throbber
Biochemical Pharmacology, Vol. 41, No. 12, pp, 1879-1886, 1991
`Printed in Great Britain.
`
`0006-2952/91 $3.00 + 0.00
`Pergamon Press pic
`
`STIMULATION OF SECRETION BY THE T,, COLONIC
`EPITHELIAL CELL LINE WITH DIETARY FLAVONOLS
`
`TOAN D. NGUYEN,* ANDREW T, CANADA,t GREGORY G. HEINTZ, THOMAS W. GETTYS
`and JONATHAN A. COHN
`
`Departments of Medicine and + Anesthesiology, Duke University Schoo! of Medicine and Durham
`V.A. Medical Center, Durham, NC, U.S.A.
`
`(Received 26 July 1990; accepted 3 January 1991)
`
`Abstract--Flavonols are dietary compounds widely distributed in plants and characterized by a 2-
`phenyl-benzo(a@)pyrane nucleus possessing hydroxy! and ketone groupsat positions 3 and 4, respectively.
`Kaempferol, quercetin, and myricetin are flavonols that are further mono-, di-, or trihydroxylated on
`the phenyl ring, respectively. To test whether these ingested flavonols might exert a direct secretory
`effect on intestinal epithelial cells, monolayers of the T,, colonocyte cell line were mounted in Ussing
`chambers and examinedfor ion transport response. Twenty minutes after addition of 100 uM quercetin
`to either the serosal or mucosal side, the short-circuit current change was maximal at 16.6 uA/cm’.
`Kaempferol wasless potent than quercetin, while myricetin and glycosylated quercetin (rutin) did not
`induce secretion. The secretion induced by quercetin did not seem to be mediated by the reactive
`oxygen species generated by quercetin through auto-oxidation and/or redox cycling (superoxide,
`hydrogen peroxide, and the hydroxyl radical) because it was neither enhanced by iron, nor inhibited
`by desferroxamine B or catalase (alone or in combination with superoxide dismutase). Like vasoactive
`intestinal peptide, quercetin induced a secretory response that was inhibited by barium chloride and
`bumetanide, and which exhibited synergism with carbachol. Quercetin also stimulated a modest increase
`in intracellular cAMP levels and the phosphorylation of endogenous protein substrates for cAMP-
`dependent protein kinase. Thus, quercetin is a potent stimulus of colonocyte secretion that resembles
`secretagogues which act via a cAMP-mediated signaling pathway.
`
`Flavonoids constitute a class of compounds which
`contain the basic structural feature of a 2-phenyl-
`benzo(@)pyrane nucleus (Fig. 1). Either as free
`aglycones or more commonly glycosylated at carbons
`3,4, or 7, these compoundsare universally distributed
`among vascular plants where they may serve as
`natural transport regulators for the plant growth
`substance auxin [1]. Flavonols are a subgroupof the
`flavonoids, characterized by a hydroxyl group at
`position 3 and a ketone at position 4. Flavonols can
`be further hydroxylated at positions 3’, 4’ or 5’ on
`the 8 phenyl ring to yield the 4’-monohydroxy-
`flavonol kaempferol,
`the 3’,4’-dihydroxy-flavonol
`quercetin, and the 3’,4’ ,5’-trihydroxy-flavonol myr-
`icetin (Fig. 1). Both quercetin and myricetin produce
`reactive oxygen species
`(superoxide, hydrogen
`peroxide, and hydroxyl
`radical)
`through auto-
`oxidation and redox cycling [2-4]. Since reactive
`oxygen species mayinduceintestinal secretion [5, 6],
`we examined the possibility that flavonols might act
`on the intestinal epithelial cell
`to stimulate ion
`transport.
`In this study, the colonic epithelial cell line Ts,
`was used as a model to study the effects of flavonols
`on the enterocyte. These cells grow as well-
`differentiated monolayers which exhibit vectorial
`chloride secretion when mounted in Ussing chambers
`and exposed to a variety of neurohormonalstimuli
`{7]. Chloride secretion, monitored by a change in
`
`
`* Correspondence: Toan D. Nguyen, M.D., Building 2,
`2nd Floor, V.A. Medical Center, 508 Fulton St., Durham,
`NC 27705.
`
`Chemicals. Quercetin, kaempferol, myricetin,
`rutin, barium chloride, superoxide dismutase (SOD)
`(from bovine erythrocytes), catalase (from bovine
`liver), desferroxamine B, carbachol, and Fe(IEH)-
`EDTAwere obtained from Sigma, St. Louis, MO.
`Bumetanide was provided by Biomol Research
`Laboratories, Plymouth Landing, PA. VIP was from
`Peninsula, Belmont, CA. P as inorganic phosphate
`1879
`
`the short-circuit current (Isc) necessary to nullify the
`potential difference across the cell monolayer,
`is
`stimulated by agents which increase cAMP, such as
`vasoactive intestinal peptide (VIP) or prostaglandin
`E, [8, 9], and also by agents which act through Ca?*-
`mediated pathways, such as carbachol, histamine
`and calcium ionophores [10, 11]. Chloride secretion
`occurs through Ci7 channels located on the apical
`membrane of confluent monolayers [12], and is the
`result of the Cl” electrochemical gradient generated
`by the concerted action of three transporters: the
`basolateral Na*,K*,Cl”
` co-transporter,
`the
`Na*t,K*-ATPase pump, and Kt channels [9]. The
`Na* and K* imported into the cell by the
`co-transporter are recycled to the extracellular
`compartment, respectively, by the Na*,Kt-ATPase
`pumpandbyatleast two distinct K* efflux channels
`(activated separately by VIP and by carbachol).
`Because the secretory response in Ts, cells is well
`characterized and reflects
`the direct action of
`secretagogues on the enterocyte, we chose this model
`to characterize the effects of dietary fiavonols on
`colonic secretion.
`
`MATERIALS AND METHODS
`
`MSNExhibit 1036 - Page 1 of 8
`MSNv. Bausch - IPR2023-00016
`
`

