Nayar Prize II Year 2 Progress Report - October 2018 Project ......Nayar Prize II Year 2 Progress...

59
Nayar Prize II Year 2 Progress Report - October 2018 Project: Microfluidic Drug-Microbiota Interaction Platform Team: Abhinav Bhushan, Genoveva Murillo, Rajendra Mehta Postdoctoral Scholar: Sonali Karnik, Students: Chengyao Wang, Rongfei Wu, Kihwan Kim, Andrea Cancino, Thao Dang, Adithya Subramanian. In the second year of the Nayar Prize II, we have accomplished our year 2 milestones as well as formulated clear strategies for making an impact on the lives of patients. Our overall project goals are to study the role microbiota play in influencing drug metabolism. We are developing a microfluidic platform to facilitate study of interactions between the drugs, intestine, and microbiota. We have made extraordinary progress to advance rapidly towards the milestones including strengthening collaborations with Dr. Ali Keshavarzian, who leads one of the leading groups on microbiome/IBD research. Challenged by the reviewers at the end of year 1 to define how the microfluidic drug- microbiota interaction platform can help patients, we talked to several key opinion leaders and collaborated with the business school to develop strategies that will directly benefit patients. We came up with several solutions. One builds upon the concepts of a drug-microbiota interaction platform towards a patient-specific (personalized) microfluidic bioassay; in other words, we will generate an IBD patient-based intestinal platform to test microbiota reprograming that can be used to query prebiotics, probiotics, and IBD therapeutics, potentially to determine the effects of these interventions before prescribing to the IBD patients. The other plan will give individuals the opportunity to generate a personal score that will represent the drug metabolizing capability of their own microbiome. The support from the Nayar Prize was the catalyst that made this possible. Phase II summary 1) Establish intestinal culture in the microfluidic device and characterize drug interactions. We developed a novel microfluidic device that incorporated micrometer resolution membranes which are synthesized out of extracellular matrix. As our results show, cells on the microfluidic device with biomembranes had excellent viability for extended periods of time than the cells in a single layer device. More importantly, the biomembranes induced expression of tight junction between the intestinal cells, which is an important factor in the cells mimicking in vivo phenotypes. This work was published in ACS Biomaterials Science and Engineering and the journal highlighted the science in a special feature (“More realistic and accurate organs-on-chips”). The intellectual property concerning the collagen membrane device was filed by the university and granted a provisional patent. We built upon this work in four directions. A. Establish primary colon cells in the device (Phase II Aim 1). Development of the primary colon culture is quite challenging as the microenvironment external to the in vivo gut environment does not have all the factors to support the growth and function of the cells. We have carried out a large number of experiments to grow the primary colon on a variety of extracellular matrices – fibronectin, collagen type I, collagen type IV, and Matrigel. We have conducted these experiments in tissue culture plates as well in the microfluidic devices. Our results indicate that Matrigel with a collagen coat provided the best results in the microfluidic devices. The colon cells express tight barrier junctions and also e-cadherin, which is one of the important cellular proteins (Figure 1). These results are being finalized in collaboration with the clinical collaborators. Figure 1. Expression of e-cadherin protein in primary colon cells.

Transcript of Nayar Prize II Year 2 Progress Report - October 2018 Project ......Nayar Prize II Year 2 Progress...

  • Nayar Prize II Year 2 Progress Report - October 2018 Project: Microfluidic Drug-Microbiota Interaction Platform Team: Abhinav Bhushan, Genoveva Murillo, Rajendra Mehta Postdoctoral Scholar: Sonali Karnik, Students: Chengyao Wang, Rongfei Wu, Kihwan Kim, Andrea Cancino, Thao Dang, Adithya Subramanian. In the second year of the Nayar Prize II, we have accomplished our year 2 milestones as well as formulated clear strategies for making an impact on the lives of patients. Our overall project goals are to study the role microbiota play in influencing drug metabolism. We are developing a microfluidic platform to facilitate study of interactions between the drugs, intestine, and microbiota. We have made extraordinary progress to advance rapidly towards the milestones including strengthening collaborations with Dr. Ali Keshavarzian, who leads one of the leading groups on microbiome/IBD research. Challenged by the reviewers at the end of year 1 to define how the microfluidic drug-microbiota interaction platform can help patients, we talked to several key opinion leaders and collaborated with the business school to develop strategies that will directly benefit patients. We came up with several solutions. One builds upon the concepts of a drug-microbiota interaction platform towards a patient-specific (personalized) microfluidic bioassay; in other words, we will generate an IBD patient-based intestinal platform to test microbiota reprograming that can be used to query prebiotics, probiotics, and IBD therapeutics, potentially to determine the effects of these interventions before prescribing to the IBD patients. The other plan will give individuals the opportunity to generate a personal score that will represent the drug metabolizing capability of their own microbiome. The support from the Nayar Prize was the catalyst that made this possible. Phase II summary 1) Establish intestinal culture in the microfluidic device and characterize drug interactions. We

    developed a novel microfluidic device that incorporated micrometer resolution membranes which are synthesized out of extracellular matrix. As our results show, cells on the microfluidic device with biomembranes had excellent viability for extended periods of time than the cells in a single layer device. More importantly, the biomembranes induced expression of tight junction between the intestinal cells, which is an important factor in the cells mimicking in vivo phenotypes. This work was published in ACS Biomaterials Science and Engineering and the journal highlighted the science in a special feature (“More realistic and accurate organs-on-chips”). The intellectual property concerning the collagen membrane device was filed by the university and granted a provisional patent. We built upon this work in four directions.

    A. Establish primary colon cells in the device (Phase II Aim 1). Development of the primary colon culture is quite challenging as the microenvironment external to the in vivo gut environment does not have all the factors to support the growth and function of the cells. We have carried out a large number of experiments to grow the primary colon on a variety of extracellular matrices – fibronectin, collagen type I, collagen type IV, and Matrigel. We have conducted these experiments in tissue culture plates as well in the microfluidic devices. Our results indicate that Matrigel with a collagen coat provided the best results in the microfluidic devices. The colon cells express tight barrier junctions and also e-cadherin, which is one of the important cellular proteins (Figure 1). These results are being finalized in collaboration with the clinical collaborators.

    Figure 1. Expression of e-cadherin protein in primary colon cells.

  • B. Characterize role drugs play in

    regulating bacterial metabolites (Phase II, Aim 2). It has been shown that metformin boosts the capacity of gut microbiota to produce certain short-chain fatty acids (butyric acid and propionic acid). We have exposed the several strains of bacteria to drugs and their selected metabolites and characterized their effects on the bacteria.

    C. Characterize role bacteria play in

    regulating activity of the colon drug metabolizing enzymes (Phase II, Aim 3). Objective 3. We systematically studied the interactions (genes and specific activity of phase I metabolism) between colon cells and bacteria. The major findings are that (1) species-specific differences exist in the regulation and activity of these enzymes, (2) regulation of the various enzymes appeared to be unique, and (3) communication between the bacteria and the colon cells is required for modulation of enzymatic activity.

    The results of Aims 2 and 3 are part of a manuscript that is under peer-review, which is provided with this report. The complexity of the gut environment includes a biphasic oxygen environment – normal oxygen for the epithelial cells and anaerobic environment for the microbiota. Creating such an environment in vitro is very challenging, and does not currently exist, especially in microfluidics. We have created a representative gas transport model and used it to create prototype microfluidic devices. The results are being finalized for a publication as well as invention disclosure to the university (Figure 2).

    D. We initiated work towards (see Appendix and below)

    a. IBD patient-based intestinal platform to test microbiota reprograming and b. microfluidic model of microbiota-intestine-brain to study microbiota mediated neural

    inflammation which is implicated in neurodegenerative disorders (Alzheimer’s).

    2) Market research. We carried out interviews of several technical leaders from the pharmaceutical industry on the use of the microfluidic drug-microbiota platform in drug discovery. The overwhelming response we received was that the industry is aware of the influence of bacteria but does not have assays to test for these interactions – any device or platform, such as ours, that can measure these interactions will be hugely beneficial. This work was corroborated by a study conducted in parallel by the students from the Stuart School of Business. During our research, we discovered another possible opportunity – the evaluation of an individual’s propensity for metabolizing drugs. The reports are attached as Appendix. The outcome of these studies is that our Nayar project has three possible market opportunities. Some of this work is included in the disclosure filed with the university. Plans to commercialize are

    Figure 2. Oxygen transport model (not drawn to scale). A is the concentration of oxygen in the middle-chamber after cellular respiration. B is the oxygen concentration in the top-chamber. C is the oxygen concentration outside the device (disclosure pending).

    PDMS top cover

    Top-chamber

    Membrane

    Middle-chamber

    Oxygen concentrationA C

    B

  • being explored. a. drug-microbiota interaction platform, b. IBD patient-based IBD bioassay, and c. personalized score of drug metabolizing capability by microbiome.

    3) Training. The project has been instrumental in exciting students about research and has helped

    tremendously in recruiting students. #4 disciplines integrated (engineering, biology, clinical, business)

    #12 students directly supported/educated #2 Armour college PURE scholarships #1 PhD fellowship (Starr Fieldhouse), #2 travel awards #5 presentations at national & local meetings

    4) Journal papers Wang C, Tanataweethum N, Karnik S, ML, Bhushan A, A Novel Microfluidic Colon with An Extracellular Matrix Membrane, ACS Biomaterials Science and Engineering, 2018, doi: 10.1021/acsbiomaterials.7b00883. Wang C, Cancino A, Dang T, Bhushan A, Regulation of drug metabolizing enzymes by microbiota, under review. Wang C, Dang T, Bhushan A, Tunable multiphasic oxygen environments for coculture of anaerobic and aerobic bacteria with intestinal cells, under preparation. Presentations Chengyao Wang, Chengyao Wang, Maliha W Shaikh, Christopher Frosyth, Ali Keshavarzian, Abhinav Bhushan, “Intestinal Monolayers on a Novel Microfluidic Device with an Extracellular Matrix Membrane”, BMES Annual Meeting, 2018. Chengyao Wang, Nida Tanataweethum, Sonali Karnik, Abhinav Bhushan, “Novel Microfluidic Colon with an Extracellular Matrix Membrane”, Rainin Foundation Innovation Symposium, 2018. Chengyao Wang, Andrea Cancino, Abhinav Bhushan, “Role of Bacteria in Regulating Activity of Enzymes Responsible for Metabolism of Drugs”, Rainin Foundation Innovation Symposium, 2018. Role of Bacteria and Bacterial Products in Regulating Activity of Enzymes Responsible for Metabolism of Drugs in Microfluidic Devices, MIT-Harvard Microbiome Symposium. Chengyao Wang, Nida Tanataweethum, Genoveva Murillo, Rajendra Mehta, Abhinav Bhushan, “A novel microfluidic device with an extracellular matrix-based membrane”, Experimental Biology, 2017.

