Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

www.acsami.

org Research Article

Flowing through Gastrointestinal Barriers with Model Nanoparticles:


From Complex Fluids to Model Human Intestinal Epithelium
Permeation
Iris Renata Sousa Ribeiro, Raquel Frenedoso da Silva, Renata Santos Rabelo, Talita Miguel Marin,
Jefferson Bettini, and Mateus Borba Cardoso*
Cite This: ACS Appl. Mater. Interfaces 2023, 15, 36025−36035 Read Online
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ACCESS Metrics & More Article Recommendations *


sı Supporting Information
Downloaded via UNIV ESTADUAL DE CAMPINAS on August 22, 2023 at 15:30:37 (UTC).

ABSTRACT: Most nanomaterial-based medicines are intravenously


applied since oral administration comprises challenging-related bio-
logical obstacles, such as interactions with distinct digestive fluids and
their transport through the intestinal barrier. Moreover, there is a lack of
nanoparticle-based studies that faithfully consider the above-cited
obstacles and boost oral-administered nanomedicines’ rational design.
In this study, the physicochemical stability of fluorescent model silica
nanoparticles (f-SiO2NPs) passing through all simulated gastrointestinal
fluids (salivary, gastric, and intestinal) and their absorption and
transport across a model human intestinal epithelium barrier are
investigated. An aggregation/disaggregation f-SiO2NPs process is
identified, although these particles remain chemically and physically
stable after exposure to digestive fluids. Further, fine imaging of f-
SiO2NPs through the absorption and transport across the human intestinal epithelium indicates that nanoparticle transport is time-
dependent. The above-presented protocol shows tremendous potential for deciphering fundamental gastrointestinal nanoparticles’
evolution and can contribute to rational oral administration-based nanomedicine design.
KEYWORDS: nanoparticles, simulated gastrointestinal fluids, intestinal epithelium model, stability, transport

■ INTRODUCTION
Nanomedicine has significantly evolved over the last 20
static in vitro digestion model based on conditions physiolog-
ically relevant to the human gastrointestinal tract.26,27 The
years,1−4 and nanoparticle (NP)-based formulations have been method comprises the three digestion steps (oral, gastric, and
successfully used for diagnosis and disease treatments.5−11 intestinal), considering pH, duration, ionic strength, compo-
While the oral route of nanomedicine administration provides sition, and enzyme concentration. In parallel, in vitro models
benefits such as reduced infection risks, low cost, patient have been developed to mimic small intestine enterocytes,
comfort, and easy use, intravenous injection has been the most allowing absorption and transport analysis with high
employed route. This is mainly due to obstacles related to accuracy.14,28−36 Optimized intestinal models involve the co-
orally administered nanomedicines which undergo aggregation cultivation of human colon carcinoma epithelial (Caco-2, non-
and degradation induced by the acidic pH in the stomach and mucus secretion) and human colorectal adenocarcinoma (HT-
digestive enzymes.2,12−20 Consequently, these structural 29, high mucus secretion) cells in a 9:1 ratio.29,32,37,38
changes can affect the absorption of NPs by the small intestine Nevertheless, most reports have scrutinized the transport of
and their ultimate fate.14,17,21,22 Therefore, the rational nanomedicines across nonideal intestine monoculture models,
development of nanomaterial-based drugs that can successfully
which cannot mimic the human intestinal epithelium
pass through the gastrointestinal fluids and be sufficiently
absorbed and transported across the intestinal epithelium is of accordingly.36,39,40
paramount importance.14,15,23−25
One of the limiting factors facing the development of Received: May 16, 2023
sustainable NP-based medicine for oral administration relies Accepted: July 6, 2023
on the lack of protocols that simultaneously deal with the NPs’ Published: July 19, 2023
stability and degradation in complex digestive fluids and their
transport and absorption through optimized human epithelial
models. The INFOGEST community recently proposed a

© 2023 American Chemical Society https://doi.org/10.1021/acsami.3c07048


36025 ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

Figure 1. Representation of the protocol to evaluate the gastrointestinal evolution of orally administered NPs. Step 1: NPs’ incubation with
simulated salivary, gastric, and intestinal fluids. Fluids’ preparation followed the INFOGEST model. Step 2: Schematic representation of NPs’
absorption and transport across the intestinal barrier (not to scale). Optimized epithelial models were prepared by co-cultivation of Caco-2 and
HT-29 cells (9:1 ratio) in a polycarbonate cell culture insert. Mucus is represented in blue. The porosity of the polycarbonate membrane was not
represented in the drawing to enhance the visualization of the intestinal epithelium.

Most NP-driven studies have only focused on the fluorescent core. Afterward, an additional raw silica shell was
permeation through the intestinal epithelium, while studies formed around the fluorescent core to prevent dye-biological
on complex digestive system-simulating fluids are still lacking environment unspecific interactions, and a set of comple-
in the literature.33,36,38,41−44 These studies in complex media mentary techniques thoroughly characterized the synthesized
are compulsory to draw a widespread understanding of their system. Scanning electron microscope (SEM) images showed a
absorption and transport in conditions that mimic real-world typical quasi-spherical morphology and indicated the average
events. Recent efforts to evaluate the impact of simulated particle size of 86.4 nm and a half-width at half-maximum of
digestive fluids on the absorption and transport of NPs 12.0 nm (Figure S2). Furthermore, these structures presented
through the intestinal model have faced physical and chemical an average hydrodynamic diameter of 106.8 ± 1.6 nm with a
stability issues.45 Consequently, robust studies that simulta- low polydispersity index (PDI, 0.06) and a ζ-potential of −49.1
neously probe the stability and degradation of NPs in complex ± 1.8 mV. Also, f-SiO2NPs showed red fluorescence with
digestive fluids and their transport and absorption through maximum emission at 580 nm, characteristic of the rhodamine
optimized human epithelium models are still lacking in the B isothiocyanate presence (Figure S3).
literature. In Vitro Digestion Experiments. f-SiO2NPs were
Herein, we present a new protocol to investigate the stability submitted to gastrointestinal digestion procedures,26,27 which
and degradation of fluorescent model silica nanoparticles (f- included the oral, gastric, and intestinal phases since they
SiO2NPs) in simulated salivary, gastric, and intestinal fluids mimic in vivo conditions, taking into account the presence of
simultaneously while in-depth probing their absorption and digestive enzymes and their concentrations, pH, digestion
transport through an optimized intestinal epithelium model time, and salt concentrations, among other factors (Table S1).
(Caco-2:HT-29) (Figure 1). The first innovative strategy First, f-SiO2NPs (final concentration of 0.5 mg mL−1) were
combines scattering and microscopy techniques to follow NPs’ incubated with simulated salivary fluid (SSF) for 2 min,
aggregation state, surface degradation, and organic material followed by 2 h incubations with simulated gastric fluid (SGF)
adsorption while passing through salivary, gastric, and and with simulated intestinal fluid (SIF), respectively. It is
intestinal fluids (step 1�Figure 1). The second meaningful essential to highlight that the transit time of the small intestine
advance is based on visualizing the intimate interaction of f- is relatively constant (2−4 h).23 INFOGEST static in vitro
SiO2NPs with the epithelial barrier model during absorption model considers 2 h of incubation not only because of the
and transport processes (step 2�Figure 1). This protocol accurate transit times in the gastrointestinal tract but also due
overcomes drawbacks found in previous research studies, the process is a static method, so no eventual products are
where the obstacles of the gastrointestinal tract are not removed, which may impair the functioning of the enzymes for
analyzed in their entirety but addressed separately and, prolonged periods.48 The conductivity measurements indi-
generally, under nonoptimized conditions. cated values of 3.60 mS cm−1 for SSF, 16.30 mS cm−1 for SGF,
and 11.80 mS cm−1 for SIF, and the magnitude of these

