents


13 download

TRANSCRIPT

Page 1: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

The Human Microbiome: Time for Effective Translation

March 29-30, 2018

California NanoSystems Institute, UCLA, Los Angeles

Page 2: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac
Page 3: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

About French American Innovation Days

French-American Innovation Days are events organized by the Office for Science and Technology of the

Embassy of France in the United States. They are high-level events where researchers and companies have the

opportunity to exchange views on a specific technological issue, start co-operative activities and develop business

transactions with a transatlantic perspective.

The goal of the program is to facilitate the development of innovation ties between France and America by bringing together

scientists, companies and other interested stakeholders from both countries.

The first FAID to be held in Los Angeles is entitled “The Human Microbiome: Time for Effective Translation”.

Microbiome research has developed exponentially in recent years and is moving from a descriptive state to one where

causality is now being examined. At the same time, almost all major pharmaceutical or nutrition companies, and a huge

number of start-ups, have invested in this field. Altogether, this is a very appropriate timing for the Los Angeles FAID

that aims to bring together scientists and companies to discuss effective translation.

Organizers

The Office for Science and Technology (OST), a team of 24 staff

members including professors, senior researchers and engineers

located in the Embassy (Washington, DC) and 6 consular offices

(Atlanta - Boston - Chicago - Houston - Los Angeles - San Francisco)

is dedicated to bilateral French-American collaborations in Science

and Technology.

The California NanoSystems Institute (CNSI) is an integrated

research center whose mission is to create a collaborative, closely-

integrated and strongly interacted environment that will foster

innovation in nanosystems research and education and bring scientific

and technological innovation into the economy and society.

Page 4: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Our Sponsors

Danone’s mission is to “bring health through food to as many people as

possible.” by helping people to adopt healthier choices and lifestyles, caring

about the health and well-being of its company and employees, of the

communities it serves, of the planet, and of current and future generations.

The Institut National de la Santé et de la Recherche Médicale is a French

public research institution focused on human health and medical research.

Abbott is dedicated to translating science into lasting contributions to health

through diverse products that encircle life, from newborns to aging adults, from

nutrition, and diagnostics through medical care and pharmaceutical therapy.

Boehringer Ingelheim is a research-driven pharmaceutical company that

creates innovative medicines for the areas of human pharmaceuticals, animal

health, and biopharmaceutical contract manufacturing.

Pfizer is a pharmaceutical company which seeks to make the best use of its

partnerships with non-profits, governments, and foundations in order to

identify and allocate the most important resources to underserved communities

around the world.

Microbiome Labs is an organization focused on providing probiotic

bacteriotherapy. Their goal is to provide the public with integrative solutions

and clinical research data to address digestive and immune health issues.

Kellogg’s is an American multinational food manufacturing company that

produces cereal and convenience foods. Their products are manufactured in 18

countries and marketed in over 180 countries.

Knobbe Martens is an agent of innovation, providing clients worldwide with

forward-focused Intellectual Property and Technology law service and

representation. They are one of the largest and fastest-growing IP law firms,

with more than 300 attorneys and scientists representing the complete spectrum

of technologies and IP practice areas.

Page 5: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Scientific Organizers

Pascale Cossart

Pascale Cossart, after studying chemistry in Lille (France) obtained a master degree at Georgetown

University, Washington, DC. Back in France, she obtained her PhD in Paris in the Institut Pasteur

where she is still now, heading the « Bacteria-Cell Interactions » unit which is also an Inserm and an

INRA unit. After studying DNA-protein interactions, she started to study the molecular and cellular

basis of infections by intracellular bacteria taking as a model the bacterium Listeria monocytogenes.

Her research has led to new concepts in infection biology but also in microbiology, cell biology and

epigenetics.

Pascale Cossart is considered as a pioneer in Cellular Microbiology. Her contributions have been recognized by a

number of international awards, including the Robert Koch Prize (2007), the Louis Jeantet Prize for Medicine (2008), the

Balzan Prize (2013). She is a member of the French Academy of Science (2002), a foreign member of the American National

Academy of Science (NAS) (2009), of the German Leopoldina (2001), of the Royal Society (2010), and of the American

National Academy of Medicine (NAM) (2014).

Jeff Miller

Jeff F. Miller, Ph.D., studies molecular mechanisms of bacterial pathogenesis and the evolution of

functional diversity in bacteria and phage. He received his bachelor’s degree in Chemistry from Case

Western Reserve University and his Ph.D. in Molecular Biology from Tufts Univ. School of Medicine.

After postdoctoral training with Dr. Stanley Falkow at Stanford, he joined the faculty at UCLA in

1990. From 2002-2014 he held the M. Philip Davis Chair in Microbiology and Immunology and served

as Chairman of the Department of Microbiology, Immunology and Molecular Genetics. In November,

2014, he was appointed as the Fred Kavli Endowed Chair in NanoSystems Sciences and Director of the

California NanoSystems Institute at UCLA. In 2004, Dr. Miller co-founded AvidBiotics Corp., a biotherapeutics company

in South San Francisco. In 2009 he was appointed by the Secretary of Health and Human Services to serve on the National

Science Advisory Board for Biosecurity and he is a voting member of the Board. From 2008-2010 he was Chair of the

General Meeting of the American Society for Microbiology (ASM), and from 2012-2014 he served as President of ASM,

which represents 40,000 members in the US and abroad. Dr. Miller is a former Pew Scholar in the Biomedical Sciences, a

member of the American Academy of Microbiology, a fellow of the American Association for the Advancement of Science,

and in April, 2015 he was elected to membership in the National Academy of Sciences

Jean Rosenbaum

Jean Rosenbaum joined the Office for Science and Technology of the French Embassy in Washington

in September, 2016 as Scientific Attaché specialized in Life Sciences. His office is based in the French

Consulate in Los Angeles, California.

Before occupying this position, he was trained as a Medical Doctor, then worked as a research scientist

for the National Institute of Health and Medical Research (Inserm, France). His first appointment was

in the Paris area (Créteil, 1990-1995). Then in 1995, he moved to Bordeaux where he became the head

of an Inserm laboratory (1995-2016). He was also the Director of the Department of Life Sciences and

Health of the University of Bordeaux (2015-2016) and worked as scientific officer at the French High Council for the

Evaluation of Research and Higher Education (HCERES, 2011-2016).

Jean Rosenbaum’s research dealt mostly with liver diseases. He contributed to over 120 scientific publications.

