1. Introduction
Oral dosing is the most common route of administration for human therapeutics, accounting for over 90% of all drug formulations[
1]. For systemically active pharmaceuticals, orally delivered dosage forms must be absorbed by the gastrointestinal (GI) tract to exert their clinical effects at the target site. Disintegration, dissolution, and permeation are three key processes involved in the GI absorption of solid dosage forms[
2,
3]. Solubilized molecules must pass through the single layer of enterocytes lining the gut lumen to be absorbed into the portal circulation. Thus, the intestinal epithelium serves both as an absorptive surface and an absorption barrier to the systemic entry of therapeutic drugs. Barrier functions of the enterocytes include metabolizing enzymes within the cells, membrane efflux transporters, and tight junctions (TJs); the latter consisting of extracellular folds of the transmembrane proteins and multiprotein junctional complexes that form pore-like structures constraining movement across the intercellular spaces[
4] (
Figure 1).
Movement of a drug (rate of flux) in a diffusion cell system from a donor compartment to a receiver compartment can be used to obtain a mathematical estimate of the apparent permeability (
Papp) (
Figure 2). The
Papp estimate, which is a function of both passive and active transport mechanisms, can be derived using a variety of
in vitro systems[
5]. Excluding the possibility of drug metabolism within the enterocyte, drug absorption can be evaluated within monolayer cells systems such as those associated with the parallel artificial membrane permeability assay (PAMPA), which typically reflects passive permeability only [
6], the human colon adenocarcinoma (Caco-2) cell line [
7,
8,
9,
10], and more recently intestinal organoids [
11].
Mathematically, the
in vitro Papp can be used to estimate
in vivo effective permeability (
Peff) by incorporating information on the fraction of drug molecules existing in their neutral form at a given pH, the surface area for absorption, the fraction of unbound drug in the unstirred boundary layer and the drug permeability across the unstirred boundary layer[
12]. Determination of a drug
Papp early within the drug product development process is critical for predicting possible challenges with oral bioavailability and setting up corresponding formulation strategies that can circumvent these issues. Cell culture systems,
e.g., Caco-2, are frequently employed to determine oral drug permeability and are currently considered the gold standard for estimating
in vitro intestinal permeability and oral absorption of candidate therapeutics[
13]. The Caco-2 cell line forms monolayers that both morphologically and functionally resemble the small intestinal (absorptive) enterocytes[
14].
One of the reasons for the extensive use of the Caco-2 assay lies in the versatility of the cell line, allowing for the study of passive diffusion processes, active drug transport, and pre-systemic drug metabolism[
15]. Caco-2 cells spontaneously differentiate into mature small intestinal enterocytes that express morphological (polarized columnar epithelium) and functional features such as intercellular TJs, efflux [
e.g., P-glycoprotein (P-gp) and member of the Multidrug Resistance-Associated Protein (MRP) family] and influx [
e.g., the Organic Anion Transporting Protein (OATP)] transporters, and the presence of enzymes such as the cytochrome P450 (CYP) isoenzymes[
15,
16,
17]. While Caco-2 cells are widely used in the pharmaceutical industry and are accepted by regulatory agencies to predict human intestinal drug permeability, caution should be exercised when extrapolating data from these
in vitro models to
in vivo physiology[
18,
19]. More specifically, in addition to issues outlined above, there have been discrepancies in the expression of influx transporters in Caco-2 cells between studies conducted in different laboratories[
20,
21,
22,
23,
24]. Additionally, one disadvantage of Caco-2 cells is that they only represent one cell type from the epithelial layer of the small intestine[
25]. Specifically, the absence of goblet cells, which are responsible for mucus production, makes it impossible to evaluate mucus-drug interactions [
26]. The presence of mucus from goblet cells represents a physiological barrier that drugs cross to enter the intestinal enterocytes. It should also be noted that Caco-2 cultures lack expression of numerous key nuclear receptors normally found in the intestine, including the pregnane X receptor (PXR), steroid X receptor (SXR), and constitutive androstane receptor (CAR)[
27]. As a result, Caco-2 cultures are unable to simulate the induction of drug transporters and enzymes by certain drugs (
e.g., rifampin) that interact with these receptors[
28].
Research in biomedical science, disease modeling, and personalized medicine has advanced since Sato
et al. (2009) first reported the
in vitro culture of intestinal organoids[
19,
29,
30,
31,
32,
33,
34,
35,
36,
37,
38,
39]. Three-dimensional (3D) organoids are derived from leucine-rich repeat containing G protein-coupled receptor 5 (Lgr5)-positive adult stem cells [
35,
38,
39]. Their 3D structure enables organoids to morphologically, physiologically, and structurally mimic endogenous epithelia. Therefore, these systems provide an opportunity to evaluate the transport and intestinal metabolism of administered orally therapeutic drugs [
19]. Although human adult-stem cell derived intestinal organoids are now used as intestinal permeability models[
40,
41], canine organoids present a promising alternative to human organoids due to ethical constraints related to research on human stem cells[
42] and the lack of availability of large human organoid bioarchives.
Canine-specific permeability tools are also needed for assessing the unique absorption challenges associated with the canine GI tract. In this regard, although Caco-2 cells are commonly used to evaluate human drug permeability, their predictive performances in modeling Papp in dogs remain to be demonstrated. The use of canine intestinal organoids in permeability studies may be more accurate in predicting canine, not human, intestinal permeability and metabolism of small pharmacological molecules as compared to that derived using Caco-2 or Madin-Darby Canine Kidney (MDCK) cells, with a potential to avoid the ethical and financial constraints associated with the use of live animal models. At last, the possibility of re-using organoids preserved in a biorepository further supports the 3R “Replacement, Reduction, and Refinement” initiative.