■ RESULTS AND DISCUSSION


Characterization of f-SiO2NPs. f-SiO2NPs were chosen
numbers correlates with the concentration of electrolytes in
solution. According to dynamic light scattering (DLS, Figure
2a), f-SiO2NPs in the presence of SSF (pH 7.4) revealed to be
due to their promising nanomedicine-related properties, such stable, with an average hydrodynamic diameter of 115.5 ± 0.3
as high area/volume ratio, physicochemical stability, easy nm and low PDI (0.08). However, NPs underwent an
detection by incorporating a fluorescent dye, biocompatibility, aggregated state in SGF (pH 3.0), likely associated with the
and low toxicity.46,47 f-SiO2NPs were synthesized (Figure S1) low pH and high concentration of electrolytes. Silica
using a modified Stöber method where a fluorescent dye nanoparticles reach their isoelectric point at pH ∼2, resulting
(rhodamine B isothiocyanate) was covalently conjugated to a in a decrease of the electric field formed in the double electric
silica precursor and subsequently condensed, forming a layer and consequent aggregation (Figure S4).49,50 Moreover,
36026 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

Figure 2. NPs’ incubation with gastrointestinal fluids. (a) Size distribution curves obtained using a Zetasizer Nano ZS instrument and (b)
transmission electron microscopy (TEM) images of f-SiO2NPs in water (control, before incubation) and after undergoing interactions with all of
the simulated fluids used in the digestion experiments (after incubation). The samples were centrifuged and redispersed in water for analysis. After
incubation, organic material can be found around the NP as well as salts (black dots). The salt was evidenced by the diffraction pattern highlighted
in the top right inset in (b). Scale bar: 50 nm (TEM images) and 20 nm−1 (diffraction pattern). (c) Schematic representation of the salts around f-
SiO2NPs after incubation in digestive fluids. (d) EFTEM micrographs of f-SiO2NPs before and after SSF, SGF, and SIF incubation. Silicon is
represented in red, oxygen in blue, and carbon in green. As observed, f-SiO2NPs passing through the digestive process have a large amount of
carbon, indicating the presence of organic material. Scale bar: 50 nm. (e) Statistical analysis of height data obtained by atomic force microscopy
(AFM). The analyzes were performed for f-SiO2NPs in water (control) and after incubation in SSF, SGF, and SIF. Results are displayed as mean ±
SD. The one-way analysis of variance (ANOVA) and Tukey’s test were used for data analysis (n = 12). Differences were considered significant
when p < 0.05. Common symbols on top of the bars indicate no statistical difference between them. (f) Representation of f-SiO2NPs surface
roughness at different length scales L (40, 25, and 10 nm) characterized by AFM. (g) Root-mean-square (RMS) roughness as a function of L
obtained from AFM measurements carried out for f-SiO2NPs in water (control) as well as for f-SiO2NPs after incubation in SSF, SGF, and SIF.
Results are displayed as mean ± SD. Two-way ANOVA and Tukey’s test were used for data analysis. Differences were considered significant when
p < 0.05. Common symbols on top of the bars indicate no statistical difference between them.

the effects of ions also play a prominent role and likely induce pH due to the deprotonated hydroxyl groups on their surface,
unspecific interaction between particles as SGF contains high inducing an increase in the electric field formed in the electric
concentrations of electrolytes. Meanwhile, certain disaggrega- double layer and consequent repulsion among NPs (Figure
tion was observed after incubating f-SiO2NPs in SIF (pH 7.4), S4).49 It is worth mentioning that SSF, SGF, and SIF present
which presented an average hydrodynamic diameter of 187.1 ± distinct ionic strengths, and the presence of different enzymes
13.2 nm. Silica nanoparticles are negatively charged at neutral can ultimately play an essential role in the NPs’ stability. The
36027 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

Figure 3. NPs’ stability and absorption and transport across the intestinal barrier. (a) Size distribution curves obtained using a Zetasizer Nano ZS
instrument of f-SiO2NPs (0.5 mg mL−1) after incubation in culture medium DMEM supplemented with 10% FBS. The incubation times were 0, 1,
2, 3, and 24 h at 37 °C. (b) Representation of the intestinal barrier (green) formed by Caco-2 and HT-29 cells in culture inserts. The DMEM
culture medium and f-SiO2NPs are represented by pink and pink spheres, respectively. (c) Percentage of absorption and transport of f-SiO2NPs
(0.5 mg mL−1) through the intestinal epithelium, where the upper compartment is represented in green, the bottom compartment in orange, and
the intestinal epithelium in pink. The concentration of f-SiO2NPs in samples was measured directly for fluorescence considering the excitation
parameter fixed at 524 nm, the gain value at 120, and the maximum emission at 580 nm. Results are displayed as mean ± SD. The ANOVA and
Tukey’s test were used for data analysis (n = 10). Differences were considered significant when p < 0.05. Common symbols on top of the bars
indicate no statistical difference between them. (d) Confocal microscopy images (intermediate slices) for the intestinal epithelium after incubation
for 4 and 24 h with f-SiO2NPs. (e, f) Magnified images of the region with SiO2NPs in (d) referring to the incubation times of 4 and 24 h,
respectively. The cells were stained with DAPI and phalloidin 488 and observed under a confocal microscope using lasers at 405 nm for DAPI
(nuclei), at 490 nm for phalloidin 488 (actin filaments), and at 561 nm for f-SiO2NPs. Scale bar for (d, e, f): 25 μm.

aggregation/disaggregation pattern has already been demon- difference in particle size and form when pristine f-SiO2NPs
strated for nano-sized silica during in vitro digestion of foods were compared with those passed through all gastrointestinal
containing silica as a food additive.49 Nonetheless, little has fluids (Figure S7). It is also worth mentioning that some TEM
been evaluated regarding the effect of digestive fluids on the images revealed the presence of small structures around f-
resulting NPs’ surface. Understanding possible NP surface- SiO2NPs (Figure 2b,c), attributed to the fluids’ salts and
induced degradation is paramount for developing oral organic material content. The diffraction pattern confirmed the
nanomedicines since it may trigger a premature drug release presence of the salts (Figures 2b,c and S8), and the organic
process and induce surface-functionalized nanocarriers mis- material by energy-filtered transmission electron microscopy
targeting. Therefore, an in-depth morphological investigation (EFTEM) images. EFTEM images indicated the deposition of
was carried out, considering the evolution of f-SiO2NPs organic moieties, especially in the carbon channel, around the
through the gastrointestinal fluids. f-SiO2NPs that passed through all gastrointestinal fluids
Transmission electron microscopy (TEM) (Figure S5) and (Figure 2d).51 These residues hampered the effective particle
atomic force microscopy (AFM) (Figure S6) images height acquisition by AFM, so a purification column was
corroborated the dynamic light scattering (DLS) results employed to remove these impurities (Figure S9).
indicating that f-SiO2NPs underwent aggregation in the AFM images revealed a statistically significant height
stomach with subsequent disaggregation in intestinal con- reduction after impurities removal for f-SiO2NPs exposed to
ditions. Moreover, TEM analysis also showed no significant SGF and SIF, which may indicate NP leaching (Figures 2e and
36028 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

Figure 4. NPs’ absorption and transport across the intestinal barrier. (a, b) Scanning electron micrographs of intestinal epithelium after incubation
for 24 h with f-SiO2NPs (0.5 mg mL−1). (c) Schematic representation of the intestinal epithelium (green), showing f-SiO2NPs (pink) on the
microvilli. (d, e) Transmission electron micrographs of resin-embedded intestinal epithelium before incubation with f-SiO2NPs. Abbreviations: DE,
desmosome; TJ, tight junction. (f) Representation of junctions formed between adjacent cell membranes and of some internal cell structures, where
1: zonule of occlusion, 2: zonule of adhesion, 3: desmosome, 4: gap junction, 5: hemidesmosome, 6: mucus granules, and 7: lipid drops. (g, h)
Transmission electron micrographs of resin-embedded intestinal epithelium after incubation for 24 h with f-SiO2NPs. (i) Schematic representation
of f-SiO2NPs located in the vicinity of the microvilli and crossing the intestinal epithelium and reaching the polycarbonate membrane. (j) EDS
analysis of resin-embedded intestinal epithelium after incubation with f-SiO2NPs for 4 h. Maps of the constituent elements of f-SiO2NPs, silicon
(pink) and oxygen (blue) are shown in panels (k) and (l), respectively.