    5) Collaborations Each of the collaborators has been excited by the strengths of our group, IIT, and the support from the Nayar Prize. It is needless to say that without the Nayar Prize, venturing into any of these areas would not have been possible. Dr. Ali Keshavarzian, Rush University - Role of microbiota along the gut-brain axis in the development of Parkinson’s disease.

  • Dr. Gokhan Dalgin, University of Chicago - Development of mini-brains for modeling neurodegenerative disorders.

    6) Grant funding

    a) Dynamics of microbial interactions in Alzheimer's using a microfluidic platform, NIH R03 under review.

    b) Patient-based intestinal platform to test microbiota reprograming in IBD, NIH R21. Scored, to be resubmitted in October 2018.

    c) A microfluidic platform to generate functionally reproducible human stem cell derived mini-

    brains, NIH R21. Scored, to be resubmitted in July 2019.

    d) Microfluidic platform to discover interactions between intestine, liver, and bacteria, NSF CAREER (positive feedback from Program Officer, to be resubmitted in 2019)

    7) Intellectual property

    a) Microfluidic Device with Extracellular Matrix Support Membrane, Provisional awarded.

    b) Modulation of drug metabolism by bacteria, Disclosure filed with the Office of Technology Development, Illinois Institute of Technology.

    Plans for Phase III

    We have a clear understanding of the path to translate this research. We are on track to accomplish our original objectives as well as some of the new directions that we have identified (Figure 3). We will explore the commercialization of the “personalized score of microbiome’s drug metabolizing capability” (MyBioScore). Our office of technology development has recommended that we participate in the NSF I-Corps program. We will work with them and possibly MATTER Chicago to develop a strategy and participate for commercialization. Our objectives for Phase III are: Objective 1. Create a biphasic oxygen environment in the microfluidic devices. Several bacteria such as VSL#3, akkermansia muciniphila, and Bifidobacterium require an anaerobic environment for growth. Although anaerobic chambers exist at the macroscale, the challenge is to culture the bacteria with the intestinal cells. We have carried out feasibility studies to model and create a microfluidic device that can simultaneously support two oxygen concentrations. In this objective, we will finish characterization of cells and bacteria in these devices and develop the devices for routine fabrication. Objective 2. Characterize intestine-bacteria influence on drug metabolism in microfluidic devices. As the results from this year indicate, bacterial strains have tremendous effects on a wide range of cellular processes, including drug metabolizing capability. We know that cells in a microfluidic environment are more physiological than those in the tissue culture plates (we had shown this in year 1). In this objective, we will carry out the next phase of our studies to characterize the role of bacteria on drug metabolism in microfluidic devices. Objective 3. Develop a personalized intestinal bioassay to test therapeutics. Inflammatory bowel diseases (IBD), affect millions of Americans and carry an economic burden of hundreds of billions of dollars. There are several experimental and human observational studies to support the current notion that IBD is a consequence of abnormal mucosal immune response to intestinal microbiota due to abnormal microbiota composition, increased exposure of bacterial product to mucosal immune cells [disrupted intestinal barrier integrity] and exaggerated mucosal immunity. Thus, microbiota

  • reprograming is a promising therapeutic approach to prevent/treat IBD. But, in spite of success of microbiota reprograming by diet/pro and prebiotics and stool transplantation in animal models of IBD, the result in patients is disappointing. This failure could be due to lack of universal response of microbiota to any given intervention and immune response to change in microbiota. For example, it is known that not all IBD patients respond to TNF antibodies or anti-IL12/23 antibody suggesting that immune pathways leading to intestinal inflammation and tissue damage is different in different patients. This limitation could explain why result of animal studies and in vitro cell line studies are not universally translatable to human studies. Thus, availability of a robust and reliable patient-based “bioassay” is a major unmet need to overcome highly individualize and complex intestinal microenvironment in order to develop new precision and personalize therapeutic intervention including microbiota reprograming in IBD. To address this critical gap, we propose to develop a discovery platform that will enable colon-microbe-immune cultures and allow query of factors that can result in favorable/unfavorable outcomes. In this objective, we will test pre-/probiotics to regulate intestinal response to inflammation or differential drug metabolism activity. We will carry out this study at Rush University. Mindful of the additional logistics involved with patient studies, we will conduct a pilot study with all the proper controls (patient +/- Crohn’s, a number of bacteria and pre-/pro-biotics). The outcome will be a personalized patient-based assay that can be queried with therapeutics that can inform the physician on the best suitable line of therapy. In later phases, we could use patient’s own microbiota in the model and be able to carry out interesting experiments by cross culturing samples from patients and controls. Examples include= - Crohn’s intestinal cells+ healthy microbiota + probiotics; Crohn’s intestinal cells + healthy microbiota+ probiotics; Healthy intestinal cells + healthy microbiota+ microbiota; Healthy intestinal cells + Crohn’s microbiota+ probiotics.

    Figure 3. Timeline for the proposed project.

    10/2016 2017 2018 2019 2020

    Phase I: Customizable microfluidic platform

    Topological biomembranes

    Microfluidic devices with biomembranes

    Phase III: Drug interactions

    Microfluidic devices with dual-O2 env

    Intestine-microbiota drug interactions in devices

    Commercialize:personalized score of microbiome’s drug metabolizing capability

    Phase II: Microfluidic intestine-on-chip

    Primary colon in microfluidic device

    Role of drugs in regulating bacterial metabolites

    Bacteria modulation of intestine activity

    Personalized intestinal bioassay

  • Confidential

  • Confidential

  • Confidential

  • 10/15/2018 Proceedings of the National Academy of Sciences

    https://www.pnascentral.org/cgi-bin/main.plex?form_type=view_ms&j_id=1&ms_id=451782&ms_rev_no=0&ms_id_key=ftdZvUogMIm1tWmQUKvzSB… 1/1

    Manuscript # 2018-17445 Current Revision # 0Submission Date 2018-10-15Manuscript Title Bacteria Regulate Activity of Enzymes Responsible for Metabolism of DrugsManuscript Type Direct SubmissionSpecial Features and Colloquia N/AClassification Biological Sciences/Applied Biological SciencesDatabase depositionCorresponding Author Abhinav Bhushan (Illinois Institute of Technology)

    Co-Authors Chengyao Wang , Andrea Cancino , Rongfei Fu , Thao Dang , Qifan He , AbhinavBhushan (corr-auth)

    AbstractThe trillions of microbes that inhabit our intestines and that are present inprobiotics affect health and regulate important biochemical factors. One of theimportant ... View full abstract

    Significance StatementThe microbes that inhabit our intestines and that are present in probiotics affecthealth and regulate important biochemical factors including metabolism of drugs.Due to ... View full Significance Statement

    Suggested Editorial Board MemberJames Collins (Massachusetts Institute of Technology)

    Suggested EditorSangeeta Bhatia (Massachusetts Institute of Technology), Minor Coon (Victor C.Vaughan Distinguished University), Philippe J. Sansonetti (Unité de PathogénieMicrobienne Moléculaire et Unité INSERM 1202, Institut Pasteur / Chaire deMicrobiologie et de Maladies Infectieuses, Collège de France)

    Author Conflict of InterestFunding Body Archiving Mandates No Funders

    Preprint ServerPlease let us know whether you have posted a version of your submission to apreprint server:

    No Electronic Forms 1 of 1 forms complete - View Electronic Forms Status

    Manuscript Items

    1. Manuscript File PDF (239KB)

    2. Parsed Citations (Parsed Citations file not available)

    3. Related Manuscript File PDF (1072KB)

    Manuscript Tasks Send Manuscript Correspondence Check Status

    Contact Feedback Subscribe For the Press Editorial Board Terms & Conditions

    MANUSCRIPT SUBMISSION

    ABOUT // AUTHORS // REVIEWERS // LOGOUT MY HOMEPAGE

    https://www.pnascentral.org/cgi-bin/main.plex?form_type=e_forms_chasers_display_ms&j_id=1&ms_id=451782&ms_rev_no=0&ms_id_key=ftdahPmh0gqo1cE7chpMWRA&from_view_ms=1&view_ms_return_to_ms_rev_no=0https://www.pnascentral.org/pnas_files/2018/10/10/00451782/00/451782_0_unknown_upload_7584164_pgnym3_sc.pdfhttps://www.pnascentral.org/pnas_files/2018/10/10/00451782/00/451782_0_unknown_upload_7584165_pgn1m4_sc.pdfhttps://www.pnascentral.org/cgi-bin/main.plex?form_type=display_contact&j_id=1&ms_id=451782&ms_rev_no=0&ms_id_key=ftdCFUN7BkQRDBPQ4wohykaghttps://www.pnascentral.org/cgi-bin/main.plex?form_type=status_details&j_id=1&ms_id=451782&ms_rev_no=0&ms_id_key=ftdoNISOVZSnz7XX7b29fMaPAhttp://www.pnas.org/page/about/contacthttp://pnas.org/cgi/feedbackhttp://pnas.org/site/subscriptions/index.xhtmlhttp://www.pnas.org/site/media/journalist.xhtmlhttp://www.pnas.org/page/about/editorial-boardhttps://www.pnascentral.org/html/terms_and_conditions.htmlhttp://www.pnas.org/https://www.pnascentral.org/html/about.htmlhttps://www.pnascentral.org/cgi-bin/main.plex?form_type=display_auth_instructionshttps://www.pnascentral.org/cgi-bin/main.plex?form_type=display_rev_instructionshttps://www.pnascentral.org/cgi-bin/main.plex?form_type=logouthttps://www.pnascentral.org/cgi-bin/main.plex?form_type=home

  • Bacteria Regulate Activity of Enzymes Responsible for Metabolism of Drugs

    Chengyao Wang, Andrea Cancino, Rongfei Fu, Thao Dang, Qifan He, Abhinav Bhushan

    Department of Biomedical Engineering, Illinois Institute of Technology,

    Chicago, IL 60616

    Abstract

    The trillions of microbes that inhabit our intestines and that are present in probiotics affect

    health and regulate important biochemical factors. One of the important factors that bacteria,

    both gut microbes and probiotics, can regulate is the activity of enzymes that are important

    for drug metabolism. Due to the challenges of an experimental system that can study

    intestinal-bacterial interactions, studies that can report these effects are lacking. We report a

    carefully designed study that systematically looks at different levels the interactions between

    the colon cells and different bacteria (gut microbes and probiotics). We find that paracrine

    signaling between the bacteria and colon cells is critical in regulating the activity of important

    enzymes responsible for drug metabolism. Pathway analysis revealed that the bacteria cause

    changes in diverse set of biological pathways, especially in those related to drug metabolism.