The potential utility of organoid systems is gaining recognition within pharmaceutical sciences, with examples being applied to exploring drug screening, cancer therapies, gene therapy, and a host of other kinds of diseases [
43,
44,
45]. Examples of cultured organoid systems include human kidney tubuloids[
46], dog prostate cancer organoids[
47], and human bladder cancer organoids[
29,
48,
49]. Although still early in its development, with many questions and challenges yet to be addressed, there are recent efforts to improve the ability of the 3D organoid to recapitulate
in vivo organ physiology through the development of the organ-on-a-chip technology[
50]. Microfluidic organ-on-chips [
50] provide a constant flow of media through the cell culture, while various other physiologically important parameters, such as oxygen saturation and shear stress, can be manipulated and mimicked more closely for physiological representation. However, this technology is still relatively new and bears a number of limitations, such as significant batch-to-batch variability causing a wide range of results for the same parameters [
51,
52]. Also, due to the complexity of the microfluidic system, experiments involving organs-on-a-chip require additional technical skill sets, thereby limiting its broader use in preclinical research [
51,
52].
Whether or not influx and efflux transporters are involved in the transmembrane movements of a drug candidate, it is essential that a model system does not introduce a bias by failure to adequately control passive transport. Accordingly, the first step in characterizing the function of cell monolayers is to assess their passive permeability. Furthermore, predictive performances of the novel canine colonoid model need to be compared with those of the current gold standard Caco-2 cell monolayer to identify possible interspecies differences in drug passive permeability. To meet this objective, the study presented herein used model drugs with well-characterized transcellular absorption (propranolol and metoprolol) and paracellular diffusion (atenolol) attributes. Since this was a proof-of-concept study, this choice was motivated based on the known
in vivo and
in vitro permeability of these drugs. Metoprolol and propranolol are human BCS Class I (high solubility, high permeability) drugs [
53], while atenolol is a BCS Class III drug (low permeability, high solubility) [
54].
In this study, we thoroughly validated the integrity of canine monolayers and their functionality by transepithelial electrical resistance (TEER) measurement and FITC-Dextran assay. We also report expression data for key intestinal epithelial cell markers, tight and
adherens junction proteins, transporters and cytochrome P450 (CYP) enzymes. At last, both the absorptive (apical to basolateral, AP→BL) and secretive (basolateral to apical, BL→AP)
Papp estimates of these model drugs were evaluated in Caco-2 cell and canine colonoid-derived monolayers under various pH conditions intended to reflect the
in vivo intestinal physiology. Our working hypothesis was that the passive permeability of the two transcellularly absorbed molecules would be similar across the two cell systems, but that differences may be observed with atenolol [
55]. Noteworthily, this first proof-of concept study focused on the number of conditions being explored rather than replicates per condition, with the option of expanding the number of replicates for studying a particular condition as needed.
4. Discussion
Identifying the permeability characteristics of an orally administered drug is one of the critical steps in the prediction of GI drug absorption. This information helps identify factors that can influence oral bioavailability and guide formulators in their efforts to optimize the fraction of the administered dose that is absorbed. While in vitro tools for exploring drug permeability and enterocyte drug metabolism are available for human therapeutics, there is no corresponding tool available that faithfully models the canine GI tract. Therefore, we studied the feasibility of using canine colonoid-derived 2D-monolayers for an apparent permeability assessment, focusing first on the passive diffusion of drugs across the canine enterocyte membrane.
Generally, we expect that passive transcellular permeability characteristics are translatable across biological membranes. This assumption is supported by the use of the MDCK cells and Lewis lung carcinoma-porcine kidney cells (LLC-PK
1) for the evaluation of passive absorption mechanisms [
75]. However, differences between cell lines may exist when assessing paracellular absorption [
55,
76]. Therefore, we explored the feasibility of the canine colonoid system using three well-characterized β-blockers with transcellular or paracellular absorption and determined the validity of our assumptions by comparing canine colonoid P
app values to those obtained when the same compounds were tested using the Caco-2 cell system. If differences in P
app estimates between systems were identified, then the use of Caco-2 data to predict canine drug intestinal passive permeability would need to be further examined.
Consistent with our expectations, we observed comparable absorption (AP→BL) and secretion (BL→AP) characteristics across the two systems for propranolol. In contrast, system dissimilarities were observed for metoprolol. For example, while the concentration of metoprolol influenced colonoid Papp values across both AP→BL and BL→AP directions, such differences were not seen when using the Caco-2 monolayer. Furthermore, Papp values in both directions were higher for the Caco-2 monolayer when the donor concentration was 0.4 mg/mL as compared to that of the colonoid. In contrast, when the donor concentration was 0.04 mg/mL, AP→BL (apical pH = 6.8), Papp values were comparable. Only movement in the BL→AP direction was greater for the Caco-2 vs canine colonoid monolayer systems.