S8). In contrast, this height reduction was not observed by in the samples’ roughness were observed at different
TEM analysis (Figure S6). In addition, no significant changes magnifications (Figure 2f,g), indicating that the f-SiO2NPs
36029 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

are likely chemically stable to the digestion simulated process. increasing incubation times (Figure 3c). The results showed
Thus, we suggest that AFM height reduction is an artifact that around 50% f-SiO2NPs could be found in the intestinal
attributed to the NPs’ passage through the purification column. epithelium after 15 and 24 h of incubation, either deposited on
Larger structures elute first during the desalination procedure, its surface or absorbed by the cells, as shown above.
and the analyzed aliquots were likely rich in smaller NPs. Electron microscopy imaging was further employed to
Stability of f-SiO2NPs in Culture Medium and Cell unriddle the intimate interaction between f-SiO2NPs and the
Viability Assays. Following in vitro digestion experiments, we intestinal epithelium. Regardless of the incubation time, SEM
attempted to evaluate the absorption and transport of f- images confirmed the presence of f-SiO2NPs outside the cells,
SiO2NPs through the model intestinal epithelium since this is mainly associated with microvilli (Figures 4a,b, and S12). A
one of the critical barriers before NPs are distributed schematic epithelium representation is presented in Figure 4c,
throughout the organism. For this purpose, we first tested where the interaction between f-SiO2NPs and the microvilli
the f-SiO2NPs colloidal stability in the epithelium culture becomes evident. TEM images allowed us to identify particular
medium (Dulbecco’s modified Eagle’s medium, (DMEM)) structures of the intestinal epithelium, such as microvilli, cell
supplemented with 10% of fetal bovine serum (FBS) and junctions (e.g., desmosomes [DE] and tight junctions [TJ]),
evaluated the toxicity of these particles against the constituent and the lipid drops produced by HT-29 cells (Figures 4d,e, and
cells of the intestinal barrier (Caco-2 and HT-29). The average S13). These structures in the intestinal epithelia validate our
hydrodynamic diameter of f-SiO2NPs (0.5 mg mL−1) remained co-culture model as a reliable strategy to mimic a lifelike
at ∼160 nm, and PDI at 0.14 after 24 h of incubation suggests epithelial barrier. A drawing containing the most relevant
the absence of massive aggregation (Figure 3a). Particularly, a epithelium structures is presented in Figure 4f and agrees well
subtle difference is observed in the 24 h curve in relation to the with the TEM images described above. The presence of f-
other incubation times, which can be likely attributed to the SiO2NPs in TEM images was ascribed to high electron density
sedimentation of the few aggregates formed at the beginning of structures within the same size range as the NPs synthesized in
the incubation process, slightly shifting the curve to smaller this work (Figures 4g,h, and S13). f-SiO2NPs are more
diameter values. This maintenance in the colloidal stability of electronically dense than those present in the cytosol or within
NPs is a favorable condition for the cellular internalization the lysosomal compartments in cells (lower mass atoms), so
process. Also, f-SiO2NPs presented a negligible influence on these NPs can be easily distinguished from cellular compart-
Caco-2 and HT-29-cells survival rates (Figures S10 and S11). ments. 55 According to TEM images, f-SiO 2NPs were
The viability always remained above 80% irrespective of the simultaneously seen in the vicinity of microvilli and crossing
tested NP concentration, indicating that f-SiO2NPs are the intestinal epithelium before reaching the polycarbonate
nontoxic to these cell lines.52 The high stability of f-SiO2NPs membrane, as depicted in Figure 4i. Energy-dispersive
associated with their nontoxicity suggests that these particles spectroscopy (EDS) images (Figure 4j−l) confirmed that the
can translocate through the intestinal epithelial model without high electron density structures seen by TEM, on the surface
causing cell damage. and inside the intestinal epithelium correspond to f-SiO2NPs.
Transport of f-SiO2NPs across the Intestinal Epithe- After 4 h of incubation, the elements silicon (Si) and oxygen
lium. After 21 days of culture, Caco-2/HT-29 intestinal (O) were identified as components of these nanometric
epithelia were exposed to f-SiO2NPs for 4, 15, and 24 h structures, confirming that these NPs can be internalized and
(Figure 3b). Fluorescence measurements were used to transported across the cells and finally reach the bottom
quantitatively estimate the transport of f-SiO2NPs across the compartment (Figure 4j−l). Osmium (Os) and uranium (U)
intestinal barrier. The incubation time of 4 h corresponds to were typically seen in the regions of resin-embedded
regular intestine transit time in humans and is an indirect epithelium, and their presence is inherent in the sample
strategy to probe NPs-epithelium interaction reliably. After 4 h, preparation. Similar EDS results were seen when f-SiO2NPs
70 ± 12% of f-SiO2NPs remained in the upper compartment, incubation time is increased to 24 h (Figure S14).
while a low transport to the bottom region was observed The results mentioned above reveal that f-SiO2NPs can be
(Figure 3c). The remaining 30 ± 11% of f-SiO2NPs were absorbed and transported through the intestinal epithelium,
either deposited on the intestinal epithelium surface or which is critical for oral drug administration. However,
absorbed by the epithelium. The discrimination between improvements in the particle are needed to increase transport
both scenarios was done qualitatively by imaging the f- efficiency across the intestinal epithelium. Using NPs surface
SiO2NPs-treated intestines with a confocal microscope (Figure functional groups is a relevant strategy that can be used to
3d,e). Using such images for quantitative assessment of f- enhance cell interaction and ultimately facilitate absorption
SiO2NPs absorption by the epithelium is inaccurate, as they do and transport.
not statistically represent the entire specimen. Intermediate Finally, although f-SiO2NPs were used as model nano-
slice images of the epithelium indicated the presence of f- particles, this entire protocol (in vitro digestion and transport
SiO2NPs, most likely around the nucleus, which is a common across intestinal epithelium) can be extended for an in-depth
intracellular destination for particles similar to the ones used understanding of the behavior of other types of nanoparticles
here (Figure 3d,e).53,54 A recent report assessed the passage of (e.g., metallic and polymeric nanoparticles) aimed at oral
SiO2NPs through a nonoptimized intestinal barrier after 6 h of administration.
incubation. Low NP absorption was observed, likely due to the
use of a mucus-free Caco-2 monoculture since HT-29 cells
were not considered in the experimental model.36 Afterward,
■ CONCLUSIONS
In summary, we presented a successful protocol that
we hypothesized that increasing the incubation time would simultaneously evaluates the NPs’ stability and degradation
allow some intracellular f-SiO2NPs to be transported through in digestive fluids (salivary, stomach, and intestinal fluids) as
the intestines. As expected, the percentage of f-SiO2NPs that well as their absorption and transport through a model human
passed through the intestine was significantly higher with intestinal epithelial barrier (co-culture of Caco-2/HT-29 cells).
36030 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