Page 6: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Agenda

Day 1 – March 29, 2018

8:00 AM Welcome breakfast

8:30 AM Opening remarks

Christophe Lemoine, Consul General of France in Los Angeles

Minh-Hà Pham, Counselor for Science and Technology of the Embassy of France in the United States,

Washington DC

Jeff Miller, Director of the California NanoSystems Institute, UCLA

8:45 AM Keynote lecture: Sarkis Mazmanian, California Institute of Technology – Introduced by Pascale Cossart

“Influence of the Gut Microbiome on Autism Spectrum Disorder”

Session I: Microbiomes & host physiology Moderator: Suzanne Devkota, Cedars-Sinai Medical Center

9:30 AM Serguei Fetissov, Inserm-Université de Rouen, co-founder of TargEDys SA, France

“Gut Bacteria and Host Feeding Behavior”

10:00 AM François Leulier, Lyon Functional Genomics Institute, CNRS-ENS-INRA-Université Claude Bernard, France

“The Microbiome and Linear Growth Promotion: Lessons from Gnotobiotic Animal Models”

10:30 AM Coffee Break - Poster viewing - Networking

Session II: Immunity, inflammation & pathogen resistance Moderator: Suzanne Devkota, Cedars-Sinai Medical Center

11:00 AM Yasmine Belkaid, National Institute of Allergy and Infectious Diseases, Bethesda

“Control of Skin Immunity and Inflammation by the Microbiota”

11:30 AM Pascale Cossart, Pasteur Institute-Inserm-INRA, France

“Intestinal Microbiota and Infection: the Listeria Paradigm”

12:00 PM Eric Pamer, Memorial Sloan Kettering Cancer Center, New-York

“Microbiota-mediated Defense Against Intestinal Infection”

12:30 PM Lunch-Poster viewing- Networking

Session III: Microbiome-associated diseases & therapeutic targets

Moderator: Jonathan Braun, UCLA

02:00 PM Karine Clément, Pitié-Salpêtrière Hospital-Sorbonne University-Inserm, France

“Acting on Gut Microbiota in Metabolic Diseases: Why, for Whom, What and When”

02:30 PM Elaine Hsiao, University of California, Los Angeles

“Microbiome-Nervous System Interactions in Health and Disease”

03:00 PM Harry Sokol, MICALIS, INRA, France

“Dangerous Liaisons Between Gene and Microbiota: the Example of Card9 in IBD”

03:30 PM Kerwyn C Huang, Stanford University

“Resilience of the Gut Microbiota to Perturbations”

04:00 PM Coffee Break - Poster viewing – Networking

Page 7: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Special Topic Moderator: Minh-Hà Pham, Counselor for Science and Technology of the Embassy of France in the United States

04:20 PM Sandrine Miller-Montgomery, Center for Microbiome Innovation, University of California San Diego

“The Center for Microbiome Innovation of UCSD”

04:35 PM Evelyne Jouvin-Marche, Inserm, France

“Inserm Transversal Program on Microbiota : The intestinal ecosystem, a key determinant in health and disease”

04:50 PM Marion Leclerc, INRA, France

“Microbiomes, food system and health. Science and opportunities at the French National Institute for Agronomy

Research (INRA)” Round Table

Moderator: Emeran Mayer, UCLA

05:00 PM Addressing the Barriers to Translation from Basic Research to Medicine

Participants:

- Miguel Freitas, Vice-President Scientific Affairs Danone North America

- Andrew Ritter, Ritter Pharmaceuticals

- Serguei Fetissov, Inserm-Université de Rouen, co-founder of TargEDys SA, France

- Momo Vuyisich, Viome

- Priscilla Kelly, Biomedicine Editor at the Science Journal

06:00 PM Reception (CNSI lobby)

Day 2 – March 30, 2018

08:00 AM Welcome breakfast

08:30 AM Keynote Lecture: Rodolphe Clerval, Enterome Bioscience, Paris-Boston – Introduced by Jeff Miller

“Mining the gut microbiome: a new source of targets and small molecules for treating chronical disorders”

Session IV: Understanding microbial communities & their products

Moderator: Pascale Cossart, Pasteur Institute-Inserm-INRA, France

09:15 AM Jeff Miller, California NanoSystems Institute, University of California Los Angeles

“Diversity-Generating Retroelements in Phage, Microbes and Microbiomes”

09:45 AM Marcelo Freire, J. Craig Venter Institute, La Jolla

“Oral Microbiome-Host Communications in Health and Disease”

10:15 AM Pieter Dorrestein, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego

“3D Surface and Volume Digitization of the Chemical World of the Microbiome”

10:45 AM Coffee Break – Poster viewing – Networking

Session V: Short talks / pitches by start-up companies

11:15 AM Moderators: Miguel Freitas, Danone North America & Jean Rosenbaum, Consulate General of France in Los Angeles

Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Stéphanie Culler, Perspephone Biome Jack Oswald, Isothrive

Iryna Dzieciuch, Omnibiome Srinivas Rao, Axial Biotherapeutics

Kiran Krishnan, Microbiome Labs Lada Rasochova, Dermala Inc

Mark Wilson, MatriSys Bioscience

Session VI: Engineering microbiomes

Moderator: Ebrahim Zandi, USC

12:15 PM David Bikard, Eligo Bioscience & Pasteur Institute, France

“CRISPR Tools to Study and Control Bacteria”

12:45 PM Emma Taylor, Naked Biome, San Francisco

“Challenges in Microbiome Drug Development”

13:15 PM Conclusions

13:30 PM End of meeting, Lunch

Page 8: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Faculty (by alphabetical order)

Full biographies available on our website: http://faid-losangeles.france-science.org/

Yasmine Belkaid - Senior Investigator, Mucosal Immunology Section - NIH

Dr. Yasmine Belkaid is a Senior Scientist at the National Institute of Allergy and Infectious Diseases (NIAID), and an

Adjunct Professor in the department of Pathology at the University of Pennsylvania. She is currently the chief of the

Mucosal Immunology section and Director of the NIAID Microbiome program. Her work explores mechanisms that

regulate host immune responses to microbes at barrier sites such as the gut and the skin and revealed key roles for the

microbiota and dietary factors in the control of immunity and inflammation.

David Bikard - Head of Lab at Pasteur Institute - Founder at Eligo Bioscience

David Bikard, PhD, is a young investigator at the Pasteur Institute in the department of Microbiology where he started

his group in 2014. He is interested in applying engineering principles to better understand and fight pathogenic bacteria.

He is a co-founder of Eligo Bioscience.

Karine Clément - Professor of Nutrition - Pitié-Salpêtrière Hospital-Sorbonne University-Inserm

Karine Clément is a Full Professor of Nutrition, Nutrition department, Pitié-Salpêtrière Hospital and at Sorbonne

University in Paris. Her team is currently exploring the link between environment changes, immune systemic changes

and functional modifications in the adipose tissue.

Rodolphe Clerval - Chief Business Officer - VP US Operations - Enterome Bioscience

Before joining Enterome, Rodolphe Clerval was VP Corporate and Business Development of TcLand Expression, a

molecular diagnostic company. Previously he was Business Development Manager at Genzyme and financial analyst for

the brokerage firm Natixis Bleichroeder. Earlier in his career, Rodolphe Clerval served as Research Scientist at Aventis

Animal Nutrition.

Pascale Cossart - Pasteur Institute - « Secrétaire Perpétuel » of the French Academy of Sciences

Pascale Cossart is the head of the « Bacteria-Cell Interactions » unit at the Pasteur Institute, which is also an Inserm and

an INRA unit. Her research has led to new concepts in infection biology but also in microbiology, in cell biology and in

epigenetics. She is a member of the French Academy of Science (2002), a foreign member of the American National

Academy of Science (NAS) (2009), of the German Leopoldina (2001), of the Royal Society (2010), and of the American

National Academy of Medicine (NAM) (2014).