Although we assumed that transcellular permeability would be similar across species and tissues, one possible reason for the observed differences between Caco-2 and colonoids could be unanticipated regional differences across the enterocytes lining the various intestinal regions and differential expression of transporters and CYP enzymes. This possibility was explored by Incecayir et al.[
77]
and Zur et al.[
78]. Incecayir et al. estimated metoprolol P
eff values using an in situ single-pass intestinal perfusion system (SPIPS) in mice and/or rats[
77]. They observed that the metoprolol intestinal permeability of both species higher in the distal ileum vs. the jejunum. Recognizing that P
eff values factor surface area into its estimate, this outcome likely reflects segmental surface area differences or dissimilarity in intestinal mucus boundary layers. However, in contrast to the aforementioned rodent study, using a SPIPS study design in human subjects, Dahlgren et al. detected no statistically significant regional differences in metoprolol P
eff in the colon vs. the ileum [
79]. Altogether, these results are not consistent with a possible influence of cellular configuration of the Caco-2 cell vs. canine colonoid.
With regard to potential transporter involvement, Incecayir et al. confirmed that metoprolol is not a substrate for P-gp transport, excluding the notion that observations were due to differences in efflux transporter expression [
77]. Moreover, if P-gp transporters were in fact involved, it would have negatively influenced movement from AP→BL (decrease in P
app values) but positively increased P
app values in the BL→AP direction. This is not consistent with what was observed experimentally, either at a donor concentration of 0.04 mg/mL or for the BL→AP P
app values (colonoid vs. Caco-2 results) at the donor metoprolol concentration of 0.4 mg/mL. Factoring this point along with our correction for drug loss during our study, we can reasonably assume that expression of efflux transporters (or losses associated with enterocyte drug metabolism) cannot explain the differences seen between the two systems.
Another point considered in the SPIPS studies by Incecayir et al.[
77] and Zur et al.[
78] was the potential influence of pH on metoprolol permeability. Although higher pH tended to be associated with higher P
eff values, this difference was not statistically significant in the rat (N=6). In contrast, in the rat SPIPS study conducted by Zur et al. markedly higher P
eff values were observed as perfusate pH was raised from 6.5 to 7.5 (N=6 per experiment) [
78]. In addition, both Incecayir et al.[
77] and Zur et al.[
78] reported a significant pH-associated change in permeability using either the Caco-2 monolayer or octanol/buffer partition coefficient (and PAMPA membrane)[
78]. In all cases, the permeability of metoprolol decreased as the donor pH decreased. The investigators attributed this pH effect to metoprolol being a basic secondary amine that serves as its only ionizable center. As a result, the fraction unionized of metoprolol is negligible at low pH (i.e., at pH values less than the corresponding pKa), and gradually increases as the pH rises. Incecayir et al. suggested that the small effect of pH on in vivo absorption vs. in vitro permeability may have been attributable to the in vivo presence of a mucous layer which retains the microclimate pH, regardless of the luminal pH [
77]. Interestingly, however, we observed the opposite effect of pH on our 0.04 mg/mL metoprolol P
app values where there tended to be a decrease rather than increase as pH was raised from 6.8 to 7.4. Therefore, again, the reasons for our observations with metoprolol are not readily apparent.
Finally, unlike that of the Caco-2 monolayer or colonoid permeability studies conducted at a donor metoprolol concentration of 0.04 mg/mL, FITC-dextran was included when the canine colonoid donor concentrations were 0.4 mg/mL. While this fluorescent probe has not been associated with changes in drug transcellular permeability, we do not have the data to exclude that possibility in our current investigation. Therefore, an impact of this design difference between permeability study conditions cannot be excluded. The permeability findings on Caco-2 monolayers from Day 14 with low TEER value[
54] indicate that the transport of hydrophilic marker (FITC-dextran) and hydrophilic drug (atenolol) was significantly increased, whereas the transport of metoprolol remained unchanged. The findings indicate that a hydrophilic drug such as atenolol can cross the leaky intercellular connection via the paracellular channel. This assessment is consistent with the observations of Yang et al. (2007), where palmitoylcarnitine (PCC), a compound that opens tight junctions, increased the transport of sotalol (a hydrophilic drug) and of FITC-dextran, produced only a small increase in metoprolol transport, but significantly decreased the TEER, the latter indicating a loosening of the tight junction[
54].
Unlike the unanswered questions associated with the metoprolol study results, reasons for the observations with atenolol may be found in the investigation by Dahlgren et al. [
79]. Using a Single-Pass Intestinal Perfusion (SPIP) study design in human subjects, atenolol exhibited >10-fold lower P
eff in the colon as compared to that of the ileum and >350-fold lower P
eff in the colon as compared to that in the jejunum[
79]. This translated to statistically significantly different drug exposure in vivo values (expressed as area under the concentration time-curve, AUC) when doses were administered to the colon vs. ileum. Extrapolating their observations to our study results and recognizing that the P
eff values were influenced by regional differences in absorptive surface area[
80], we cannot exclude the possibility that differences observed between atenolol P
app in colonoids vs. Caco-2 cells may reflect differences in the TJ expression between these two systems, since they represent different intestinal segments showing both enterocyte and colonocyte features[
7,
8]. Under normal culture conditions, Caco-2 cells can spontaneously undergo morphological and biochemical enterocytic differentiation[
7]. The cells become polarized, forming a cell monolayer with apical brush boundary microvilli, tight intercellular junctions, villin expression, and dome formation. When cells approach confluence, the number of proteins characteristic of the colon decreases while the number of proteins characteristic of the enterocytes increases[
7]. Consistent with this interpretation is the results of the TEER values we obtained to determine the integrity of the intercellular junctional complex.