    This was confirmed by measuring specific activities of important cytochrome P450s. The

    major findings are that (1) species-specific differences exist in the regulation and activity of

    these enzymes, (2) regulation of the various enzymes appeared to be unique, and (3)

    communication between the bacteria and the colon cells is required for modulation of

    enzymatic activity. This knowledge can help a priori design of therapeutics.

    Significance statement

    The microbes that inhabit our intestines and that are present in probiotics affect health and

    regulate important biochemical factors including metabolism of drugs. Due to the challenges

    of an experimental system that can study intestinal-bacterial interactions, studies that report

    these effects are lacking. This gap has limited our ability to a priori use this knowledge in drug

    development. We systematically studied the interactions (genes and specific activity of phase

    I metabolism) between colon cells and bacteria. The major findings are that (1)

    species-specific differences exist in the regulation and activity of these enzymes, (2)

    regulation of the various enzymes appeared to be unique, and (3) communication between

    the bacteria and the colon cells is required for modulation of enzymatic activity.

  • Introduction

    The intestine is the site for processing of food, drugs, absorption of nutrients, and generation

    of waste; chronic intestinal ailments affect our overall physiological health. Microbiota that

    colonizes the intestinal microenvironment contribute to all these functions. The microbiota is

    perturbed by change in diet, onset of diseases, and consumption of drugs (1, 2). Almost 24%

    of the common drugs modulated the growth of 40 representative human gut bacterial strains

    (3). The microbiota themselves are causally linked to metabolic disorders (4) and to the

    metabolism of drugs (5). In fact, the microbiota may contribute to the metabolism of at least

    fifty drugs (6, 7). Recent studies have shown the effect gut microbes have on the activity of

    drugs such as acetaminophen and digoxin. For example, metronidazole, a drug used to treat

    Crohn’s disease, has antimicrobial properties which puts selective pressure on the intestinal

    microbiota that leads to inactivation of the drug and hence reduced efficiency (8, 9). Bacteria

    can inactivate a drug as in the case of Actinobacterium Eggerthella lenta which converts the

    cardiac drug digoxin into an inactive metabolite dihydrodigoxin (10). In another example,

    the drug sulfasalazine is hydrolyzed by an intestinal bacterium to 5-aminosalicylic acid, the

    active compound, and sulfapyridine. The parent drug, sulfasalazine, can inhibit the NF-κB

    inflammatory pathway whereas sulfapyridine cannot, illustrating a situation where the parent

    drug and its bacterial metabolites can have different mechanisms of action and presumably

    different targets (11). With the trillions of bacteria in the microbiome, the inter-individual

    diversity in the microbiome composition can have broad impact on the potential variation in

    PK/PD of drugs. Even probiotics, the usage of which is on the rise (4), modify the

    pharmacokinetics of orally administered drugs (12–14).

    Cytochrome P450 enzymes (CYPs) are proteins that are used as substrates for phase I

    drug metabolism and account for 75% of the metabolism of drugs (15). They are a major

    source of variability in drug pharmacokinetics and response (15). Fifty-seven human CYPs,

    belonging to the CYP1, 2, and 3 families, have been identified to be responsible for the

    biotransformation of most foreign substances including 70–80% of all drugs in clinical use

    and involved in over 90% of the reactions in drug metabolism (16). It has been shown that

    approximately 70% of human drug metabolism and pharmacokinetics can be attributed to six

    major CYP450s, which include CYP1A2, 2C9, 2C19, 2D6, 2E1, 3A4 and 3A5 . The CYP3A4

    in the intestine and the liver is involved in the metabolism of majority of the drugs on the

    market (23–27). The ability of gut bacteria to metabolize drugs is comparable to that of the

  • liver (28). CYP450 3A4 enzyme is abundant in the human intestine (29) and its activity is

    markedly lower in patients with Crohn's disease (30); enterocytic CYP3A4 influences

    coeliac disease and cystic fibrosis in pediatric populations (31). CYP1A2, another major

    cytochrome in the intestine, influences the oxidative metabolism of the exogenous and

    endogenous molecules including caffeine (32) as well as is involved in several drug

    interactions (33).

    CYP activity is modulated by many factors such as inflammation (34, 35), food (36), and

    bacteria (37–39). Blockade of pro-inflammatory cytokines and lipopolysaccharide (LPS) with

    aminoguanidine both extended down-regulation of CYP2A5 to the high dose range while not

    affecting LPS-induced depression of CYP1A and CYP2B (34, 40). The anti-inflammatory

    effect and modulation of cytochrome P450 activities by Artemisia annua tea infusions

    inhibited the activity of CYP3A4 and of CYP1A1 (36). Probiotics can also affect drug

    metabolism, for example, gut microbiota can influence the metabolism of nifedipine through

    affecting CYP3A in the intestinal mucosa (12). VSL#3, a common probiotic (41–44), which

    is a combination of eight strains, contains facultative or obligate, anaerobic, probiotic strains

    including L. acidophilus, L. plantarum, L. paracasei, L. delbrueckii subsp. bulgaricus, B. breve,

    B. longum, B. infantis, and Streptococcus thermophiles, modifies CYP activity (45–47).

    VSL#3 probiotic regulates the intestinal epithelial barrier in vivo and in vitro (42), induces

    mucin expression and secretion in colonic epithelial cells (48), and modulates human

    dendritic cell phenotype and function (49). Lactobacillus and Bifidobacterium have effects on

    gut disorders and drug metabolism where they can affect the metabolism of acetaldehyde

    and ethanol (45–47). All the Lactobacillus and Bifidobacterium strains from VSL#3 revealed

    a modification to oxidize ethanol and acetaldehyde which suggested a beneficial impact on

    high gastrointestinal acetaldehyde levels following alcohol intake (45).

    As we discover more and more the causative roles of bacteria on human health (4), the

    need for an assay to study the role of bacteria on the potential metabolism of drugs has never

    been more. However, there are limited experimental tools to screen drugs for potential

    metabolism by the gut microbiome. The major bottleneck to understanding impact of

    microbes (both intestinal and probiotics) on drug metabolism is the lack of fundamental

    analytical approaches to quantifying the enzymes that are responsible for metabolizing the

    drugs. Although the interactions between several drugs and intestinal bacterial strains were

  • recently reported (3), such studies ignore the role of intestinal cells, which we know play an

    important role in regulating the activity of the microbes. Thus, there remains a need for

    efficient approach for characterizing the role of bacteria on potential regulation of drug

    metabolism. In this paper, we have systematically studied the role of three different bacteria

    on the regulation of intestinal drug metabolizing enzymes. We have selected a probiotic

    (VSL#3), and two intestinal microbes E-coli Nissle 1917 and Bifidobacterium adolescentis (B.

    adolescentis). E-coli Nissle 1917, an anaerobic bacteria, is predominantly found in the human

    gut is another bacteria, has been shown to regulate drug metabolism (49–53). E. coli Nissle

    1917 administration has been shown responsible for better drug absorption from the

    gastrointestinal tract (54) as well as for maintaining tight epithelial junction and repair of the

    barrier function (55, 56). B. adolescentis, is a bacterial component seen in the gut

    microbiota and plays a significant role in influencing the prevention or/and treatment of

    different gastrointestinal infections, intestinal disorders such as Crohn’s disease (57), which

    could be attributed to the effects of mechanisms of B. adolescentis consumption of lactate

    and acetate (58) . Due to the challenges of in vitro culture of intestinal cells, we used Caco2

    cells because of their ability to form stable monolayers with the expression of relevant human

    drug transporters and CYP activity (59, 60). When grown on semi-permeable membranes,

    these cells can grow into 3D structures with tight barrier junctions as others and we have

    shown (59, 61) . When grown on semi-permeable membranes, these cells can grow into 3D

    structures with tight barrier junctions as others and we have shown (59, 61) . We studied

    more than 80 genes related to different aspects of drug metabolism under different culture of

    Caco2 cells and bacteria (Figure S2). We quantified not only the changes in relative mRNA

    but also the activity of important CYPs.

    Materials and Methods

    Cell Culture

    Human intestinal epithelial Caco2 cells were obtained from ATCC (Manassas, VA) and grown

    in 1X Eagle’s Modified Eagle Medium (EMEM) (VWR) containing 4.5 g/l glucose and 25 mM

    HEPES supplemented with 20% fetal bovine serum (FBS) (Gibco), 1% pen-strep (Gibco).

    The cells were cultured at 37°C in a humidified incubator with 5% CO2. Cells were cultured in

    a 75cm2 cell culture flask until the cells were 80% confluent. The cells were then seeded in

    well plates and on Transwell inserts (Corning, NY) and grown until confluent.