`

`1880
`
`T. D. NGUYENef al.
`
`C KAEMPFEROL
`
`penicillin, and 5000 ug/L streptomycin sulfate) to
`bathe both sides of
`the cell monolayer. The
`confluent monolayers used for secretory studies were
`maintained for at least 7 days after the filters were
`seeded.
`Secretory studies. The cell monolayers on the
`filter/ring units were mounted in a modified Ussing
`chamberas previously described [7], and both sides
`of the monolayer were bathed with a Ringer's
`solution containing 115mM NaCl, 1.2mM CaCh,
`1.2mM MgCh, 0.4mM KH>PO,, 2.5mM K,HPO,,
`25mM NaHCO, and 10mM glucose. Quercetin,
`myricetin, and kaempferol were dissolved in ethanol
`and addedto the Ringer’s solution at a 1: 100 dilution
`(final concentrations of ethanol 1%). The medium
`was warmed to 37° with a circulating water jacket
`and gently mixed and oxygenated with a constant
`inflow of 95%O,/5%CO,. During secretory studies,
`spontaneoustissue potential differences were short-
`circuited by an automatic voltage clamp (model
`DVC-1000, World Precision Instruments, New
`Haven, CT) with Ag-AgCl, electrodes, and the
`current necessary to maintain this short circuit (Isc)
`recorded at 1-min intervals. Instrument calibration
`was performed prior to each experiment using a
`filter/ring unit without cells. All comparative studies
`used matched pairs of monolayers seeded at the
`same time and studied concurrently. Spot checks of
`the Ts, monolayers
`after
`completion of
`the
`experiments showed that cells exposed to 100 uM
`quercetin for > 1 hr could still exclude trypan blue.
`Cellular protein phosphorylation. Tg, cell protein
`phosphorylation responses were
`studied using
`methods previously described [13]. Briefly, cells
`were labeled with **P; in a phosphate-free buffer,
`exposed to 100 uM quercetin for 5 min, scraped from
`the filters, and homogenized with a glass—Tefion
`homogenizer. Phosphoproteins contained in the
`supernatantfraction after centrifugation at 436,000 g
`for 15 min wereprecipitated with 10% trichloroacetic
`acid and resolved by two-dimensionalpolyacrylamide
`gel electrophoresis. Phosphoproteins were detected
`by autoradiography using X-OMAT ARS film
`exposed at room temperature.
`cAMP Assay. Ty, cells grown to confluence on
`filters were washed twice with Ringer’s solution and
`immersed in 15 mLofRinger’ssolutionsupplemented
`with 0.2 mM 3-isobutyl-1-methyixanthine and 10 mM
`glucose, warmedto 37°, and equilibrated with 95%
`OQ, 5% CO. Quercetin (final concentration 100 nM)
`or VIP (final concentration 1 nM) was added to the
`solution, and after different
`time periods,
`the
`filters containing the cells were transferred into
`12mm Xx 75mm plastic tubes and placed in liquid
`nitrogen. The cAMP wasthen extracted by boiling
`the cells for 7 min in 1 mL of 5mM KH,PO,, 5mM
`K,HPO,, 1mM EDTA, and 0.1mM 3-isobutyl-1-
`methylxanthine. The supernatant
`resulting from
`centrifugation at 15,000g for 7.5 min was assayed
`for cAMP according to the method described by
`Gettys et al. [14].
`
`RESULTS
`
`Stimulation of secretion with flavonols. As shown
`in Fig. 2, addition of 100 uM quercetin to the mucosal
`
`MSNExhibit 1036 - Page 2 of 8
`MSNv. Bausch - IPR2023-00016
`
`A FLAVONOIDS
`
`B FLAVONOLS
`
`D QUERCETIN
`
`Ho
`
`On
`
`OH
`
`9:
`
`i