Pieter Dorrestein - Professor - UC San Diego

Pieter Dorrestein is Professor at the University of California – San Diego and the Director of the Collaborative Mass

Spectrometry Innovation Center and a Co-Director, Institute for Metabolomics Medicine in the Skaggs School of

Pharmacy & Pharmaceutical Sciences, and Departments of Pharmacology and Pediatrics.

Page 9: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Serguei Fetissov - Professor of physiology - Rouen University-Inserm and co-founder of TargEDys SA, France

Sergueï O. Fetissov MD, PhD, is a Professor of physiology at Rouen University, France. The focus of his current research

is the identification of bacterial mimetic proteins of neuropeptides and peptide hormones involved in regulation of

motivated behavior as well as functional role of immunoglobulins cross-reactive with such peptides. He is a co-founder

of TargEDys, SA a company developing solutions for eating dysfunction and obesity based on the modulation of the

microbiome.

Marcelo Freire - Associate Professor - J. Craig Venter Institute

Marcelo Freire, DDS, PhD, DMSc, is an Associate Professor at J. Craig Venter Institute in the Department of Genomic

Medicine and Infectious Diseases. Freire’s research focuses on biological communications between host immune system

and microbiome. In particular, Freire’s expertise is in the host response to the oral microbiome in the context of chronic

inflammatory diseases.

Elaine Hsiao - Assistant Professor - UCLA

Dr. Elaine Y. Hsiao is an Assistant Professor in the Department of Integrative Biology & Physiology at UCLA, where

she leads a laboratory studying fundamental interactions between the microbiome, brain and behavior, and their

applications to neurological disorders.

Kerwyn C. Huang – Associate Professor – Stanford University

Kerwyn C Huang’s lab is currently situated in the departments of Bioengineering and Microbiology and Immunology

at Stanford, and his current interests include cell division, membrane organization, cell wall biogenesis, and collective

motility of bacterial communities.

Evelyne Jouvin Marche – Director of research - Inserm

Evelyne Jouvin Marche, Ph.D, Director of research at Inserm, has developed fundamental and translational research

focused on the role of immune cells. She is the Deputy Director of the multi-organization thematic institute

“Immunology, Inflammation, Infectious Disease and Microbiology”.

Marion Leclerc – Team leader - INRA

Marion Leclerc works for INRA microbiology department strategy and is a scientific advisor for INRAScience for Food

and Bioproduct Engineering. In their new INRA team, Phylogeny and Physiology of the Human Microbiome, Marion

Leclerc and Patricia Lepage combine omics and anaerobic microbiology to unravel the functionalities of the human

microbiota, in clinical (Immunotherapy, CD) or nutritional context (Fiber, Food structure).

François Leulier - Lyon Functional Genomics Institute, CNRS-ENS-INRA-Université Claude Bernard

François Leulier is a geneticist by training. The Leulier lab is currently developing research programs in bacteriology and

animal physiology and nutrition to decipher the functional interplays between the intestinal microbiota, lactobacillus

strains and chronic undernutrition and their influence on juvenile growth dynamics (including wasting and stunting).

Page 10: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Emeran Mayer – Executive Director - G. Oppenheimer Center for Neurobiology of Stress and Resilience

Emeran A Mayer is a Gastroenterologist, Neuroscientist and Professor in the Departments of Medicine, Physiology and

Psychiatry at the David Geffen School of Medicine at UCLA. In addition to his ongoing research in chronic visceral pain,

his most recent work in the area of brain gut interactions has focused on the role of the gut microbiota in influencing

different aspects of the brain gut axis, including food addiction in obesity, and gastrointestinal symptoms in functional

and inflammatory bowel disorders.

Sarkis Mazmanian - Professor of Microbiology - California Institute of Technology

Sarkis K. Mazmanian, PhD, is the Luis and Nelly Soux Professor of Microbiology in the Division of Biology and

Biological Engineering at the California Institute of Technology (Caltech), and an Investigator of the Heritage Medical

Research Institute. Dr. Mazmanian’s laboratory focuses on the study of beneficial bacterial molecules from the human

gut microbiome as novel therapies for immunologic and neurologic disorders, with a specific focus on developing

probiotic treatments for inflammatory bowel disease, autism and Parkinson’s disease.

Jeff F. Miller - Director - CNSI

Jeff F. Miller, Ph.D., studies molecular mechanisms of bacterial pathogenesis and the evolution of functional diversity

in bacteria and phage. Miller is a former Pew Scholar in the Biomedical Sciences, a member of the American Academy

of Microbiology, a fellow of the American Association for the Advancement of Science, and in April, 2015 he was

elected to membership in the National Academy of Sciences.

Sandrine Miller-Montgomery - Executive Director - Center for Microbiome Innovation – University of California San Diego

Dr. Sandrine Miller-Montgomery is the Executive Director of the Center for Microbiome Innovation, led by Pr. Rob

Knight at UC San Diego. In this position, she is leading a team focused on fostering and expanding industry and

academic collaborations, with the mission of accelerating Microbiome discovery and creating innovative

technologies that will support this emerging but exploding field in the consumer world as well as enabling major

clinical breakthroughs.

Eric Pamer - Head, Division of Subspecialty Medicine - Memorial Sloan Kettering Cancer Center

Eric G. Pamer, MD is the Head of the Division of Subspecialty Medicine, Enid A. Haupt Chair in Clinical

Investigation and Director of the Lucille Castori Center for Microbes, Inflammation & Cancer at Memorial Sloan

Kettering Cancer Center (MSKCC).

Harry Sokol - Professor of Gastroenterology - Sorbonne University-Inserm-INRA

Harry Sokol is a Professor of Gastroenterology and is the head of the Gut Microbiota and Immunity lab (Inserm

U1157/UMR CNRS 7203, UPMC, Paris and INRA Micalis). Harry Sokol is an internationally recognized expert in

IBD and in gut microbiota fields. Harry Sokol is now exploring particularly the role of the microbiota in tryptophan

metabolism for which he is recipient of an ERC starting grant.

Emma Taylor - CEO & Co-founder - Naked Biome

Dr. Emma Taylor is a board-certified Dermatologist and Dermatopathologist. She completed her residency and

fellowship training at UCLA and stayed on as an assistant professor of Dermatology and Dermatopathology, when

she co-founded Naked Biome. She is also an inventor of 2 published patents, Emma Taylor has published in notable

journals such as Nature and the Journal of the American Academy of Dermatology, and is the author on more than 40

book chapters.