  • Bacteria Culture

    Three different types of bacteria strands were used which included VSL#3 (Alphasigma USA,

    Covington, LA), Bifidobacterium adolescentis (B. adolescentis ) (Manassas, VA), and E-Coli

    Nissle 1917 (Ardeypharm GmbH, Germany). All three bacteria strains were grown following

    the manufacturer’s instructions. The respective mediums included 1:1 mixture of Difco

    Lactobacilli MRS Broth (BD) and Reinforced Clostridial Medium (BD), Reinforced Clostridial

    Medium, and LB which contains 1% (W/V) Tryptone, 0.5% (W/V) yeast extract, and 1% (W/V)

    sodium hloride. The bacteria were first plated on agar plates to determine the standard curve

    of bacteria colonies and optical density (OD). The gradient concentrations of original bacteria

    suspensions (½, 1/5, 1/10, 1/25, 1/50, 1/100, 1/250, 1/500, 1/1000) were used to determine

    the standard curve. Twenty μl of bacteria suspension and a control of the medium were

    seeded at agar plate. The OD of each concentration was measured using the Spectra Max

    M2 (Molecular Devices) by measuring the absorbance at 600nm. After the bacteria colonies

    were visible, an image was taken on the microscope and the number of colonies was counted

    using Image J (NIH, Bethesda, MD). Once that information was obtained, a standard curve

    was made (Figure S1). The standard curve was used to quantify the MOI for the cocultures.

    Experimental Design

    We carried out three sets of experiments (Figure S2). (1) Caco2 cells and bacteria were

    cocultured (a) in well plates and (b) in (0.4 um) Transwell inserts. In well plates, the cells and

    bacteria were in contact with each other. In Transwell inserts, cells were seeded on the

    inserts in the top chamber and bacteria were seeded at bottom where caused the interactions

    to be paracrine. Three different Multiplicity of Infection (MOI) of bacteria and Caco2 cells were

    tested - 1:1, 10:1, and 100:1. The viability of Caco2 cells seeded with the bacteria for 2, 4, 6,

    and 8 hours was estimated. Live/dead images were used to calculate the cell viability.

    (2) We also used the bacterial supernatant to test its effect on intestinal enzymatic activity.

    The bacteria supernatant was added to the Caco2 cell culture media in three different ratios -

    1:1, 1:10, and 1:100. The conditioned media was introduced into (a) well plates and (b)

    bottom well of Transwell inserts. The bacteria supernatant was filtered by 0.22 um syringe

    filter (Corning) to clear all the bacteria effect. The viability of Caco2 cells seeded with the

    different ratio of bacteria supernatant for 8 hours was estimated.

    (3) We then used a few of the specific metabolites that the bacteria are known to produce.

    Three metabolites (butyrate (Sigma), propionate (Sigma), p-cresol (Sigma)) were selected.

  • Bacterial metabolic products, such as short chain fatty acids and their relative ratios, may be

    causal in disturbed immune and metabolic signaling (62). Metabolites were mixed with cell

    culture media to result appropriate concentration - 2 mM butyrate (63, 64), 3 mM propionate

    (64, 65), and 1 mM p-cresol (66, 67). The conditioned metabolites were introduced into (a)

    tissue culture plates and (b) bottom chamber of Transwell inserts. The viability of Caco2 cells

    seeded with the different metabolites for 8 hours was estimated.

    cDNA Synthesis and Quantitative Polymerase Chain Reaction (qPCR)

    Caco2 cells and the bacteria co-cultures were prepared as described above. After a 4 hour

    incubation, the cells were washed with sterile 1X PBS (Sigma) and 1X trypsin (Gibco) was

    added to detach the cells from the walls. The RNA was isolated using the RNeasy Mini Kit

    (QIAGEN, Germantown, MD). RNA concentration of each sample was quantified, and cDNA

    samples prepared by using the First Stand Kit (Qiagen) and SYBR Green (Qiagen) using

    Thermo Cycler C1000 Touch (BIO-RAD, Hercules, CA). qPCR was performed using the RT2

    Profiler PCR Arrays (QIAGEN) on the ABI ViiA7 (Thermo Fisher, Skokie, IL).

    Pathway analysis

    Heatmap was generated using the log2 (fold change) of the different genes by FunRich (68).

    Data were also analyzed with FunRich, Gene Ontology (69, 69), and Reactome (70) to study

    biological pathways with the following analysis settings: the fold change that larger than 3 and

    less than 0.3 were selected. Fold change was calculated as below: first, the ΔCt value, which

    is the difference between Ct of target gene and of HKG genes of each treatment and control,

    was calculated. By subtracting the ΔCt values of experimental group from the control, ΔΔCt

    was calculated for each gene which was used to calculate the fold change = 2 (-ΔΔCt). Heatmap

    was generated from 28 genes with the p-value < 0.05. In the heatmap, log2 fold change was

    used to represent gene expression and the color map indicates the downregulation of gene

    expression (log2 fold change < 0) as blue and up regulation as red (log2 fold change > 0).

    The Ct values of seven important CYP 450s were normalized to same housekeeping genes

    (ΔCt = Ct of target gene - Ct of HKG gene). Fold change was calculated following the Qiagen

    protocol based on the difference between control group and experimental group and log2 fold

    change was plotted to show the genes that were up and down regulated. (n>=3)

  • CYP 450 Analysis

    Caco2 cells were seeded in the 48-well plate and a 48 Transwell inserts for 2-3 days,

    following which the bacteria was added as described above and cultured together in a 37°C,

    5% CO2 incubator. P450-Glow CYP3A4 Assay with Luciferin-IPA, P450-Glo™ CYP1A2 and

    CYP3A4 were purchased from Promega (Madison, WI) and were used according to the

    manufacturer’s instructions. The luminescence of each experiment was read using the

    Spectra Max M2.

    Data analysis

    Unless otherwise indicated, data are expressed as means ± SEM. Differences between

    experimental groups were compared using t test. Statistical analyses were done with Excel

    and FunRich. Differences with a p < 0.05 were considered statistically significant.

    Results and Discussions

    Cell viability in coculture

    We used VSL#3 as model bacteria to determine the time for which the co-culture could

    be carried out without significant loss of Caco2 cell viability. Viability of Caco2 cells

    co-cultured with VSL#3 bacteria in a well plate was stable at first 4 hours, which stayed at

    least at 97%. When the coculture was taken to 8 hours, the viability of Caco2 cells decreased

    to 65% (Figure 1A, D). In contrast, the coculture in Transwell insert could be taken beyond 8

    hours without significant drop in viability (Figure 1B, E). Similar high viability was observed

    when the cells were cultured under different ratios of bacterial supernatant and cell culture

    media (Figure 1C, F). Based on these results, we selected the four-hour time point for the

    subsequent co-culture studies.

    Gene expression analysis

    The heatmap shows that in the selected gene set, the upregulation of genes by B.

    adolescentis was higher than that by VSL#3 and E.coli Nissle1917 (Figure 2A, Figure S3).

    These genes belong to CYP450, aldehyde dehydrogenases, and flavin containing

    monooxygenases. Surprisingly, B. adolescentis upregulated most of the genes whereas the

    other two bacteria led to a downregulation of the same genes. We discovered that for VSL#3,

    CYP1A2, 3A4, 2C9, and 2E1 expression was highly upregulated, whereas 2C19, 2D6 and

  • 3A5 was downregulated. E.coli Nissle 1917 upregulated CYP2C9, 2E1, 3A4, and 3A5 and

    downregulated CYP1A2, 2C19 and 2D6. B. adolescentis downregulated CYP 2D6 and 3A5

    while upregulating the other five. If we look at just the ΔCt values (lower ΔCt value reports

    higher gene expression), we find that while B. adolescentis and VSL#3 upregulated several

    of the CYP450 genes significantly (p < 0.05), E.coli Nissle 1917 did not (Figure 2C). VSL#3

    and B. adolescentis significantly upregulated the expression of CYP 1A2 and 3A4, whereas

    only B. adolescentis had an effect (downregulation) on the expression of CYP2D6. The data

    suggests that B. adolescentis and VSL#3 will potentially affect the pharmacokinetics of drugs,

    at least of those that rely on phase I metabolism which could be significant as these include

    the CYPs that contribute to almost 70% of human drug metabolism and pharmacokinetics

    (16).

    Pathway analysis

    We carried out analysis to identify the major pathways that corresponded to the

    upregulated and downregulated genes for each of the bacteria (Figure 3). The data suggest

    that all three bacteria regulate drug metabolism and inflammatory signals. The analysis

    showed that for VSL#3 the upregulated genes contributed primarily to processes for

    metabolism of xenobiotics and synthesis of HETE whereas the downregulated genes to

    modification of inert chemicals into biologically reactive species (69) (Figure 3A). Synthesis

    of HETE is associated with inflammatory signals and cancers as well as with the ability to

    facilitate tumor cell motility (70). E.coli Nissle 1917 contributed to pathways important for

    xenobiotics, endogenous sterols, fatty acids, and the synthesis of epoxy,

    dihydroxyeicosatrienoic acids, and HETE (Figure 3B). Excessive storage of fatty acids can

    lead to obesity related dyslipidemia, hypertension, and hypertriglyceridemia and also alters

    the lipid metabolism (71). EET and DHET are involved in by formation and metabolism of

    CYP450s (71). The genes up- and down- regulated by B. adolescentis contributed to

    pathways similar to E.coli Nissle 1917 (Figure 3C). The pathways affected by E.coli Nissle

    1917 and B. adolescentis were primarily related to the modification of xenobiotic metabolism

    and inflammatory signals, but the proportion that the genes contribute to each pathway was

    less than VSL#3. The differences among VSL#3 and the other two bacteria may occur

    because VSL#3 comprises eight different strains. While it may be interesting to study the

    effect of each of these stains, our results are important because consumers take VSL#3 as a

    compound-probiotic. The knowledge can help a priori design of therapeutics.

  • CYP 450 activity

    The bacteria we tested modify the activity of drug metabolizing enzymes CYP3A4 and

    1A2. Three different Multiplicity of Infection (MOI) of bacteria and Caco2 cells were used - 1:1,

    10:1, and 100:1 which were derived from the standard curve (Figure S1). Bringing the

    bacteria in contact with the cells suppressed the CYP activity for in all three cases (Figure 4).

    Both VSL#3 and its supernatant up-regulate CYP3A4 and 1A2 in Transwell inserts, but not in

    well plate (Figure 4A). We observed that for MOI =1:1, 10:1 and 100:1, the activity of

    CYP3A4 was more than 3-fold over the control and more than 2-fold over control for CYP1A2.