`
`|
`
`(O}
`
`on
`
`OH
`
`E MYRICETIN
`
`Fig. 1. Chemical structures of flavonoids (A), flavonols
`(B), kaempferol (C), quercetin (D), and myricetin (E).
`
`wasobtained from ICN, Irvine, CA. Culture medium
`was obtained from the Tissue Culture Facility of the
`University of North Carolina, Chapel Hill, NC, or
`from Gibco, Grand Island, NY.
`Growth and maintenance of Tgq cells. Tg4 cells
`were provided by Dr. K. Dharmsathaphorn
`(University of California, San Diego). These cells
`were cultured at 5% CO, and 37° in a 1:1 mixture
`of Dulbecco’s modified Eagle’s medium and Ham’s
`F-12 medium supplemented with 5% (v/v) newborn
`calf serum. Cells were seeded onto collagen-coated
`Nucleporefilters previously glued onto plastic rings
`(surface area: 2.9cm*, approximately 10° cells/
`filter). These filters were then set on glass beads to
`allow medium (supplemented with 5000 units/L
`
`

`

`1881
`
`te
`
`10 uM
`100 uM
`
`{B)
`
`a
`
`J
`
`
`
`
`Quercetin
`,
`
`15
`
`10
`
`5 Q
`
`a
`Ss
`E
`
`2<=
`
`a
`
`o2
`
`0
`
`10
`
`40
`
`20
`
`Quercetin
`
`Stimulation of T,, cell secretion with flavonols
`25
`
`80
`
`60
`
`40
`
`20
`
`g
`
`2 &
`
`B
`
`£&
`
`= F
`
`isc(uA/em2) 8
`{C) Isc(uA/em
`
`10
`
`20
`
`40
`30
`Time (min)
`
`680
`
`60
`
`)
`
`2
`
`
`
`
`0
`
`10
`
`20
`
`30
`
`40
`
`50
`
`60
`
`Time (min)
`
`30
`20
`Time (min)
`ee
`100
`29=Quercetin
`
`100 pM
`green
`2
`Mucosal
`300 1M

`~~~ Serosal

`3
`£&
`g
`
`
`
`
`
`Quercetin
`100 pM
`
`
`
`0
`
`20
`
`40
`Time (min)
`
`60
`
`Fig. 2. Secretory effect of quercetin. T,, monolayers were grown to confluence on semipermeable
`membranes, mounted on modified Ussing chambers, and stimulated with quercetin as described in
`Materials and Methods. The resulting Isc’s were recorded every minute and the resulting means and
`SEM (alternated for clarity in panels C and D) shown, Panel A (top left): Incubation with 100 uM
`quercetin added to the mucosal compartment(1-36 min: N = 47; 37-66 min: N = 20-46). Panel B (top
`right): Incubationwith either 10 or 100 uM quercetin addedto the mucosal compartment(three matched
`pairs). Panel C (bottom left): Incubation with either 100 or 300 4M quercetin added to the mucosal
`compartment (three matched pairs). Panel D (bottom right): Incubation with 100 uM quercetin added
`to either the serosal or the mucosalside of the T,, cell monolayer. Within each of the seven matched
`pairs, the Isc increases were normalized using the maximal Isc increase; the resulting means and SEM
`are shown [mean maximalIsc response: 53 + 8.7 wA (18 wA/cm’)).
`
`side of the cell monolayer produced an increase in
`Effects of modulators of the metabolism ofreactive
`Isc which peaked after 15-20 min to a maximum
`oxygen species on quercetin-stimulated chloride
`value above baseline of 16.6 #A/cm?(SEM = 0.9 wA/
`secretion. It
`is possible that the reactive oxygen
`cm?, N = 48, 48.1 wA/filter). The Isc was unaffected
`species (superoxide, hydrogen peroxide, or hydroxyl
`in controlfilters exposed to ethanol aloneatafinal
`radical) produced by quercetin upon auto-oxidation
`concentration of 1%.
`and/or redox cycling may mediate its secretory
`A threshold response to quercetin was obtained
`effect. In this case, the Isc response should bealtered
`at 104M (Fig. 2B). The maximal
`Isc increase
`by compounds or enzymes which modulate the
`produced by 300 uM quercetin was the sameas that
`production or degradation of these species. Quer-
`produced by 100 uM, but the response was more
`cetin-induced secretion was therefore studied after
`rapid in onset and shorter in duration (Fig. 2C). As
`cells were preincubated with superoxide dismutase
`shown in Fig. 2D, quercetin stimulated a similar
`(SOD)(to enhance the conversion of superoxide to
`secretory response whether added to the serosal or
`hydrogen peroxide), catalase
`(to enhance the
`the mucosal side of the monolayer. However, the
`conversion of hydrogen peroxide to water and
`maximal response elicited from the serosal side was
`oxygen), iron (to facilitate the Haber~Weiss reaction
`only 68% of the maximal response obtained from
`in which hydrogen peroxide is converted to hydroxyl
`the mucosal side (P = 0.01 for a smaller serosal
`radical), or desferroxamine B (to chelate iron and
`response, paired two-tailed t+test with 6df, mean
`prevent the Haber-~Weiss reaction). If quercetin-
`maximal mucosal Isc response: 11.9 + 1.9 uA/cm?).
`inducedsecretion is mediated by hydrogen peroxide,
`The effects of flavonols structurally related to
`then catalase should inhibit the secretory response
`quercetin were also investigated. The maximal Isc
`to quercetin. Similarly, if secretion is mediated by
`increase observed with 100 uM kaempferol was
`hydroxyl radical, then the Isc response should be
`26 42% of the maximal
`increase obtained with
`inhibited by desferroxamine and enhanced by Fe**.
`100 uM quercetin (three paired experiments). For
`Finally,
`if extracellular superoxide is
`the key
`myricetin, secretion was detected only at 300 uM
`mediator, then secretion should be blocked by the
`combination of SOD and catalase. As shown in
`and not at 100 uM. A minimalresponse wasobtained
`with a 3004M concentration of the glycosylated
`Table 1, little effect on quercetin-induced secretion
`quercetin-rutinoside (rutin) (data not shown).
`was observed after preincubation with 504M
`
`MSNExhibit 1036 - Page 3 of 8
`MSNv.Bausch - IPR2023-00016
`
`