Page 11: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Abstracts (in the order of the program)

Sarkis Mazmanian

Influence of the Gut Microbiome on Autism Spectrum Disorder

Commensal bacteria influence a variety of complex behaviors, including social, emotional and anxiety-like

behaviors, and contribute to brain development in mice and neuronal activation in humans. However, specific connections

between the gut and the brain that impact neurologic function remain poorly described. Autism spectrum disorder (ASD)

is a neurodevelopmental disease that affects 1 million children and their families, and is characterized by 3 cardinal

symptoms: repetitive and stereotyped behaviors, impaired communication, and abnormal social interactions. The incidence

of autism has increased at an alarming rate in recent decades, suggesting a possible role for the environment and making

ASD one of the most significant medical and social burdens of our time. Intriguingly, studies report that children with

autism exhibit dysbiosis of the gut microbiome. Using a validated mouse model that displays the 3 core symptoms of ASD,

we show that animals with behavioral deficits also display an altered gut microbiome and increased intestinal permeability,

similar to features reported in humans. Remarkably, treatment with a specific member of the human microbiome reduces

ASD-related anxiety and improves communicative, stereotyped and sensorimotor abnormalities. Consistent with the role

of gut microbes in regulating intestinal permeability and metabolic homeostasis, we show that probiotic treatment corrects

altered metabolite profiles of “autistic” mice. In a proof-of concept test, we reveal that injection of one identified metabolite

into naïve mice is sufficient to induce anxiety-like behavior. Further, we demonstrate that transplant of gut microbiota from

children with ASD into mice is sufficient to transfer core behaviors of autism. These data suggest that microbiome-

regulated metabolomic changes contribute to behavioral abnormalities. We propose the provocative concept that ASD may

be, at its core, a disease of the gut that manifests in altered behaviors partially mediated by bacterial metabolites.

Page 12: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Serguei Fetissov

Gut Bacteria and Host Feeding Behavior

Feeding behavior is driven by visceral feelings of hunger and satiety. Their daily changes follow the stable rhythms

consisting of about 20 min of satiation after beginning of a meal and 5 hours of satiety before feeling of hunger triggering

the onset of a next meal. The regularity of appetite changes is fundamental for structuring daily human activity. While

several hormonal and neuronal circuitries are known to regulate feeding behavior, recent data also support a fundamental

role for gut bacteria. In fact, when animals eat they also feed the numerous bacteria inhabiting their gastro-intestinal tract.

Such bacterial feeding occurs both immediately after nutrient ingestion as well as several hours later after the intestinal

transit of non-absorbed fibers. Indeed, as for the process of digestion, food intake triggers a pavlovian reflex of intestinal

secretion setting-up the timing of the hormonal and metabolic changes with resulting rhythms of feeding behavior.

Providing nutrients to bacteria triggers their growth which undergoes a specific dynamic under conditions of a regular

nutrient provision and rich bacterial density in a limited volume such as in the lower gut. These dynamics include an

immediate bacterial duplication lasting for 20 min before the onset of the bacterial stationary phase. E.coli grown in such

conditions, imitating the daily feeding rhythms in humans, produces bacterial proteins which reduce food intake and

activate intestinal satiety pathways, depending on bacterial growth phase. An independent from the host regulation of

bacterial growth dynamics, involving the quorum sensing, may underlie an orchestrating role of gut microbiota in the

specific alternation and duration of the host appetite cycles.

An important task is to understand molecular mechanisms linking the gut bacteria with the host pathways regulating

appetite and feeding behavior. One such molecular target has been identified in Enterobacteriaceae by linking these

common gut bacteria with the host melanocortin system, critically involved in the regulation of energy balance. Indeed, a

bacterial protein homologue of caseinolytic protease B (ClpB) has been identified as a conformational mimetic of α-

melanocyte-stimulating hormone (α-MSH), an anorexigenic neuropeptide. E.coli ClpB is present in gut lumen and plasma

and is able to activate directly both intestinal and central satiety pathways, respectively. On the other hand, autoimmune

reaction against ClpB can be involved in the pathophysiology of eating disorders via production of IgG cross-reactive with

α-MSH. Thus, these findings reveal mechanisms linking specific gut bacteria with the host specific molecular pathways

regulating appetite in both normal and pathological conditions. Author proposes a new theoretical model of appetite control

integrating the energy needs of both the host and gut bacteria. These data are actively exploited by TargEDys SA, a start-

up company developing probiotic-based therapy for altered feeding behavior and body weight.

Page 13: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

François Leulier

The Microbiome and Linear Growth Promotion: Lessons from Gnotobiotic Animal Models

The interaction that binds animals to their microbiota profoundly shapes many aspects of their biology. One such

aspect is linear growth at the juvenile stage, especially initiated by undernutrition. We aim to elucidate the mechanisms

that forge the beneficial interaction between the juvenile host and its gut microbiota. Using gnotobiotic flies and mice, we

have demonstrated the ability of selected lactobacilli strains to promote linear growth following chronic undernutrition.

These discoveries suggest that the still unknown molecular mechanisms underlying microbiota-mediated linear growth

promotion are likely conserved during evolution. We are now probing the mechanistic basis of such phenomena using

genetic approaches in Drosophila coupled to functional studies in monoxenic and conventional mice. We believe that this

basic knowledge carries a lot of potential for industrial applications and will pave the way for clinical studies that will

evaluate the potential of lactobacilli-based interventions to support child growth and health upon undernutrition.

Yasmine Belkaid

Control of Skin Immunity and Inflammation by the Microbiota

Page 14: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Pascale Cossart

Intestinal Microbiota and Infection: the Listeria Paradigm

Bacterial enteropathogens encounter in the intestine a number of microorganims that in various ways can protect

the host against the invading microbe. Listeria monocytogenes is a food borne pathogen which invades the intestinal

epithelium mainly at the level of the goblet cells. We have identified in Listeria genomes, genes encoding bacteriocins,

i.e. toxins that are active against bacteria. The first bacteriocin named LLS which is present in most epidemic strains is

poorly expressed in vitro but is highly expressed in vivo. Its presence is critical for oral infection and correlates with a

decrease in the amount of Allobaculum and Alloprevotella in the intestine but it is still unknown whether this effect is direct

or not. The second bacteriocin is expressed in the majority of clinical strains. Quite strikingly it targets

specifically Prevotella copri in both the mouse and the human gut microbiota. Expression of the bacteriocin lowers

infection, revealing for the first time an unsuspected role for Prevotella copri in exacerbating intestinal bacterial infection.

Eric Pamer

Microbiota-mediated Defense Against Intestinal Infection

Infections caused by antibiotic-resistant bacteria generally begin with colonization of mucosal surfaces, in

particular the intestinal epithelium. The intestinal microbiota provides resistance to infection with highly antibiotic-

resistant bacteria, including Vancomycin Resistant Enterococcus (VRE), Klebsiella pneumoniae and Clostridium difficile,

the major cause of hospitalization-associated diarrhea. Metagenomic sequencing of the murine and human microbiota

following treatment with different antibiotics is beginning to identify bacterial taxa that are associated with resistance to

VRE and C. difficile infection. We demonstrate that reintroduction of a diverse intestinal microbiota to densely VRE

colonized mice eliminates VRE from the intestinal tract. While oxygen-tolerant members of the microbiota are ineffective

at eliminating VRE, administration of obligate anaerobic commensal bacteria to mice results in a billion-fold reduction in

the density of intestinal VRE colonization. Recent studies have identified specific bacterial species, including Blautia

producta and Clostridium bolteae that prevent intestinal colonization with VRE. By treating mice with different antibiotics

that result in distinct microbiota changes and lead to varied susceptibility to C. difficile, we correlated loss of specific

bacterial taxa with development of infection. Using a workflow involving mouse models, clinical studies, metagenomic

analyses and mathematical modeling, we have identified a probiotic candidate that corrects a clinically relevant

microbiome deficiency. Our studies indicate that obligate anaerobic bacteria enable clearance of intestinal VRE

colonization and may provide novel approaches to prevent the spread of highly antibiotic-resistant bacteria.

