You are on page 1of 15

Articles

https://doi.org/10.1038/s41551-019-0397-0

Corrected: Author Correction

A complex human gut microbiome cultured in an


anaerobic intestine-on-a-chip
Sasan Jalili-Firoozinezhad   1,2,11, Francesca S. Gazzaniga1,3,11, Elizabeth L. Calamari1,11,
Diogo M. Camacho1, Cicely W. Fadel1, Amir Bein   1, Ben Swenor   1, Bret Nestor1, Michael J. Cronce1,
Alessio Tovaglieri1,4, Oren Levy1, Katherine E. Gregory5,6, David T. Breault6,7,8, Joaquim M. S. Cabral2,
Dennis L. Kasper   3, Richard Novak1 and Donald E. Ingber   1,9,10*

The diverse bacterial populations that comprise the commensal microbiome of the human intestine play a central role in health
and disease. A method that sustains complex microbial communities in direct contact with living human intestinal cells and
their overlying mucus layer in vitro would thus enable the investigation of host–microbiome interactions. Here, we show the
extended coculture of living human intestinal epithelium with stable communities of aerobic and anaerobic human gut micro-
biota, using a microfluidic intestine-on-a-chip that permits the control and real-time assessment of physiologically relevant
oxygen gradients. When compared to aerobic coculture conditions, the establishment of a transluminal hypoxia gradient in the
chip increased intestinal barrier function and sustained a physiologically relevant level of microbial diversity, consisting of over
200 unique operational taxonomic units from 11 different genera and an abundance of obligate anaerobic bacteria, with ratios
of Firmicutes and Bacteroidetes similar to those observed in human faeces. The intestine-on-a-chip may serve as a discovery
tool for the development of microbiome-related therapeutics, probiotics and nutraceuticals.

A
major recent paradigm shift in medicine is the recognition of complex populations of human aerobic and anaerobic microbiota
the central role that the microbiome, composed of host-spe- in contact with living human tissues to analyse dynamic and physi-
cific communities of commensal microorganisms, plays in ologically relevant human host−microbiome interactions.
human health and disease1. While human microbiota colonize the Existing in  vitro models, such as Transwell inserts, have been
mucosal surfaces of various tissues, the gastrointestinal tract sup- used to study human host−microorganism interactions; however,
ports the greatest mass and diversity of microorganisms2. Aerobic these studies can be carried out only over a period of hours before
and anaerobic commensal gut microorganisms are essential for the bacterial overgrowth leads to cell injury and death11–13. Organoid
maintenance of normal nutrient absorption, drug metabolism and cultures showed great promise for studying host−microbiome
immune responses, as well as protection against infectious patho- interactions, but they also cannot be cocultured with living micro-
gens3. Conversely, changes or imbalances in the microbial commu- organisms for more than ~1 d; furthermore, they do not provide a
nity in the intestine can contribute to the development of a broad vascular interface nor can they sustain luminal oxygen levels below
range of pathological disorders within and beyond the gastrointes- 0.5%, which is required for the coculture of certain obligate anaer-
tinal system, including inflammatory bowel disease, colorectal can- obes14,15. Specialized bioreactor models, such as the mucosal-sim-
cer, radiation enteropathy, diabetes, hepatic steatosis, obesity and ulator of the human intestinal microbial ecosystem (M-SHIME),
rheumatoid arthritis4,5. Thus, the establishment and preservation of have been developed to sustain the growth of luminal and mucosal
balanced host–intestinal microbiome interactions are key require- gut microorganisms in vitro, but they do not include living human
ments for maintaining gut homeostasis and human health. intestinal epithelium16. A human−microbiota interaction module
The analysis of gut−microbiome crosstalk has almost exclusively was developed that permits the analysis of aerobic and anaerobic
relied on the genomic or metagenomic analysis of samples col- microorganisms, including a complex living microbiome derived
lected in vivo because no method exists to establish stable complex from a SHIME reactor, when cocultured with human Caco2 intesti-
communities of gut commensal microorganisms in direct contact nal epithelial cells under an oxygen gradient; however, the microor-
with intestinal epithelium and its overlying mucus layer in vitro6,7. ganisms need to be separated from the human cells by a nanoporous
Although animal models have been used to analyse host−micro- membrane with an artificial mucus layer and, even under these con-
biome interactions and their contributions to pathophysiology8–10, ditions, the cocultures were maintained for only 48 h17–19. We pre-
there are no in  vitro systems available to verify these interactions viously described a two-channel microfluidic Organ Chip device
in human cells cultured with a complex human microbiome. Thus, lined with human Caco2 intestinal epithelial cells cultured under
there is a great need for experimental models that can sustain dynamic fluid flow and peristalsis-like mechanical deformations,

1
Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA. 2Department of Bioengineering and Institute for Bioengineering
and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal. 3Department of Immunology, Harvard Medical School, Boston,
MA, USA. 4Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland. 5Department of Pediatric Newborn Medicine, Brigham and
Women’s Hospital, Boston, MA, USA. 6Department of Pediatrics, Harvard Medical School, Boston, MA, USA. 7Department of Endocrinology, Boston
Children’s Hospital, Boston, MA, USA. 8Harvard Stem Cell Institute, Cambridge, MA, USA. 9Vascular Biology Program and Department of Surgery,
Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA. 10Harvard John A. Paulson School of Engineering and Applied Sciences,
Cambridge, MA, USA. 11These authors contributed equally: Sasan Jalili-Firoozinezhad, Francesca S. Gazzaniga, Elizabeth L. Calamari.
*e-mail: don.ingber@wyss.harvard.edu

520 Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng


Nature Biomedical Engineering Articles
which enabled the establishment of stable cocultures of a mucus- permeable PDMS membrane from the well-oxygenated medium
producing human villus intestinal epithelium with up to eight dif- flowing through the lower endothelium-lined vascular channel
ferent strains of human commensal gut microorganisms for weeks from the external oxygenated medium reservoirs (Supplementary
in  vitro under aerobic conditions20–22. However, the human gut Fig.  2a,b). Using this method, physiologically relevant anaero-
microbiome contains hundreds of different types of bacteria, many bic conditions (<0.5%) can be generated in less than 30 min
of which are obligate anaerobes that will not grow in this environ- at 243 ml min−1 of nitrogen flow into the anaerobic chamber
ment. Thus, no existing in vitro model enables the analysis of direct (Fig. 1d). The chamber can also sustain low oxygen levels (<5.0%)
interactions among complex communities of anaerobic and aerobic for about 15 min after it is disconnected from the nitrogen source
gut bacteria, human intestinal epithelium and its overlying mucus (Fig. 1d). This allows the chamber to be temporarily moved from
layer when cultured for multiple days in vitro, which is crucial for the incubator for imaging or into a bacterial glove box (to replen-
analysing gut health and disease. ish the culture medium or add microbiota, for example) without
In this study, we therefore set out to develop an experimen- disturbing the low-oxygen environment.
tal intestine-on-a-chip (Intestine Chip) system that can support When human Caco2 intestinal epithelial cells are cultured for
dynamic interactions between living, mucus-producing human 5−7 d under aerobic conditions and dynamic flow, they undergo vil-
intestinal epithelium and a directly apposed complex community lus differentiation and express multiple features of the ileal portion
of living human aerobic and anaerobic commensal gut microorgan- of the human small intestine, including the secretion of a mucus
isms with a population diversity similar to that observed in living layer overlying the apical surface of the epithelium and the estab-
human intestine. To meet this challenge, we first modified human lishment of barrier function20,25. Endothelial cells can also be cocul-
Caco2 Intestine Chips by integrating microscale oxygen sensors into tured on the bottom of the central porous membrane in the lower
the devices for in situ oxygen measurements and placing the chips channel of the same device, where they form a hollow vascular
in an engineered anaerobic chamber to establish a physiologically lumen lined with cells joined by VE-cadherin-containing cell−cell
relevant oxygen gradient across a human intestinal epithelium, as junctions under aerobic conditions25. The coculture of endothelium
well as a microvascular endothelium, which are cultured in parallel has been shown to enhance barrier function and mucus produc-
channels separated by a porous matrix-coated membrane within the tion (for example, expression of MUC2 and MUC5AC), as well as
device. To ensure a stable source of complex human intestinal gut influence villus development and cytokine production by intestinal
microbiota, we used complex microbiota originally derived from Caco2 epithelium under these conditions25,26.
healthy human stool specimens, which were maintained stably in When we cultured Intestine Chips lined with Caco2 intestinal
gnotobiotic mice for multiple years and closely resemble the relative epithelial cells and human intestinal microvascular endothelial cells
abundance patterns of the major bacterial phyla in their respective (HIMECs) under a hypoxia gradient using our chamber, differential
inocula23,24. We also used the same method to coculture a fresh gut interference contrast (DIC) and immunofluorescence microscopic
microbiome isolated from human infant stool samples in a primary analysis confirmed that the cells again formed a villus intestinal epi-
human Intestine Chip lined with cells isolated from normal human thelium containing polarized cells joined by ZO-1-containing tight
ileum. Here, we describe how establishing a hypoxia gradient across junctions (Fig. 1e, top; Supplementary Fig. 3a−d) that was underlaid
the engineered tissue−tissue (endothelium−epithelium) interface by a confluent HIMEC monolayer with cells linked by VE-cadherin-
of the Intestine Chip allows for the stable coculture of highly com- containing tight junctions, even in the presence of this hypoxia gra-
plex communities of anaerobic and aerobic human commensal gut dient (Fig. 1e, bottom). Both cell types also remained viable under
bacteria in the same channel as mucus-producing human villus these conditions, as measured by quantifying the release of the
intestinal epithelium while simultaneously monitoring oxygen lev- intracellular enzyme lactate dehydrogenase (LDH), which remained
els and intestinal barrier function for at least 5 d in vitro. relatively unchanged compared to the aerobic control during 7 d of
anaerobic culture (Supplementary Fig.  4a). The quantification of
Results the apparent permeability (Papp) of the intestinal epithelial barrier
Establishing an oxygen gradient across the lumen of the Intestine similarly revealed no changes in the paracellular barrier function
Chip. To recapitulate a physiologically relevant intestinal oxy- and these human Intestine Chips displayed Papp values of about
gen gradient profile in Intestine Chips (Fig.  1a), we fabricated 1 × 10−7 cm s−1 after 7 d (Supplementary Fig.  4b), which are simi-
an oxygen-sensing, dual channel human Organ Chip composed lar to those previously reported25. Importantly, we also confirmed
of optically clear and flexible poly(dimethyl siloxane) (PDMS) that both the human intestinal epithelium and endothelium expe-
polymer (Fig. 1b; Supplementary Fig. 1a), as well as an anaerobic rienced oxygen gradients by analysing the expression of hypoxia-
chamber (Supplementary Fig. 2). Six sensor spots containing oxy- inducible factor-1α (HIF-1α). HIF-1α is a key mediator of oxygen
gen-quenched fluorescent particles were embedded in the top and homeostasis and intestinal epithelial cell adaptation to oxygen
bottom portions of the chip beneath the central microchannels for deprivation27, which is stabilized in a graded fashion in response
the real-time, non-invasive monitoring of oxygen tension (Fig. 1b; to decreasing oxygen concentrations28. Nuclear HIF-1α expres-
Supplementary Fig.  1b). Changes in the fluorescent intensities of sion levels were significantly higher (~threefold; P < 0.01) in the
these sensors in response to oxygen tension were captured by a lumen of the anaerobically cultured epithelium than in the adjacent
VisiSens camera (Supplementary Fig. 1b) and translated into oxy- oxygenated endothelium-lined channel (Supplementary Fig. 5a,b),
gen concentrations by comparison with a standard Oxy-4 probe which is where our sensors indicated the maintenance of a hypoxic
system (Supplementary Fig. 1c). As both the chips and sensors are environment for up to 7 d in culture (Fig.  1f and Supplementary
composed of highly gas-permeable PDMS, the sensors respond rap- Fig.  4c). Similar nuclear HIF-1α expression has been previously
idly (<30 s) to changes in oxygen concentrations (Fig. 1c). shown in both mouse and human cells cultured under hypoxic
To simultaneously provide adequate oxygen to maintain human (1% O2) conditions29.
cells and an anaerobic microenvironment suitable for culturing
complex human microbiota, while establishing a functional host− Coculture of human intestinal epithelium with an obligate anaer-
microbiome interface, we flushed the custom anaerobic chamber obe on-chip. We next explored whether the hypoxic environment can
(Supplementary Fig.  2a,b) continually with humidified 5% CO2 support the coculture of the intestinal epithelium with the obligate
in nitrogen gas. This set-up enabled us to maintain low oxygen anaerobe Bacteroides fragilis (strain NCTC 9343), which is a human
levels within the lumen of the upper chamber (Fig. 1d) while the commensal symbiotic bacterium that cannot grow under aerobic
epithelium was sustained via the diffusion of oxygen through the (>0.5% oxygen) conditions30,31. During the coculture procedure

Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng 521


Articles Nature Biomedical Engineering
a Mucus Microbiome Epithelium b

PDMS
T T B
Sensor
B B T Epithelium
Endothelium
Sensor

150
µm
O2
Endothelium
PDMS

c d
25 0% 1% 12.5%
20
20

stopped
N2 flow
15
15
O2 (%)

O2 (%)
10 10

5 5

0 0
0 75 150 225 300 375 450 0 60 120 180 240
Time (s) Time (min)

e Epithelium Endothelium f

Inlet Middle Outlet Aerobic


25

20 1

15
O2 (%)

Sensor
Sensor 2 2

*
1

*
4
0
0 1 2 3 4 5 6 7
Time (d)

Fig. 1 | Oxygen-sensitive human Intestine Chip microfluidic culture device. a, A schematic of the two-channel microfluidic Organ Chip device with an
oxygen gradient (colour scale). The human intestinal epithelium, which is overlaid with its own mucus layer and complex gut biota, is positioned over an
extracellular matrix (ECM)-coated porous membrane (grey). The vascular endothelium lies below the porous membrane. b, A schematic representation
of the Intestine Chip with six oxygen-quenched fluorescent particles embedded in the inlets, middles and outlets of the top and bottom channels (T and B,
respectively). c, A sensitivity analysis of oxygen spots located in the Intestine Chip in response to defined, standard oxygen concentrations. d, Anaerobic
chamber validation at various N2 inflow pressures; N2 was introduced into the chamber at 81 ml min−1, 162 ml min−1 or 243 ml min−1 for 1 h before gas flow
was stopped and the chamber was allowed to recover (n = 3; line and shaded region show mean ± s.d., respectively). e, Microscopy images showing the
villus morphology of human Caco2 intestinal epithelium (top left; scale bar, 100 μm) and vascular endothelium (top right; scale bar, 100 μm) cultured for
6 d in the Intestine Chip under anaerobic conditions, when viewed from above by DIC and phase contrast imaging, respectively, or by immunofluorescence
staining for the tight junction protein, ZO-1 (red, bottom left; scale bar, 100 μm), and endothelial cell junction-associated protein, VE-cadherin (red, bottom
right; scale bar, 20 μm). Grey indicates 4,6-diamidino-2-phenylindole (DAPI)-stained nuclei; white dashed lines indicate the border of the oxygen sensor
spot. f, Oxygen concentration profiles in aerobically and anaerobically cultured Intestine Chips. Representative pseudocolour insets indicate the average
oxygen concentration in the aerobic chip (1) and the inlet (2), middle (3) and outlet (4) of the anaerobically cultured epithelium channel, at day 7 of
culture. Scale bar, 200 μm; n = 3 individual chips; data are presented as mean ± s.d.; significance was calculated by one-way analysis of variance
(ANOVA); *P = 0.046.

that began after the epithelium had been cultured and differenti- maintained a low-oxygen environment that decreased from ~1%
ated on-chip for 7 d under anaerobic conditions (Supplementary oxygen to 0.3% in the presence of B. fragilis (Fig. 2a). Yet, the intes-
Fig. 6a), 2.5 × 105 colony forming units (CFU) of B. fragilis bacte- tinal epithelium maintained its ZO-1-containing tight junctions
ria that were fluorescently labelled with HCC-amino-d-alanine and apical brush border polarity when cocultured in direct contact
(HADA)32(Supplementary Fig. 6b) were introduced into the lumen with B. fragilis under these highly anaerobic conditions (Fig.  2b).
of the upper channel, where they became distributed across the sur- Interestingly, the presence of this obligate anaerobe enhanced,
face of the intestinal epithelium (Supplementary Fig. 6c). They were rather than decreased, barrier function (reduced Papp by 1.8-fold
subsequently cultured under either aerobic or anaerobic conditions, compared to aerobic conditions; *P = 0.033) after 3 d in anaerobic
while being flushed daily to remove both luminal and tissue-associ- culture (Fig. 2c) and this barrier was maintained for up to at least 8 d
ated microorganisms, and CFU counts were carried out by plating. in culture (Supplementary Fig. 6d). As expected, the B. fragilis bac-
The continuous monitoring of oxygen concentrations from inoc- teria continued to grow in the anaerobic chips over 3 d (P < 0.001),
ulation to day 3 of coculture revealed that our anaerobic chip set-up whereas they started to die off by day 2 and appeared at significantly

522 Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng


Nature Biomedical Engineering Articles
a b A mucus layer separates the commensal microorganisms from
Aerobic Anaerobic Villin B. fragilis Zo-1
the epithelium. One of the characteristic features of host−micro-
18 biome interactions in the living intestine is that they are mediated
12 through an intervening mucus layer that is secreted by the epithe-
O2 (%)

Merge
lium along its apical surface. Live staining using wheat germ aggluti-
1.2 nin (WGA), which has been previously used for mucus visualization
0.6 in vitro33 and in vivo34, confirmed that B. fragilis resides on top of
0.0
the mucus layer (Fig. 2e), which is secreted by the intestinal Caco2
0 1 2 3 epithelium, as previously demonstrated33,35. This was independently
Time (d) confirmed by scanning electron microscopy (SEM), which clearly
revealed a continuous and dense mucus blanket that completely
c d
Aerobic Aerobic covered the surface of the differentiated villus epithelium, separat-
10 Anaerobic * Anaerobic ing it from overlying bacteria after 12 d of culture (Fig. 2f), much
* ***
Papp (10–7 cm sec−1)

8
106 * as is observed in vivo36,37. This was in contrast to SEM analysis of
6
Caco2 Intestine Chips, which were cultured for only 4 d before
CFU ml−1

104 full differentiation occurred and mucus accumulated, where the


4
102 microvilli-lined surface of the apical epithelium remained clearly
2 detectable (Fig. 2f). Using these images, the thickness of the mucus
100 layer was estimated at ~10 μm, which is similar to that reported with
0
0 1 2 3 1 2 3 30-d-old mouse ileum36.
Time (d) Time (d)
Sustaining a complex human intestinal microbiome in  vitro.
e f
B. fragilis To optimize the growth of a complex microbiome in our culture
WGA system, we searched for a source of complex human gut microor-
DAPI
ganisms that remains stable over time. We therefore inoculated the
anaerobic Intestine Chips with a sample of complex gut microbiota
that was originally isolated from human faeces; the gut microbiota
had been stably maintained in gnotobiotic mice in isolators for
over 30 generations23,24 and thus maintained a composition closely
resembling the original human stool inoculum at the genera and
species levels23. To identify a medium composition that would pro-
mote the growth of a complex set of commensal bacteria, we first
Fig. 2 | Coculture of human intestinal epithelium and B. fragilis on-chip. inoculated 13 different types of culture medium with the healthy
a, Oxygen concentration profiles in aerobic and anaerobic Intestine Chips human microbiome (Hmb) stock in standard culture tubes, placed
cocultured with B. fragilis (n = 4 individual chips; data are presented the cultures in an anaerobic chamber at 37 °C and then carried out
as mean ± s.d.). b, Representative vertical cross-sectional confocal 16S ribosomal RNA sequencing after 3 d of culture (Supplementary
micrographic views through the intestinal epithelium−microbiome interface Fig. 7a). Samples of these 13 types of medium were also added to
within the Intestine Chip cultured under anaerobic conditions, when cultured human Caco2 intestinal epithelial cells to test for toxic-
immunostained for villin (cyan) and ZO-1 (magenta). Scale bar, 50 μm; ity (Supplementary Fig. 7b). The medium that promoted the most
B. fragilis was HADA-labelled (yellow); representative images from four diverse set of viable microorganisms without injuring the epithe-
intestine chips are shown. c, Changes in Papp measured by quantitating lium contained DMEM medium, 20% fetal bovine serum (FBS), 1%
Cascade blue transport across the tissue−tissue interface within the glutamine, 1 mg ml−1 pectin, 1 mg ml−1 mucin, 5 μg ml−1 haemin and
Intestine Chip microdevices cocultured with B. fragilis under aerobic 0.5 μg ml−1 vitamin K1. The microbiota stock was introduced into
and anaerobic conditions (n = 4 individual chips; data are presented as this medium (0.1 mg ml−1) and perfused through the upper Caco2
mean ± s.d.; significance was calculated by one-way ANOVA; *P = 0.042 epithelium-lined channel of the Intestine Chip while oxygenated
and 0.033 for anaerobic day 2 versus day 0 and aerobic versus anaerobic endothelial culture medium flowed through the lower channel.
day 3, respectively). d, CFU counts per millilitre of B. fragilis cocultured in The epithelial channels of the chips were flushed daily with a short
the Intestine Chip under aerobic and anaerobic conditions (n = 3 individual (2 min) fluid pulse at a higher flow rate (50 μl min−1) to remove
chips; data are presented as mean ± s.d.; significance was calculated by one- adherent and luminal bacteria and 16S rRNA sequencing was car-
way ANOVA; *P = 0.031 for day 2; ***P < 0.001 for day 3). e, Cross-sectional ried out using samples from the effluent to assess bacterial diversity
fluorescence microscopic view of the Caco2 epithelium (nuclei stained with in each condition over 3 d of culture (n = 4 for each condition).
DAPI), the mucus layer stained with Alexa Fluor 488-conjugated WGA and After data processing, we identified a total of 938 operational
B. fragilis bacteria (GalCCP-labelled) when cocultured in the Intestine Chip. taxonomic units (OTUs) among all the samples, corresponding to
Scale bar, 10 μm. f, SEM views of the apical surface of the Caco2 epithelium approximately 200 unique OTUs shared between the samples from
in the Intestine Chip comparing the morphology on day 4 of culture, before it each chip after filtering and removing singletons. This is similar in
accumulates a mucus layer and when the surface microvilli are visible (left), scale to the number of OTUs previously observed in human intes-
versus when B. fragilis have been added on day 12, after the mucus layer has tinal aspirates (280 OTUs)38, although, as expected, it has a differ-
accumulated, which can be seen as a dense mat that separates the bacteria ent phylum distribution. Analysis of the alpha diversity between the
from the epithelial cell surface (right). Scale bar, 2 μm. two conditions showed that the species diversity in anaerobic chips
was statistically different (permutational multivariate ANOVA,
lower levels at day 3 under aerobic culture conditions (Fig.  2d). ***P < 0.001) from that in aerobic chips (Fig.  3a). Although the
These data confirm that our chips, which experience a hypoxia observed diversity and Shannon index are lower than those
gradient, support the growth of an obligate anaerobic bacterial spe- observed in human stool samples (Supplementary Fig.  8a,b), we
cies in the same channel as living human intestinal epithelial cells, observed an increase in richness compared to our starting inoculum
whereas these bacteria would have otherwise died in a conventional over the course of the 3 d experiment (Supplementary Fig. 9a,b). We
aerobic culture system. identified 11 well-characterized genera, including Eubacterium,

Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng 523


Articles Nature Biomedical Engineering

Oscillospira, Blautia, Sutterella, Bilophila, Akkermansia, recently implicated as an enhancer of gut barrier function43–45, had
Ruminococcus, Bacteroides, Parabacteroides, Enterococcus and a considerably higher total count number in the anaerobic culture
Citrobacter (Fig.  3b), and an additional 8 OTUs of unknown system compared to human stools (Supplementary Figs. 12 and 13).
genera from the phyla Firmicutes (5 OTUs) and Proteobacteria The genus Enterococcus was also found to be present at higher levels
(3 OTUs) were present in our chips (phylum level analysis is shown in both chip culture systems compared to the stool samples, sug-
in Supplementary Fig. 10). Interestingly, in comparison to aerobic gesting that some gut microbial species may grow better under con-
conditions, co-culturing diverse microbiota under anaerobic con- ditions that more closely mimic regions of the living intestine than
ditions for 3 d in direct contact with the human intestinal epithe- in stools. Taken together, these data confirm that this anaerobic
lium did not compromise intestinal barrier integrity; instead, it led human Intestine Chip system enables living human intestinal epi-
to an increase in barrier function of almost twofold (a decrease in thelium to be cocultured in the same channel as a complex human
Papp from 3.1 × 10−7 to 1.6 × 10−7 cm s−1 in aerobic versus anaerobic gut microbiome that contains a range of bacterial genera much
chips, respectively; *P = 0.048;Fig.  3c). In contrast, epithelial bar- closer to that observed in healthy human donors.
rier function decreased (***P < 0.001) after day 3 of coculture under To determine the stability of the microbial communities in the
aerobic conditions when cocultured with the same complex gut anaerobic Intestine Chip system, we analysed their change in abun-
microbiome (Fig.  3c). To explore the durability of these cultures, dance over 3 d of coculture with human Caco2 intestinal epithelium
we then carried out an additional experiment with an extended and underlying endothelium. Our results show that genera com-
duration of 5 d on aerobic and anaerobic chips with and without posed of obligate anaerobes, such as Akkermansia, Blautia, Bilophila
the same Hmb stock. Sterile chips maintained their barrier func- and Sutterella, increased in abundance over time, presumably due
tion over the 5 d culture under both anaerobic and aerobic condi- to the maintenance of low oxygen concentrations (Fig.  4a, top).
tions (Supplementary Fig. 11a,b). Again, we observed that although Bacteroides, the most abundant genus in the anaerobic Intestine
the barrier function decreased in aerobic chips with Hmb stock Chips, remained relatively stable over time. Furthermore, in a sub-
at day 3 (Supplementary Fig. 11a), no loss of barrier function was sequent experiment, we cultured anaerobic Intestine Chips with
seen in anaerobic chips with Hmb stock, even over the 5 d culture the same Hmb microbiome stock for 5 d and plated serial dilutions
(Supplementary Fig. 11b), and the duration of these cultures could of the flush on anaerobic Brucella culture plates in the presence or
be extended if experimental needs required. Moreover, in the pres- absence of vancomycin. Our results show that the bacterial load
ence of complex microbiota, the intestinal epithelium remained increased at day 1, compared to the seeding inoculum, and then
viable, as measured by quantifying the release of the intracellular remained constant throughout the experiment (Supplementary
enzyme LDH, which remained unchanged compared to the sterile Fig.  11e). Vancomycin kills Gram-positive bacteria and thus the
control chip cultures (Supplementary Fig. 11c). difference in counts between Brucella plates in the presence ver-
To further assess the physiological mimicry obtained using the sus absence of vancomycin suggests that both Gram-positive and
anaerobic Intestine Chip lined with Caco2 epithelium, we com- Gram-negative bacteria (that survived in the vancomycin plates)
pared the genera identified in this study with publicly available data remained viable over 5 d of coculture with the intestinal epithelium
from studies of human stools generated by the Human Microbiome on-chip. Thus, although serial dilutions of a mixed population of
Project39 (Fig.  3d). We did not expect the composition of the bacteria do not provide accurate total counts, these data show that
microbiome grown on-chip to precisely recapitulate that of stools both Gram-positive and Gram-negative bacteria remained viable
because the microbiome of the human intestine is known to show and proliferated (increased in number) over time in the human
regional differences40,41. For example, previous reports have shown Intestine Chips.
different ratios of the Firmicutes, Bacteroidetes and Actinobacteria Importantly, when we compared the growth of microbiota cul-
phyla in stool samples compared to small intestine aspirates40,42. tured for 3 d in the anaerobic Intestine Chip with that produced by
Nevertheless, our results show that the anaerobic culture system culturing the same microbiome samples in a conventional liquid
can sustain a diverse bacterial community that falls within the medium culture in an anaerobic chamber, we observed significantly
range of abundances reported in the Human Microbiome Project. different growth responses for multiple genera (Fig.  4a, bottom).
Furthermore, the relative abundances of the phyla that dominate Notably, the genus Akkermansia shows preferential growth in the
the human gut, Bacteroidetes (Bacteroides and Parabacteroides anaerobic Intestine Chip, presumably due to the presence of high
genera) and Firmicutes (Blautia, Enterococcus, Ruminococcus and levels of mucus produced by the Caco2 intestinal epithelium on-
Oscillospira genera), were higher in the anaerobic chips than in the chip45 (Fig.  2e,f), which complements the mucins already present
aerobic chips with some genera (Blautia and Oscillospira) entirely in the medium. Bacteria in the Blautia, Bilophila, Oscillospira and
absent in the aerobic chips (Fig.  3d). Oxygen sensor readouts in Sutterella genera also showed enhanced growth in the anaerobic
aerobic and anaerobic chips cultured with a viable microbiome or chips containing living human intestinal epithelium compared to
under sterile (microorganism-free) conditions confirmed that the anaerobic liquid culture, whereas the Gram-negative obligate aer-
oxygen concentration was maintained below 1% throughout the obe Citrobacter was less abundant on-chip. Thus, the presence of
5 d coculture period in the anaerobic cocultures (Supplementary human intestinal epithelium that secretes a natural mucus layer
Fig. 11d). Moreover, these results showed a decrease in oxygen con- above its apical surface20 (Fig. 2e,f) is crucial for culturing and sus-
centration in aerobic chips cultured with the microbiome over time taining the complex features of the human microbiome in vitro.
(Supplementary Fig. 11d), which is similar to what we observed in To complement these analyses, we calculated the differential
cocultures with B. fragilis. This was probably due to the increased abundance of the genera over time in the anaerobic versus aero-
vertical growth of villi that we observed in these chips relative to bic Intestine Chips. Our results show that the obligate anaerobes
anaerobic chips, as well as concomitant oxygen use by the bacte- Eubacterium, Oscillospira, Blautia and Sutterella were significantly
ria, which became more numerous by day 1 in both aerobic and more abundant in anaerobic chips compared to aerobic chips over
anaerobic chips. Although the oxygen concentration in the aero- our time course (false discovery rate: q < 0.05), whereas Citrobacter
bic chip never reached the low levels obtained in anaerobic chips, consistently showed a lower abundance in the anaerobic chips
this decrease in oxygen could explain the presence of some obligate (Fig. 4b). Quantification of the abundance of the various genera in
anaerobes, such as Akkermansia, that we observed in the aerobic either the anaerobic liquid culture (Supplementary Fig. 12) or the
chips; however, the constant anaerobic conditions are more optimal Hmb microbiome stock (Supplementary Fig. 13) confirmed that the
for the maintenance of cocultures of anaerobic bacteria with viable total numbers of obligate anaerobes, including Sutterella, Blautia,
human cells. Interestingly, the genus Akkermansia, which has been Oscillospira, Bilophila and Akkermansia, were significantly higher

524 Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng


Nature Biomedical Engineering Articles
a Day 1 Day 2 Day 3 b
300 300 300 1.00
Genes
Unknown

Observed richness

Relative abundance
250 250 250 Sutterella
0.75 Bilophila
Akkermansia
Blautia
200 200 200 Oscillospira
0.50 Ruminococcus
Enterococcus
150 150 150 (Eubacterium)
0.25 Parabacteroides
Bacteroides
100 100 100 Citrobacter

0
c

c
An bic

An bic

An bic
bi

bi

bi
ro

ro

ro
ro

ro

ro
ae

ae

ae
Ae

Ae

Ae

3
1

b
m
c

c
c

bi

bi

bi
bi

bi

bi

H
ro

ro

ro
ro

ro

ro

ae

ae

ae
Ae

Ae

Ae

An

An

An
c d Aerobic Anaerobic Human stool

Aerobic
60 Anaerobic
*** 4
Papp (10–7 cm s–1)

45

log10[Total counts]
***
30 3
NS
10
2
5 *
0 1
0 1 2 3
Time (d) 0

s
us
a

ira

lla
ila

tia

de

cu
te
de
si

re
cc
ph

sp
au
an

ac

oc
oi
oi

tte
co
lo

illo
Bl

er
ob
rm

er

oc
Bi

Su
ro

ct
ct

sc
itr
ke

in
te

ba
Ba

um
Ak

En

ra

R
Pa
Fig. 3 | Analysis of the diversity and relative abundance of microbiota cocultured in Intestine Chips under aerobic and anaerobic conditions. a, Observed
alpha diversity (richness) in our complex gut microbiome samples when cultured for 1−3 d in direct contact with human Caco2 intestinal epithelium (each
data point represents one Intestine Chip; error bars represent the s.d.; data are presented as box plots with individual data points overlaid, where the lower
or upper edges of the box represent the 25th or 75th percentiles and the middle bar is the median). b, The relative abundance of genera measured across
all samples highlighting changes in the abundance of the different genera observed over time. Data represent each of the 3 replicates cultured under
aerobic or anaerobic conditions at 0, 1, 2 or 3 d of culture (left to right, respectively); Hmb indicates abundance in the complex microbiome stock at time
0 d. Bacterial phylogenies were obtained from GreenGenes 1.7. c, Changes in Papp measured by quantifying Cascade blue transport across the tissue−tissue
interface within the Intestine Chip after coculture with a complex gut microbiome under aerobic and anaerobic conditions (n = 4 individual chips; data
are presented as mean ± s.d.; significance was calculated by one-way ANOVA; *P = 0.048; ***P < 0.001; NS, not significant). d, Differences in microbial
abundance between Intestine Chip samples and a human microbiome stool sample from the Human Microbiome Project. Data are shown as log10 of the
total number of reads; each data point corresponds to a single sample (error bars represent the s.d.; data are presented as box plots with individual data
points overlaid, where the lower or upper edges of the box represent the 25th or 75th percentiles and the middle bar is the median).

in the anaerobic chips. Taken together, these results confirm that and confocal fluorescence microscopic (Fig. 5b and Supplementary
the hypoxia gradient system combined with the presence of a living Fig.  14a) imaging of the primary human Ileum Chips confirmed
human intestinal epithelium provides a preferential environment the presence of a villus intestinal epithelium lined with a con-
for the sustained culture of anaerobic as well as aerobic gut bacteria tinuous polarized epithelium with F-actin- and villin-containing
from diverse genera. brush borders along its apical membrane, MUC2-producing cells
and basal nuclei. Importantly, when we measured the produc-
Culture of a fresh gut microbiome with primary intestinal epi- tion of secreted mucus using alcian blue staining (Supplementary
thelium on-chip. Finally, we explored whether this experimen- Fig. 14b), we observed blue-stained mucus over the apical surface
tal approach can be used to coculture a complex gut microbiome of the epithelium and we detected up to 600 µg ml−1 of mucin in the
obtained from fresh human stool specimens in direct contact with chip outflow (Supplementary Fig. 14c). As expected, the bacterial
primary human intestinal epithelium (instead of using the estab- richness was reduced in the infant stool stock (586 OTUs) com-
lished Caco2 intestinal cell line). To do this, we engineered human pared to adult human-derived stool (938 OTUs) at the same dilu-
Intestine Chips lined with intestinal epithelial cells isolated from tion per gram of materials, and these differences in richness were
organoids derived from normal regions of surgical biopsies of accurately recapitulated on-chip. We again found that the primary
human ileum that exhibit multilineage differentiation, villus for- human intestinal epithelium could be cocultured in direct contact
mation and mucus production when grown on-chip26. We then with this complex gut microbiome without compromising epithe-
inoculated the epithelial channels of four different chips with a lial barrier function and this coculture was stably maintained for
complex microbiome isolated from fresh human stool samples col- at least 5 d on-chip (Fig.  5c), much as we had observed with the
lected from four different infants (one with a corrected gestational Caco2 epithelium. Importantly, the microbiome cultured in these
age of 30 weeks and three with an age of 36 weeks). DIC (Fig. 5a) primary Intestine Chips also maintained a high bacterial richness,

Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng 525


Articles Nature Biomedical Engineering
a Discussion
4
Given the importance of the commensal gut microbiome for human
health and the lack of an in vitro model that can faithfully mimic
log10[Counts]

3 Day 1 the complex epithelial−microorganism interactions that occur


Day 2 across the host−microbiome interface, we leveraged human Organ
Day 3
2 Chip technology to develop a device that enables human intesti-
nal epithelium to be cocultured with the highly diverse community
1
of commensal microorganisms that forms the human gut micro-
biome under aerobic and anaerobic conditions. Our results show
that the anaerobic human Intestine Chip offers a robust modular
system for recapitulating the human intestinal−microbiome inter-
4 face in vitro. Using this device, it is now possible to stably coculture
a complex living microbiome in direct contact with living mam-
log10[Counts]

3 Aerobic chip
Anaerobic chip
malian cells and their naturally produced overlying mucus layer
Anaerobic liquid culture for 5 d or more in vitro. This is substantially longer than past stud-
2
ies using the human−microbiota interaction model that sustained
1 cocultures of intestinal epithelium with a complex microbiome for
only 48 h and in which the bacteria had to be physically restricted
0 from contacting the epithelium by a semi-permeable membrane
to ensure epithelial viability17,46. Importantly, providing a physi-
ct sia

Bi es

B la

En ob ia
er

ra illo s
R cte ira

oc s

tte s
lla
rm )

ologically relevant low-oxygen microenvironment on-chip also


Pa sc cu

in ide

Su cu
ke ium

t
te act
ph

re
d

C lau

ba sp
Ba an

O oc

oc
oi

um ro
lo
Ak cter

er

sustained a higher level of microbial diversity (~200 unique OTUs)


ro
itr
a
ub

and increased the abundance of obligate anaerobic microbiota


(E

compared to aerobically cultured chips, in addition to maintaining


b a diverse community of commensal microorganisms that closely
30
resembled that of the human gut microbiome in vivo. For example,
log2[Fold difference]

20 when the complex gut microbiome was cultured in the anaerobic


Day 1 Intestine Chip, an abundance of obligate anaerobic bacteria was
10 Day 2
maintained, with ratios of Firmicutes and Bacteroidetes similar to
Day 3
those observed in human faeces43. Furthermore, we found that co-
0
culturing intestinal epithelium with either single commensal gut
–10
microorganisms (for example, B. fragilis) or over 200 different bac-
terial OTUs under physiologically relevant anaerobic conditions
actually enhanced epithelial barrier function compared to aerobic
Bi lla

um rma a
oc ia

ra ter s

En ter s
ro es

ob us

r
Bl a

tte a
illo )

te

chips, which is again consistent with in vivo findings47. This is in


Pa ac ccu

e
l
ir
Su ti
sc m

ke hi

in ns
re

ba oid

te oid
C occ
au
sp

ac
O eriu

Ak lop

B o

direct contrast to past studies in which the coculture of pathogenic


t

itr
ac

c
ub

bacteria (for example, enteroinvasive Escherichia coli) was shown to


(E

rapidly compromise barrier function in the same Caco2 cell-lined


Intestine Chips21. This ability to discriminate between the healthy
Fig. 4 | Anaerobic conditions in the Intestine Chip enhance the growth
and injury responses of human intestinal epithelium in the pres-
of multiple genera compared to the aerobic chip and conventional liquid
ence of commensal and pathogenic bacteria, respectively, and to
culture. a, The differential abundance of bacterial genera in the Caco2
study these direct intercellular interactions under controlled con-
Intestine Chip measured under anaerobic conditions over 3 d of culture
ditions in  vitro, is an important advantage of this model versus
(top) or at day 3 in the anaerobic chip compared to the aerobic chip or
approaches that separate the microorganisms from the epithelium
anaerobic liquid culture (bottom). Data are presented as log10 of the total
by nanoporous membranes.
read counts for each genus; each data point represents one chip.The total
Oxygen tension is one of the main regulators of intestinal func-
read counts for all genera are normalized to their counts in liquid culture
tion and the pathogenesis of gastrointestinal diseases48,49. By inte-
(data are presented as box plots with individual data points overlaid, where
grating non-toxic oxygen sensors into our devices, we were able
the lower and upper edges of the box represent 25th and 75th percentiles
to control and measure oxygen levels throughout the microfluidic
and the middle bar is the median; error bars represent the s.d.). b, The
Intestine Chips without interfering with microscopic imaging,
differential abundance in bacterial genera measured over 1−3 d of coculture
device fabrication or cell culture. Using these sensors, instead of
in the anaerobic versus aerobic Intestine Chip (data are represented as
incorporating multiple external oxygen-detecting probes, enables
log2 fold change; each data point corresponds to the differential abundance
this approach to be more easily scaled so that many miniaturized
for a given genus on a given day, comparing anaerobic to aerobic cultures;
Organ Chips can be created. The anaerobic chamber that we engi-
n = 4 chips for each group on each day). Bacterial phylogenies were
neered also generates radial oxygen gradients across the endothe-
obtained from GreenGenes 1.7.
lium−epithelium−microbiome interface that allow the oxygenation
of human tissues while providing an anaerobic environment for
the growth of obligate anaerobes. Anaerobic incubators or glove
ranging from 118 to 135 OTUs (Supplementary Table 2) corre- boxes can be used to maintain hypoxic conditions for bacterial
sponding to 6 phyla (Actinobacteria, Bacteroidetes, Cyanobacteria, cultures, but they commonly provide a single, uniform low oxy-
Firmicutes, Proteobacteria and Tenericutes) and 32 unique genera. gen concentration, as opposed to physiologically relevant oxygen
Thus, the hypoxic Intestine Chip method can be used to sustain a gradients directed across tissue−tissue interfaces. In contrast, our
complex community of human microorganisms in direct contact anaerobic chamber is portable, highly customizable, compatible
with normal, patient-derived human intestinal epithelial cells for with imaging and, most importantly, capable of engineering oxygen
many days in culture, which could be valuable for personalized gradients across the endothelial−epithelial interface of any Organ
medicine in the future. Chip on demand.

526 Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng


Nature Biomedical Engineering Articles
a b More importantly, we found that the hypoxic human Intestine
F-actin Muc2 DAPI
Chip supports the coculture of complex human microbiota com-
posed of over 200 unique OTUs and at least 11 different gen-
era of bacteria for multiple days in coculture. Bacterial members
of the Bacteroidetes and Firmicutes phyla, and to a lesser degree
Verrucomicrobia and Proteobacteria, which comprise the human
intestinal microbiome in vivo55, also colonized our Caco2 Intestine
Chips at similar ratios. Anaerobic chips had increased levels of
anaerobic Clostridia, Bacteroides and Akkermansia, whereas
Proteobacteria, which accumulate mainly at more oxygenated
regions of the proximal gastrointestinal tract52,56, dominated the
aerobic chips. One limitation of our approach is the need to dilute
c the complex microbiome inoculum to avoid rapid unrestrained
15 Anaerobic
Anaerobic + Microbiome bacterial overgrowth. This may result in the exclusion of some
rare bacteria; however, this could be ameliorated by using larger
Papp (10–5 cm s–1)

10 Intestine Chips, optimizing the lumen perfusion rate, applying


cyclic (peristalsis-like) mechanical deformations that can suppress
5 the growth of some commensals21 or altering medium conditions
to limit bacterial overgrowth in the future. Nevertheless, these data
0 show that the Intestine Chip with a hypoxia gradient and anaerobic
0 1 2 3 4 5 conditions in the lumen of the epithelial channel promoted more
Time (d) bacterial diversity than the aerobic system. Moreover, the anaerobic
human Intestine Chip supported a wide range of bacterial genera
Fig. 5 | Anaerobic coculture of a gut microbiome obtained from fresh similar to those found in human stools, which is much more com-
human patient-derived stool with primary human ileal epithelium in the plex than microbiome communities that have been cultured previ-
Intestine Chip. a,b, Microscopic views showing the villus morphology of ously in direct contact with mammalian cells, and we could sustain
the primary ileal epithelium cultured for 5 d in the Intestine Chip under these cocultures for at least 5 d in vitro.
anaerobic conditions. a, Viewed from above by DIC (scale bar, 50 μm). Others have maintained complex microbiota in test tube cul-
b, A confocal immunofluorescence cross-section image for MUC2, F-actin tures57; however, our results indicate that the presence of a more
and DAPI (scale bar, 50 μm; the inset shows the area highlighted by the in vivo-like intestinal tissue microenvironment significantly influ-
white dashed rectangle at a higher magnification; representative images ences the composition of the microbial community. For example,
from 4 intestine chips are shown). c, Changes in Papp were measured by the mucus degrading, obligate anaerobe genus Akkermansia was
quantifying Cascade blue transport across the tissue interface in the found in greater abundance in the anaerobic Intestine Chips, which
primary Intestine Chip during coculture with or without a complex contain human intestinal epithelial cells that secrete mucus, than
human gut microbiome under anaerobic conditions (bacteria from in liquid cultures maintained under similar anaerobic conditions,
patient-derived stool samples were added on day 0; n = 3 individual which were artificially supplemented with mucin. In contrast to liq-
chips; data are presented as mean + s.d.). See Supplementary Table 2 uid cultures, the anaerobic Intestine Chip also allows inferences to
for the richness and Shannon diversity of bacteria in effluent samples of be made regarding the effects of commensal microorganisms on the
primary Intestine Chips. host epithelium and vice versa. It is interesting that the enhanced
growth of Akkermansia in the anaerobic Intestine Chip was accom-
panied by increased intestinal barrier function, as the high abun-
Oxygen concentrations in the lumen of the human intestine dance of this organism has been previously suggested to enhance
are known to affect the spatial distribution and metabolism of gut gut barrier function in vivo43–45.
flora50 and most intestinal bacteria are obligate anaerobes, many HIF-1α is believed to control barrier integrity by regulating mul-
of which fail to grow at oxygen concentrations greater than ~0.5% tiple barrier-protective genes and its dysregulation may be involved
(ref. 51). Any culture system designed to recapitulate the host gut− in gastrointestinal disorders58,59. Although we observed elevated lev-
microbiome interface must therefore be able to achieve and sustain els of nuclear HIF-1α in anaerobic Intestine Chips, we did not detect
oxygen concentrations at these low levels. A microfluidics-based any changes in barrier function unless we also cocultured complex
anaerobic culture system that maintains oxygen levels as low as 0.8% microbiota. Thus, this experimental system allows us to parse which
in the presence of a facultative anaerobe has been described previ- physiological effects are due to changes in oxygen levels alone and
ously46, but this level is still too high to support obligate anaerobes. which are due to the presence of commensal microorganisms under
Moreover, this past model used both a synthetic mucus layer and low-oxygen conditions. Defining the causative relationship between
a nanoporous membrane to physically separate bacteria from the the abundance of each individual genus of bacteria and the distinct
intestinal epithelium46. Using our custom anaerobic chamber, we functions of the cocultured human intestinal epithelium is beyond
were able to attain an oxygen concentration of less than 0.3% in the the scope of this study. However, this system could be harnessed
epithelial channel where we cultured the commensal microorgan- to address these types of questions, as well as how different com-
isms, which is much closer to that found in the gut lumen in vivo52. mensal microorganisms contribute to the pathophysiology of vari-
We also validated the relevance of these hypoxic culture conditions ous gastrointestinal diseases60 in the future. This model could also
by showing that they support the growth of an obligate anaerobe, be used to analyse fluctuations in microbiome abundance, function
B. fragilis, that cannot grow in the presence of greater than ~0.5% and location within the chip (for example, under conditions of anti-
dissolved oxygen31,53. Furthermore, the finding that the coculture of biotic exposure or hormonal stimulation) and their effects on intes-
human intestinal epithelium with B. fragilis under anaerobic con- tinal epithelial physiology. Moreover, the clinical relevance of these
ditions also increased (rather than decreased) intestinal barrier studies could be further enhanced by developing patient-specific
function on-chip is consistent with the finding that oral delivery Intestine Chips with both host cells and the gut microbiome isolated
of B. fragilis corrects intestinal permeability defects in a mouse from the same patients, as well as by obtaining microbiome isolates
autism model54. from different regions of the intestine (for example, duodenum,

Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng 527


Articles Nature Biomedical Engineering

jejunum, ileum, colon) and culturing them on chips lined with cells chips and sensors with thicknesses between 150 and 300 μm did not affect the
from each of these regions. oxygen readouts.
The purpose of this study was to develop a method for co-cul- Anaerobic chamber fabrication and validation. To fabricate the anaerobic
turing a complex living human gut microbiome, including obligate chamber, acrylic parts were cut using a laser cutter (Epilog) and assembled
anaerobes that require strict anaerobic conditions (<0.5–1% O2) to using an acrylic solvent (SciGrip Acrylic Cement). Gaskets were laser cut from
survive, in direct contact with human intestinal epithelial cells and adhesive-backed silicone rubber sheets (20 Shore A hardness, McMaster−Carr)
their overlying mucus layer for extended times in vitro. Although and magnetic clasps were attached using adhesive-backed magnets. The anaerobic
chamber was tested using a calibrated Oxy-4 optical probe system (PreSens
we did not intend to model any specific region of the gastroin- GmbH) to verify the hypoxic conditions. To do this, the chamber was purged with
testinal system using our chips, it should be noted that it is pos- 5% CO2 in N2 bubbled through deionized water at 81 ml min−1, 162 ml min−1 or
sible to create Organ Chips lined with cells from different regions 243 ml min−1 for 1 h, at which point N2 flow was stopped and the chamber allowed
of the intestine (for example, duodenum, jejunum, ileum, colon), to recover to atmospheric oxygen levels for 3 h.
use oxygen tensions appropriate for each region (5−0.5%, moving
from duodenum to colon) and potentially introduce the microbi- Oxygen sensing in the Intestine Chip. To visualize and quantify the concentration
of oxygen throughout the chip, oxygen measurements were taken by non-invasive
ome aspirates from each of these regions. We have previously shown fluorescence readout using a VisiSens system (PreSens GmbH). Using a CCD
that a primary Intestine Chip better recapitulates the morphology, (charge-coupled device) camera and the VisiSens software (V1.1.2.10), the amount
multicellular composition and gene expression patterns of the intes- of oxygen was detected at sensor spots and displayed using a computer code in
tinal segment from which it was derived than other in vitro intesti- pseudocolours. The software was designed to calculate oxygen levels on the sensor
nal culture systems, such as the Caco2 chip and three-dimensional spots via the calibration of the fluorescence reading with defined oxygen levels
at 0 and 100% air saturation (that is, 20.9% O2 of all dissolved gas by volume). In
intestinal organoids26. Furthermore, by integrating primary epithe- all the experiments, oxygen levels were quantified after comparing the readings
lial cells from intestinal biopsies, as we did here, or patient-derived with the calibration values. Air-saturated water and oxygen-free solution (Oakton,
induced pluripotent stem cells61, in combination with microbiomes WD-00653-00) were used to calibrate the sensor spots. As the field-of-view of
obtained from the same patients, it should be possible to develop the VisiSens camera is inherently small, we designed a linear positioning system
that can position the camera directly beneath the Intestine Chips in the hypoxia
patient-, disease- and location-specific host−microbiome cocul-
chamber. This allows indexed movements of the camera to any sensor spot along
ture models and thus pursue a personalized medicine approach the chip or between the chips, and thus facilitates the reproducible imaging of
in the future. The Caco2 Intestine Chips also recapitulate many multiple chips in one run. The sensors do not obscure the regular imaging of the
features of human intestinal physiology and pathophysiology, and chips as they cover only a small portion of the culture area (~3 mm2), allowing for
these cells can be obtained commercially (rather than requiring a regular monitoring of the cultures throughout the experiment. We also designed a
black opaque box that covers the entire chip culture chamber and VisiSens camera
patient biopsy), which would enable their widespread use by aca- to block extraneous light. To analyse the accuracy of sensor spots inside the chips,
demic and industrial laboratories, as well as regulatory agencies oxygen concentrations were measured in the gas phase using calibrated gas tanks
(for example, the FDA). In addition, the modular nature of Organ with known oxygen concentration: 0, 1 and 12.5% O2. The VisiSens imaging system
Chip technology allows for the incorporation of additional cell was validated using an Oxy-4 optical probe system (PreSens GmbH) with optical
types. In this study, we incorporated intestinal endothelium in our fibres (POF-L2.5, PreSens GmbH).
Intestine Chips because it enhances intestinal barrier function, vil-
Oxygen sensor analysis. Images of oxygen sensors were processed in MATLAB
lus development and mucus production21,25,26, but other cell types— (Mathworks, version 9.2) and binarized using the method developed by Otsu66.
such as immune cells and pathogens that play crucial roles in host Morphological erosion and dilation were performed to eliminate any spurious
gut−microbiome interactions62,63—could be incorporated as well. artefacts created during binarization. Simulated annealing was applied to find the
Our oxygen-sensing chips also have the potential to be combined correct assignment of sensors in each image regardless of the chip alignment. The
with on-chip trans-epithelial electrical resistance technology64 for sum of the distances between the centroid of each sensor in the current image
and the centroid of the nearest sensor in the original image was minimized. After
real-time monitoring of intestinal barrier function in the pres- aligning the images, the sensors in the current image registered consistently with
ence of different cell types and individual strains of bacteria. Thus, the sensors in the former image and colorimetric analyses were computed. The
this methodology could be used in the future to unravel complex average intensities were calculated for each of the red, blue and green channels in
functional links between intestinal epithelial cells, immune cells each sensor. The uncalibrated signal from each sensor was taken to be the average
green intensity divided by the average red intensity. The uncalibrated signal was
and gut microorganisms to understand the mechanisms of human
then fitted to a calibration curve. We used a modified Michaelis−Menten two-
disease, discover new therapeutics and advance personalized medi- point calibration as the most generalizable model, Coxy = kmin + (kmax − kmin) × [xg:r/
cine. It may be possible to modify the Organ Chip to model and (krate + xg:r)]; kmax = α × katm, where xg:r denotes the ratio of average green intensity
study host−microbiome interactions in other organs (for example, to average red intensity, Coxy is the fraction of atmospheric oxygen, kmin is the
vagina, skin, lung) as well. sensor signal in anaerobic conditions, kmax is the sensor signal when saturated
with oxygen. krate explains the effect that xg:r has on the concentration of oxygen.
The atmospheric oxygen concentration does not fully saturate the sensor with
Methods oxygen. To overcome this, the actual maximum possible signal from a sensor, kmax,
Fabrication of the oxygen-sensing Organ Chip. Oxygen sensor spots were is estimated by multiplying the uncalibrated signal at atmospheric concentration,
prepared by mixing oxygen-sensitive and optical isolating particles (PreSens katm, by a scale factor α. The Michaelis−Menten curve was approximately linear
GmbH) at a weight ratio of 1:1 in methanol (Sigma, 50 mg ml−1) for 2 h with between xg:r = katm and xg:r = kmax and scaling by a linear coefficient did not hamper
constant stirring. PDMS prepolymer (Sylgard 184, Dow Corning) was then the equation’s ability to generalize between sensors. The curve was fitted using
added to the mixture at 1 g ml−1 and solvent was subsequently removed by images acquired at known oxygen concentrations. The known concentrations
applying a −70 kPa vacuum at 55 °C for 2 h. The PDMS prepolymer was then were measured by an Oxy-4 optical probe system (PreSens GmbH). The oxygen
mixed with curing agent (Sylgard 184, Dow Corning) at a weight ratio of 10:1 concentrations were also validated using oxygen flow at known concentrations
for 4 min under vacuum, spin-coated (150 μm thick) onto a 5 cm silanized over the probe and sensor. Both krate and α were fitted using the data. The model
silicon wafer at 800 r.p.m. for 2 min and cured at 60 °C for at least 30 min. produced a suitable fit for the data (coefficients of multiple correlation: R2 = 0.990
The wafer was removed and the 150 µm thick film was punched into 1 mm for training; R2 = 0.997 and 0.998 for testing). The fitted model generalized well for
diameter sensor discs using a biopsy punch. The sensor discs were dip-coated trials repeated in different chips and on different days.
in uncured PDMS (PDMS prepolymer:curing agent, 10:1), embedded in the
PDMS channels of a 2-channel Organ Chip by placing them in moulds at the Cell culture procedures. Before cell seeding, microfluidic sensor chips were
inlet, middle and outlet of both the upper (epithelium) and lower (endothelium) activated using oxygen plasma (Diener ATTO) and functionalized with
channels and cured in place at 60 °C for 30 min. Organ Chip fabrication was (3-aminopropyl) trimethoxysilane (Sigma, 281778), as reported previously25. Chips
then performed as described previously65. Using this two-step moulding process, were then washed with ethanol, oven-dried at 80 °C and coated with 30 μg ml−1
these sensors were placed directly on the surface of both the vascular and collagen (Gibco, A10483-01) and 100 μg ml−1 Matrigel (BD Biosciences, 356237)
epithelial channels of the Organ Chips at their inlet, middle and outlet regions. in serum-free DMEM (Gibco, 10564011) for 1 h at 37 °C. To fabricate the Caco2
The chip fabrication and sensor integration steps involving plasma treatment Intestine Chip, HIMECs (ScienCell) were first seeded (1.5 × 105 cells cm−2) in the
did not interfere with sensor function or the functionality of the microfluidic bottom channel of the chips, on the opposite side of the porous membrane. Chips