    However, culturing the cells with different amounts of the bacteria supernatant conditioned

    media recapitulated only partial activity of the two CYP enzymes (Figure 4B), suggesting that

    signaling between the bacteria and colon cells in an important factor in determining the

    activities. Paracrine signaling between VSL#3 and Caco2 cells upregulated activity of both

    CYP enzymes CYP3A4 and 1A2. The modification of CYP3A4 activity was higher than that of

    CYP1A2. VSL#3 supernatant also upregulated the activities although at a lower level than the

    co-culture, again, suggesting the important of bidirectional paracrine signaling. In well plate

    cultures, surprisingly, the supernatant did not affect CYP3A4 activity but downregulated

    CYP1A2 activity. E.coli Nissle 1917 modified the activity of CYP3A4 and 1A2 in the well plate

    as well as the Transwell inserts (Figure 4C). In plates, CYP3A4 activity was upregulated

    when for MOIs 1:1 and 100:1, whereas there were no changes for MOI=10:1. E.coli Nissle

    1917 did not affect the activity of CYP1A2 in the well plate. In the Transwell insert, CYP3A4

    activity went up and the highest fold change was 1.9 that occurred at MOI=10:1 while the fold

    change for MOI=1:1 and 100:1 was 1.4 and 1.5. However, the E.coli’s effect on CYP1A2 was

    the opposite. The highest reduction in fold change, which was 0.75 compared to control, was

    at MOI=10:1 (Figure 4C). E.coli Nissle 1917 supernatant did not change CYP3A4 activity but

    slightly decreased the activity of CYP1A2 when the ratio of supernatant and media was 1:100

    (Figure 4D). By decreasing the bacterial supernatant from 1:1 to1:100, the CYP1A2 activity

    fold change decreased from 1.1 to 0.85 compare to control. E.coli Nissle 1917 supernatant

    caused the activity of CYP3A4 to increase, while decreasing that of CYP1A2 (Figure 4D).

    When the ratio of E.coli Nissle 1917 supernatant to cell culture media was 1:10, the

    upregulation on CYP3A4 activity was the highest. The other two treatments, ratio of 1:10 and

    1:100, just showed upregulation in the activity of CYP3A4. Unlike CYP3A4, the activity of

    CYP1A2 was downregulated under all three ratios. B. adolescentis slightly lowered CYP3A4

    activity at all MOIs in cell culture plate (Figure 4E). The CYP1A2 activity was also slightly

  • downregulated. In the Transwell insert, CYP3A4 activity was increased only at MOIs 10:1 and

    100:1. When the MOI decreased to 1:1, there was no influence on 3A4 activity any more. In

    contrast, CYP1A2 activity was slightly upregulated at MOI of 10:1. There was no influence on

    activity when the MOIs were 1:1 and 100:1. The effect of B. adolescentis supernatant on the

    activity of both CYPs was similar to that in the tissue culture plate – higher ratio of the

    supernatant corresponded to lower activity. In Transwell inserts, B. adolescentis supernatant

    slightly increased the CYP3A4 activity (Figure 4F). There was no significant change in the

    activity of CYP1A2 at 1:1; however, when the ratio decreased to 1:10 and 1:100, the fold

    change value of CYP1A2 activity was higher. Our data suggests that the bacteria have

    significant effect on CYP activity when the cells and bacteria are not in contact. Bacterial

    communication with the colon cells appears to be important in modulating the CYP activity.

    Although bacterial supernatant can modify CYP activity as well, the levels are not the same

    as those by the bacteria. In contrast, tests with individual bacterial metabolites (2mM butyrate,

    3mM propionate and 2mM p-cresol) indicated no significant influence on CYP3A4 activity and

    a downregulation of CYP1A2 activity (Figure 4G), although at levels lower than the other

    cases. The results indicate that the individual metabolites do not significantly regulate CYP

    activity as compared to the co-culture and the supernatant.

    Conclusions

    Despite advances in our understanding a bacteria’s (gut microbe and probiotics)

    contribution to human health, one factor of the host-microbe interactions that remains

    understudied is the influence of bacteria on drugs. These interactions could have an

    important role in explaining the variation in human responses to drugs or the variable

    response to therapy. However, there is a serious lack of available data that one could use to

    determine how bacteria would potentially regulate a drug’s metabolism. As a first step to fill

    that gap, we have established and thoroughly characterized a simple co-coculture between

    colon cells and different types of bacteria. We have systematically dissected the role of

    colon-bacteria interactions versus paracrine signaling through co-cultures of human colon

    cells with three different strains of gut bacteria. We have used this setup to study alterations

    in general phase I enzymes and more importantly, measured the activity of human CYP450.

    Our findings indicate that the bacteria and their supernatant both modify the activity of drug

    metabolizing enzymes. We found species specific differences in the regulation and activity of

    the phase I enzymes uniquely. Our results indicate that communication between the bacteria

  • and the colon cells is required for modulation of metabolic activity. The regulation of the

    various CYP450’s appeared to be different, which suggested that studies are needed to

    uncover the regulatory mechanisms.

    Supplementary

    Figures are included in the supplementary material.

    Abbreviations

    TV: the colon cells co-cultured with VSL#3 in Transwell inserts, MOI=1:10

    TN: the colon cells co-cultured with E.coli Nislle 1917 in Transwell inserts, MOI=1:10

    TB: the colon cells co-cultured with B. adolescentis in Transwell inserts, MOI=1:10

    TC: the colon cells cultured in Transwell inserts, without bacteria.

    MOI: Multiplicity of Infection

    P.MOI: the Multiplicity of Infection bacteria and cells tissue culture plate, ranging from 1:1 to

    100:1

    T.MOI: the Multiplicity of Infection bacteria and cells Transwell inserts, ranging from 1:1 to

    100:1

    P.CM: the bacteria supernatant conditioned media in tissue culture plate with different ratio

    range from 1:1 to 1:100

    P.CM: the bacteria supernatant conditioned media in Transwell inserts with different ratio

    range from 1:1 to 1:100.

    Acknowledgments

    This project was partly funded by the Nayar Prize II and student scholarships from the Armor

    College of Engineering. We acknowledge technical help from Dr. Hong lab.

    Author contributions

    CW designed the study, conducted experiments, and wrote the manuscript. AC conducted

    experiment and wrote the manuscript. RF, TD, and QH conducted experiment. AB designed

    the study and wrote the manuscript.

  • References

    1. Flowers SA, Evans SJ, Ward KM, McInnis MG, Ellingrod VL (2017) Interaction Between

    Atypical Antipsychotics and the Gut Microbiome in a Bipolar Disease Cohort.

    Pharmacotherapy 37 (3):261–267.

    2. Forslund K, et al. (2015) Disentangling type 2 diabetes and metformin treatment

    signatures in the human gut microbiota. Nature 528 (7581):262–266.

    3. Maier L, et al. (2018) Extensive impact of non-antibiotic drugs on human gut bacteria.

    Nature 555 (7698):623–628.

    4. Scheithauer TPM, Dallinga-Thie GM, de Vos WM, Nieuwdorp M, van Raalte DH (2016)

    Causality of small and large intestinal microbiota in weight regulation and insulin resistance.

    Mol Metab 5 (9):759–770.

    5. Fuller AT (1937) IS p-aminobenzenesulphonamide the active agent in prontosil therapy?

    The Lancet 229 (5917):194–198.

    6. Li H, He J, Jia W (2016) The influence of gut microbiota on drug metabolism and toxicity.

    Expert Opin Drug Metab Toxicol 12 (1):31–40.

    7. Spanogiannopoulos P, Bess EN, Carmody RN, Turnbaugh PJ (2016) The microbial

    pharmacists within us: a metagenomic view of xenobiotic metabolism. Nat Rev Microbiol 14

    (5):273–287.

    8. Yoo DH, et al. (2014) Gut microbiota-mediated drug interactions between lovastatin and

    antibiotics. Drug Metab Dispos 42 (9):1508–13.

    9. Arndt H, Palitzsch KD, Grisham MB, Granger DN (1994) Metronidazole inhibits

    leukocyte-endothelial cell adhesion in rat mesenteric venules. Gastroenterology 106

    (5):1271–6.

    10. Saha JR, Butler J VP, Neu HC, Lindenbaum J (1983) Digoxin-inactivating bacteria:

    identification in human gut flora. Science 220 (4594):325–7.

    11. Wahl C, Liptay S, Adler G, Schmid RM (1998) Sulfasalazine: a potent and specific

    inhibitor of nuclear factor kappa B. J Clin Invest 101 (5):1163–74.

    12. Kato R, et al. (2007) Effect of Lactobacillus casei on the absorption of nifedipine from rat

    small intestine. Drug Metab Pharmacokinet 22 (2):96–102.

    13. Lee HJ, Zhang H, Orlovich DA, Fawcett JP (2012) The influence of probiotic treatment on

    sulfasalazine metabolism in rat. Xenobiotica 42 (8):791–7.

    14. Stojancevic M, Bojic G, Salami HA, Mikov M (2014) The Influence of Intestinal Tract and

    Probiotics on the Fate of Orally Administered Drugs. Curr Issues Mol Biol 16:55–68.

  • 15. Pikuleva IA, Waterman MR (2013) Cytochromes P450: Roles in Diseases. J Biol Chem

    288 (24):17091–17098.

    16. Lin JH (2007) Pharmacokinetic and pharmacodynamic variability: a daunting challenge in

    drug therapy. Curr Drug Metab 8 (2):109–136.

    17. Meyer UA, Skoda RC, Zanger UM (1990) The genetic polymorphism of

    debrisoquine/sparteine metabolism-molecular mechanisms. Pharmacol Ther 46 (2):297–308.

    18. von Moltke LL, et al. (1998) Multiple human cytochromes contribute to biotransformation

    of dextromethorphan in-vitro: role of CYP2C9, CYP2C19, CYP2D6, and CYP3A. J Pharm

    Pharmacol 50 (9):997–1004.

    19. Uttamsingh V, Lu C, Miwa G, Gan L-S (2005) Relative contributions of the five major

    human cytochromes P450, 1A2, 2C9, 2C19, 2D6, and 3A4, to the hepatic metabolism of the

    proteasome inhibitor bortezomib. Drug Metab Dispos Biol Fate Chem 33 (11):1723–1728.