`

`1882
`
`T. D. NGUYENetal.
`
`Table 1. Effects of modulators of the metabolism of reactive oxygen species on quercetin-
`induced secretion in T,, cells
`Maximal Isc
` Agent (% control) N P (#100)
`
`
`
`
`
`
`0.78
`6
`104 + 12.6
`SOD (470 units/mL) + catalase (450 units/mL)
`0.08
`6
`7+ 7.8
`Catalase (450 units/mL)
`0.68
`6
`97 + 5.6
`Desferroxamine B (50 uM)
`
`
`
`75+ 9.2 6Fe3* (50 uM) 0.04
`
`T,, cells mounted in modified Ussing chambers were incubated withthe different modulators
`of reactive oxygen species metabolism for 10-15 min, and quercetin was added toafinal
`concentration of 100 uM. In each matchedpair, the maximal short-circuit current (Isc) change
`induced by quercetin in the presence of the modulator is expressed as the percentage of the
`maximal Isc change induced by quercetin in the absence of the modulator. Values are means
`+ SEM. The mean maximal Isc changes in the control monolayers were, respectively,
`21.9 + 3.3, 15+ 2.5, 15.8 + 1.66, and 18.3 + 2.7 uA/cm? for the experiments studying the
`effects of SOD pluscatalase, catalase, desferroxamine B, and Fe**. P values were calculated
`using two-tailed t-tests with 5 df.
`
`desferroxamine B or a combination of 470 units/
`mL SOD and 450Qunits/mL catalase. A_
`slight
`enhancement
`in secretion, which did not
`reach
`statistical significance (0.1 > P > 0.05), was noted
`with 450 units/mL catalase, while 50 uM Fe(III)-
`EDTAproduceda significant (P < 0.05) inhibition of
`quercetin-induced secretion. However, as discussed
`previously, these last two effects were opposite of
`whatwas expected should either hydrogen peroxide
`or the hydroxyl radical mediate secretion. In the
`aggregate, these findings do not support a role for
`reactive oxygen species in the secretory response of
`quercetin.
`Intracellular mechanism ofsecretion. Tg, cell apical
`chloride secretion is dependent on the chloride
`gradient across the mucosal membrane ofthe cell.
`This gradient
`is generated by the basolateral
`Na*,K*,Cl” co-transporter with the imported Nat
`and K* recycled outside the cell by the Nat,K*-
`ATPase pump and K* efflux channels [15]. The role
`of
`these transport systems in quercetin-induced
`chloride secretion was studied using bumetanide,
`which inhibits the Na*,K*,Cl~ co-transporter, and
`barium chloride, which inhibits a VIP-responsive K*
`channel [10, 16]. Table 2 demonstratesthe inhibitory
`effects of 0.3 mM bumetanide and 6mM BaCl, on
`the secretory responseelicited by 100 uM quercetin.
`Compared with matched controls, the Isc response
`obtained 20 min after addition of quercetin was only
`19 + 1% and 31 + 5% of the expected response for
`bumetanide and barium chloride, respectively. Thus,
`quercetin-inducedsecretion appearsto require active
`Na*,K*,Cl co-transport and K* efflux mechanisms.
`One approach to determining which intracellular
`signalling
`pathway(s) mediates
`the
`quercetin
`secretory response is to evaluate whether quercetin
`exhibits synergism when administered with other
`secretagogues. The interactions between carbachol,
`VIP, and quercetin were therefore studied. In Fig.
`3A,
`cells were
`exposed either
`to carbachol
`(final concentration 10 uM) or to quercetin (final
`concentration 50 uM). After 15 min, quercetin was
`added to the cells previously exposed to carbachol
`and vice versa. In these matchedpairs, the effect of
`
`Table 2. Effects of ion transport inhibitors on quercetin-
`induced secretion in Tg, cells
`
`Isc
`
`Agent
`(% control) NP (# 100)
`
`0.0001
`4
`19+1
`Bumetanide (0.3 mM)
`
`
`
`31+5 4Barium chloride (6 mM) 0.0007
`
`Ty, cells were incubated for 15 min with either bumetanide
`or barium chloride, and quercetin was added to a final
`concentration of 100 4M. In each matched pair, the Isc
`change induced by quercetin after 20 min in the presence
`of the inhibitor is expressed as the percentage of the Isc
`change induced by querecetin alone. Bumetanide was
`dissolved in 0.1M NaOH and added to thecells at a 1: 100
`dilution; the corresponding control also contained 1: 100
`dilution of 0.1 M NaOH. Values are means + SEM. The
`maximal Isc responses in the control monolayers were
`12.1 + 0.59 and 13.4 + 1.2 wA/cm’, respectively, for the
`experiments studying the effects of bumetanide and barium
`chloride. P values were calculated using two-tailed ¢-tests
`with 3 df.
`
`(or carbachol
`quercetin followed by carbachol
`followed by quercetin) in one monolayer can be
`compared to the initial effect of quercetin (or
`carbachol) alone in the other. Analyzed in this
`fashion, quercetin alone produced anIscincrease of
`9.4 + 0.4 wA/cm? (27 + 1.2 uA, N = 3) after 15 min,
`while carbachol produced an Isc
`increase of
`2.3+1pA/em? (7+3.5uA, N=3) after 3 min.
`Carbachol added to cells responding maximally to
`quercetin produced an additional Isc increase of
`34.9+ SuA/cem?
`(101+ 14.4 WA)
`resulting in a
`combined total
`Isc increase of 44.4 +5 A/cm?
`(129 = 14.4 pA). Quercetin addedtocells previously
`exposed to carbachol produced an Isc increase of
`19.1 + 1.7 wA/em? (55 + 4.8 vA). However, in the
`latter case, quercetin was added after completion of
`the carbachol response andthe interaction between
`quercetin and carbachol may be suboptimal.
`In
`additional experiments,
`the
`secretory response
`produced by a simultaneous dose of carbachol and
`
`MSNExhibit 1036 - Page 4 of 8
`MSNv. Bausch - IPR2023-00016
`
`