528 Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng


Nature Biomedical Engineering Articles
were then placed in a 37 °C incubator for 1.5 h. For the HIMEC culture, endothelial at Brigham and Women’s Hospital. All methods were carried out in accordance
growth medium (EGM2-MV) containing human epidermal growth factor, with a protocol approved by the Partner’s Human Research Committee, the
hydrocortisone, vascular endothelial growth factor, human fibroblastic growth Institutional Review Board (IRB) of Partners HealthCare overseeing research at
factor-B, R3-insulin-like growth factor-1, ascorbic acid and 5% FBS (Lonza, CC- Brigham and Women’s Hospital (protocol no. 2016-P-001020). Faecal samples were
3202) was used. Human intestinal epithelial cells (Caco2 BBE human colorectal collected from preterm infants born before 32 weeks of gestation from birth until
carcinoma cells, Harvard Digestive Disease Center) were then seeded into the top discharge. Briefly, diapers with faecal samples were collected daily by the bedside
microchannel of the chip (1.5 × 105 cells cm−2) and incubated for 1.5 h. Epithelial nurse, placed in a specimen bag and stored at 4 °C for no more than 24 h. Faecal
cells were fed with DMEM (Gibco, 10564011) containing penicillin/streptomycin material was extracted from diapers using sterile procedures and immediately
and 20% FBS (Gibco, 10082-147). After washing with 200 μl of medium, chips frozen at −80 °C. Selected samples were suspended in brain heart infusion media
were cultured statically overnight to allow cells to form monolayers on both sides (100 mg ml−1) to create a stock solution.
of the membrane. A day after seeding, the top and bottom channels were perfused
(at 60 μl h−1) with epithelial medium and reduced-FBS endothelial medium, Coculture with gut microorganisms and a complex microbiome in the Intestine
respectively. Chips were kept in this condition until villus-like intestinal epithelium Chips. One day before adding the bacteria, media reservoirs were washed with PBS
spontaneously appeared. For the anaerobic culture, the same procedure was and antibiotic-free media was then added to the Intestine Chips in a tissue culture
followed except that after 1 d of perfusion in aerobic conditions, chips were placed hood (aerobic conditions) or in an anaerobic chamber (anaerobic conditions).
in an anaerobic chamber and continuously perfused with 5% CO2 in N2 flowing at The next day, 25 μl of B. fragilis (1 × 107 CFU ml−1) or diluted microbiota stock was
243 ml min−1. added to the apical side of differentiated Intestine Chips in a tissue culture hood
To create primary Intestine Chips, we used cells isolated from human (aerobic conditions) or in an anaerobic chamber (anaerobic conditions). Chips
intestinal organoids that were derived from the resected tissue of a 15-yr-old were left static for 30 min and then perfused at 1 μl min−1. Every 24 h, a 2 min flush
patient diagnosed with ulcerative colitis and collected from grossly unaffected at 50 μl min−1 was performed, the flush outflow was collected and serial dilutions
(macroscopically normal) areas of the terminal ileum during an endoscopy were plated on Brucella plates incubated at 37 °C in an anaerobic chamber (B.
procedure, as described previously26. Informed consent and developmentally fragilis cultures), Brucella plates ±6 μg ml−1 vancomycin (Hmb cultures) or sent for
appropriate assent were obtained at Boston Children’s Hospital from the donor’s 16S rRNA sequencing (Diversigen).
guardian and the donor, respectively. All methods were carried out in accordance
with a protocol approved by the Institutional Review Board of Boston Children’s Morphological analyses. For each experiment, regions of three different Intestine
Hospital (protocol no. IRB-P00000529). Chips were analysed morphologically at each interval. The villus structures formed
Organ Chips with the same two-channel design, but obtained from Emulate by the intestinal epithelium were evaluated using DIC microscopy (Zeiss Axio
Inc., were used to create the primary human Intestine Chip. The chips were Observer Z1 2, AXIO2) or immunoflorescence microscopy with a laser scanning
chemically activated using ER1 and ER2 solutions (Emulate Inc.) before confocal microscope (Leica SP5 X MP DMI-6000 and Zeiss TIRF/LSM 710).
introducing type I collagen (200 µg ml−1) and Matrigel (1% in PBS) into the High-resolution horizontal or vertical cross-sectional images were obtained using
channels and incubating in a humidified 37 °C incubator for 2 h to coat the deconvolution (Huygens) followed by a two-dimensional projection process.
porous membrane with ECM before washing with PBS. Primary human intestinal IMARIS (version 8.1; Bitplane Scientific Software) and ImageJ (PMID 22930834)
epithelial cells isolated from ileal organoids using collagenase and mechanical were used to analyse the obtained images.
dissociation, as described previously26, were then resuspended in expansion For the SEM analysis, Intestine Chips were designed such that the top channel
medium consisting of Advanced DMEM/F12 supplemented with L-WRN- was not irreversibly bonded to the membrane, which permitted the device to be
conditioned medium (50% volume/volume, ATCC), GlutaMAX, HEPES buffer, dismantled manually without disturbing the cultured cells. Chips were first treated
murine epidermal growth factor (50 ng ml−1), N2 supplement, B27 supplement, with Carnoy’s solution for mucus fixation and then fixed with paraformaldehyde
human (Leu15)-gastrin I (10 nM), N-acetyl cysteine (1 mM), nicotinamide (4%; Electron Microscopy Sciences, 157-4) and glutaraldehyde (2.5%; Sigma,
(10 mM), SB202190 (10 µM) and A83-01 (500 nM). A small volume (30 µl) of G7776) and incubated in osmium tetroxide (0.5%; Electron Microscopy Sciences,
the expansion medium solution containing the primary intestinal cells (6 × 106 19152) before serial dehydration in ethanol. Samples were then dried using a
cells ml−1) was used to infuse and fill the apical chamber of each chip resulting critical point drier and imaged using field emission SEM (Hitachi S-4700).
in ~180,000 cells chip−1; these chips were then incubated overnight under static
conditions at 37 °C to promote cell adhesion. The following day, expansion Immunofluorescence microscopy. Epithelial and endothelial cells were washed
medium without cells was perfused (at 60 µl h−1) through the top and bottom with PBS, fixed with 4% paraformaldehyde (Electron Microscopy Sciences, 157-4)
channels and cyclic suction was applied to the hollow side chambers using a and subsequently washed with additional PBS. Permeabilization of cells was
vacuum pump controlled by an electronic vacuum regulator (ITV009, SMC Corp.) accomplished with 0.25% Triton X-100 (Sigma, T8787), followed by incubation
and an Arduino microcontroller to exert peristalsis-like stretching motions (10% in blocking buffer containing 1% BSA (Sigma, A4503) and 10% donkey serum
cell strain, 0.15 Hz frequency) on the porous membrane and attached epithelium. (Sigma, D9663) at room temperature. Primary antibodies against ZO-1 (Life
Chips were maintained under these conditions until villus-like structures were Technologies, 33–9100, dilution 1:200), VE-cadherin/CD144 (BD Biosciences,
detected visually under phase contrast imaging (~14 d). Before adding the 555661, dilution 1:200), villin (Life Technologies, PA5-29078, dilution 1:100),
microbiota, peristalsis-like cyclic stretching was stopped and the apical medium Muc2 (Santa Cruz Biotechnology, sc-15334, dilution 1:100) or HIF-1α (Abcam,
was then replaced with antibiotic-free differentiation medium containing ab16066, dilution 1:100) were added and incubated overnight at 4 °C, followed
microbial supplements (1 mg ml−1 pectin, 1 mg ml−1 mucin, 5 μg ml−1 haemin and by multiple PBS washes. Cells were then incubated with secondary fluorescent
0.5 μg ml−1 vitamin K1) and the basal medium was replaced with antibiotic-free antibodies (Life Technologies) at room temperature and washed with PBS;
expansion medium. nuclei were co-stained with DAPI (Invitrogen, D1306). Microscopy was
performed with a laser scanning confocal microscope (Leica SP5 X MP DMI-6000
Bacterial and microbiota culture. B. fragilis (9343 strain) was grown overnight or Zeiss TIRF/LSM 710).
at 37 °C under anaerobic conditions (80% N2, 10% H2, 10% CO2) in rich media
containing yeast extract (5 g l−1), proteose peptone (20 g l−1), NaCl (5 g l−1), haemin Mucus detection. Mucus was visualized using a WGA−Alexa Fluor 488 conjugate
(5 mg l−1), vitamin K1 (0.5 mg l−1), K2HPO4 (5 g l−1) and HADA (emission maximum (Thermo Fisher Scientific) for live cell imaging, as described previously33. Briefly,
450 nm; 0.8 mM) or N-cyclopropenyl galactosaminyl carbamate (GalCCP; 250 μm). WGA solution (25 μg ml−1 in culture medium) was allowed to flow through the
Haemin, vitamin K1, K2HPO4 and HADA32 were added through a 0.22 μm filter epithelium channel for 30 min. The top channel was subsequently washed with PBS
after autoclaving the other ingredients. For the HADA labelling, B. fragilis was in the dark and counter-stained with DAPI to visualize the nuclei. To stain acidic
pelleted at 5,000g, washed once in DMEM and resuspended in Caco2 media mucopolysaccharides within the intestinal mucus, Intestine Chips were stained
(DMEM, 20% FBS, 1% glutamine, 1 mg ml−1 pectin, 1 mg ml−1 mucin, 5 μg ml−1 with 0.1% (weight/volume) alcian blue solution (pH 2.5; 8GX, Sigma) in 3% acetic
haemin, 0.5 μg ml−1 vitamin K1) at 1 × 107 CFU ml−1. For the GalCCP labelling, acid (Sigma) by allowing the solution to flow into the microchannels at 50 μl h−1 for
B. fragilis was pelleted at 5,000g, washed twice with PBS, once with 1% BSA in PBS, 12 h and then washing with PBS.
then incubated with 5 μM tetrazine−Cy3 for 1 h at 37 °C. Bacteria were pelleted
and washed twice with 1% BSA in PBS, once with PBS32 and resuspended in Caco2 Paracellular permeability measurements. 50 μg ml−1 of Cascade blue (5.9 kDa;
media. For the microbiota coculture, colon and caecum content from five mice Thermo Fisher, C687) was introduced to the epithelium channel (at 60 ml h−1)
colonized with healthy human microbiota (Hmb)23 were collected and resuspended and the fluorescence intensities (390 nm/420 nm) of the top and bottom channel
in sterile PBS inside an anaerobic chamber (100 mg of content ml−1). The slurry effluents were measured using a multimode plate reader (BioTek NEO). The apical-
was then filtered (40 μm) and aliquoted and stored at −80 °C as the Hmb stock, to-basolateral flux of the paracellular marker was calculated using the following
which was diluted 1:100 in epithelial medium when added to the Intestine Chips. equation: Papp = (dQ/dt)/AdC. dQ/dt (g s−1) is molecular flux, A (cm2) is the total
Animal experiments were performed under IACUC approval (ID no. IS00000187-3; area of diffusion and dC (mg ml−1) is the average gradient. In some studies, Lucifer
Microbial Effect on Mammalian Host). For microbiota coculture with patient- yellow was used instead of Cascade blue, per a previously published method26.
derived specimens, faecal samples were collected from infants born at Brigham
and Women’s Hospital and cared for in a single-centre Newborn Intensive Care Cellular toxicity. A CytoTox 96 Non-Radioactive Cytotoxicity Assay (LDH;
Unit. For the infant faecal samples, informed consent from guardians was obtained Promega, G1780) was used in accordance with the manufacturer’s instructions

Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng 529


Articles Nature Biomedical Engineering
to measure epithelium and endothelium viability over time under aerobic and 11. Sadabad, M. S. et al. A simple coculture system shows mutualism
anaerobic culture conditions. In brief, effluents were collected from the top and between anaerobic Faecalibacteria and epithelial Caco-2 cells. Sci. Rep. 5,
bottom channels, mixed with LDH substrate reagent and incubated for 30 min. The 17906 (2015).
enzymatic reaction was terminated using stop solution (containing acetic acid) and 12. Dutta, D. & Clevers, H. Organoid culture systems to study host–pathogen
the absorbance at 492 nm was recorded using a multimode plate reader (BioTek interactions. Curr. Opin. Immunol. 48, 15–22 (2017).
NEO). LDH activity was assessed using quadruplicates of each group, calculated 13. Fatehullah, A., Tan, S. H. & Barker, N. Organoids as an in vitro model of
after subtracting the background absorbance values and reported as a fold change human development and disease. Nat. Cell Biol. 18, 246–254 (2016).
of the total LDH values from the control group. 14. Williamson, I. A. et al. A high-throughput organoid microinjection platform
to study gastrointestinal microbiota and luminal physiology. Cell. Mol.
16S rRNA sequencing analysis. Read counts were obtained after sequencing by Gastroenterol. Hepatol. 6, 301–319 (2018).
processing the FASTQ files using QIIME 1.0, following standard protocols (PMID 15. Bein, A. et al. Microfluidic organ-on-a-chip models of human intestine.
20383131); resulting joined reads were aligned to the Greengenes database. A total Cell. Mol. Gastroenterol. Hepatol. 5, 659–668 (2018).
of 938 OTUs were identified. During our analyses of the 16S sequencing data, 16. Van den Abbeele, P. et al. Incorporating a mucosal environment in a dynamic
we removed OTUs that did not meet certain criteria in terms of representation gut model results in a more representative colonization by lactobacilli.
across all the samples. The data were loaded into R and the phyloseq (version Microb. Biotechnol. 5, 106–115 (2012).
1.26) package67 was used for further processing. Data from a sequencing control 17. Marzorati, M. et al. The HMITM module: a new tool to study the
(medium only) were used to correct the count data in all of our samples. For Host-Microbiota Interaction in the human gastrointestinal tract in vitro.
any given OTU, the read counts for that OTU in the control sample (a medium BMC Microbiol. 14, 133 (2014).
sample without microorganisms that was run through the sequencer, allowing 18. Van de Wiele, T., et al. in The Impact of Food Bioactives on Health: In Vitro
for the control of sequencing errors) was effectively subtracted. If the number of and Ex Vivo Models (eds Verhoeckx, K. et al.) 305–317 (Springer, 2015).
reads for a given OTU in a sample of interest was negative (the number of reads 19. Van den Abbeele, P. et al. Microbial community development in a dynamic
in the control was higher than that of the sample), we listed the number of reads gut model is reproducible, colon region specific, and selective for
of that OTU as 0. After performing diversity analyses, all singletons were removed Bacteroidetes and Clostridium cluster IX. Appl. Environ. Microbiol. 76,
from the dataset and the OTUs were summarized to the genus level, resulting 5237–5246 (2010).
in a total of 42 unique OTUs, corresponding to 9 well-characterized genera (34 20. Kim, H. J., Huh, D., Hamilton, G. & Ingber, D. E. Human gut-on-a-chip
OTUs) and 8 OTUs of unknown genus (Proteobacteria and Firmicutes phyla). inhabited by microbial flora that experiences intestinal peristalsis-like
The differential abundance of these genera between the two culture conditions motions and flow. Lab Chip 12, 2165–2174 (2012).
(aerobic and anaerobic) was calculated using the DESeq2 (version 1.22) package68. 21. Kim, H. J., Li, H., Collins, J. J. & Ingber, D. E. Contributions of microbiome
OTUs showing a differential abundance with a false discovery rate corrected P and mechanical deformation to intestinal bacterial overgrowth and
value q < 0.05 were considered significant. The permutational multivariate ANOVA inflammation in a human gut-on-a-chip. Proc. Natl Acad. Sci. USA 113,
test was run in R using the adonis function in the vegan package between aerobic E7–E15 (2016).
and anaerobic conditions, as well as between the two oxygen conditions across 22. Park, G.-S. et al. Emulating host-microbiome ecosystem of human
the different days. Comparisons to the human stool microbiome were performed gastrointestinal tract in vitro. Stem Cell Rev. 13, 321–334 (2017).
against the Human Microbiome Project HMP1 dataset (dataset contains 44,740 23. Chung, H. et al. Gut immune maturation depends on colonization with a
OTUs across 4,743 samples from 18 different body sites.) host-specific microbiota. Cell 149, 1578–1593 (2012).
24. Surana, N. K. & Kasper, D. L. Moving beyond microbiome-wide associations
Statistical analysis. All experiments were carried out at n = 3–6 (see figure to causal microbe identification. Nature 552, 244–247 (2017).
captions) and results and error bars indicate mean ± s.d. Data analysis was 25. Jalili-Firoozinezhad, S. et al. Modeling radiation injury-induced cell death
performed with a one-way ANOVA with a Tukey honestly significant difference and countermeasure drug responses in a human Gut-on-a-Chip. Cell Death
post hoc test using Prism (GraphPad, version 7.05). Statistical analysis between the Dis. 9, 223 (2018).
two conditions was performed using an unpaired Student’s t-test. P values less than 26. Kasendra, M. et al. Development of a primary human Small Intestine-on-a-
0.05 were considered significant. Chip using biopsy-derived organoids. Sci. Rep. 8, 2871 (2018).
27. Zheng, L., Kelly, C. J. & Colgan, S. P. Physiologic hypoxia and oxygen
Reporting Summary. Further information on research design is available in the homeostasis in the healthy intestine. A review in the theme: cellular
Nature Research Reporting Summary linked to this article. responses to hypoxia. Am. J. Physiol. Cell Physiol. 309, C350–C360 (2015).
28. Jiang, B. H., Semenza, G. L., Bauer, C. & Marti, H. H. Hypoxia-inducible
Data availability factor 1 levels vary exponentially over a physiologically relevant range of O2
The main data supporting the findings of this study are available in the Article and tension. Am. J. Physiol. Cell Physiol. 271, C1172–C1180 (1996).
Supplementary Information. The raw data generated in this study are available 29. Chilov, D. et al. Induction and nuclear translocation of hypoxia-inducible
from the corresponding author on reasonable request. factor-1 (HIF-1): heterodimerization with ARNT is not necessary for nuclear
accumulation of HIF-1alpha. J. Cell Sci. 112, 1203–1212 (1999).
30. Surana, N. K. & Kasper, D. L. The yin yang of bacterial polysaccharides:
Received: 11 September 2018; Accepted: 4 April 2019; lessons learned from B. fragilis PSA. Immunol. Rev. 245, 13–26 (2012).
Published online: 13 May 2019 31. Patrick, S., Reid, J. H. & Larkin, M. J. The growth and survival of capsulate
and non-capsulate Bacteroides fragilis in vivo and in vitro. J. Med. Microbiol.
References 17, 237–246 (1984).
1. Cho, I. & Blaser, M. J. The human microbiome: at the interface of health and 32. Hudak, J. E., Alvarez, D., Skelly, A., von Andrian, U. H. & Kasper, D. L.
disease. Nat. Rev. Genet. 13, 260–270 (2012). Illuminating vital surface molecules of symbionts in health and disease.
2. Donaldson, G. P., Lee, S. M. & Mazmanian, S. K. Gut biogeography of the Nat. Microbiol. 2, 17099 (2017).
bacterial microbiota. Nat. Rev. Microbiol. 14, 20–32 (2016). 33. Shin, W. & Kim, H. J. Intestinal barrier dysfunction orchestrates the onset of
3. Pickard, J. M., Zeng, M. Y., Caruso, R. & Núñez, G. Gut microbiota: role in inflammatory host–microbiome cross-talk in a human gut inflammation-on-
pathogen colonization, immune responses, and inflammatory disease. a-chip. Proc. Natl Acad. Sci. USA 115, E10539–E10547 (2018).
Immunol. Rev. 279, 70–89 (2017). 34. Kuo, J. C.-H. et al. Detection of colorectal dysplasia using fluorescently
4. Sommer, F. & Bäckhed, F. The gut microbiota—masters of host development labelled lectins. Sci. Rep. 6, 24231 (2016).
and physiology. Nat. Rev. Microbiol. 11, 227–238 (2013). 35. Kim, H. J. & Ingber, D. E. Gut-on-a-Chip microenvironment induces
5. Walter, J. & Ley, R. The human gut microbiome: ecology and recent human intestinal cells to undergo villus differentiation. Integr. Biol. 5,
evolutionary changes. Annu. Rev. Microbiol. 65, 411–429 (2011). 1130–1140 (2013).
6. Sommer, M. O. Advancing gut microbiome research using cultivation. 36. Rozee, K. R., Cooper, D., Lam, K. & Costerton, J. W. Microbial flora of the
Curr. Opin. Microbiol. 27, 127–132 (2015). mouse ileum mucous layer and epithelial surface. Appl. Environ. Microbiol.
7. Eain, M. M. G. et al. Engineering solutions for representative models of the 43, 1451–1463 (1982).
gastrointestinal human-microbe interface. Engineering 3, 60–65 (2017). 37. Lock, J. Y., Carlson, T. L., Wang, C.-M., Chen, A. & Carrier, R. L. Acute
8. Fritz, J. V., Desai, M. S., Shah, P., Schneider, J. G. & Wilmes, P. From exposure to commonly ingested emulsifiers alters intestinal mucus structure
meta-omics to causality: experimental models for human microbiome and transport properties. Sci. Rep. 8, 10008 (2018).
research. Microbiome 1, 14 (2013). 38. Villmones, H. C. et al. Species level description of the human ileal bacterial
9. Arrieta, M.-C., Walter, J. & Finlay, B. B. Human microbiota-associated mice: microbiota. Sci. Rep. 8, 4736 (2018).
a model with challenges. Cell Host Microbe 19, 575–578 (2016). 39. The Human Microbiome Project ConsortiumStructure, function and diversity
10. Nguyen, T. L. A., Vieira-Silva, S., Liston, A. & Raes, J. How informative of the healthy human microbiome. Nature 486, 207–214 (2012).
is the mouse for human gut microbiota research? Dis. Model. Mech. 8, 40. Stearns, J. C. et al. Bacterial biogeography of the human digestive tract.
1–16 (2015). Sci. Rep. 1, 170 (2011).

530 Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng


Nature Biomedical Engineering Articles
41. Guarner, F. & Malagelada, J.-R. Gut flora in health and disease. Lancet 361, 60. Cirstea, M., Radisavljevic, N. & Finlay, B. B. Good bug, bad bug: breaking
512–519 (2003). through microbial stereotypes. Cell Host Microbe 23, 10–13 (2018).
42. Wang, M., Ahrné, S., Jeppsson, B. & Molin, G. Comparison of bacterial 61. Workman, M. J. et al. Enhanced utilization of induced pluripotent stem
diversity along the human intestinal tract by direct cloning and sequencing of cell–derived human intestinal organoids using microengineered chips. Cell.
16S rRNA genes. FEMS Microbiol. Ecol. 54, 219–231 (2005). Mol. Gastroenterol. Hepatol. 5, 669–677 (2018).
43. Fujio-Vejar, S. et al. The gut microbiota of healthy Chilean subjects reveals 62. Hooper, L. V., Littman, D. R. & Macpherson, A. J. Interactions between the
a high abundance of the phylum Verrucomicrobia. Front. Microbiol. 8, microbiota and the immune system. Science 336, 1268–1273 (2012).
1221 (2017). 63. Rimoldi, M. et al. Intestinal immune homeostasis is regulated by the crosstalk
44. Schneeberger, M. et al. Akkermansia muciniphila inversely correlates with the between epithelial cells and dendritic cells. Nat. Immunol. 6, 507–514 (2005).
onset of inflammation, altered adipose tissue metabolism and metabolic 64. Henry, O. Y. F. et al. Organs-on-chips with integrated electrodes for
disorders during obesity in mice. Sci. Rep. 5, 16643 (2015). trans-epithelial electrical resistance (TEER) measurements of human
45. Everard, A. et al. Cross-talk between Akkermansia muciniphila and intestinal epithelial barrier function. Lab Chip 17, 2264–2271 (2017).
epithelium controls diet-induced obesity. Proc. Natl Acad. Sci. USA 110, 65. Huh, D. et al. Microfabrication of human organs-on-chips. Nat. Protoc. 8,
9066–9071 (2013). 2135–2157 (2013).
46. Shah, P. et al. A microfluidics-based in vitro model of the gastrointestinal 66. Otsu, N. A threshold selection method from gray-level histograms. IEEE
human–microbe interface. Nat. Commun. 7, 11535 (2016). Trans. Syst. Man Cybern. 9, 62–66 (1979).
47. Pedicord, V. A. et al. Exploiting a host-commensal interaction to promote 67. McMurdie, P. J. & Holmes, S. phyloseq: an R package for reproducible
intestinal barrier function and enteric pathogen tolerance. Sci. Immunol. 1, interactive analysis and graphics of microbiome census data. PLoS ONE 8,
eaai7732 (2016). e61217 (2013).
48. Sheridan, W. G., Lowndes, R. H. & Young, H. L. Intraoperative tissue 68. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and
oximetry in the human gastrointestinal tract. Am. J. Surg. 159, dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
314–319 (1990).
49. He, G. et al. Noninvasive measurement of anatomic structure and Acknowledgements
intraluminal oxygenation in the gastrointestinal tract of living mice with This research was supported by a US FDA grant (HHSF223201310079C), a DARPA
spatial and spectral EPR imaging. Proc. Natl Acad. Sci. USA 96, THoR grant (W911NF-16-C-0050), the Bill & Melinda Gates Foundation, the Wyss
4586–4591 (1999). Institute for Biologically Inspired Engineering at Harvard University and Fundação
50. Ohland, C. L. & Jobin, C. Microbial activities and intestinal homeostasis: a para a Ciência e a Tecnologia Portugal (project PD/BD/105774/2014 at the Institute for
delicate balance between health and disease. Cell. Mol. Gastroenterol. Hepatol. Bioengineering and Biosciences). We thank D. E. Achatz (PreSens GmbH) for providing
1, 28–40 (2015). oxygen-sensing particles and for her expert technical advice and T. Ferrante for his
51. Flint, H. J., Scott, K. P., Louis, P. & Duncan, S. H. The role of the gut assistance with imaging.
microbiota in nutrition and health. Nat. Rev. Gastroenterol. Hepatol. 9,
577–589 (2012).
52. Albenberg, L. et al. Correlation between intraluminal oxygen gradient and Author contributions
radial partitioning of intestinal microbiota in humans and mice. S.J.-F., F.S.G., E.L.C., J.M.S.C., R.N. and D.E.I. designed the research. S.J.-F., E.L.C., F.S.G.,
Gastroenterology 147, 1055–1063 (2014). B.N., C.W.F., A.T., A.B., B.S. and M.J.C. performed experiments. S.J.-F., F.S.G., D.M.C.,
53. Baughn, A. D. & Malamy, M. H. The strict anaerobe Bacteroides fragilis grows E.L.C., B.N., D.L.K., R.N. and D.E.I. analysed and interpreted the data. K.E.G. helped to
in and benefits from nanomolar concentrations of oxygen. Nature 427, prepare infant microbiota. D.T.B. established and prepared human ileal organoids. S.J.-F.,
441–444 (2004). F.S.G., E.L.C., D.M.C. and D.E.I. wrote the Article with input from B.N., O.L., J.M.S.C.
54. Hsiao, E. Y. et al. Microbiota modulate behavioral and physiological and R.N. All authors reviewed, discussed and edited the manuscript.
abnormalities associated with neurodevelopmental disorders. Cell 155,
1451–1463 (2013). Competing interests
55. Clemente, J. C., Ursell, L. K., Parfrey, L. W. & Knight, R. The impact D.E.I. holds equity in Emulate, Inc., consults for the company and chairs its scientific
of the gut microbiota on human health: an integrative view. Cell 148, advisory board.
1258–1270 (2012).
56. Shin, N.-R., Whon, T. W. & Bae, J.-W. Proteobacteria: microbial signature of
dysbiosis in gut microbiota. Trends Biotechnol. 33, 496–503 (2015). Additional information
57. Goodman, A. L. et al. Extensive personal human gut microbiota culture Supplementary information is available for this paper at https://doi.org/10.1038/
collections characterized and manipulated in gnotobiotic mice. Proc. Natl s41551-019-0397-0.
Acad. Sci. USA 108, 6252–6257 (2011). Reprints and permissions information is available at www.nature.com/reprints.
58. Karhausen, J. et al. Epithelial hypoxia-inducible factor-1 is protective in Correspondence and requests for materials should be addressed to D.E.I.
murine experimental colitis. J. Clin. Invest. 114, 1098–1106 (2004).
59. Manresa, M. C. & Taylor, C. T. Hypoxia inducible factor (HIF) hydroxylases Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in
as regulators of intestinal epithelial barrier function. Cell. Mol. Gastroenterol. published maps and institutional affiliations.
Hepatol. 3, 303–315 (2017). © The Author(s), under exclusive licence to Springer Nature Limited 2019