The TEER of the monolayers used to assess pre-assay integrity of the monolayers in the present study is comparable to previous reports in Caco-2 cells (
Figure 5). In the present study, TEER values greater than 500 Ω×cm
2 were required in order for Caco-2 cell monolayers to be considered appropriate for use in the transmembrane transport studies [
62]. Studies have identified that TEER values in the range of 300-600 Ω×cm
2 imply the establishment of robust TJs between cells (which are essential for maintaining good monolayer integrity) [
9,
62,
63]. However, TEER of the Caco-2 cell line is generally higher than that of the in vivo human intestine. Therefore, the passive paracellular pathway in the Caco-2 cells is generally lower than what would be observed in vivo [
10].
Based on their relative permeability to small ions, epithelia can be classified as "leaky" or "tight," as proposed by Machen et al. (1972) [
81] and Fromter and Diamond (1972) [
82], and this designation is still in use at present [
83]. "Leaky" epithelia have higher paracellular small ion permeability than transcellular permeability and low transepithelial resistance, while "tight" epithelia have similar or better transcellular small ion permeability than paracellular small ions with higher transepithelial resistance [
81,
82]. A “leaky” epithelium [
83] has a TEER of less than 100 Ω×cm
2, indicating greater paracellular permeability, whereas a “tight” epithelium [
83] has a TEER of about 2000 Ω×cm
2, indicating lower paracellular permeability [
61,
84]. We observed that monolayers obtained from the canine colonoids have eight times higher TEER values than that of the Caco-2 monolayer, reflecting tighter intercellular junctional complexes than monolayers derived from Caco-2 cells (
Figure 4 and
Figure 5). That observation is consistent with the higher atenolol P
app values observed in Caco-2 monolayer as compared with that of the canine colonoids when the 2 mg/mL atenolol concentrations was used. Regarding the difference in P
app estimates obtained with 2 mg/mL vs. 0.2 mg/mL atenolol concentrations, we cannot determine if some movement did in fact occur across the Caco-2 monolayer when testing 0.2 mg/mL atenolol, because all receiver compartment concentrations were below the analytical limit of quantification. Higher P-gp (Mdr1) expression in colonoid-derived monolayers relative to MDR1 in Caco-2 cell monolayers is also a potential explanation for the absence of quantifiable atenolol concentrations in colonoid-derived receiver compartments as atenolol is a substrate for P-gp. This was evidenced in studies showing that co-administration of P-gp inhibitors such as verapamil and zosuquidar decreased the efflux ratio of atenolol[
85,
86] and that co-administration of another P-gp inhibitor, cyclosporine (a non-specific inhibitor of both efflux and influx transporters) increased the absorption rate of atenolol[
85,
86]. On a final note, we recognize that the P
app values estimated in our study tended to be lower than those reported by others [
7,
8,
10,
54,
87]. This underscores the importance of comparing permeability results obtained with both systems from the same laboratory. In that regard, variations in permeability results have been observed between laboratories and different Caco-2 culture batches [
7,
24]. Possible reasons for this observation are the differences in culture and transport protocols between the different research groups [
7,
42].
Intestinal stem cells (ISCs) can differentiate into progenitor cells that develop into a diverse range of cell lineages. While a variety of secretory cells (including enteroendocrine cells, goblet cells, and tuft cells), and M cells within Peyer's patches are all present in the normal canine large intestine [
88], tuft cells are only infrequently observed [
89]. Therefore, it is important to confirm that the monolayer used in our studies was in fact colonic enterocytes.
In the present study, while the expression of mucin 2 (MUC2), intestinal alkaline phosphatase (ALP) and neurogenin 3 (NEUROG3) in the canine colonoid-derived monolayers validate the differentiation of intestinal epithelial cells (IECs) into different cell subtypes, the expression of olfactomedin 4 (OLFM4), HOP homeobox (HOPX), prominin 1 (PROM1), SRY-box transcription factor 9 (SOX9), and leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) genes suggests the presence of intestinal epithelial stem cells[
35,
38]. PROM1 is a marker for stem cells and early progenitors in the intestine [
90]. Both microvilli and brush border enzyme ALP expression also indicate the presence of differentiated enterocytes[
35] OLFM4 is also highly expressed in the crypt base columnar cells in the colon [
91]. In our previous research, monolayers generated from canine colonoids were also comprehensively studied for differentiated cell lineages displaying both Ki67 expression and the expression of LGR5, an important marker for adult intestinal stem cells. These monolayers showed NEUROG3 and Chromogranin A (CgA) expression further confirming the presence of differentiated neuroendocrine cells. Moreover, monolayers contained epithelia covered with mucus-like substances, indicating mucus production[
36]. The presence of differentiated goblet cells containing mucus visualized on transmission electron microscopy (TEM) and Alcian blue staining in the present study confirm findings from our prior research[
36].
Continuation and expansion of this preliminary work is based upon the importance of using species-specific monolayers to assess factors such as active influx and efflux transporters, as well as intracellular metabolism on intestinal drug permeability. Moreover, this information should be generated across the various intestinal segments. A first step in supporting the use of such systems is their ability to adequately reflect the passive transport properties (transcellular and paracellular) of the enterocyte membrane.
Despite the limited number of replicates per condition, our study succeeded in providing promising results regarding the utility of canine-derived organoid monolayers for species-relevant assessments of drug passive permeability processes. It also highlighted potential sources of error and the challenges remaining to be addressed. Furthermore, can influence the reliable extrapolation of passive permeability estimates from the Caco-2 cell line to that associated with the canine colon. Clearly, these observed differences warrant future investigations.
Figure 1.