`

`Stimulation of Tg, cell secretion with flavonols
`
`1883
`
`Isc(uA/em)
`
`50
`
`(A) 40
`
` (OQ
`{o)C
`(QC
`
`304
`
`20
`
`0
`
`10
`
`20
`
`30
`
`40
`
`50
`
`60
`
`Time (min)
`
`30
`
`iy°
`
`Isc(uA/em)
`
`(0) Q
`v (@) VIP
`
`(0) VIP
`(e}Q
`
` (B)
`
`0
`
`10
`
`20
`
`40
`30
`Time (min)
`
`50
`
`60
`
`Fig. 3. Interactions between quercetin, carbachol and VIP. Pairs of confluent T,, monolayers were
`mounted in modified Ussing chambers and their secretory responses (mean Isc and SEM) compared.
`Panel A (left panel): In each pair, one monolayer was exposed to 10 uM carbachol[C](serosal surface)
`or to 50 #M quercetin [Q] (mucosal surface); after 15 min, 50 uM quercetin was added to the monolayer
`previously exposed to carbachol and vice versa. Additional monolayers were also exposed to a combined
`concentration of 10 uM carbachol and 50 uM quercetin [Q&C]. The following symbols are used: (@)
`quercetin followed by carbachol (N = 3), (O) carbachol followed by quercetin (N = 3), (A) quercetin
`and carbachol added simultaneously (N = 5). Panel B (right panel): In each pair, one monolayer was
`exposed to 1 nM VIP(serosal surface) and the other to 50 uM quercetin [Q] (mucosal surface). After
`15 min, 50 uM quercetin was added to the monolayer previously exposed to VIP and vice versa. The
`following symbols are used: (@) quercetin followed by VIP (N = 3), (©) VIP followed by quercetin
`(N = 3).
`
`quercetin was also evaluated. A peak Isc with an
`intermediate value of 23.6+2.2uA/cm* (69+
`6.5 uA, N = 5) was obtained 14 min after addition
`of the combined secretagogues. In all the different
`sequences,
`the Isc increases produced by cells
`exposed to the combination of quercetin and
`carbacholweregreater than the sum ofthe individual
`Isc changes produced by quercetin and carbachol
`(9.4+2.3=11.7uA/cm’). The difference in the
`degrees of synergism probably reflects the different
`timing of the maximal effect of quercetin and
`carbachol. In contrast, as shown in Fig. 3B, when
`the interaction between quercetin and VIP was
`analyzed in the same fashion, no such synergism,
`but a possible inhibitory effect was demonstrated.
`The synergism between carbachol and quercetin,
`but not between VIP and quercetin, suggests that
`quercetin may induce secretion through pathways
`related to the ones activated by VIP, but not by
`carbachol. This possibility was explored further in
`the following phosphorylation studies.
`Phosphorylation and intracellular cAMPstudies.
`Previous studies have shown that Tg, cells exhibit
`distinct phosphorylation responses to stimuli acting
`via CAMPorvia Ca?* [13, 17]. Phosphoproteins p83,
`p29, and p23 are examples of proteins exhibiting
`increased phosphorylation in cells stimulated by
`agents which act via Ca’*,
`such as carbachol,
`histamine,
`and ionomycin. By contrast, phos-
`phoproteins p37, p18, and p23 exhibit
`increased
`phosphorylation in cells exposed to agents which act
`via cAMP, such as VIP and forskolin. Each of these
`five phosphoproteins showed increased labeling in
`monolayers stimulated with forskolin plus carbachol
`(Fig. 4, comparing panels A and B). When
`monolayers were stimulated with 100 uM quercetin,
`only three of these five phosphorylation responses
`were observed: quercetin stimulated the phos-
`phorylation of p37, p18, and p23, but not p29 or p83
`(Fig. 4, comparing panels C and D). These results
`
`suggest that quercetin activates intracellularsignaling
`mechanisms mediated by cAMP, but not by Ca?*.
`In an attempt to study whether quercetin induces
`the generation of cAMP, this second messenger was
`measuredin cells exposed to 100 uM quercetin for
`different time periods ranging from 1 to 20min.
`Surprisingly, as shown in Table 3, only a modest
`increase in cAMP levels was detected. These
`increases were minimal when compared with the
`mean 85-fold increase in cAMP demonstrated with
`control monolayers exposed to 1nM VIP for the
`same time periods (data not shown).
`
`DISCUSSION
`
`We have demonstrated that quercetin is a potent
`stimulator of ion transport in Tg, cells. When added
`to either the mucosal or serosal side of the cell
`monolayer, quercetin produced a concentration-
`dependentincrease in Isc, which peaked 15-20 min
`after the addition of 100 uM quercetin. The observed
`Isc response is consistent with the possibility that
`quercetin can act directly on the enterocyte to
`stimulate electrogenic Cl~ secretion. Of the related
`hydroxylated flavonols, kaempferol was less potent
`than quercetin, while myricetin and the glycosylated
`quercetin-rutinoside (rutin) produced minimalres-
`ponses. Considering the effective mucosal-serosal
`barrier, the observation that quercetin can act from
`either side of the monolayer suggests that its effect
`is not
`initially mediated by
`cellular
`surface
`componentsselectively present on either side of the
`cell (e.g. receptors). It is also possible that quercetin,
`being a small hydrophobic molecule, penetrated the
`cell and produced its effect
`intracellularly. The
`similar
`time courses of the secretory responses
`produced by the addition of quercetin to either side
`of the cell do not support the possibility that the
`effect of quercetin is localized to oneside of the cell
`
`MSN Exhibit 1036 - Page 5 of 8
`MSNv. Bausch - IPR2023-00016
`
`