    20. Turpault S, et al. (2009) Pharmacokinetic assessment of a five-probe cocktail for CYPs

    1A2, 2C9, 2C19, 2D6 and 3A. Br J Clin Pharmacol 68 (6):928–935.

    21. Yenilmez ED, Tamam L, Karaytug O, Tuli A (2018) Characterization CYP1A2, CYP2C9,

    CYP2C19 and CYP2D6 Polymorphisms Using HRMA in Psychiatry Patients with

    Schizophrenia and Bipolar Disease for Personalized Medicine. Comb Chem High Throughput

    Screen 21 (5):374–380.

    22. Paine MF, et al. (2006) THE HUMAN INTESTINAL CYTOCHROME P450 “PIE.” Drug

    Metab Dispos Biol Fate Chem 34 (5):880–886.

    23. Labroo RB, Paine MF, Thummel KE, Kharasch ED (1997) Fentanyl Metabolism by

    Human Hepatic and Intestinal Cytochrome P450 3A4: Implications for Interindividual

    Variability in Disposition, Efficacy, and Drug Interactions. Drug Metab Dispos 25 (9):1072–

    1080.

    24. Fang J, Bourin M, Baker GB (1999) Metabolism of risperidone to 9-hydroxyrisperidone by

    human cytochromes P450 2D6 and 3A4. Naunyn Schmiedebergs Arch Pharmacol 359

    (2):147–151.

    25. Khan KK, He YQ, Correia MA, Halpert JR (2002) Differential Oxidation of Mifepristone by

    Cytochromes P450 3A4 and 3A5: Selective Inactivation of P450 3A4. Drug Metab Dispos 30

    (9):985–990.

    26. Zhou S-F (2008) Drugs behave as substrates, inhibitors and inducers of human

    cytochrome P450 3A4. Curr Drug Metab 9 (4):310–322.

    27. Yamazaki H, Nakamoto M, Shimizu M, Murayama N, Niwa T (2010) Potential impact of

  • cytochrome P450 3A5 in human liver on drug interactions with triazoles. Br J Clin Pharmacol

    69 (6):593–597.

    28. Kim DH (2015) Gut Microbiota-Mediated Drug-Antibiotic Interactions. Drug Metab Dispos

    43 (10):1581–9.

    29. Lin JH, Chiba M, Baillie TA (1999) Is the Role of the Small Intestine in First-Pass

    Metabolism Overemphasized? Pharmacol Rev 51 (2):135–158.

    30. Wilson A, Tirona RG, Kim RB (2017) CYP3A4 Activity is Markedly Lower in Patients with

    Crohn’s Disease. Inflamm Bowel Dis 23 (5):804–813.

    31. Johnson TN, Tanner MS, Taylor CJ, Tucker GT (2001) Enterocytic CYP3A4 in a

    paediatric population: developmental changes and the effect of coeliac disease and cystic

    fibrosis. Br J Clin Pharmacol 51 (5):451–460.

    32. Härtter S, et al. (2003) Effects of caffeine intake on the pharmacokinetics of melatonin, a

    probe drug for CYP1A2 activity. Br J Clin Pharmacol 56 (6):679–682.

    33. Chen H, et al. (2014) Bioactive components of Glycyrrhiza uralensis mediate drug

    functions and properties through regulation of CYP450 enzymes. Mol Med Rep 10 (3):1355–

    1362.

    34. De-Oliveira ACAX, Poça KS, Totino PRR, Paumgartten FJR (2015) Modulation of

    Cytochrome P450 2A5 Activity by Lipopolysaccharide: Low-Dose Effects and Non-Monotonic

    Dose-Response Relationship. PLoS ONE 10 (1). doi:10.1371/journal.pone.0117842.

    35. De-Oliveira ACA, et al. (2010) Up- and down-modulation of liver cytochrome P450

    activities and associated events in two murine malaria models. Malar J 9 (1):81.

    36. Melillo de Magalhaes P, et al. (2012) Anti-inflammatory effect and modulation of

    cytochrome P450 activities by Artemisia annua tea infusions in human intestinal Caco-2 cells.

    Food Chem 134:864–71.

    37. Claus SP, et al. (2011) Colonization-Induced Host-Gut Microbial Metabolic Interaction.

    mBio 2 (2):e00271-10.

    38. Bezirtzoglou EEV (2012) Intestinal cytochromes P450 regulating the intestinal microbiota

    and its probiotic profile. Microb Ecol Health Dis 23 (1):18370.

    39. Boer J, et al. (2016) The Role of UGT, CYP and Gut Microbiota in the Metabolism of

    Epacadostat (EPA) in Humans. Drug Metab Dispos:dmd.116.070680.

    40. Li-Masters T, Morgan ET (2001) Effects of Bacterial Lipopolysaccharide on

    Phenobarbital-Induced CYP2B Expression in Mice. Drug Metab Dispos 29 (3):252–257.

    41. Selwyn FP, Cheng SL, Klaassen CD, Cui JY (2016) Regulation of Hepatic

  • Drug-Metabolizing Enzymes in Germ-Free Mice by Conventionalization and Probiotics. Drug

    Metab Dispos Biol Fate Chem 44 (2):262–274.

    42. Dai C, Zhao D-H, Jiang M (2012) VSL#3 probiotics regulate the intestinal epithelial barrier

    in vivo and in vitro via the p38 and ERK signaling pathways. Int J Mol Med 29 (2):202–208.

    43. Cho SS, Finocchiaro T (2009) Handbook of Prebiotics and Probiotics Ingredients: Health

    Benefits and Food Applications (CRC Press).

    44. Gionchetti P, et al. (2002) Probiotics — Role in inflammatory bowel disease. Dig Liver Dis

    34:S58–S62.

    45. Nosova T, Jousimies-Somer H, Jokelainen K, Heine R, Salaspuro M (2000)

    ACETALDEHYDE PRODUCTION AND METABOLISM BY HUMAN INDIGENOUS AND

    PROBIOTIC LACTOBACILLUS AND BIFIDOBACTERIUM STRAINS. Alcohol Alcohol 35

    (6):561–568.

    46. Presti I, et al. (2015) Evaluation of the probiotic properties of new Lactobacillus and

    Bifidobacterium strains and their in vitro effect. Appl Microbiol Biotechnol 99 (13):5613–5626.

    47. Paturi G (2007) Probiotic characteristics of Lactobacillus acidophilus and Lactobacillus

    paracasei and their effects on immune response and gene expression in mice. Available at:

    https://researchdirect.westernsydney.edu.au/islandora/object/uws%3A2504/ [Accessed

    October 12, 2018].

    48. Caballero-Franco C, Keller K, De Simone C, Chadee K (2007) The VSL#3 probiotic

    formula induces mucin gene expression and secretion in colonic epithelial cells. Am J

    Physiol-Gastrointest Liver Physiol 292 (1):G315–G322.

    49. Lammers KM, et al. (2002) Effect of probiotic strains on interleukin 8 production by

    HT29/19A cells. Am J Gastroenterol 97 (5):1182–1186.

    50. Wassenaar TM (2016) Insights from 100 Years of Research with Probiotic E. Coli. Eur J

    Microbiol Immunol 6 (3):147–161.

    51. Schultz M, et al. (2004) Preventive Effects of Escherichia coli Strain Nissle 1917 on Acute

    and Chronic Intestinal Inflammation in Two Different Murine Models of Colitis. Clin Diagn Lab

    Immunol 11 (2):372–378.

    52. Maltby R, Leatham-Jensen MP, Gibson T, Cohen PS, Conway T (2013) Nutritional basis

    for colonization resistance by human commensal Escherichia coli strains HS and Nissle 1917

    against E. coli O157:H7 in the mouse intestine. PloS One 8 (1):e53957.

    53. Schultz M (2008) Clinical use of E. coli Nissle 1917 in inflammatory bowel disease.

    Inflamm Bowel Dis 14 (7):1012–1018.

  • 54. Matuskova Z, et al. (2014) Administration of a Probiotic Can Change Drug

    Pharmacokinetics: Effect of E. coli Nissle 1917 on Amidarone Absorption in Rats. PLOS ONE

    9 (2):e87150.

    55. Ukena SN, et al. (2007) Probiotic Escherichia coli Nissle 1917 Inhibits Leaky Gut by

    Enhancing Mucosal Integrity. PLOS ONE 2 (12):e1308.

    56. Zyrek AA, et al. (2007) Molecular mechanisms underlying the probiotic effects of

    Escherichia coli Nissle 1917 involve ZO-2 and PKCζ redistribution resulting in tight junction

    and epithelial barrier repair. Cell Microbiol 9 (3):804–816.

    57. Belenguer A, et al. (2006) Two Routes of Metabolic Cross-Feeding between

    Bifidobacterium adolescentis and Butyrate-Producing Anaerobes from the Human Gut. Appl

    Env Microbiol 72 (5):3593–3599.

    58. Kim HJ, Huh D, Hamilton G, Ingber DE (2012) Human gut-on-a-chip inhabited by

    microbial flora that experiences intestinal peristalsis-like motions and flow. Lab Chip 12

    (12):2165–2174.

    59. Zhan T, et al. (2018) Metabolic Susceptibility of 2-Chlorothioxanthone and Its Toxic

    Effects on mRNA and Protein Expression and Activities of Human CYP1A2 and CYP3A4

    Enzymes. Environ Sci Technol. doi:10.1021/acs.est.8b04643.

    60. Wang C, Tanataweethum N, Karnik S, Bhushan A (2018) Novel Microfluidic Colon with an

    Extracellular Matrix Membrane. ACS Biomater Sci Eng 4 (4):1377–1385.

    61. Zhao L, et al. (2018) Gut bacteria selectively promoted by dietary fibers alleviate type 2

    diabetes. Science 359 (6380):1151–1156.

    62. Barcenilla A, et al. (2000) Phylogenetic Relationships of Butyrate-Producing Bacteria

    from the Human Gut. Appl Env Microbiol 66 (4):1654–1661.

    63. Gibson SA, Sewell GW (1992) Stimulation of Reductive Dechlorination of

    Tetrachloroethene in Anaerobic Aquifer Microcosms by Addition of Short-Chain Organic Acids

    or Alcohols. Appl Env Microbiol 58 (4):1392–1393.