Summary diagram of the transport of a therapeutic drug through the intestine facilitated by transcellular transport, carrier-mediated (CM) and passive diffusion pathways such as passive lipoidal diffusion, CM influx, CM efflux, paracellular diffusion, endocytosis, and transcytosis. The figure was produced with BioRender.com.
Figure 1.
Summary diagram of the transport of a therapeutic drug through the intestine facilitated by transcellular transport, carrier-mediated (CM) and passive diffusion pathways such as passive lipoidal diffusion, CM influx, CM efflux, paracellular diffusion, endocytosis, and transcytosis. The figure was produced with BioRender.com.
Figure 2.
Representative diagram of a dual-chamber culture apparatus showing a cell monolayer grown on filter membrane with apical and basolateral chambers and validation of monolayer integrity by transepithelial electrical resistance (TEER) measurement. Absorption is measured by adding drug to the apical chamber and measuring its appearance in the basolateral chamber over time. Conversely, secretion (efflux) is measured by adding drug to the basolateral chamber and measuring its appearance in the apical chamber over time. The figure was produced with BioRender.com.
Figure 2.
Representative diagram of a dual-chamber culture apparatus showing a cell monolayer grown on filter membrane with apical and basolateral chambers and validation of monolayer integrity by transepithelial electrical resistance (TEER) measurement. Absorption is measured by adding drug to the apical chamber and measuring its appearance in the basolateral chamber over time. Conversely, secretion (efflux) is measured by adding drug to the basolateral chamber and measuring its appearance in the apical chamber over time. The figure was produced with BioRender.com.
Figure 3.
Formation of the canine colonoid-derived monolayer. (A) Representative phase-contrast micrograph of a differentiated colonoid on Day 5 used for monolayer preparation. Scale Bar, 500 μm. (B) Representative phase-contrast micrographs of colonoid-derived monolayers on Days 3, 9, and 11 and representative micrographs of eosin-stained colonoid-derived monolayers on Day 11. Scale Bar, as indicated.
Figure 3.
Formation of the canine colonoid-derived monolayer. (A) Representative phase-contrast micrograph of a differentiated colonoid on Day 5 used for monolayer preparation. Scale Bar, 500 μm. (B) Representative phase-contrast micrographs of colonoid-derived monolayers on Days 3, 9, and 11 and representative micrographs of eosin-stained colonoid-derived monolayers on Day 11. Scale Bar, as indicated.
Figure 4.
Evaluation of canine colonoid-derived monolayer integrity. Continuous TEER analysis over 15 days was used to assess the integrity of each colonoid monolayer. Monolayers were prepared for propranolol, atenolol, and metoprolol studies. Values are expressed as the arithmetic mean of the data and one standard deviation of N = 18 monolayers.
Figure 4.
Evaluation of canine colonoid-derived monolayer integrity. Continuous TEER analysis over 15 days was used to assess the integrity of each colonoid monolayer. Monolayers were prepared for propranolol, atenolol, and metoprolol studies. Values are expressed as the arithmetic mean of the data and one standard deviation of N = 18 monolayers.
Figure 5.
Evaluation of Caco-2 cell monolayer integrity. Continuous TEER analysis of the monolayer over 23 days was used to assess the permeability of propranolol, atenolol, and metoprolol. Values are expressed as the arithmetic mean of the data and one standard deviation of N = 12 monolayers.
Figure 5.
Evaluation of Caco-2 cell monolayer integrity. Continuous TEER analysis of the monolayer over 23 days was used to assess the permeability of propranolol, atenolol, and metoprolol. Values are expressed as the arithmetic mean of the data and one standard deviation of N = 12 monolayers.
Figure 7.
Expression of tight and adherens junction proteins, intestinal epithelial cell differentiation and stem cell markers in canine colonoid-derived monolayers. The gene expression results are displayed as normalized Ct (dCt) to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (see
Supplemental Table 4 for Ct values). Values are expressed as the arithmetic mean of the data and one standard deviation of N = 3 monolayers. Cadherin 1 (CDH1); occludin (OCLN); tight junction protein 1 (TJP1); mucin 2 (MUC2); neurogenin 3 (NEUROG3); intestinal alkaline phosphatase (ALP); olfactomedin 4 (OLFM4); HOP homeobox (HOPX); prominin 1 (PROM1); SRY-box transcription factor 9 (SOX9); leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5).
Figure 7.
Expression of tight and adherens junction proteins, intestinal epithelial cell differentiation and stem cell markers in canine colonoid-derived monolayers. The gene expression results are displayed as normalized Ct (dCt) to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (see
Supplemental Table 4 for Ct values). Values are expressed as the arithmetic mean of the data and one standard deviation of N = 3 monolayers. Cadherin 1 (CDH1); occludin (OCLN); tight junction protein 1 (TJP1); mucin 2 (MUC2); neurogenin 3 (NEUROG3); intestinal alkaline phosphatase (ALP); olfactomedin 4 (OLFM4); HOP homeobox (HOPX); prominin 1 (PROM1); SRY-box transcription factor 9 (SOX9); leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5).
Figure 8.
Visualization of goblet cells containing mucus in canine colon tissue and colonoids. (A) Histological images of hematoxylin and eosin (H&E) staining of colon tissue. (B) Alcian blue staining of colon tissue reveal goblet cells containing mucus. (C) The H&E staining of colonoids at 5 days after passage. (D) Colonoids at 5 days after passage with mucus production are shown by Alcian blue staining. (E) A representative transmission electron microscopy (TEM) image of colonoid shows a goblet cell with mucin granules. (F) A TEM image of the intercellular junctional complex in canine colonoid. The microvilli of the canine colonoid are visible at higher magnifications using TEM than with a standard optical light microscope [
74]. (G) A zoom-in that shows a high-power magnification of the red dashed area in “F”.