f-SiO2NPs exhibited no irreversible aggregation and degrada- In Vitro Digestion Experiments. The prepared fluids (Table S1,
tion when exposed to simulated gastrointestinal fluids, and Supporting Information) were designed as SSF, SGF, and SIF. The
their absorption and permeation through the epithelial cells conductivity of each fluid was measured using a pH/Cond 3320
conductivity meter (WTW). f-SiO2NPs were added to the respective
were time-dependent. The in vitro protocol described here
fluids at the final concentration of 0.5 mg mL−1. Between each studied
provides a comprehensive picture of the behavior of NPs when phase, NPs were centrifuged (23,940g for 5 min) and suspended in
exposed to the obstacles of oral administration. We suggest the subsequent medium. In addition, a sample of f-SiO2NPs in water,
that this protocol can be employed to evaluate the effect of without incubation in the simulated fluids, was analyzed as a control.
NPs bioaccumulation on the intestinal barrier over time as well The particle size was evaluated after the incubation time of the f-
as to deeply investigate the ultimate behavior of degradable SiO2NPs in each medium using a Zetasizer Nano ZS instrument.
and functional NPs in the gastrointestinal tract. The above- Besides, TEM, EFTEM, and AFM images were obtained to assess
described strategy can easily be expanded to other nano- changes in the particles’ surface along the digestion process in
structured systems and might be employed as a quality control simulated fluids.
TEM images of the f-SiO2NPs were obtained using TALOS F200C
platform that supports complex oral methods and in vivo (Thermo Fisher Scientific) microscope operating at 200 kV
assays. (conventional mode, room temperature) with FEI Ceta 16 M Pixel
CMOS 4 K × 4 K camera (Thermo Fisher Scientific). The samples
■ MATERIALS AND METHODS
Materials. Ammonium hydroxide (28−30%) and tetraethyl
were centrifuged (23,940g for 5 min) and redispersed in water for
analysis. Drop casting was used to previously deposit the sample on
the holey carbon-coated copper grid.
orthosilicate (TEOS) were obtained from Sigma-Aldrich, and ethanol Core-loss EFTEM analyses were carried out at JEOL JEM2100F
(absolute) was purchased from Merck. All reagents and chemicals (200 kV). After incubation of f-SiO2NPs (0.5 mg mL−1) in SSF, SGF,
were used as received without further purification. Water used in and SIF fluids, the samples were centrifuged (23,940g for 5 min) and
described procedures was obtained from the Water purification redispersed in water for analysis. In addition, a sample of f-SiO2NPs in
system (Purelab from ELGA, resistivity of 18.2 MΩ·cm−1). Cell water, without incubation in the simulated fluids, was analyzed as a
culture supplies were obtained from Gibco (Thermo Fisher control. Drop casting was used to previously deposit the sample on
Scientific). the holey carbon-coated copper grid. For EFTEM analysis, the
f-SiO2NPs Synthesis. The dye precursor was synthesized by elemental maps of silicon, oxygen, and carbon were acquired.
conjugation of the isothiocyanate group of rhodamine B isothiocya- High-resolution AFM images were obtained using a Bruker
nate to (3-aminopropyl)triethoxysilane (APTES) at a molar ratio of MultiMode 8 instrument with Peak Force Tapping mode. The
50:1 (dye:APTES) in absolute ethanol under nitrogen flow. The analyses were performed with a ScanAsyst-Air cantilever, charac-
resulting solution was continuously stirred for 24 h. The conjugate terized by a nominal spring constant of 0.4 N m−1 and resonance
was then hydrolyzed in a basic ethanolic solution with TEOS and frequency of 70 kHz. The f-SiO2NPs, after incubation in SSF, SGF,
catalyzed by ammonia.56 In summary, 0.02 mL of dye precursor and and SIF, were centrifugated and resuspended in ultrapure water for
0.51 mL of TEOS were added to 30 mL of ethanol under continuous AFM characterization or submitted to the illustra NAP-25 column for
stirring for 30 min. Then, 1.50 mL of ammonia solution 28% was desalting and buffer exchange of NPs dispersions. Then, the samples
added and the resulting mixture was stirred for 3 h. Subsequently, were deposited in a mica substrate and overnight dried under a gentle
0.02 mL of conjugate and 0.51 mL of TEOS were added once again nitrogen flow. The heights and the surface roughness of the f-
and the reaction was stirred overnight. A silica shell was synthesized SiO2NPs as the root-mean-square (RMS) values Rq of the distribution
around the preformed fluorescent core by the addition of ethanol (60 of heights in the AFM high-resolution topographical images were
mL), ammonia solution (1.40 mL), and TEOS (1.70 mL). TEOS was evaluated with open-source software available at http://gwyddion.
added at a flow rate of 1.00 mL min−1 using a syringe pump (New Era net/.
Pump Systems, NE-8000). The resulting suspension was continually Stability of f-SiO2NPs in Biological Medium. The stability of f-
stirred for 24 h and, then the f-SiO2NPs were centrifuged (10,640g for SiO2NPs (0.5 mg mL−1) in the biological medium was verified
15 min) and, then washed once with ethanol and four times with considering 0, 1, 2, 3, and 24 h of incubation at 37 °C. The medium
deionized water. f-SiO2NPs were dispersed in aqueous solution with used was DMEM supplemented with 10% of FBS, and the
the final concentration adjusted to 5.70 g L−1. measurements were performed using a Zetasizer Nano ZS instrument.
f-SiO2NPs Characterization. The particle size (Z-average), PDI, Three measurements (each consisting of at least 10 scans) were taken
and ζ-potential measurement of the dispersions were determined for each sample.
using a Zetasizer Nano ZS instrument (Malvern Instruments Ltd.). Cell Viability Assays. Alamar Blue. Caco-2 and HT-29 cells were
The size and PDI measurements were taken at 25 °C at an angle of seeded in 96-well microplates at a density of 5 × 105 cells mL−1 and
173°, and the data were calculated as the mean of three successive incubated for 24 h in a humidified atmosphere at 37 °C and 5% CO2
analyses (100 s) for at least three independent samples. The to obtain a subconfluent monolayer. Cells were exposed to f-SiO2NPs
dispersions were diluted with water to adjust the signal level. ζ- at concentrations of 0.05, 0.10, 0.50, and 1.00 mg mL−1. After 24 h
Potential measurements were used to evaluate the residual charge of incubation, the cells were washed three times with phosphate-buffered
the f-SiO2NPs. The measurements were taken using a He−Ne (633 saline (PBS), Alamar Blue (Invitrogen, 10% in DMEM) was added to
nm) laser. A dispersion of nanoparticles (1 mg mL−1) was prepared in each well, and samples were incubated for 3 h at 37 °C. The
10 mM phosphate buffer (pH 7.4) for the analysis. Three supernatant was posteriorly collected from the wells, put in a fresh 96-
measurements (each consisting of at least 10 scans) were taken for well plate, and analyzed with a spectrophotometer (1420 Victor,
each sample. PerkinElmer) at 560 nm excitation wavelength and 590 nm emission
Electron microscopy was performed using a SEM-FEM HR (FEI wavelength. Results are displayed as mean ± standard deviation (SD).
Inspect) operating at an accelerating voltage of 10 kV. Images were LIVE/DEAD. Caco-2 and HT-29 cells were incubated for 24 h under
acquired in scanning mode. The samples were deposited onto 400 standard conditions. Culture medium was removed, and the viability
mesh holey carbon-coated copper grids. was assessed by LIVE/DEAD Viability/Cytotoxicity Kit assays
Fluorescence measurements were performed using a TECAN (Invitrogen). LIVE/DEAD kit quickly discriminates live from dead
spectrophotometer (Infinite 2000). The analyses were performed on a cells by simultaneously staining with green fluorescent calcein-AM to
96-well plate. The excitation parameter was fixed at 524 nm, and indicate intracellular esterase activity and red-fluorescent ethidium
emission spectra were generated at 560−700 nm. The gain value was homodimer-1 to indicate loss of plasma membrane integrity.
kept constant at 120, and three measurements were taken for each However, to carry out these experiments, ethidium was replaced by
sample. DAPI, since f-SiO2NPs, like ethidium homodimer-1, show fluo-