    64. Güven D, et al. (2005) Propionate Oxidation by and Methanol Inhibition of Anaerobic

    Ammonium-Oxidizing Bacteria. Appl Env Microbiol 71 (2):1066–1071.

    65. Shields MS, Montgomery SO, Chapman PJ, Cuskey SM, Pritchard PH (1989) Novel

    Pathway of Toluene Catabolism in the Trichloroethylene-Degrading Bacterium G4. Appl Env

    Microbiol 55 (6):1624–1629.

    66. Beller HR, Spormann AM, Sharma PK, Cole JR, Reinhard M (1996) Isolation and

    characterization of a novel toluene-degrading, sulfate-reducing bacterium. Appl Env Microbiol

  • 62 (4):1188–1196.

    67. Pathan M, et al. (2015) FunRich: An open access standalone functional enrichment and

    interaction network analysis tool. PROTEOMICS 15 (15):2597–2601.

    68. Ashburner M, et al. (2000) Gene ontology: tool for the unification of biology. The Gene

    Ontology Consortium. Nat Genet 25 (1):25–29.

    69. Joshi-Tope G, et al. (2005) Reactome: a knowledgebase of biological pathways. Nucleic

    Acids Res 33 (suppl_1):D428–D432.

    70. Patterson AD, Gonzalez FJ, Idle JR (2010) Xenobiotic Metabolism: A View through the

    Metabolometer. Chem Res Toxicol 23 (5):851–860.

    71. Powell WS, Rokach J (2015) Biosynthesis, biological effects, and receptors of

    hydroxyeicosatetraenoic acids (HETEs) and oxoeicosatetraenoic acids (oxo-ETEs) derived

    from arachidonic acid. Biochim Biophys Acta 1851 (4):340–355.

    72. Hanif A, Edin ML, Zeldin DC, Morisseau C, Nayeem MA (2016) Effect of Soluble Epoxide

    Hydrolase on the Modulation of Coronary Reactive Hyperemia: Role of Oxylipins and PPARγ.

    PLOS ONE 11 (9):e0162147.

  • List of figures and tables

    Figure 1. Viability of colon cells co-cultured with VSL#3 bacteria in (A) well plate and (B)

    Transwell inserts, in (C) different proportion of VSL#3 bacteria supernatant and cell culture

    media mixture in tissue culture plate for 4 hr. Live/dead stain of colon cells co-cultured with

    VSL#3 for 4 hr and 8 hr in (D) well plate and (E) Transwell inserts; (F) Live/dead stain of colon

    cells cocultured with VSL#3 supernatant conditioned media for 4 hr, in ratio of 1:1 and 1:10. S:

    VSL3 supernatant, M: cell culture media; Green live, Red dead.

    Figure 2. qPCR analysis of the colon cells co-cultured with VSL#3, E.coli Nissle 1917 and B.

    adolescentis bacteria in Transwell inserts. (A) Heatmap generated from 28 genes (p-value <

    0.05). The gene expression is plotted as log2 fold change. (B) Log2 fold change plot of seven

    CYPs. Fold change was calculated based on the control group. (n>=3). (C) Ct values of

    seven primary CYP450s genes normalized to same housekeeping gene. Values represent

    mean ± SEM (Student’s t test; *p < 0.05). TV: colon cells co-cultured with VSL#3 in Transwell

    inserts, MOI=1:10; TN: colon cells co-cultured with E.coli Nissle 1917 in Transwell inserts,

    MOI=1:10; TV: colon cells co-cultured with B. adolescentis in Transwell inserts, MOI=1:10;

    TC: colon cells cultured in Transwell inserts, without bacteria.

    Figure 3. (A) – (C) Biological pathway analysis of colon cells co-cultured with VSL#3, E.coli

    Nissle 1917 and B. adolescentis in Transwell inserts for upregulated (fold change >3) and

    downregulated genes (fold change

  • cells Transwell inserts, ranging from 1:1 to 100:1; P.CM: bacterial supernatant conditioned

    media in well plate with different ratios ranging from 1:1 to 1:100; P.CM: bacterial supernatant

    conditioned media in Transwell inserts in different ratios ranging from 1:1 to 1:100.

    Figure S1. Bacteria standard curve of OD and density (CFU/mL)

    Figure S2. Schematic diagram of co-culture between colon cells and bacteria in well plate

    and Transwell inserts.

    Figure S3. qPCR analysis of the colon cells co-cultured with VSL#3, E.coli Nissle 1917 and B.

    adolescentis bacteria in Transwell inserts. Heatmap generated from 84 genes.

    Figure S4. Log2 Fold Change plot of the all 84 genes in the kit. Fold change was calculated

    based on the control group and separated to 6 different enzymes group. CYP450:

    Cytochrome P450s; ADH:Alcohol Dehydrogenases; ALDH: Aldehyde Dehydrogenases; FMO:

    Flavin Containing Monooxygenases.

    Figure S5. Biological pathways analysis of colon cells co-cultured with VSL#3 bacteria in

    Transwell inserts based on genes fold change >3 and 3 and 3 was considered as upregulated and < 0.3 as downregulated.

  • A

    Figure 1. Viability of colon cells co-cultured with VSL#3 bacteria in (A) well plate and (B) Transwell inserts, in(C) different proportion of VSL#3 bacteria supernatant and cell culture media mixture in tissue culture platefor 4 hr. Live/dead stain of colon cells co-cultured with VSL#3 for 4 hr and 8 hr in (D) well plate and (E)Transwell inserts; (F) Live/dead stain of colon cells cocultured with VSL#3 supernatant conditioned media for4 hr, in ratio of 1:1 and 1:10. S: VSL3 supernatant, M: cell culture media; Green live, Red dead.

    B C

    0

    25

    50

    75

    100

    Ctrl 2 4 8

    % V

    iab

    ility

    Coculture time (h)

    0

    25

    50

    75

    100

    Ctrl 2 4 8

    % V

    iab

    ility

    Coculture time (h)

    D E F

    4 hrs 4 hrs S:M=1:18 hrs 8 hrs S:M=1:10

    0

    25

    50

    75

    100

    0 0.5 0.1 0.04 0.02 0.01

    % V

    iab

    ility

    proportion of VSL#3 bacteria supernatant

  • Figure 2. qPCR analysis of the colon cells co-cultured with VSL#3, E.coli Nissle 1917 and B.adolescentis bacteria in Transwell inserts. (A) Heatmap generated from 28 genes (p-value < 0.05).The gene expression is plotted as log2 fold change. (B) Log2 fold change plot of seven CYPs. Foldchange was calculated based on the control group. (n>=3). (C) Ct values of seven primaryCYP450s genes normalized to same housekeeping gene. Values represent mean ± SEM(Student’s t test; *p < 0.05). TV: colon cells co-cultured with VSL#3 in Transwell inserts, MOI=1:10;TN: colon cells co-cultured with E.coli Nissle 1917 in Transwell inserts, MOI=1:10; TV: colon cellsco-cultured with B. adolescentis in Transwell inserts, MOI=1:10; TC: colon cells cultured inTranswell inserts, without bacteria.

    ΔCt

    0

    2

    4

    6

    8

    10

    12

    14

    16

    18

    20

    TV

    TN

    TB

    TC

    *

    ** * *

    -3

    -2

    -1

    0

    1

    2

    3

    4

    5

    TV

    TN

    TBLog2

    (Fo

    ld C

    han

    ge)

    AB

    C

  • Figure 3. (A) – (C) Biological pathway analysis of colon cells co-cultured with VSL#3,E.coli Nissle 1917 and B. adolescentis in Transwell inserts for upregulated (foldchange >3) and downregulated genes (fold change

  • Figure 4. Enzymatic activity of CYP3A4 andCYP1A2 in colon cells co-cultured with (A)VSL3; (C) E.coli Nissle 1917; (E) B.adolescentis; and (B, D, F) their respectivesupernatant conditioned media in tissueculture plate and Transwell inserts. (G)Enzymatic activity of CYP3A4 and CYP1A2activity of colon cells when cultured withspecific bacterial metabolites (butyrate,propionate and p-cresol). P.MOI: theMultiplicity of Infection bacteria and cellstissue culture plate, ranging from 1:1 to 100:1;T.MOI: the Multiplicity of Infection bacteria andcells Transwell inserts, ranging from 1:1 to100:1; P.CM: bacterial supernatantconditioned media in well plate with differentratios ranging from 1:1 to 1:100; P.CM:bacterial supernatant conditioned media inTranswell inserts in different ratios rangingfrom 1:1 to 1:100.

    0

    1

    2

    3

    4

    VSL#3 3A4

    1A2

    0

    1

    2

    3

    4

    VSL#3 supernatant 3A4

    1A2

    0

    1

    2

    3

    4

    E.coli Nissle 19173A4

    1A2

    0

    1

    2

    3

    4

    E.coli Nissle 1917 supernatant 3A4

    1A2

    0

    1

    2

    3

    4

    B. adolescentis3A4

    1A2

    0

    1

    2

    3

    4

    B. Adolescentis supernatant3A4

    1A2

    0

    1

    2

    3

    4

    caco2+2mMbutyrate

    caco2+3mMpropionate

    caco2+2mMp-cresol

    Metabolites

    3a4

    1a2

    Fold

    ch

    ange

    Fold

    ch

    ange

    Fold

    ch

    ange

    Fold

    ch

    ange

    Fold

    ch

    ange

    Fold

    ch

    ange

    Fold

    ch

    ange

    C

    BA

    D

    EF

    G

  • Table 1. Up- and down- regulated genes influenced by VSL#3, E.coli Nissle 1917, and B.adolescentis. Fold change >3 was considered as upregulated and < 0.3 as downregulated.