Figure 8.
Visualization of goblet cells containing mucus in canine colon tissue and colonoids. (A) Histological images of hematoxylin and eosin (H&E) staining of colon tissue. (B) Alcian blue staining of colon tissue reveal goblet cells containing mucus. (C) The H&E staining of colonoids at 5 days after passage. (D) Colonoids at 5 days after passage with mucus production are shown by Alcian blue staining. (E) A representative transmission electron microscopy (TEM) image of colonoid shows a goblet cell with mucin granules. (F) A TEM image of the intercellular junctional complex in canine colonoid. The microvilli of the canine colonoid are visible at higher magnifications using TEM than with a standard optical light microscope [
74]. (G) A zoom-in that shows a high-power magnification of the red dashed area in “F”.
Figure 9.
Comparative expression of transporters in Caco-2-derived and canine colonoid-derived monolayers. Organic anion transporting polypeptide (OATP2B1, human; Oatp2b1, dog); multidrug resistance p-glycoprotein (MDR1, human; Mdr1, dog).
Figure 9.
Comparative expression of transporters in Caco-2-derived and canine colonoid-derived monolayers. Organic anion transporting polypeptide (OATP2B1, human; Oatp2b1, dog); multidrug resistance p-glycoprotein (MDR1, human; Mdr1, dog).
Figure 10.
Comparative expression of CYP enzymes in Caco-2-derived and their orthologs in canine colonoid-derived monolayers. Gene expression results are displayed as normalized Ct (dCt) to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (see
Supplemental Table 4 for Ct values). Values are expressed as the arithmetic mean of the data and one standard deviation of N = 3 monolayers. CYP enzymes in human Caco-2 (CYP3A4, CYP2B6, CYP2C9 and CYP2C19); CYP enzymes in canine colonoids (Cyp3a12, Cyp2b11 and Cyp2c21). Group comparisons were performed with GraphPad Prism 9 using a two-tailed Student’s t-test. P-values < 0.05 (*) were considered as statistically significant for all analyses. **** P < 0.0001. ns = no significance difference.
Figure 10.
Comparative expression of CYP enzymes in Caco-2-derived and their orthologs in canine colonoid-derived monolayers. Gene expression results are displayed as normalized Ct (dCt) to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (see
Supplemental Table 4 for Ct values). Values are expressed as the arithmetic mean of the data and one standard deviation of N = 3 monolayers. CYP enzymes in human Caco-2 (CYP3A4, CYP2B6, CYP2C9 and CYP2C19); CYP enzymes in canine colonoids (Cyp3a12, Cyp2b11 and Cyp2c21). Group comparisons were performed with GraphPad Prism 9 using a two-tailed Student’s t-test. P-values < 0.05 (*) were considered as statistically significant for all analyses. **** P < 0.0001. ns = no significance difference.
Table 1.
Primers used for real-time PCR expression of tight and adherens junction proteins, intestinal epithelial cell markers, transporters, and cytochrome P450 (CYP) enzymes.
Table 1.
Primers used for real-time PCR expression of tight and adherens junction proteins, intestinal epithelial cell markers, transporters, and cytochrome P450 (CYP) enzymes.
Species |
Category |
Gene Full Name |
Symbol |
Forward Primer Sequence (5'-3') |
Reverse Primer Sequence (5'-3') |
Canis lupus familiaris (dog) |
Housekeeping gene |
Glyceraldehyde-3-phosphate dehydrogenase |
GAPDH |
TCAACGGATTTGGCCGTATTGG |
TGAAGGGGTCATTGATGGCG |
Tight and adherens junction proteins |
Cadherin 1 |
CDH1 |
GACCCAGTAACTAACGACG |
CTTCATTCACATCTTCCACG |
Occludin |
OCLN |
CACTACTGTGTGGTGGATCC |
CCTTGTCCCACAATATATTCG |
Tight junction protein 1 |
TJP1 |
GAGGGTGATCAAATTCTCAGG |
CTGATTCTACAATGCGACG |
Intestinal epithelial cell differentiation markers |
Mucin 2 |
MUC2 |
CCTGTGCCCCATATTCTGC |
GAGATGTTGGAATGGATGCC |
Neurogenin 3 |
NEUROG3 |
GAATGCACAACCTCAACTCG |
GTAGAGGCTGTGGTCCGC |
Intestinal alkaline phosphatase |
ALP |
CGTAGTAAACCGCAACTGG |
GGAAACATGTACTTTCGGC |
Stem cell markers |
Olfactomedin 4 |
OLFM4 |
GTATCATGAATGTCAGCAAGC |
CTGTAATATTCCAGAATTCTTCC |
HOP homeobox |
HOPX |
GACCAGGTGGAGATTCTGG |