`

`
`
`Fig. 4. Protein phosphorylation induced by quercetin. As described in Materials and Methods,
`monolayers were labeled with *P and exposedto different stimuli for 5 min. Soluble phosphoproteins
`were then resolved using two-dimensional gel electrophoresis by isoelectric point from pH 4 to pH 7.5
`(from left to right) and by size (from top to bottom), and detected by radioautography. Panels A and
`B: Compared to a control incubation (panel A), at least five proteins showed increased labeling after
`stimulation with 10 uM forskolin and 100 uM carbachol (panel B): p18, p23, p29, p37, and p83. Panels
`C and D: Comparedto a control incubation (panel C), exposure to 100 uM quercetin for 5 min (panel
`D) resulted in the increased labeling of p18, p23 and p37. The labeling of p29 and p83 wasnotincreased.
`Similar results were obtained in three additional experiments.
`MSNExhibit 1036 - Page 6 of 8
`MSNv. Bausch - IPR2023-00016
`
`

`

`Stimulation of T,, cell secretion with flavonols
`
`1885
`
`Table 3. Effect of 100 uM quercetin on cAMPproduction in T,, cells
`
`Incubation time
`(min)
`
`cAMPproduction
`(% control)
`
`N (df)
`
`P (# 100)
`
`1
`5
`10
`15
`20
`
`94+6
`152 +31
`143 + 13
`116 + 12
`126 + 13
`
`6 (5)
`6 (5)
`8 (7)
`8 (7)
`6 (5)
`
`0.19
`0.07
`0.006
`0.11
`0.05
`
`Confluent T,, cell monolayers were exposed to 100 uM quercetin for the indicated
`time periods, and the levels of cAMPin the correspondingcell homogenates were
`determined as outlined in Materials and Methods. In each paired experiment,
`cAMPproductionin cells exposed to quercetin is expressed as the percentage of
`the cAMPproductionin control cells not exposed to quercetin. Values are means
`+ SEM. The mean cAMPproduction values in control cells for the time periods
`1, 5, 10, 15, and 20min were respectively, 10.5+0.8, 7.4+0.9, 15.9 + 1.3,
`12.1 + 0.8, and 11.4 + 1.2 pmol/filter. P values werecalculated using one-tailed t
`tests with the indicated df.
`
`transported
`be
`to
`and that quercetin needs
`transcellularly when added to the otherside.
`Weinitially postulated that the reactive oxygen
`species produced by flavonol auto-oxidation and/or
`redox cycling may be the ultimate mediators of
`quercetin-induced secretion. However, because the
`secretory response to quercetin was not enhanced
`by Fe?* and was notinhibited by desferroxamine B
`or catalase (alone or in combination with SOD), we
`were unable to substantiate a
`role for either
`superoxide, hydrogen peroxide, or the hydroxyl
`radical in the action of quercetin. The possibility
`that quercetin-induced secretion is independent of
`reactive oxygen species is further supported by the
`observation that kaempferol, which doesnotgenerate
`reactive oxygen species, stimulates secretion, while
`the converseis true of myricetin, a potent generator
`of reactive oxygen species [4]. When studies were
`performed to identify the intracellular mechanism
`responsible for quercetin-induced secretion, quer-
`cetin resembled other secretagogues known to
`stimulate Tg, cells via cAMP: (a) quercetin-induced
`secretion was inhibited by barium chloride and
`bumetanide,
`(b) quercetin was synergistic with
`carbachol, but not with VIP, and (c) exposure of