36031 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

rescence in the red region, which could interfere with the result. The the samples were post-fixed with 1% osmium tetroxide, 0.8%
samples were incubated with calcein-AM (1.8 μM) and DAPI (1.8 potassium ferricyanide, 3 mM calcium chloride, and 0.1 M sodium
μM) for 20 min. The reading of fluorescence was carried out in a cacodylate buffer for 30 min on ice. After post-fixation, the sample was
macro-confocal microscope (Leica TCS LSI, Leica). washed with ultrapure water and stained with 2% uranyl acetate
Small Intestine Model Production. Caco-2 and HT-29 cells overnight at 4 °C. The dehydration process was performed in 15−
were cultivated in high-glucose DMEM, supplemented with 10% FBS, 100% ethanol ascending series. Then, the samples were gradually
100 units per mL penicillin, 100 μg mL−1 streptomycin, and 1% infiltrated in EMbed812 and polymerized at 60 °C for 72 h. Finally,
nonessential amino acids. Cell cultures were incubated at 37 °C in a the samples were cut into ultrathin sections with an ultramicrotome
fully humid, 5% CO2 environment. The medium was renewed every and imaged in a TEM 1400-JEOL (Jeol) operating at 120 kV. The
two or three days. Polycarbonate cell culture inserts were used to microscope is equipped with a OneView CMOS 4 k × 4 k camera
produce a functional intestinal cell barrier model (intestine (GATAN) for digital image acquisition.
equivalent), with a 0.40 μm pore size and an area of 0.60 cm2 EDS. The samples prepared for TEM imaging were also evaluated
(Millicell, Millipore, Merck KGaA). The inserts were integrated in a for their chemical composition using EDS. The images were obtained
24-well cell culture plate (Sarstedt) with 800 μL of DMEM per well in in a double-beam electron microscope Helios Nanolab 660 (Thermo
the bottom compartment (which represents the human intestinal Scientific), in which an Oxford Instruments X-Maxn EDS detector is
bloodstream). To produce the intestine equivalent, 2.50 × 105 cells in installed. For the analysis, an acceleration voltage of 10 kV, a current
200 μL of DMEM57,58 were seeded in the insert’s upper compartment of 1.6, and a working distance of approx. 4 mm were used.
(which represents the human intestinal lumen), considering the 9:1 Statistical Analysis. The Shapiro−Wilk test evaluated the
ratio of Caco-2:HT-29, corresponding to 2.25 × 105 Caco-2 and 2.5 × normality of data. The Student t-test was used to make comparisons
104 HT-29 per insert. About 21 days are required for Caco-2 cells to between two sets of data with a normal distribution (parametric data),
fully differentiate and form confluent and tight monolayers in cell while the Mann−Whitney test was used to make comparisons
culture inserts.59 The media were changed every two or three days for between two nonparametric data sets. One-way and two-way analysis
cell differentiation and barrier formation during the culture period. of variance (ANOVA) and Tukey’s test were used to compare three
Trans-epithelial electrical resistance (TEER) was used to further or more parametric data. Differences were considered significant
confirm barrier formation and to test the cell monolayer health and when p < 0.05. All statistical calculations were carried out using the R
confluence, besides the absence of leaks before. The measurements program version 3.6.2 (R Development Core Team, 2019).
were performed every three days with the Millicell ERS-2 meter
(Millipore, Merck KGaA).60 Intestinal models with a TEER value of
150−300 Ω·cm2 were chosen for nanoparticle transport assays.61
Transport of f-SiO2NPs across the Intestinal Epithelium. For

*
ASSOCIATED CONTENT
sı Supporting Information

transport studies, 200 μL nanoparticles (0.5 mg mL−1) in DMEM The Supporting Information is available free of charge at
containing 10% FBS were added to the upper compartment of the https://pubs.acs.org/doi/10.1021/acsami.3c07048.
insert and 800 μL of the medium was added to its bottom
compartment. The analyses were performed at the incubation times Representation of nanoparticles core−shell synthesis;
of 4, 15, and 24 h. The concentration of f-SiO2NPs in samples was nanoparticle characterization in water (By SEM and
measured directly for fluorescence using a Tecan Infinite M200 Pro fluorescence measurements); description of the prep-
multimode reader (Tecan US, Inc, microplate reader). The results aration of the simulated fluids used in the in vitro
were obtained from 5 batches of intestines produced on different digestion experiments; nanoparticle characterization
days, and for each batch, duplicates were made for the incubation submitted to in vitro digestion assays (by TEM and
times of 4, 15, and 24 h. Results are displayed as mean ± standard AFM analyses); viability of Caco-2 and HT-29 cells
deviation (SD). It is noteworthy that TEER measurements were using Alamar Blue and LIVE/DEAD assays; and
performed to test the cell monolayer health after incubation with f-
SiO2NPs.
absorption and transport of NPs across the intestinal
Confocal Microscopy. After 4, 15, and 24 h of incubation with f- epithelium (by SEM, TEM, and EDS analyses) (PDF)
SiO2NPs, the upper and bottom compartments of the insert were
washed three times with PBS 1× and 500 μL of paraformaldehyde
(4.0% w/v) was added on each side for fixing the cells. After 1 h of
fixation, the inserts were again washed with PBS 1× and 500 μL of
■ AUTHOR INFORMATION
Corresponding Author
permeabilization and blocking solution (0.1% (v/v) Triton X-100 and Mateus Borba Cardoso − Institute of Chemistry (IQ),
1.0% (w/v) bovine serum albumin) was added on each side for 1 h. University of Campinas (UNICAMP), Campinas, SP 13083-
Following incubation and washing inserts, the membrane with the 970, Brazil; Brazilian Synchrotron Light Laboratory
cells was cut out, stained with DAPI (2.5 μM, 10 min, Thermo Fisher
Scientific) to mark nucleic acids and phalloidin 488 (0.2 μM, 1 h 30
(LNLS), Brazilian Center for Research in Energy and
min, Thermo Fisher Scientific) to bind actin filaments, and then Materials (CNPEM), Campinas 13083-970, Brazil;
washed with PBS 1×. The cells were observed under a confocal orcid.org/0000-0003-2102-1225; Email: cardosomb@
microscope Leica TCS SP8 from the Brazilian Biosciences National lnls.br
Laboratory - LNBio/CNPEM using lasers at 405 nm for DAPI, at 490
nm for phalloidin 488 and 561 nm for f-SiO2NPs. Authors
SEM. At the end of incubation times, f-SiO2NPs were fixed for 30 Iris Renata Sousa Ribeiro − Institute of Chemistry (IQ),
min with 2.5% glutaraldehyde in cacodylate buffer (0.1 M, pH 7.2− University of Campinas (UNICAMP), Campinas, SP 13083-
7.4) with calcium chloride (3 mM) and rinsed three times with wash 970, Brazil; Brazilian Synchrotron Light Laboratory
buffer for 5 min in an ice bath. Then, the samples were dehydrated (LNLS), Brazilian Center for Research in Energy and
through a series of increasing concentrations (15, 30, 50, 70, 90, and Materials (CNPEM), Campinas 13083-970, Brazil
100%) of ethanol and dried in a desiccator. They were then sputtered
Raquel Frenedoso da Silva − Brazilian Synchrotron Light
with a 5 nm thick gold coat and analyzed in an FEI Inspect
microscope (FEI Company) operated at 10−20 kV. Laboratory (LNLS), Brazilian Center for Research in Energy
TEM. After incubation with f-SiO2NPs, the intestinal epithelium and Materials (CNPEM), Campinas 13083-970, Brazil
samples were fixed for 30 min with 2.5% glutaraldehyde in cacodylate Renata Santos Rabelo − Brazilian Synchrotron Light
buffer (0.1 M, pH 7.2−7.4) with calcium chloride (3 mM) and rinsed Laboratory (LNLS), Brazilian Center for Research in Energy
three times with wash buffer for 5 min in an ice bath. Subsequently, and Materials (CNPEM), Campinas 13083-970, Brazil
36032 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