GCCAGACGCTGCTTAAACC |
Prominin 1 |
PROM1 |
GATTATTATTTGTGCTGTCC |
GAGACTGTAAAGTATTTCCTC |
SRY-box transcription factor 9 |
SOX9 |
GTCATCTCCAACATAGAGACC |
CTGCTTGGACATCCACACG |
Leucine-rich repeat-containing G protein-coupled receptor 5 |
LGR5 |
GCTAGATCTGTCTTACAACC |
GTTCCAGGCTAAATTCAGC |
Transporters |
Organic anion transporting polypeptide |
Oatp2b1 |
GATGACTTTGCCCACAACAGC |
CAGCAGCAGAGATGAGGAAGC |
Multidrug resistance p-glycoprotein |
Mdr1 |
GTAGCTGAAGAAGTCTTAGCAGC |
GCGGCACCAATAGAAATGTTGGC |
Cytochrome P450 (CYP) enzymes |
Cytochrome P-450 3a12 |
Cyp3a12 |
GATCATGAACATGAAACTTGC |
CTTTTCAGGTTGAATAATCCC |
Cytochrome P450 2b11 |
Cyp2b11 |
CTGAGGGAGTCCTCCAGGACCC |
CACATAGAACAAGTTCATCAGG |
Cytochrome P450 2C21 |
Cyp2c21 (Cyp2c18) |
CAAGCACCTCCTGGATACAGC |
CTTCGTGTTCTTTTATTTTTTCC |
Homo sapiens (human) |
Housekeeping gene |
Glyceraldehyde-3-phosphate dehydrogenase |
GAPDH |
TGCACCACCAACTGCTTAGC |
GGCATGGACTGTGGTCATGAG |
Transporters |
Organic anion transporting polypeptide 2B1/ solute carrier organic anion transporter family member 2B1 |
OATP2B1 (SLCO2B1) |
CAAACCTGACTGTGATCCAG |
GAGCAGGTTGGCGTATGAGG |
ATP binding cassette subfamily B member 1 |
ABCB1 (MDR1) |
CAGTAGCTGAAGAGGTCTTGGC |
CTGTAATAGCTTTCTTTATCCC |
Cytochrome P450 (CYP) enzymes |
Cytochrome P450 family 3 subfamily A member 4 |
CYP3A4 |
GAGATGGTCCCTATCATTGCC |
GATGTTCACTCCAAATGATGTGC |
Cytochrome P450 family 2 subfamily B member 6 |
CYP2B6 |
GAAACCGCTGGAAGGTGCTTCG |
CTCCTCTATCAGACACTGAGC |
Cytochrome P450 family 2 subfamily C member 9 |
CYP2C9 |
GAAGGAGATCCGGCGTTTCTCC |
CTTGGTTTTTCTCAACTCCTCC |
Cytochrome P450 family 2 subfamily C member 19 |
CYP2C19 |
GATCTGCTCCATTATTTTCC |
GTTTTTAAGTAATTTGTTATGG |
Table 2.
Physicochemical characteristics of metoprolol, atenolol, and propranolol.
Table 2.
Physicochemical characteristics of metoprolol, atenolol, and propranolol.
Drug |
Molecular Formula (MF) |
Molecular Weight (MW) |
Aqueous solubility (25°C) |
Log P |
Dissociation Constants (Basic pKa) |
Metoprolol |
C15H25NO3
|
267.36 g/mol |
>1000 mg/mL |
2.15 |
9.56 |
Atenolol |
C14H22N2O3
|
266.34 g/mol |
13.3 mg/mL |
0.16 |
9.58 |
Propranolol |
C16H21NO2
|
259.339 g/mol |
0.0617 mg/L |
3.48 |
9.53 |
Table 3.
Transport of FITC–dextran (expressed in percentage) across the colonoid-derived monolayer. Values are expressed as the arithmetic mean of the data and one standard deviation of 3 monolayers.
Table 3.
Transport of FITC–dextran (expressed in percentage) across the colonoid-derived monolayer. Values are expressed as the arithmetic mean of the data and one standard deviation of 3 monolayers.
Compound |
Time (min) |
Donor (%) |
Receiver (%) |
FITC-Dextran (200 µg/mL) |
0 |
100 ± 0.0 |
0.020 ± 0.001 |
|
15 |
|
0.021 ± 0.003 |
|
30 |
|
0.024 ± 0.004 |
|
45 |
|
0.022 ± 0.002 |
|
60 |
|
0.024 ± 0.003 |
|
90 |
|
0.023 ± 0.004 |
|
120 |
93.7 ± 7.8 |
0.022 ± 0.002 |
Metoprolol (0.4 mg/mL) + |
0 |
100 ± 0.0 |
0.021 ± 0.001 |
FITC-Dextran (200 µg/mL) |
15 |
|
0.023 ± 0.001 |
|
30 |
|
0.020 ± 0.00 |
|
45 |
|
0.020 ± 0.00 |
|
60 |
|
0.022 ± 0.001 |
|
90 |
|
0.023 ± 0.00 |
|
120 |
94.6 ± 1.4 |
0.020 ± 0.001 |
Table 4.
Adjusted Papp value estimates as a function of drug/concentration, direction, experimental pH and the cell culture system. BLQ = below the analytical limit of quantification. *Metoprolol (0.4 mg/mL) ± FITC-dextran (200 µg/mL) testing was exclusively performed in canine colonoid monolayers. Values presented herein were aggregated from both Metoprolol (0.4 mg/mL) ± FITC-dextran (200 µg/mL) experiments. Transport of metoprolol, atenolol and propranolol across the Caco-2-derived (human) and canine colonoid-derived (dog) monolayers. BL→AP and AP→BL transports of the drugs were studied in two different pH conditions i.e., 6.8 and 7.4 (AP site). Two different monolayers (N = 2) were used for each system for specific directions and pH conditions (for dog: AP→BL, pH 6.8/7.4 or BL→AP, pH 7.4/7.4 or AP→BL, pH 7.4/7.4 and for human: AP→BL, pH 6.8/7.4 or BL→AP, pH 7.4/7.4). In total, 20 Caco-2 monolayers and 34 colonoid monolayers were used throughout the study.