`intact Tg cells
`to quercetin resulted in
`the
`phosphorylation of endogenous protein substrates
`for cAMP-dependent protein kinase. However,
`quercetin, at concentrations capable of stimulating
`cellular secretion, did not increase cAMPlevels to
`the extent demonstrated by VIP.It is possible that,
`similar to the case of the adenosine analogue 5’-(N-
`ethyl)-carboxamido-adenosine [18], there may be a
`shift
`in the concentration-response curve when
`stimulation of cAMPis studied instead of secretion.
`However, we have not been able to explore this
`possibility becauseof the poorsolubility and potential
`toxicity of high concentrations of quercetin [19]. It
`still remains possible that the modest increase in
`cAMP produced by quercetin was sufficient
`to
`stimulate the phosphorylation of the substrates for
`cAMP-dependent protein kinase responsible for
`controlling chloride secretion. That the interaction
`between quercetin and the cAMP pathway may be
`
`complexis further suggested by a possible inhibition
`of VIP-induced secretion by quercetin (e.g.
`less
`potent activation by quercetin of a pathway shared
`with VIP). Should this be the case, quercetin may
`prove to be useful as a probe for further studies of
`the intracellular mechanisms regulating intestinal
`secretion.
`Additional studies will be required to judge
`whether
`flavonols are physiologically important
`stimuli of intestinal secretion. Flavonols are found
`in the edible portions of many fruits and vegetables
`and the average intake is estimated to be 100 mg/
`day (20, 21], with vegetarians consumingsignificantly
`larger amounts. Quercetin is the most abundant
`dietary flavonol, and it therefore seems plausible
`that
`luminal
`concentrations of quercetin may
`reach 50-100 uM (15-30 mg/L) depending on the
`disposition of this flavonol after ingestion. Should
`these concentrations of quercetin be obtained in the
`intestinal lumen, the findings presented in this study
`indicate that this flavonolcould stimulatea significant
`secretory response.
`In this
`regard,
`it
`seems
`noteworthy that dietary supplementation withfruits
`and vegetables has provedto be usefulin theclinical
`management of chronic constipation [22]. Even
`though it has been assumedthat the benefit of fruits
`and vegetables results from secretory as well as
`osmotic mechanisms, and from the containedfiber,
`scant information exists concerning the identity of
`the substances that may functionas secretory stimuli.
`The present study raises the possibility that
`the
`secretory action of quercetin and other flavonols
`may account for the beneficial effects of fruits and
`vegetables for patients with constipation.
`
`Acknowledgements—This research was funded in part by
`the Department of Veterans Affairs, by NIH Grants DK
`40506 (T.D.N.), DK 40701 (J.A.C.) and DK 42486
`(T.W.G.), by NIEHS Grant IS 04752 (A.T.C.), and by a
`grant from the Cystic Fibrosis Foundation (J.A.C.). The
`authors are indebted to Margaret Wolfe, Jolanta Kole, and
`Wei Wangfortheir technical assistance, and to Dr. Helen
`Berschneider forher help in setting up the Ussing chambers.
`MSNExhibit 1036 - Page 7 of 8
`MSNv. Bausch - IPR2023-00016
`
`