Talita Miguel Marin − Brazilian Biosciences National Nanoparticles Targeted to the Site of Infection Enhance Antibacterial
Laboratory (LNBio), Brazilian Center for Research in Efficacy. Nat. Biomed. Eng. 2018, 2, 95−103.
Energy and Materials (CNPEM), Campinas 13083-970, (6) Capeletti, L. B.; Oliveira, J. F. A.; Loiola, L. M. D.; Galdino, F.
Brazil; Department of Biochemistry and Tissue Biology, E.; Santos, D. E. S.; Soares, T. A.; Freitas, R. O.; Cardoso, M. B.
Gram-Negative Bacteria Targeting Mediated by Carbohydrate−
Institute of Biology, University of Campinas (UNICAMP),
Carbohydrate Interactions Induced by Surface-Modified Nano-
Campinas, SP 13083-970, Brazil particles. Adv. Funct. Mater. 2019, 29, No. 1904216.
Jefferson Bettini − Brazilian Nanotechnology National (7) Mitchell, M. J.; Billingsley, M. M.; Haley, R. M.; Wechsler, M. E.;
Laboratory (LNNano), Brazilian Center for Research in Peppas, N. A.; Langer, R. Engineering Precision Nanoparticles for
Energy and Materials (CNPEM), Campinas 13083-970, Drug Delivery. Nat. Rev. Drug Discovery 2021, 20, 101−124.
Brazil (8) Jing, Z.; Du, Q.; Zhang, X.; Zhang, Y. Nanomedicines and
Complete contact information is available at: Nanomaterials for Cancer Therapy: Progress, Challenge and
Perspectives. Chem. Eng. J. 2022, 446, No. 137147.
https://pubs.acs.org/10.1021/acsami.3c07048
(9) García-Fernández, A.; Aznar, E.; Martínez-Máñez, R.; Sancenón,
F. New Advances in In Vivo Applications of Gated Mesoporous Silica
Author Contributions as Drug Delivery Nanocarriers. Small 2020, 16, No. 1902242.
M.B.C. directed the research. M.B.C., I.R.S.R., and R.F.S. (10) Liu, X.; Tang, I.; Wainberg, Z. A.; Meng, H. Safety
designed the experiments. I.R.S.R. carried out all of the Considerations of Cancer Nanomedicine�A Key Step toward
nanoparticle synthesis and characterization tests. I.R.S.R. and Translation. Small 2020, 16, No. 2000673.
R.F.S. performed all of the biological tests. T.M.M. (11) de Oliveira, J. F. A.; Scheffer, F. R.; Landis, R. F.; Neto, É . N.;
collaborated with the preparation of the intestinal epithelium Rotello, V. M.; Cardoso, M. B. Dual Functionalization of Nano-
model. R.S.R. processed the samples for analysis by TEM, particles for Generating Corona-Free and Noncytotoxic Silica
Nanoparticles. ACS Appl. Mater. Interfaces 2018, 10, 41917−41923.
SEM, and AFM. J.B. performed the TEM and EFTEM analyses (12) D’Mello, S. R.; Cruz, C. N.; Chen, M. L.; Kapoor, M.; Lee, S.
and processed the images. I.R.S.R., R.F.S., R.S.R., and M.B.C. L.; Tyner, K. M. The Evolving Landscape of Drug Products
analyzed the data and wrote the manuscript with contributions Containing Nanomaterials in the United States. Nat. Nanotechnol.
from all authors. All authors have given approval to the final 2017, 12, 523−529.
version of the manuscript. (13) Fattal, E.; Tsapis, N. Nanomedicine Technology: Current
Notes Achievements and New Trends. Clin. Transl. Imaging 2014, 2, 77−87.
(14) Reinholz, J.; Landfester, K.; Mailänder, V. The Challenges of
The authors declare no competing financial interest. Oral Drug Delivery via Nanocarriers. Drug Delivery 2018, 25, 1694−

■ ACKNOWLEDGMENTS
This work was supported by Fundação de Amparo à Pesquisa
1705.
(15) Kim, K. S.; Suzuki, K.; Cho, H.; Youn, Y. S.; Bae, Y. H. Oral
Nanoparticles Exhibit Specific High-Efficiency Intestinal Uptake and
Lymphatic Transport. ACS Nano 2018, 12, 8893−8900.
do Estado de São Paulo (processes 2015/25406-5, 2017/ (16) Wahlich, J.; Desai, A.; Greco, F.; Hill, K.; Jones, A. T.; Mrsny,
21318-0, and 2021/12071-6). The authors acknowledge ME R. J.; Pasut, G.; Perrie, Y.; Seib, F. P.; Seymour, L. W.; Uchegbu, I. F.
laboratory of Brazilian Nanotechnology National Laboratory Nanomedicines for the Delivery of Biologics. Pharmaceutics 2019, 11,
(LNNano) for the use of electron microscopy facility No. 210.
(proposal TEM-C2-27225, TEM-FT-28036, and SEM-FT- (17) Abeer, M. M.; Rewatkar, P.; Qu, Z.; Talekar, M.; Kleitz, F.;
28100). LNBio and Brazilian Biorenewables National Labo- Schmid, R.; Lindén, M.; Kumeria, T.; Popat, A. Silica Nanoparticles:
ratory (LNBr) are acknowledged for laboratory infrastructure A Promising Platform for Enhanced Oral Delivery of Macromolecules.
for biological experiments and for fluorescence measurements J. Controlled Release 2020, 326, 544−555.
(18) Gupta, R.; Badhe, Y.; Mitragotri, S.; Rai, B. Permeation of
(MAC-27630). The authors thank Nathalia de Carvalho
Nanoparticles across the Intestinal Lipid Membrane: Dependence on
Indolfo for assistance in the preparation of the intestinal Shape and Surface Chemistry Studied through Molecular Simulations.
model and Tiago M. dos Santos for trimming and slicing Nanoscale 2020, 12, 6318−6333.
samples embedded in resin to acquire TEM images. They (19) Lamson, N. G.; Berger, A.; Fein, K. C.; Whitehead, K. A.
acknowledge Carla Pollo and Florian Meneau for ptychog- Anionic Nanoparticles Enable the Oral Delivery of Proteins by
raphy tests, Raul Freitas for nano-IR imaging, and Francesco Enhancing Intestinal Permeability. Nat. Biomed. Eng. 2020, 4, 84−96.
Lena and Helio Tolentino for fluorescent experiments done, (20) Sadeghi, S.; Lee, W. K.; Kong, S. N.; Shetty, A.; Drum, C. L.
respectively, at Cateretê, Imbuia, and Carnaúba beamlines at Oral Administration of Protein Nanoparticles: An Emerging Route to
Sirius-LNLS. Disease Treatment. Pharmacol. Res. 2020, 158, No. 104685.
(21) Wang, L.; Yang, J.; Li, S.; Li, Q.; Liu, S.; Zheng, W.; Jiang, X.