Page 15: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Karine Clément

Acting on Gut Microbiota in Metabolic Diseases: Why, for Whom, What and When

The high prevalence of metabolic disorders is a global health problem resulting in considerable health care costs.

Lifestyle changes are seen as cornerstones in the management of these tightly-linked but heterogeneous disorders that

progress over an individual’s lifetime. Some patients with metabolic disorders will progress rapidly toward complications

while others will progress more slowly.

The development and progression of metabolic diseases is linked to changes in a myriad of environmental factors

interacting with individuals’ genetic background and epigenetic factors. The gut microbiota is now viewed as a key player

at the interface between these environmental changes and host biology. These diseases are associated with reduced gut

microbiota diversity and modified composition, with an aggravation of dysbiosis with the progression of the disease,

particularly in the obesity context. Gut microbiota profile is also associated with specific dietary patterns and is modified

with dietary and therapeutic interventions (either medical or surgical) known to reduce cardiometabolic risks. These

interventions can improve gut microbiota diversity and richness together with the amelioration of metabolic and other

variables linked to low-grade inflammation.

Whether live microorganisms (i.e. probiotics) eventually combined with prebiotics can impact significantly gut microbiota

composition and may induce a beneficial and significant effect on human metabolism and inflammation is still an open

question. Moreover, which population or patient might benefit the most from these interventions impacting on the gut

microbiome must be determined. This presentation will examine whether modification of the microbiome can help in

improving metabolic health and if so, in which individuals.

Elaine Hsiao

Microbiome-Nervous System Interactions in Health and Disease

The gut microbiota is emerging as an important modulator of brain function and behavior, as several recent

discoveries reveal substantial effects of the microbiome on neurophysiology, neurogenesis, blood brain barrier

permeability, neuroimmunity, brain gene expression and animal behavior. Despite these findings supporting a

“microbiome-gut-brain axis”, the molecular and cellular mechanisms that underlie interactions between the gut microbiota

and brain remain poorly understood. To uncover these, my laboratory and others are mining the human microbiota for

microbial modulators of host neuroactive molecules, investigating the impact of microbiota-immune system interactions

on neurodevelopment and examining the microbiome as an interface between gene-environment interactions in

neurological diseases. We aim to dissect biological circuits for communication between the gut microbiota and nervous

system, toward understanding fundamental biological pathways that influence brain and behavior.

Page 16: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Harry Sokol

Dangerous Liaisons Between Gene and Microbiota: the Example of Card9 in IBD

The pathogenesis of the inflammatory bowel disease (IBD) is linked to an activation of the gastro-intestinal

immune system toward the gut microbiota in genetically susceptible hosts and under the influence of the environment. The

microbial community in the human gastrointestinal tract is fundamental to the health. Loss of the fragile equilibrium within

this complex ecosystem, termed dysbiosis, is involved in numerous pathologies, including IBD. Patients with IBD exhibit

an altered gut microbiota composition with a notable decrease in the abundance of anti-inflammatory bacteria such

as Faecalibacterium prausnitzii. We also observed alteration in the fungal microbiota composition in these patients. The

association of several polymorphisms of innate immunity genes involved in microbial sensing with IBD is another

argument for the involvement of the gut microbiota in the IBD pathogenesis. Some genetic factors involved in IBD might

indeed act through a microbiota effect. We notably demonstrated that this is the case for the IBD susceptibility gene

CARD9. Based on its demonstrated role in IBD pathogenesis, the gut microbiota is now considered as a potential

therapeutic target and next generation probiotics as well as fecal microbiota transplantation are actively investigated.

Kerwyn C Huang

Resilience of the Gut Microbiota to Perturbations

The use of antibiotics to resolve bacterial infections has saved countless lives since the discovery of sulfa drugs

nearly a century ago. Unfortunately, the overzealous and often inappropriate prescription of antibiotics has led to a rise in

antibiotic resistance and has wide-ranging effects on the communities of commensal bacteria that live on and within the

host. Antibiotic perturbation alters the composition and function of these communities and increases susceptibility to future

infection, underscoring the need to design antibiotics that target pathogens and minimize damage to commensals. In

previous efforts to understand how antibiotics affect communities of commensal bacteria, antibiotic sensitivity has been

studied in pure cultures of individual bacteria grown in laboratory media. These in vitro results are not always mirrored in

studies of clinical and animal models, which comprise a body of literature that is in and of itself also contradictory and

inconsistent. Understanding the factors that underlie a bacteria’s response to antibiotics within a complex host-associated

community will require integration of in vitro and in vivo data and careful dissection of quantifiable aspects of the host

environment and community. We show that the gut microbiota has a robust response to antibiotic perturbation, even after

a massive 105-fold drop in bacterial load, showing recovery during antibiotic treatment in a housing-dependent manner.

The coarse-grained taxonomic kinetics of this recovery are relatively independent of the microbiota context, the antibiotic

target, multiple antibiotic treatments, or dietary shifts, although dietary shifts result in a much larger perturbation to the

bacterial load. We also show that the recovery is dominated by a few species, and the final state after stabilization exhibits

signs of substantial extinction that are not predictable based on in vitro growth in pure culture. These results highlight the

importance of community context in the sensitivity of bacteria to antibiotics by modulating the initial composition of the

microbiota in humanized mice through diet and multiple rounds of antibiotics.

Page 17: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Sandrine Miller-Montgomery

The Center for Microbiome Innovation of UCSD

In this presentation, we will be covering the many unique ways the UC San Diego Center for Microbiome

Innovation has been set-up in order to increase synergies between Academia and Industry in the field of microbiome.

We will highlight our structures, as well as share a couple of projects which showcase how our organization can

lead to a meaningful win-win partnership, where key performance indicators can be exceeded by both parties, despite being

widely different.

Evelyne Jouvin-Marche

Inserm transversal program on microbiota : The intestinal ecosystem, a key determinant in health and disease

The “Institut National de la Santé et de la Recherche Médicale (INSERM)” has recently launched a new program named

“Programmes Transversaux “ to fund National research consortia in three major fields: “Microbiota”, “Ageing” and

“Variability of the genomes within cohorts”. This cutting- edge program has been built to facilitate interdisciplinary

approaches, to reinforce interfaces in biomedical research and to strengthen continuum towards economic development

and societal improvement. Thus, each program will result in an interdisciplinary National consortium, providing added

value compared to existing collaborations, particularly on specific areas of innovative research or technological

developments. The implementation of these national consortia should allow the French biomedical research to become a

key player in Europe in these fields and get the community ready for future calls.