    TV TN TB

    Upregulated (fold change > 3) CYP1A2, CYP3A4, CYP4F2, AADAC

    CYP1A1, CYP1B1, CYP2A13, CYP4F2, ADH4,

    FMO1, FMO2

    CYP11A1, CYP11B2, CYP17A1, CYP19A1, CYP1A1, CYP1B1, CYP21A2, CYP26A1, CYP26C1, CYP2A13, CYP2C8, CYP2C9, CYP2F1, CYP3A4,

    CYP3A43, CYP4A11, CYP4A22, CYP4B1, CYP4F2, CYP4F8, CYP7A1, CYP7B1,

    CYP8B1, ADH1A, ADH4, ADH7, AADAC, GZMB,

    UCHL1, ALDH1A2, ALDH1A3, ALDH3A1, ALDH8A1, FMO1, FMO2, FMO3, FMO4, MAOB,

    PTGS2

    Downregulated (fold change < 1/3)CYP11B1, CYP2C18,

    CYP4F11, ADH1B, GZMA, MAOA, PTGS1

    CYP11B1, CYP2C19, CYP4F11, ADH1B

    CYP2B6, CYP2D6, CYP3A5, CYP4F11, CYP4F12, ADH1C,

    ADH6, HSD17B10, PTGS1

  • Figure S1. Bacteria standard curve of OD and density (CFU/mL)

    y = 3.8371x + 0.0413R² = 0.9549

    0

    0.05

    0.1

    0.15

    0.2

    0 0.01 0.02 0.03 0.04C

    FU/m

    l (*1

    06

    )OD

    E.coli Nissle 1917

    y = 126.7x - 0.1084R² = 0.9754

    0

    0.5

    1

    1.5

    2

    2.5

    3

    3.5

    0 0.01 0.02 0.03

    CFU

    /ml (

    *10

    6)

    OD

    VSL#3

    y = 1.7638x - 0.5133R² = 0.9842

    0

    0.5

    1

    1.5

    2

    2.5

    0 0.5 1 1.5 2

    CFU

    /ml (

    *10

    6 )

    B. adolescentis

    OD1 Linear (OD1)

  • Figure S2. Schematic diagram of co-culture between colon cells andbacteria in well plate and Transwell inserts.

    Cell culture media

    Colon cells

    Colon cells

    Colon cells

    Microbiome

    Cell culture media

    Colon cells

    Cell culture media

    Colon cells

    Conditioned media

    Cell culture media

    Colon cells

    Microbiome

    Cell culture media

    Conditioned media

  • Figure S3. qPCR analysis of the colon cellsco-cultured with VSL#3, E.coli Nissle 1917 andB. adolescentis bacteria in Transwell inserts.Heatmap generated from 84 genes.

  • -4.0

    -2.0

    0.0

    2.0

    4.0

    6.0

    8.0

    10.0C

    YP

    11

    A1

    CY

    P1

    1B

    1

    CY

    P1

    1B

    2

    CY

    P1

    7A

    1

    CY

    P1

    9A

    1

    CY

    P1

    A1

    CY

    P1

    A2

    CY

    P1

    B1

    CY

    P2

    1A

    2

    CY

    P2

    4A

    1

    CY

    P2

    6A

    1

    CY

    P2

    6B

    1

    CY

    P2

    6C

    1

    CY

    P2

    7A

    1

    CY

    P2

    7B

    1

    CY

    P2

    A1

    3

    CY

    P2

    B6

    CY

    P2

    C1

    8

    CY

    P2

    C1

    9

    CY

    P2

    C8

    CY

    P2

    C9

    CY

    P2

    D6

    CY

    P2

    E1

    CY

    P2

    F1

    CY

    P2

    R1

    CY

    P2

    S1

    CY

    P2

    W1

    CY

    P3

    A4

    CY

    P3

    A4

    3

    CY

    P3

    A5

    CY

    P3

    A7

    CY

    P4

    A1

    1

    CY

    P4

    A2

    2

    CY

    P4

    B1

    CY

    P4

    F11

    CY

    P4

    F12

    CY

    P4

    F2

    CY

    P4

    F3

    CY

    P4

    F8

    CY

    P7

    A1

    CY

    P7

    B1

    CY

    P8

    B1

    CYP450

    TV

    TN

    TB

    -4

    -2

    0

    2

    4

    6

    8

    10

    AD

    H1

    A

    AD

    H1

    B

    AD

    H1

    C

    AD

    H4

    AD

    H5

    AD

    H6

    AD

    H7

    DH

    RS2

    HSD

    17

    B1

    0

    ADH

    TV

    TN

    TB

    -4

    -2

    0

    2

    4

    6

    8

    10

    Esterases

    TV

    TN

    TB

    -4

    -2

    0

    2

    4

    6

    8

    10

    ALD

    H1

    A1

    ALD

    H1

    A3

    ALD

    H2

    ALD

    H3

    A2

    ALD

    H3

    B2

    ALD

    H5

    A1

    ALD

    H7

    A1

    ALD

    H9

    A1

    ALDH

    TV

    TN

    TB

    -4

    -2

    0

    2

    4

    6

    8

    10

    FMO

    1

    FMO

    2

    FMO

    3

    FMO

    4

    FMO

    5

    FMO

    TV

    TN

    TB

    Figure S4. Log2 Fold Change plot of the all 84 genes in the kit. Fold change was calculated based on the control group and separated to 6 different enzymes group. CYP450: Cytochrome P450s; ADH:AlcoholDehydrogenases; ALDH: Aldehyde Dehydrogenases; FMO: FlavinContaining Monooxygenases.

  • 50.0

    50.0

    33.3

    33.3

    33.3

    33.3

    33.3

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    100 80 60 40 20 0 20 40 60 80 100

    Xenobiotics

    Synthesis of (16-20)-hydroxyeicosatetraenoic…

    Aflatoxin activation and detoxification

    Synthesis of epoxy (EET) and…

    Biosynthesis of protectins

    Biosynthesis of maresin-like SPMs

    Miscellaneous substrates

    Phase I - Functionalization of compounds

    PPARA activates gene expression

    Synthesis of Leukotrienes (LT) and Eoxins (EX)

    Methylation

    Aromatic amines can be N-hydroxylated or N-…

    Fatty acids

    Eicosanoids

    Glucocorticoid biosynthesis

    Ethanol oxidation

    Endogenous sterols

    Synthesis of Prostaglandins (PG) and…

    Interleukin-4 and Interleukin-13 signaling

    Metabolism of serotonin

    Biogenic amines are oxidatively deaminated to…

    Norepinephrine Neurotransmitter Release Cycle

    Enzymatic degradation of dopamine by COMT

    Enzymatic degradation of Dopamine by…

    Defective MAOA causes Brunner syndrome…

    Defective CYP11B1 causes Adrenal hyperplasia 4…

    COX reactions

    TV (%)

    Bio

    logi

    cal P

    ath

    way

    s

    UP DOWN

    Figure S5. Biological pathways analysis of colon cells co-cultured withVSL#3 bacteria in Transwell inserts based on genes fold change >3 and

  • 14.3

    14.3

    14.3

    14.3

    14.3

    14.3

    14.3

    14.3

    14.3

    14.3

    14.3

    28.6

    28.6

    28.6

    28.6

    42.9

    16.7

    33.3

    16.7

    16.7

    16.7

    16.7

    33.3

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    16.7

    100 80 60 40 20 0 20 40 60 80 100

    PPARA activates gene expression

    Ethanol oxidation

    Aflatoxin activation and detoxification

    RA biosynthesis pathway

    Endogenous sterols

    Synthesis of Leukotrienes (LT) and Eoxins (EX)

    Biosynthesis of protectins

    Defective CYP1B1 causes Glaucoma

    CYP2E1 reactions

    Miscellaneous substrates

    Eicosanoids

    Xenobiotics

    Synthesis of epoxy (EET) and…

    Fatty acids

    FMO oxidises nucleophiles

    Synthesis of (16-20)-hydroxyeicosatetraenoic…

    Glucocorticoid biosynthesis

    Synthesis of bile acids and bile salts via 7alpha-…

    Synthesis of bile acids and bile salts via 24-…

    Synthesis of bile acids and bile salts via 27-…

    Defective CYP11B1 causes Adrenal hyperplasia…

    Defective CYP27A1 causes Cerebrotendinous…

    TN (%)

    Bio

    logi

    cal P

    ath

    way

    s

    UP DOWN

    Figure S6. Biological pathways analysis of colon cells co-cultured with E.coli Nissle1917 bacteria in Transwell inserts based on genes fold change >3 and

  • 21.1 18.4 18.4 18.4

    15.8 15.8

    13.2 13.2

    10.5 10.5

    7.9 7.9 7.9 7.9 7.9 7.9 7.9

    5.3 5.3 5.3 5.3 5.3 5.3 5.3

    2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6 2.6

    11.1 33.3

    44.4

    22.2 11.1

    22.2

    22.2

    22.2 11.1

    11.1 11.1

    11.1 11.1 11.1 11.1 11.1

    100 80 60 40 20 0 20 40 60 80 100

    Endogenous sterols

    Xenobiotics

    Synthesis of (16-20)-hydroxyeicosatetraenoic acids (HETE)

    Synthesis of Leukotrienes (LT) and Eoxins (EX)

    Synthesis of epoxy (EET) and dihydroxyeicosatrienoic acids…

    Glucocorticoid biosynthesis

    Synthesis of bile acids and bile salts via 27-hydroxycholesterol

    Ethanol oxidation

    FMO oxidises nucleophiles

    Phase I - Functionalization of compounds

    Synthesis of Prostaglandins (PG) and Thromboxanes (TX)

    Vitamins

    Androgen biosynthesis

    Abacavir metabolism

    Pregnenolone biosynthesis

    Estrogen biosynthesis

    Biogenic amines are oxidatively deaminated to aldehydes by…

    Defective CYP11B2 causes Corticosterone methyloxidase 1…

    Defective CYP17A1 causes Adrenal hyperplasia 5 (AH5)

    Biosynthesis of protectins

    Defective CYP21A2 causes Adrenal hyperplasia 3 (AH3)

    Defective CYP7B1 causes Spastic paraplegia 5A, autosomal…

    NOTCH2 intracellular domain regulates transcription

    Synthesis of 15-eicosatetraenoic acid derivatives

    Biosynthesis of electrophilic ω-3 PUFA oxo-derivatives

    Biosynthesis of DPAn-3 SPMs

    tRNA modification in the mitochondrion

    rRNA processing in the mitochondrion

    TB (%)

    Bio

    logi

    cal P

    ath

    way

    s

    UP DOWN

    Supplementary Figure S5. Biological pathways analysis of colon cells co-cultured with B. adolescentis bacteria in Transwell inserts based on genes fold change >3 and