■ REFERENCES
(1) Nance, E. Careers in Nanomedicine and Drug Delivery. Adv.
Oral Administration of Starting Materials for in Vivo Synthesis of
Antibacterial Gold Nanoparticles for Curing Remote Infections. Nano
Lett. 2021, 21, 1124−1131.
Drug Delivery Rev. 2019, 144, 180−189. (22) Hu, S.; Yang, Z.; Wang, S.; Wang, L.; He, Q.; Tang, H.; Ji, P.;
(2) Bobo, D.; Robinson, K. J.; Islam, J.; Thurecht, K. J.; Corrie, S. R. Chen, T. Zwitterionic Polydopamine Modified Nanoparticles as an
Nanoparticle-Based Medicines: A Review of FDA-Approved Materials Efficient Nanoplatform to Overcome Both the Mucus and Epithelial
and Clinical Trials to Date. Pharm. Res. 2016, 33, 2373−2387. Barriers. Chem. Eng. J. 2022, 428, No. 132107.
(3) Lammers, T.; Ferrari, M. The Success of Nanomedicine. Nano (23) Lundquist, P.; Artursson, P. Oral Absorption of Peptides and
Today 2020, 31, No. 100853. Nanoparticles across the Human Intestine: Opportunities, Limitations
(4) MaHam, A.; Tang, Z.; Wu, H.; Wang, J.; Lin, Y. Protein-Based and Studies in Human Tissues. Adv. Drug Delivery Rev. 2016, 106,
Nanomedicine Platforms for Drug Delivery. Small 2009, 5, 1706− 256−276.
1721. (24) Xu, Y.; Shrestha, N.; Préat, V.; Beloqui, A. An Overview of in
(5) Hussain, S.; Joo, J.; Kang, J.; Kim, B.; Braun, G. B.; She, Z. G.; Vitro, Ex Vivo and in Vivo Models for Studying the Transport of
Kim, D.; Mann, A. P.; Mölder, T.; Teesalu, T.; Carnazza, S.; Drugs across Intestinal Barriers. Adv. Drug Delivery Rev. 2021, 175,
Guglielmino, S.; Sailor, M. J.; Ruoslahti, E. Antibiotic-Loaded No. 113795.

36033 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

(25) Shan, W.; Zhu, X.; Tao, W.; Cui, Y.; Liu, M.; Wu, L.; Li, L.; Epithelial Cell Model: Effect of Size and Surface Charge. Mol. Pharm.
Zheng, Y.; Huang, Y. Enhanced Oral Delivery of Protein Drugs Using 2014, 11, 4363−4373.
Zwitterion-Functionalized Nanoparticles to Overcome Both the (40) Zhou, G.; Zhang, J.; Pan, C.; Liu, N.; Wang, Z.; Zhang, J.
Diffusion and Absorption Barriers. ACS Appl. Mater. Interfaces Enhanced Uptake of Fe3O4 Nanoparticles by Intestinal Epithelial
2016, 8, 25444−25453. Cells in a State of Inflammation. Molecules 2017, 22, No. 1240.
(26) Minekus, M.; Alminger, M.; Alvito, P.; Ballance, S.; Bohn, T.; (41) Strugari, A.; Stan, M.; Gharbia, S.; Hermenean, A.; Dinischiotu,
Bourlieu, C.; Carrière, F.; Boutrou, R.; Corredig, M.; Dupont, D.; A. Characterization of Nanoparticle Intestinal Transport Using an In
Dufour, C.; Egger, L.; Golding, M.; Karakaya, S.; Kirkhus, B.; Le Vitro Co-Culture Model. Nanomaterials 2019, 9, No. 5.
Feunteun, S.; Lesmes, U.; Macierzanka, A.; Mackie, A.; Marze, S.; (42) Yang, D.; Liu, D.; Qin, M.; Chen, B.; Song, S.; Dai, W.; Zhang,
McClements, D. J.; Ménard, O.; Recio, I.; Santos, C. N.; Singh, R. P.; H.; Wang, X.; Wang, Y.; He, B.; Tang, X.; Zhang, Q. Intestinal Mucin
Vegarud, G. E.; Wickham, M. S. J.; Weitschies, W.; Brodkorb, A. A Induces More Endocytosis but Less Transcytosis of Nanoparticles
Standardised Static in Vitro Digestion Method Suitable for Food − an across Enterocytes by Triggering Nanoclustering and Strengthening
International Consensus. Food Funct. 2014, 5, 1113−1124. the Retrograde Pathway. ACS Appl. Mater. Interfaces 2018, 10,
(27) Brodkorb, A.; Egger, L.; Alminger, M.; Alvito, P.; Assunçaõ , R.; 11443−11456.
Ballance, S.; Bohn, T.; Bourlieu-Lacanal, C.; Boutrou, R.; Carrière, F.; (43) Gao, Y.; He, Y.; Zhang, H.; Zhang, Y.; Gao, T.; Wang, J. H.;
Clemente, A.; Corredig, M.; Dupont, D.; Dufour, C.; Edwards, C.; Wang, S. Zwitterion-Functionalized Mesoporous Silica Nanoparticles
Golding, M.; Karakaya, S.; Kirkhus, B.; Le Feunteun, S.; Lesmes, U.; for Enhancing Oral Delivery of Protein Drugs by Overcoming
Macierzanka, A.; Mackie, A. R.; Martins, C.; Marze, S.; McClements, Multiple Gastrointestinal Barriers. J. Colloid Interface Sci. 2021, 582,
D. J.; Ménard, O.; Minekus, M.; Portmann, R.; Santos, C. N.; 364−375.
Souchon, I.; Singh, R. P.; Vegarud, G. E.; Wickham, M. S. J.; (44) Martins, J. P.; Figueiredo, P.; Wang, S.; Espo, E.; Celi, E.;
Weitschies, W.; Recio, I. INFOGEST Static in Vitro Simulation of Martins, B.; Kemell, M.; Moslova, K.; Mäkilä, E.; Salonen, J.;
Gastrointestinal Food Digestion. Nat. Protoc. 2019, 14, 991−1014. Kostiainen, M. A.; Celia, C.; Cerullo, V.; Viitala, T.; Sarmento, B.;
(28) Hilgendorf, C.; Spahn-Langguth, H.; Regårdh, C. G.; Lipka, E.; Hirvonen, J.; Santos, H. A. Neonatal Fc Receptor-Targeted Lignin-
Amidon, G. L.; Langguth, P. Caco-2 versus Caco-2/HT29-MTX Co- Encapsulated Porous Silicon Nanoparticles for Enhanced Cellular
cultured Cell Lines: Permeabilities Via Diffusion, Inside- and Outside- Interactions and Insulin Permeation across the Intestinal Epithelium.
Directed Carrier-Mediated Transport. J. Pharm. Sci. 2000, 89, 63−75. Bioact. Mater. 2022, 9, 299−315.
(29) Mahler, G. J.; Shuler, M. L.; Glahn, R. P. Characterization of (45) Abdelkhaliq, A.; van der Zande, M.; Undas, A. K.; Peters, R. J.
Caco-2 and HT29-MTX Cocultures in an in Vitro Digestion/Cell B.; Bouwmeester, H. Impact of in Vitro Digestion on Gastrointestinal
Fate and Uptake of Silver Nanoparticles with Different Surface
Culture Model Used to Predict Iron Bioavailability☆. J. Nutr.
Modifications. Nanotoxicology 2020, 14, 111−126.
Biochem. 2009, 20, 494−502.
(46) Li, Z.; Mu, Y.; Peng, C.; Lavin, M. F.; Shao, H.; Du, Z.
(30) Chen, X.-M.; Elisia, I.; Kitts, D. D. Defining Conditions for the
Understanding the Mechanisms of Silica Nanoparticles for Nano-
Co-Culture of Caco-2 and HT29-MTX Cells Using Taguchi Design.
medicine. WIREs Nanomed. Nanobiotechnol. 2021, 13, No. e1658.
J. Pharmacol. Toxicol. Methods 2010, 61, 334−342.
(47) Ow, H.; Larson, D. R.; Srivastava, M.; Baird, B. A.; Webb, W.
(31) Béduneau, A.; Tempesta, C.; Fimbel, S.; Pellequer, Y.; Jannin,
W.; Wiesner, U. Bright and Stable Core − Shell Fluorescent Silica
V.; Demarne, F.; Lamprecht, A. A Tunable Caco-2/HT29-MTX Co-
Nanoparticles. Nano Lett. 2005, 5, 113−117.
Culture Model Mimicking Variable Permeabilities of the Human (48) Mackie, A.; Rigby, N. InfoGest Consensus Method. In The
Intestine Obtained by an Original Seeding Procedure. Eur. J. Pharm. Impact of Food Bioactives on Health: In Vitro and Ex Vivo Models;
Biopharm. 2014, 87, 290−298. Springer International Publishing, 2015; pp 13−22.
(32) Pan, F.; Han, L.; Zhang, Y.; Yu, Y.; Liu, J. Optimization of (49) Peters, R.; Kramer, E.; Oomen, A. G.; Rivera, Z. E. H.; Oegema,
Caco-2 and HT29 Co-Culture in Vitro Cell Models for Permeability G.; Tromp, P. C.; Fokkink, R.; Rietveld, A.; Marvin, H. J. P.; Weigel,
Studies. Int. J. Food Sci. Nutr. 2015, 66, 680−685. S.; Peijnenburg, A. A. C. M.; Bouwmeester, H. Presence of Nano-
(33) Banerjee, A.; Qi, J.; Gogoi, R.; Wong, J.; Mitragotri, S. Role of Sized Silica during in Vitro Digestion of Foods Containing Silica as a
Nanoparticle Size, Shape and Surface Chemistry in Oral Drug Food Additive. ACS Nano 2012, 6, 2441−2451.
Delivery. J. Controlled Release 2016, 238, 176−185. (50) Metin, C. O.; Lake, L. W.; Miranda, C. R.; Nguyen, Q. P.
(34) Lozoya-Agullo, I.; Araújo, F.; González-Á lvarez, I.; Merino- Stability of Aqueous Silica Nanoparticle Dispersions. J. Nanopart. Res.
Sanjuán, M.; González-Á lvarez, M.; Bermejo, M.; Sarmento, B. 2011, 13, 839−850.
Usefulness of Caco-2/HT29-MTX and Caco-2/HT29-MTX/Raji B (51) Galdino, F. E.; Picco, A. S.; Capeletti, L. B.; Bettini, J.; Cardoso,
Coculture Models To Predict Intestinal and Colonic Permeability M. B. Inside the Protein Corona: From Binding Parameters to
Compared to Caco-2 Monoculture. Mol. Pharm. 2017, 14, 1264− Unstained Hard and Soft Coronas Visualization. Nano Lett. 2021, 21,
1270. 8250−8257.
(35) Akbari, A.; Lavasanifar, A.; Wu, J. Interaction of Cruciferin- (52) International Organization for Standardization, ISO 10993-5
Based Nanoparticles with Caco-2 Cells and Caco-2/HT29-MTX Co- Biological Evaluation of Medical Devices -Part 5: Tests for in Vitro
Cultures. Acta Biomater. 2017, 64, 249−258. Cytotoxicity; (Revision of ISO 10993−5: 2009), 2006.
(36) Ye, D.; Bramini, M.; Hristov, D. R.; Wan, S.; Salvati, A.; Åberg, (53) Ahlinder, L.; Ekstrand-Hammarström, B.; Geladi, P.; Ö sterlund,
C.; Dawson, K. A. Low Uptake of Silica Nanoparticles in Caco-2 L. Large Uptake of Titania and Iron Oxide Nanoparticles in the
Intestinal Epithelial Barriers. Beilstein J. Nanotechnol. 2017, 8, 1396− Nucleus of Lung Epithelial Cells as Measured by Raman Imaging and
1406. Multivariate Classification. Biophys. J. 2013, 105, 310−319.
(37) Bao, C.; Liu, B.; Li, B.; Chai, J.; Zhang, L.; Jiao, L.; Li, D.; Yu, (54) Kusi-Appiah, A. E.; Mastronardi, M. L.; Qian, C.; Chen, K. K.;
Z.; Ren, F.; Shi, X.; Li, Y. Enhanced Transport of Shape and Rigidity- Ghazanfari, L.; Prommapan, P.; Kübel, C.; Ozin, G. A.; Lenhert, S.
Tuned α-Lactalbumin Nanotubes across Intestinal Mucus and Enhanced Cellular Uptake of Size-Separated Lipophilic Silicon
Cellular Barriers. Nano Lett. 2020, 20, 1352−1361. Nanoparticles. Sci. Rep. 2017, 7, No. 43731.
(38) Liu, Y.; Shen, J.; Shi, J.; Gu, X.; Chen, H.; Wang, X.; Wang, L.; (55) Malatesta, M. Transmission Electron Microscopy for Nano-
Wang, P.; Hou, X.; He, Y.; Zhu, C.; Wang, Z.; Guo, T.; Guo, S.; Feng, medicine: Novel Applications for Long-Established Techniques Eur. J.
N. Functional Polymeric Core−Shell Hybrid Nanoparticles Over- Histochem. 2016; Vol. 60 DOI: 10.4081/ejh.2016.2751.
come Intestinal Barriers and Inhibit Breast Cancer Metastasis. Chem. (56) Larson, D. R.; Ow, H.; Vishwasrao, H. D.; Heikal, A. A.;
Eng. J. 2022, 427, No. 131742. Wiesner, U.; Webb, W. W. Silica Nanoparticle Architecture
(39) Bannunah, A. M.; Vllasaliu, D.; Lord, J.; Stolnik, S. Mechanisms Determines Radiative Properties of Encapsulated Fluorophores.
of Nanoparticle Internalization and Transport Across an Intestinal Chem. Mater. 2008, 20, 2677−2684.