Table 4.
Adjusted Papp value estimates as a function of drug/concentration, direction, experimental pH and the cell culture system. BLQ = below the analytical limit of quantification. *Metoprolol (0.4 mg/mL) ± FITC-dextran (200 µg/mL) testing was exclusively performed in canine colonoid monolayers. Values presented herein were aggregated from both Metoprolol (0.4 mg/mL) ± FITC-dextran (200 µg/mL) experiments. Transport of metoprolol, atenolol and propranolol across the Caco-2-derived (human) and canine colonoid-derived (dog) monolayers. BL→AP and AP→BL transports of the drugs were studied in two different pH conditions i.e., 6.8 and 7.4 (AP site). Two different monolayers (N = 2) were used for each system for specific directions and pH conditions (for dog: AP→BL, pH 6.8/7.4 or BL→AP, pH 7.4/7.4 or AP→BL, pH 7.4/7.4 and for human: AP→BL, pH 6.8/7.4 or BL→AP, pH 7.4/7.4). In total, 20 Caco-2 monolayers and 34 colonoid monolayers were used throughout the study.
Drug |
Species |
Direction |
Well |
Papp × 10-6
|
Avg |
SD |
%CV |
Metoprolol (0.4 mg/mL or 584.1 μM) |
Human |
AP→BL pH 6.8/7.4 |
1 |
19.34 |
|
|
|
2 |
16.45 |
17.89 |
2.05 |
11.43 |
BL→AP pH 7.4/7.4 |
1 |
13.79 |
|
|
|
2 |
12.37 |
13.08 |
1.01 |
7.72 |
Dog* |
AP→BL pH 6.8/7.4 |
1 |
8.33 |
|
|
|
2 |
9.10 |
8.72 |
0.55 |
6.32 |
AP→BL pH 7.4/7.4 |
1 |
11.19 |
|
|
|
2 |
9.90 |
10.54 |
0.91 |
8.66 |
BL→AP pH 7.4/7.4 |
1 |
9.88 |
|
|
|
2 |
9.94 |
9.91 |
0.05 |
0.46 |
Metoprolol (0.04 mg/mL or 58.4 μM) |
Human |
AP→BL pH 6.8/7.4 |
1 |
17.54 |
|
|
|
2 |
13.13 |
15.33 |
3.12 |
20.38 |
BL→AP pH 7.4/7.4 |
1 |
16.30 |
|
|
|
2 |
19.03 |
17.67 |
1.93 |
10.92 |
Dog |
AP→BL pH 6.8/7.4 |
1 |
16.83 |
|
|
|
2 |
18.52 |
17.68 |
1.19 |
6.75 |
AP→BL pH 7.4/7.4 |
1 |
13.44 |
|
|
|
2 |
13.50 |
13.47 |
0.04 |
0.32 |
BL→AP pH 7.4/7.4 |
1 |
11.19 |
|
|
|
2 |
12.67 |
11.93 |
1.04 |
8.76 |
Atenolol (0.2 mg/mL or 750.9 μM) |
Human |
AP→BL pH 6.8/7.4 |
1 |
BLQ |
|
|
|
2 |
BLQ |
|
|
|
BL→AP pH 7.4/7.4 |
1 |
BLQ |
|
|
|
2 |
BLQ |
|
|
|
Dog |
AP→BL pH 6.8/7.4 |
1 |
BLQ |
|
|
|
2 |
BLQ |
|
|
|
AP→BL pH 7.4/7.4 |
1 |
BLQ |
|
|
|
2 |
BLQ |
|
|
|
BL→AP pH 7.4/7.4 |
1 |
BLQ |
|
|
|
2 |
BLQ |
|
|
|
Atenolol (2 mg/mL or 7,509 μM) |
Human |
AP→BL pH 6.8/7.4 |
1 |
7.25 |
|
|
|
2 |
8.01 |
7.63 |
0.54 |
7.09 |
BL→AP pH 7.4/7.4 |
1 |
3.69 |
|
|
|
2 |
4.97 |
4.33 |
0.90 |
20.89 |
Dog |
AP→BL pH 6.8/7.4 |
1 |
BLQ |
|
|
|
2 |
BLQ |
|
|
|
AP→BL pH 7.4/7.4 |
1 |
BLQ |
|
|
|
2 |
BLQ |
|
|
|
BL→AP pH 7.4/7.4 |
1 |
BLQ |
|
|
|
2 |
BLQ |
|
|
|
Propranolol (1 mg/mL or 3,380 μM) |
Human |
AP→BL pH 6.8/7.4 |
1 |
12.12 |
|
|
|
2 |
12.75 |
12.44 |
0.45 |
3.59 |
BL→AP pH 7.4/7.4 |
1 |
11.45 |
|
|
|
2 |
12.43 |
11.94 |
0.70 |
5.84 |
Dog |
AP→BL pH 6.8/7.4 |
1 |
12.23 |
|
|
|
2 |
11.05 |
11.64 |
0.83 |
7.15 |
AP→BL pH 7.4/7.4 |
1 |
11.99 |
|
|
|
2 |
11.97 |
11.98 |
0.01 |
0.09 |
BL→AP pH 7.4/7.4 |
1 |
10.94 |
|
|
|
2 |
12.03 |
11.49 |
0.77 |
6.70 |