Concerning, the research program dedicated to microbiota, the key challenges are to go beyond correlations, to demonstrate

causal effects and to identify molecular mechanisms in order to translate knowledge into diagnostic tools and rationale-

based therapeutic strategies. The Program is divided into three main Work Packages (WP). The first WP intends to develop

standardized metabolomics approaches to identify serum or tissue metabolites, derived from microbiota metabolism, which

may influence onset or severity of several diseases studied by members of the consortium. The second WP aims at getting

insight into functional intestinal ecology using innovative animal models, clinical trials or cohorts challenged by disrupting

factors such as antibiotics and food interventions. The third WP aims at developing therapeutic strategies in metabolic and

liver diseases, and will highlight the impact of bile salts metabolism in the outcome of host-microbiota interactions.

Marion Leclerc

Microbiomes, food system and health. Science and opportunities at the French National Institute for Agronomy Research

(INRA)

Page 18: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Rodolphe Clerval

Mining the gut microbiome: a new source of targets and small molecules for treating chronical disorders

Gut microbiome plays a crucial role in the building-up of our immunity system at birth. It is a life-long personal

and natural protection but increasing evidence suggests that dysregulated immune responses against commensal microbes

in the gut are strongly involved in Inflammatory Bowel Diseases, Oncology, Metabolic disorders, Depression, Allergies

amongst numerous diseases.

Our target and drug discovery platform is based on full sequencing and mapping of total fecal bacterial gene content that

leads to the characterization of the gut metagenome associated with a disease phenotype. This technology enables the

identification of genes and pathways of interest associated with the development of diseases. The metagenomics dataset

generated is then associated with a high throughput screening platform that allows the discovery of news drugs and targets.

Two lead programs were discovered through this platform :

EB8018 is an oral and non-systemic small molecule drug candidate that is designed to block FimH, a key virulence

factor expressed by pro-inflammatory Enterobacteria. EB8018 prevents colonization of gut wall, mucosal inflammatory

cytokine production and entry of such bacteria into the gut wall.

E0510 : this program aims to discover novel bacterial-derived cancer antigens that are expressed in human gut

microbiome. The first preclinical candidate (E02401) has demonstrated strong potency to induce T-cell activation

against solid tumors.

Enterome has entered clinical development stage, with EB8018 being currently evaluated in Crohn’s patients and E02401

will be tested in cancer patients in 2018.

Jeff Miller

Diversity-Generating Retroelements in Phage, Microbes and Microbiomes

Diversity-generating retroelements (DGRs) have evolved to increase host fitness by diversifying ligand-binding

domains of variable proteins in bacteria, archaea, and their viruses. DGRs function through a mutagenic retrohoming

mechanism in which an RNA intermediate serves as both the primer and template for a unique form of reverse transcriptase-

dependent cDNA synthesis in which adenines are specifically miscopied, producing adenine-mutagenized cDNA that

replaces parental sequences in genes that encode variable proteins. Naturally-occurring DGRs have the potential to generate

astronomical levels of diversity, corresponding in many cases to >1026 unique DNA sequences in diversified genes. DGRs

focus their vast mutagenic potential to evolve protein function by positioning variable nucleotides at sites that encode

solvent exposed resides in ligand-binding domains. We have discovered an enrichment of DGRs in prominent members of

the human microbiome, including numerous Bacteroides species. Bacteroides DGRs are encoded on distinct yet related

integrative and conjugative elements that undergo chromosomal excision and transfer in the GI tracts of gnotobiotic mice,

and recent evidence suggests they diversify a family of proteins that function as tip adhesins on a newly discovered pilus

family with highly modular components. Our results demonstrate the horizontal transfer of accelerated evolvability,

highlighting the dynamic nature of host-associated microbial communities.

Page 19: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Marcelo Freire

Oral Microbiome-Host Communications in Health and Disease

At the interface of oral tissues, the microbiome and immune crosstalk are very dynamic. Chronic inflammation in

the oral cavity is influenced by environmental changes, genomic and microbiome-immune interactions leading to dysbiosis

of oral tissues including, periodontal diseases, peri-implant conditions, caries, endodontic lesions and oral cancers.

Periodontal diseases are microbiome-induced inflammatory diseases with disruptions of tissues surrounding the tooth and

irreversible connective and bone tissue loss. This is aggravated when chronic systemic conditions are present as clusters

of inflammation, including type 2 diabetes. Unresolved inflammation is a key regulator of the initiation, progression, and

severity of periodontal diseases. This process is tightly regulated by a new genus of molecules called immunoresolvents

(e.g., resolvins, lipoxins, maresins and protectins). As pro-resolution molecules, immunoresolvents increase phagocytosis

of bacteria, clearance of dead cells and turn on signals to return to homeostasis. Metagenomic sequencing of human and

mice oral microbiota has revealed that P. gingivalis is an important pathobiont in different forms of periodontal and

equivalent per-implant dysbiosis. The next frontier of microbiome-host integration requires mechanistic studies to validate

and to understand the details of health and disease spectrum. We demonstrate that in a murine chronic inflammatory model

for type 2 diabetes (db/db) and periodontal diseases, P. gingivalis induced inflammation and tissue loss which is controlled

by the specific interaction of immunoresolvents and its cognate receptors present in innate immune cells. Using a workflow

of human, murine transcriptomic innate immune evaluations, we have identified g-protein receptor control of inflammation

and leading to tissue homeostasis. Also, imaging approaches guided understanding of the kinetics of local response to host-

microbial interactions in periodontal tissues. Our studies provide novel insights into the pathogenesis of periodontal

diseases and offer unique insights to therapeutic opportunities.

Pieter Dorrestein

3D Surface and Volume Digitization of the Chemical World of the Microbiome

Page 20: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

David Bikard

CRISPR Tools to Study and Control Bacteria

Over the past few years CRISPR systems have been derived into powerful tools to edit genomes, control gene

expression, visualize nucleic acids in vivo, modify epigenetic marks, kill cells and more. Most of these tools rely on the

RNA-guided nuclease known as Cas9 which can be easily reprogrammed to bind and cut almost any position in a genome.

We focus on the application of these technologies to Bacteria. We have recently investigated the properties of Cas9 as well

as the catalytic dead variant known as dCas9 in E. coli. The introduction of double strand breaks by Cas9 at a specific

genomic position leads to cell death as such breaks cannot be repaired through homologous recombination, the main DNA

repair pathway in bacteria. We used this property as a selection tool in both E. coli and S. pneumoniae, and to develop

sequence-specific antimicrobials against S. aureus. Work on the dCas9 protein has revealed how it can be used to precisely

tune the expression level of several target genes independently and with low noise levels. Finally, the ability to knockdown

gene expression with dCas9 can also be used in high-throughput screens to unravel gene function and interaction. All in

all, CRISPR technology provides a fantastic toolkit to study and fight pathogenic bacteria.