36034 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035
ACS Applied Materials & Interfaces www.acsami.org Research Article

(57) Marin, T. M.; de Carvalho Indolfo, N.; Rocco, S. A.; Basei, F.


L.; de Carvalho, M.; de Almeida Gonçalves, K.; Pagani, E.
Acetaminophen Absorption and Metabolism in an Intestine/Liver
Microphysiological System. Chem. - Biol. Interact. 2019, 299, 59−76.
(58) Cadena-Herrera, D.; Esparza-De Lara, J. E.; Ramírez-Ibañez, N.
D.; López-Morales, C. A.; Pérez, N. O.; Flores-Ortiz, L. F.; Medina-
Rivero, E. Validation of Three Viable-Cell Counting Methods:
Manual, Semi-Automated, and Automated. Biotechnol. Rep. 2015, 7,
9−16.
(59) Hubatsch, I.; Ragnarsson, E. G. E.; Artursson, P. Determination
of Drug Permeability and Prediction of Drug Absorption in Caco-2
Monolayers. Nat. Protoc. 2007, 2, 2111−2119.
(60) Millipore M. User Guide Millicell ERS-2 Electrical Resistance
System 2016.
(61) Srinivasan, B.; Kolli, A. R.; Esch, M. B.; Abaci, H. E.; Shuler, M.
L.; Hickman, J. J. TEER Measurement Techniques for In Vitro Barrier
Model Systems. SLAS Technol. 2015, 20, 107−126.

Recommended by ACS
Mechanisms of Nanoparticle Transport across Intestinal
Tissue: An Oral Delivery Perspective
Sarfaraz Ahmad Ejazi, Katharina Maisel, et al.
JULY 06, 2023
ACS NANO READ

Biopharmaceutical Modeling of Food Effect─Exploring the


Role of Dietary Fat
Hariharan Ramasubramanian and Steven Castleberry
MARCH 31, 2023
MOLECULAR PHARMACEUTICS READ

Mucus Penetration of Surface-Engineered Nanoparticles in


Various pH Microenvironments
Yiyang Guo, Bingbing Sun, et al.
JANUARY 31, 2023
ACS NANO READ

Influence of Pore Size and Surface Functionalization of


Mesoporous Silica Nanoparticles on the Solubility and
Antioxidant Activity of Confined Coenzyme Q10
Anand Kumar Meka, Amirali Popat, et al.
MAY 22, 2023
MOLECULAR PHARMACEUTICS READ

Get More Suggestions >

36035 https://doi.org/10.1021/acsami.3c07048
ACS Appl. Mater. Interfaces 2023, 15, 36025−36035

You might also like