Nature Biomedical Engineering | VOL 3 | JULY 2019 | 520–531 | www.nature.com/natbiomedeng 531


nature research | reporting summary
Corresponding author(s): Donald E. Ingber
Last updated by author(s): Apr 2, 2019

Reporting Summary
Nature Research wishes to improve the reproducibility of the work that we publish. This form provides structure for consistency and transparency
in reporting. For further information on Nature Research policies, see Authors & Referees and the Editorial Policy Checklist.

Statistics
For all statistical analyses, confirm that the following items are present in the figure legend, table legend, main text, or Methods section.
n/a Confirmed
The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement
A statement on whether measurements were taken from distinct samples or whether the same sample was measured repeatedly
The statistical test(s) used AND whether they are one- or two-sided
Only common tests should be described solely by name; describe more complex techniques in the Methods section.

A description of all covariates tested


A description of any assumptions or corrections, such as tests of normality and adjustment for multiple comparisons
A full description of the statistical parameters including central tendency (e.g. means) or other basic estimates (e.g. regression coefficient)
AND variation (e.g. standard deviation) or associated estimates of uncertainty (e.g. confidence intervals)

For null hypothesis testing, the test statistic (e.g. F, t, r) with confidence intervals, effect sizes, degrees of freedom and P value noted
Give P values as exact values whenever suitable.

For Bayesian analysis, information on the choice of priors and Markov chain Monte Carlo settings
For hierarchical and complex designs, identification of the appropriate level for tests and full reporting of outcomes
Estimates of effect sizes (e.g. Cohen's d, Pearson's r), indicating how they were calculated
Our web collection on statistics for biologists contains articles on many of the points above.

Software and code


Policy information about availability of computer code
Data collection Microsoft Excel, PRISM-ImageJ (FIJI).
PreSens (OXY-4 mini version 2.30FB) and VisiSens software.

Data analysis IMARIS (IMARIS 7.6 F1 workstation; Bitplane Scientific Software)


MATLAB
ImageJ (FIJI)
Graphpad Prism software
R: A language and environment for statistical computing
Bioinformatics: Raw reads were analyzed using QIIME 1.0 under standard protocols, and resulting joined reads were aligned to the
Greengenes database. The data were loaded into R, and the phyloseq package was used for further processing. Differential abundance of
these genera between the culture conditions was quantified by using the DESeq2 package.
For manuscripts utilizing custom algorithms or software that are central to the research but not yet described in published literature, software must be made available to editors/reviewers.
We strongly encourage code deposition in a community repository (e.g. GitHub). See the Nature Research guidelines for submitting code & software for further information.

Data
October 2018

Policy information about availability of data


All manuscripts must include a data availability statement. This statement should provide the following information, where applicable:
- Accession codes, unique identifiers, or web links for publicly available datasets
- A list of figures that have associated raw data
- A description of any restrictions on data availability

The main data supporting the findings of this study are available within the Article and its Supplementary Information. The raw data generated in this study are
available from the corresponding author upon reasonable request.

1
nature research | reporting summary
Field-specific reporting
Please select the one below that is the best fit for your research. If you are not sure, read the appropriate sections before making your selection.
Life sciences Behavioural & social sciences Ecological, evolutionary & environmental sciences
For a reference copy of the document with all sections, see nature.com/documents/nr-reporting-summary-flat.pdf

Life sciences study design


All studies must disclose on these points even when the disclosure is negative.
Sample size All experiments were carried out with n=3–6 independent biological replicates per experiment (details are provided in Methods and in the
figure legends). Sample sizes were determined on the basis of previous experimental experience as well as the power of the statistical test
performed.

Data exclusions No data were excluded from the analyses.

Replication 16S rRNA sequencing data that has been done once with n=4. All other experiments have been reproduced at least twice, with similar results.
All experiments were reproducible as assessed in multiple Intestine Chip culture devices per experiment. We did not have cases of
irreproducibility.

Randomization Intestine Chips were randomly allocated into the experimental groups.

Blinding All 16S rRNA sequencing data collection was done blindly. For other assays, investigators were not blinded to the identities of the samples.
However, all experimental and control samples were collected/analyzed at the same time under the same condition, and the quantification
was carried out with the same software settings.

Reporting for specific materials, systems and methods


We require information from authors about some types of materials, experimental systems and methods used in many studies. Here, indicate whether each material,
system or method listed is relevant to your study. If you are not sure if a list item applies to your research, read the appropriate section before selecting a response.

Materials & experimental systems Methods


n/a Involved in the study n/a Involved in the study
Antibodies ChIP-seq
Eukaryotic cell lines Flow cytometry
Palaeontology MRI-based neuroimaging
Animals and other organisms
Human research participants
Clinical data

Antibodies
Antibodies used Mouse monoclonal to ZO1 (1:200; Life Technologies 33-9100, Clone: ZO1-1A12; Lot: Q1215680)
Mouse monoclonal to VE-cadherin/CD144 (1:200; BD Biosciences 555661, Clone: 55-7H1; Lot: 6273648)
Rabbit polyclonal to Villin (1:100; Life Technologies PA5-29078, Lot: T12636343A)
Rabbit polyclonal to Muc2 (1:100; Santa Cruz Biotechnology sc-15334, Clone: H-300; Lot: F1915)
Mouse monoclonal to HIF-1a (1:100; Abcam ab16066; Clone: mgc3; Lot: GR271167)

For immunofluorescence, Donkey anti-Mouse IgG (H+L) Cross-Adsorbed Secondary Antibody, DyLight 488 (1:500; ThermoFisher
SA5-10166, Lot: SC2352413A), Donkey anti-Rabbit IgG (H+L) Cross-Adsorbed Secondary Antibody, DyLight 488
(1:500; ThermoFisher SA5-10038, Lot: SC2357051A), Donkey anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody,
Alexa Fluor 555 (1:500; ThermoFisher A31570, Lot: 1850121) and Donkey anti-Rabbit IgG (H+L) Highly Cross-Adsorbed Secondary
Antibody, Alexa Fluor 555 (1:500; ThermoFisher A31572, Lot: 1837922) secondary antibodies were used.
October 2018

Validation ZO1: recommended for immunofluorescence staining by the provider. we performed control experiments, including no primary
antibody (negative) controls and comparison to experimental group staining patterns.

VE-Cadherin/CD144: recommended for immunofluorescence staining by the provider, and the vendor demonstrated positive
staining for CD144 in Human Vein Endothelial Cells (HUVECs).

Villin: recommended for immunofluorescence staining by the provider. we performed control experiments, including no primary
antibody (negative) controls and comparison to experimental group staining patterns.

2
Muc2: recommended for detection of Mucin 2 of mouse, rat and human origin by immunofluorescence and

nature research | reporting summary


immunohistochemistry. The vendor demonstrated positive staining of methanol-fixed SW480 cells showing cytoplasmic
localization.

HIF-1a: recommended for immunofluorescence staining by the provider. The vendor demonstrated positive staining for U251,
A2058 and HeLa cells.

Eukaryotic cell lines


Policy information about cell lines
Cell line source(s) Caco2 BBE human colorectal carcinoma cell; human intestinal microvascular endothelial cells (HIMECs).

Authentication Cells were obtained from the Harvard Digestive Disease Center and from ScienCell (company), and validated via phenotype
analysis.

Mycoplasma contamination Caco2 cells received from the Harvard Digestive Disease Center were negative for mycoplasma contamination; HIMECs were
negative for mycoplasma, as described in the ScienCell data sheet.

Commonly misidentified lines No commonly misidentified cell lines were used.


(See ICLAC register)

Animals and other organisms


Policy information about studies involving animals; ARRIVE guidelines recommended for reporting animal research
Laboratory animals Five C57/Bl6 4–6-week-old mice of mixed sex.

Wild animals The study did not involve wild animals.

Field-collected samples The study did not involve samples collected from the field.

Ethics oversight Animal experiments were performed under IACUC approval (ID# IS00000187-3; Microbial Effect on Mammalian Host)

Note that full information on the approval of the study protocol must also be provided in the manuscript.

Human research participants


Policy information about studies involving human research participants
Population characteristics Cells isolated from human intestinal organoids were derived from resected tissue of a 15-year-old patient diagnosed with
ulcerative colitis, and collected from grossly unaffected (macroscopically normal) areas of the terminal ileum during an
endoscopy procedure.

Infant fecal samples were collected throughout hospitalization from premature infants born prior to 32 weeks of gestation.

Recruitment The ileal biopsies were derived from resected tissue of a patient undergoing endoscopy for gastrointestinal complaints. Infants
were recruited from those who were admitted to the Brigham and Women’s Newborn Intensive Care Unit at birth. Infants were
followed until discharge.

Ethics oversight For the ileal biopsies, informed consent and developmentally-appropriate assent were obtained at Boston Children’s Hospital
from the donors’ guardian and the donor, respectively. All methods were carried out in accordance with the Institutional Review
Board of Boston Children’s Hospital (Protocol# IRB-P00000529) approval.

For infant fecal samples, informed consent from guardians was obtained at Brigham and Women’s Hospital. All methods were
carried out in accordance with a protocol approved by the Partner’s Human Research Committee, the Institutional Review Board
(IRB) of Partners HealthCare overseeing research at Brigham and Women’s Hospital (protocol no. 2016-P-001020).
Note that full information on the approval of the study protocol must also be provided in the manuscript.
October 2018

You might also like