Emma Taylor

Challenges in Microbiome Drug Development

Despite the overwhelming data that microbial therapies can impact clinical disease, evidenced by the success of

fecal microbiome transplants, translating these approaches into meaningful and reproducible therapeutic strategies remains

a challenge. There still remains an industry-wide dependence on antibiotics and there are many company-specific

challenges in working with live biologic material, namely relating to securing intellectual property, identifying optimal

candidate strains, developing a GMP manufacturing process, establishing a regulatory strategy, and designing clinical

trials. I hope to shed light on how our company, Naked Biome, has navigated many of these challenges in developing a

topical live biologic therapy for acne.

Page 21: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Start-ups presentation

Blue Turtle Bio is a therapeutic probiotic company focused on microbial enzyme

delivery for rare diseases. Their engineered microbe can be ingested by the patient

and would provide a timely release of said protein. This platform technology

would thus be an efficient alternative to current costly intravenous infusions.

Persephone Biome is developing live biotherapeutics to improve the efficacy of

cancer immunotherapy. Leveraging tools and experience in systems and synthetic

biology, they want to unravel the complexities of the microbial ecosystem in the

human gut; specifically, the interactions among microbes, checkpoint inhibitors,

and the human immune system will be elucidated. This knowledge will be used to

design and develop live biotherapeutics that will restore the functions needed to

ensure the efficacy of cancer immunotherapy.

SciBac is a pre-clinical startup based near San Francisco, CA. They design live

biotherapeutics to target antibiotic resistant disease in the microbiome using their

DRIVE platform. DRIVE is a non-GMO platform technology that allows them to

mate different species of microbes together in a type of directed evolution. Their

first indication is for Clostridium difficile infection, a type of deadly diarrhea often

contracted in hospitals, and our second indication treats and prevents

Pseudomonas and non-tuberculous Mycobacterium infections in the lungs of

cystic fibrosis patients.

Axial Biotherapeutics is a biopharmaceutical company harnessing the link

between the human gut microbiome and the Central Nervous System (CNS) to

develop a new class of therapeutics to improve the quality of life for people with

CNS diseases and disorders. Axial’s scientific co-founder Sarkis K. Mazmanian,

Ph.D., and researchers from the California Institute of Technology (Caltech) have

discovered a novel, causal-link between the gut microbiome and Autism Spectrum

Disorder (ASD) and Parkinson’s Disease (PD). Axial’s scientific co-founder has

demonstrated that oral treatment of a mouse model of ASD with a human

commensal bacterial therapy corrects gut permeability, restores a healthy

microbial composition and ameliorates core behavioral symptoms of ASD. In a

PD mouse model, gut bacteria were shown to promote hallmark disease processes

including inflammation of the nervous system and motor dysfunction. These

findings suggest that targeting the gut microbiome using gut-selective therapies

may provide a new approach for treating ASD and PD.

Page 22: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Dermala Inc. is a consumer dermatology startup utilizing the human microbiome

and data to develop the next generation treatments for the most prevalent chronic

skin diseases and conditions.

OmniBiome, Inc. is the world’s first company focused on therapeutic / Rx

approaches to either utilize or intervene with the systemic effects of the vaginal,

lactal-duct and oral microbiomes for improving maternal healthcare and resulting

birth outcomes. The Company will focus initially on developing CLIA Dx

services for both pre-pregnancy-associated and pregnancy-associated conditions

or diseases where there is a substantive link with microbiome dys biosis

(disruption or imbalance), as well as on restoring eubiosis (proper balance). In

parallel OmniBiome will build a database of aggregated patient data that will later

inform development of Rx / therapeutic and medical device & drug-device

combination approaches for treating the same conditions or diseases.

Microbiome Labs was originally established as Physicians Exclusive in 2013 as

an organization focused on providing probiotic bacteriotherapy. In the past several

years their business model has grown and so has public awareness for gut health

issues. Their goal is to provide integrative solutions and clinical research data to

address indications that stem from digestive and immune health issues.

ISOThrive provides gastroenterologist recommended microFood that

nourishes specific bacteria in the gut to resolve imbalances that are at the root

of chronic health conditions. It is a lightly sweet nectar of non-digestible

soluble fiber called MIMOTM (maltosyl-iso-malto-oligosaccharides).

MatriSys Bio is a clinical stage Specialty Biopharmaceutical Company focused

on developing and commercializing rational microbiome therapies for the top five

dermatology and skin care conditions.

Page 23: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Posters presentation

P01- A novel method for accurate profiling of human microbiome using 16S amplicon sequencing

Authors: Igor Segota, Tao Long

Institution: Sanford Burnham Prebysterian Medical Discovery Institute, La Jolla, CA

P02- Curated Biobank of Preserved, Whole Human Fecal Samples, Sequences, Metagenomes, Metadata and

Bacterial Strains for Research and Development Authors: Raul J Cano, Naseer Sangwan and Martha Carlin

Institution: The BioCollective Inc., Denver, CO

P03- Microbiome and bile acid profiles in duodenal aspirates from cirrhotics vary by cirrhosis etiology, hepatic

encephalopathy, and ethnicity

Authors: Jonathan P. Jacobs1, Tien S. Dong1, Vatche Agopian1, Venu Lagishetty1, Vinay Sundaram2, Mazen

Noureddin2, Walid Ayoub2, Francisco Durazo1, Jihane Benhammou1, Pedram Enayati2, David Elashoff1, Marc T.

Goodman2, Joseph Pisegna1, Shehnaz Hussain1,2

Institutions: 1UCLA, 2Cedars-Sinai Medical Center, Los Angeles, CA

P04- Functionally characterizing bacterial protein homologs at large scales using multiplex gene synthesis

Authors: Calin Plesa, Angus M. Sidore, Nathan Lubock, Di Zhang, Sriram Kosuri

Institution: UCLA, Los Angeles, CA

P05- Microbiome shifts after daily HIV pre-exposure prophylaxis (PrEP)

Authors: Michael P. Dubé, Sung Yong Park, Emily Johnson, Tanzy M. T. Love, Sheldon R. Morris, and Ha Youn Lee

Institutions: Keck School of Medicine, University of Southern California, Los Angeles, CA, University of Rochester,

Rochester, NY, University of California San Diego, San Diego, CA.

P06- Bacterial uptake of serotonin regulates intestinal colonization of the indigenous gut bacterium, Turicibacter

sanguinis

Authors: Thomas C. Fung, Cristopher D. Garduno Luna, Helen E. Vuong, Anastasia Vavilina, Antoniya Aleksandrova,

Lucy R. Forrest, Elaine Y. Hsiao

Institution: Department of Integrative Biology and Physiology, University of California, Los Angeles, CA

Page 24: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

List of Participants (by alphabetical order)

Last Name First Name Affiliation

Ahn In Sook UCLA

Ahuja Umesh UCLA

Aljahmi Adham Blue Turtle Bio Inc.

Amaral Wellington UCLA

Angles Frederic The Scripps Research Institute

Archambeau Eric Quadia Impact Investing

Ballard Zachary UCLA

Bayne Tom Microbiome Labs

Belkaid Yasmine NIAID

Bentolila Laurent UCLA

Bikard David Eligo Bioscience

Birkett Anne Kellogg’s

Bortone Kara JLABS

Botelho Dinis Marcia UCLA

Braun Jonathan UCLA

Brown Calvin UCLA

Brubaker Linda UCSD

Caremoli Filippo UCLA

Casero David UCLA

Cauchy Tim USC

Cervino Alessandra Luxia Scientific

Ceylan Koydemir Hatice UCLA

Chen Lisa UCLA

Christey Peter GALT Inc.

Clément Karine Sorbonne-University/INSERM

Clerval Rodolphe Enterome

Cossart Pascale Pasteur Institute

Cozen Wendy USC

Cuff Callie UC Berkeley

Culler Stephanie Persephone Biome

Dai Lei UCLA

Degregori Samuel UCLA

Devkota Suzanne UCLA, Cedars-Sinai

DeVries Amber UCLA

Dhir Raja Seed

Diamante Graciel UCLA

Ding Ching JLABS

Ding Zhaoqing JLABS

Dong Tien UCLA

Dormant Valérie Medtronic

Dorrestein Pieter UCSD

Dubois Geraud IBM Research

Dubois Raphaël Consulate General of France

Dzieciuch Iryna Omnibiome

Erudaitius Anastassia UC Riverside

Erudaitius Dieanira UCSD

Faris Mary Pfizer

Fetissov Serguei University of Rouen Normandy

Fluke Dara CalState University Northridge

Freire Marcelo J. Craig Venter Institute

Freitas Miguel Danone

Fung Thomas UCLA

Gagnon Jeff Microbiome Labs

Giglio Laurence UCLA

Gonzalez Angel Zymo Research

Gruffaz Marion USC

Ha Connie Cedars-Sinai Medical Center

Halec Gordana UCLA

Hambor John Boehringer Ingelheim

Hawkins Jr. Stewart UCLA

Hendrick Gustaf Cedars-Sinai Medical Center

Henry Kurt Pepperdine University

Herman Dena UCLA

Hsiao Elaine UCLA

Huang Kerwyn C Stanford University

Hwang Loris UCLA

Ifkirne Rachid Biogaz Ecoconcept Valley

Jacobs Jonathan UCLA

Janiyani Kamala UCSD

Jee Noel Illumina Ventures

Jin Cathy Yan UCLA

Johnson Emily USC Keck School of Medicine

Johnson Neal UC Berkeley

Joshi Swapna UCLA

Juarez Betsy UC Irvine

Kalu Michelle UC Irvine

Karic Alda UCSF

Kazantsev Maria UCLA

Kelly Priscilla Science Journal

Kelly Denise Seventure Partners

Kim Dokyun USC

Knight Jamie UCSD

Koon Hon Wai UCLA

Krishnan Kiran Microbiome Labs

Kuhn Cyrille Boehringer Ingelheim

Lagishetty Venu UCLA

Lakritz Ania UCLA

Larauche Muriel UCLA

Leclerc Marion INRA

Lee Carol UCLA

Lee Ha Youn USC Keck School of Medicine

Lee Sungeun UCLA

Leulier François ENS de Lyon

Li Huiying UCLA

Li Rongsong UCLA

Lipman Arye BioBuilt Inc.

Liu Jing Knobbe Martens

Lo Richard USC Keck School of Medicine

Long Tao Sanford Burnham

Luethy Lauren ASM Journal

Luna Brian USC Keck School of Medicine

Lux Renate UCLA

Maiello Michelle JLABS

Maeusli Marlene USC

Martin Anthony Cedars Sinai Medical Center

Mayer Emeran UCLA

Mazmanian Sarkis Caltech

Mehrabian Margarete UCLA

Mercer Frances UCLA

Michels Karin UCLA

Miller Jeff UCLA

Miller Matthew UCLA

Miller-Montgomery Sandrine UCSD

Morin Manon UCSD

Moscicki Anna-Barbara UCLA

Mucha Jeanette SciBac

Nakamura Reine UCLA

Ngo Olivier Consulate General of France

Nguyen Jennifer UCLA

Nonaka Taichiro UCLA

Oberholzer Michael Illumina

Olson Christine UCLA

Onstead Jonathan

Onstead Zsuzsa

Orchanian Stephanie UCSD

Ormancey Xavier Beautycounter

Oswald Jack ISOThrive LLC

Ozdemir Aslihan Hacettepe University

Paing Bruno CEA

Pamer Eric Memorial Sloan Kettering CC

Pan Jun UCLA

Park Sung Yong USC

Peara Chris CDP International

Page 25: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac

Pham Minh-Hà French Embassy in the US

Plesa Calin UCLA

Pronovost Geoffrey UCLA

Radin Arielle UCLA

Ragsdale Amy UCLA

Ramler Ellie UCLA

Rao Srinivas Axial Biotherapeutics

Rasochova Lada Dermala Inc.

Rastegar Gazelle Caprea Biotechnology

Ray Aniruddha UCLA

Razavi Reza Caltech

Rehbinder Sharon AEPI

Ritter Andrew University of Pennsylvania

Roche George SF Microbiome Meetup

Rosenbaum Jean Consulate General of France

Ross Heather USC

Rosse Gerard Dart NeuroScience

Sabbah Brittney JLABS

Schiestl Robert UCLA

Seo Gil Ju USC

Shafiei Jahani Pedram USC Keck School of Medicine

Shi Baochen UCLA

Shishko Catherine University of Chambery

Shokeen Bhumika UCLA

Siera Scott Knobbe Martens

Singh Paramjit University of British Columbia

Sokol Harry INSERM, Sorbonne, INRA

Spears Isabelle UCLA

Stieg Adam UCLA

Stiemsma Leah UCLA

Sun Argus UCLA

Tahara Stanley USC

Taylor Emma Naked Biome

Teng Bobby CalState University Northridge

Thomas-White Krystal Loyola University Chicago

Thornton Shantae USC

Tran Nini UCLA

Tripp Arielle UCLA

Urtecho Guillaume UCLA

Van Dien Stephen Persephone Biome

Vandenberghe Louise UCLA

Vesna Victoria UCLA

Voreades Noah The BioCollective

Vuong Helen UCLA

Vuyisich Momo Viome Inc.

Young Thomas UCLA

Walters Dee Alumnus

Wang Shenshen UCLA

Wang Zoey UCLA

Williams Drake UCLA

Wilson Mark MatrisysBio

Xia Tian UCLA

Yee Lauren UCLA

Yoon Lara UCLA

Young Erick Boehringer Ingelheim

Young Thomas UCLA

Yu Yang USC

Zandi Ebrahim USC

Page 26: The Human Microbiome: Time for Effective Translationfaid-losangeles.france-science.org/wp-content/uploads/2018/04/FAID... · Adham Aljahmi, Blue Turtle Bio Jeanette Mucha, Sci Bac