`

`1886
`
`T. D. NGUYEN etal.
`
`REFERENCES
`
`—
`
`. Jacobs M and Rubery PH, Naturally occurring auxin
`transport regulators. Science 241: 346-349. 1988.
`. Hodnick WF, Kung FS, Roettger WJ, Bohmont CW
`and Pardini RS, Inhibition of mitochondrial respiration
`and production of toxic oxygen radicals by flavonoids.
`A structure-activity study. Biochem Pharmacol 35:
`2345-2357, 1986.
`. Hodnick WF, Kalyanaraman B, Pritsos CA and Pardini
`RS, The production of hydroxyl and semiquinonefree
`radicals during the autoxidation of
`redox active
`flavonoids.
`In: Oxygen Radicals
`in Biology and
`Medicine (Eds. Simic MG, Taylor KA, Ward JF and
`VonSonntag C), pp. 149-153. Plenum Press. New
`York, 1988.
`. Canada A, Gianella E. Nguyen TD and Mason RP.
`The production of reactive oxygen species by dietary
`flavonols. Free Radic Biol Med 9: 441-449, 1990.
`. Bern MJ, Sturbaum CW, Karayalcin SS, Berschneider
`HM, Wachsman JT and Powell DW. Immunesystem
`control of rat and rabbit colonic electrolyte transport:
`Role of prostaglandins and enteric nervous system. J
`Clin Invest 83: 1810-1820, 1989.
`. Karayalcin SS, Sturbaum CW. Dixon MU and Powell
`DW. Hydrogen peroxideis the reactive oxygen species
`(ROS) which stimulates colonic electrolyte secretion.
`Gastroenterology 94: A216(Abstract), 1988.
`. Dharmsathaphorn K. McRoberts JA, Mandel KG,
`Tisdale LD and Masui H, A human colonic tumorcell
`line that maintains vectorial electrolyte transport. Am
`J Physiol 246: G204-G208, 1984.
`. Weymer A, Huott P. Liu W, McRoberts JA and
`Dharmsathaphorn K, Chloride secretory mechanism
`induced by prostaglandin E, in a colonic epithelial cell
`line. J Clin Invest 76: 1828-1836, 1985.
`. Dharmsathaphorn K, Mandel KG. Masui H and
`McRoberts JA, Vasoactive intestinal polypeptide-
`induced chloride secretion by a colonic epithelial cell
`line: Direct participation of a basolaterally localized
`Na*,K*.Cl cotransport system. J Clin Invest 75: 462~
`471, 1985.
`Dharmsathaphorn K and Pandol SJ, Mechanism of
`
`10.
`
`il.
`
`12.
`
`16.
`
`chloride secretion induced by carbachol in a colonic
`epithelial cell line. J Clin Invest 77: 348-354, 1986.
`Dharmsathaphorn K. Cohn J and Beuerlein G. Multiple
`calcium-mediated
`effector mechanisms
`regulate
`chloride secretory responses in T,, cells. Am J Physiol
`256: C1224-C1230, 1989,
`Mandel KG, Dharmsathaphorn K and McRoberts JA,
`Characterization of
`a
`cyclic AMP-activated Cl”
`transport pathway in the apical membrane of a human
`colonic epithelial cell line. J Biol Chem 261: 704-712,
`1986.
`. Cohn J

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket