induction of heme oxygenase in intestinal epithelial cells: studies in caco-2 cell cultures

6
Molecularand CellularBiochemistry 129:93-98,1993. 0 1993 KluwerAcademic Publishers. Printedin the Netherlands. Induction of heme oxygenase in intestinal epithelial cells: studies in Caco-2 cell cultures Julia W. Cable, Edward E. Cable and Herbert L. Bonkovsky Departments of Medicine, Biochemistry and Molecular Biology and the Center for Study of Disorders of Iron and Porphyrin Metabolism, University of Massachusetts Medical School, Worcester, MA 01655, USA Received 30 June 1993;accepted 28 September 1993 Abstract Enterally administered, heme is a good source of iron in humans and other animals, but the metabolism of heme by enterocytes has not been fully characterized. Caco-2 cells in culture provide a useful model for studying cells that resemble small intestinal epithelium, both morphologically and functionally. In this paper we show that heme ox- ygenase, the rate-controlling enzyme of heine catabolism, is present in abundance in Caco-2 cells, and that levels of its mRNA and activity can be increased by exposure of the cells to heme or metal ions (cadmium, cobalt). Caco-2 cells also contain biliverdin reductase activity which, in the basal state, is similar to that of heme oxygenase (approximately 40 pmole of product per mg protein per minute); however, when heme oxygenase is induced, biliverdin reductase may become rate-limiting for bilirubin production. (Mol Cell Biochem 129: 93-98, 1993) Key words: biliverdin reductase, Caco-2 cells, heme, heme oxygenase, intestinal cells Abbreviations: BVR - biliverdin reductase; DMEM - Dulbecco's modified Eagles medium; DMSO - dimethyl sulfoxide; HO - heme oxygenase; 1 x SSC - a solution of 0.015 M sodium citrate/0.15 sodium chloride Introduction In many tissues, heme oxygenase (HO) (EC.1.14.88.3) is the rate-controlling enzyme for the breakdown of heine to biliverdin and subsequently bilirubin [1-4]. The prod- ucts of the HO reaction are iron, carbon monoxide, and biliverdin. In liver, spleen, and kidney, activities of biti- verdin reductase are higher than those of HO, such that the rate of bilirubin formation is chiefly a function of the oxygenase [5-7]. HO can be induced by its substrate heme, as well as by other chemical agents or physical changes (e.g., heat) [8-16]. HO activities can be induced by at least two different mechanisms. One is dependent Addressfor offprints: H.L. Bonkovsky,Divisionof Digestive Disease 01655, USA on heme, or nascent heme synthesis, and the other, pro- duced by certain metal ions (e.g., Cd, Co, Fe), is inde- pendent of heine synthesis [8]. Intestinal epithelial cells are known to absorb metal ions, including those known to induce HO. They also are known to absorb the iron of heme; indeed net GI ab- sorption of heme iron generally exceeds that of non- heme or inorganic iron absorption [17-21], indicating that heine may be a preferred iron source for iron reple- tion, or, conversely, a source of dietary iron to be avoid- ed in those with hemochromatosis. It is unknown wheth- and Nutrition, Universityof Massachusetts Medical Center, Worcester,MA

Upload: julia-w-cable

Post on 10-Jul-2016

220 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Induction of heme oxygenase in intestinal epithelial cells: studies in Caco-2 cell cultures

Molecular and Cellular Biochemistry 129: 93-98,1993. 0 1993 Kluwer Academic Publishers. Printed in the Netherlands.

Induction of heme oxygenase in intestinal epithelial cells: studies in Caco-2 cell cultures

Julia W. Cable, Edward E. Cable and Herbert L. Bonkovsky Departments o f Medicine, Biochemistry and Molecular Biology and the Center for Study of Disorders of Iron and Porphyrin Metabolism, University of Massachusetts Medical School, Worcester, MA 01655, USA

Received 30 June 1993; accepted 28 September 1993

Abstract

Enterally administered, heme is a good source of iron in humans and other animals, but the metabolism of heme by enterocytes has not been fully characterized. Caco-2 cells in culture provide a useful model for studying cells that resemble small intestinal epithelium, both morphologically and functionally. In this paper we show that heme ox- ygenase, the rate-controlling enzyme of heine catabolism, is present in abundance in Caco-2 cells, and that levels of its m RNA and activity can be increased by exposure of the cells to heme or metal ions (cadmium, cobalt). Caco-2 cells also contain biliverdin reductase activity which, in the basal state, is similar to that of heme oxygenase (approximately 40 pmole of product per mg protein per minute); however, when heme oxygenase is induced, biliverdin reductase may become rate-limiting for bilirubin production. (Mol Cell Biochem 129: 93-98, 1993)

Key words: biliverdin reductase, Caco-2 cells, heme, heme oxygenase, intestinal cells

Abbreviations: BVR - biliverdin reductase; D M E M - Dulbecco's modified Eagles medium; DMSO - dimethyl sulfoxide; H O - heme oxygenase; 1 x SSC - a solution of 0.015 M sodium citrate/0.15 sodium chloride

Introduction

In many tissues, heme oxygenase (HO) (EC.1.14.88.3) is the rate-controlling enzyme for the breakdown of heine to biliverdin and subsequently bilirubin [1-4]. The prod- ucts of the H O reaction are iron, carbon monoxide, and biliverdin. In liver, spleen, and kidney, activities of biti- verdin reductase are higher than those of HO, such that the rate of bilirubin formation is chiefly a function of the oxygenase [5-7]. H O can be induced by its substrate heme, as well as by other chemical agents or physical changes (e.g., heat) [8-16]. H O activities can be induced by at least two different mechanisms. One is dependent

Address for offprints: H.L. Bonkovsky, Division of Digestive Disease 01655, USA

on heme, or nascent heme synthesis, and the other, pro- duced by certain metal ions (e.g., Cd, Co, Fe), is inde- pendent of heine synthesis [8].

Intestinal epithelial cells are known to absorb metal ions, including those known to induce HO. They also are known to absorb the iron of heme; indeed net GI ab- sorption of heme iron generally exceeds that of non- heme or inorganic iron absorption [17-21], indicating that heine may be a preferred iron source for iron reple- tion, or, conversely, a source of dietary iron to be avoid- ed in those with hemochromatosis. It is unknown wheth-

and Nutrition, University of Massachusetts Medical Center, Worcester, MA

Page 2: Induction of heme oxygenase in intestinal epithelial cells: studies in Caco-2 cell cultures

94

er heme take up by enterocytes is absorbed intact or bro- ken down by HO, such that its iron enters intracellular non-heme iron pools.

Caco-2 cells, cloned from a human colon cancer, spon- taneously undergo differentiation in culture to form po- larized monolayers with structural and functional fea- tures that resemble small intestinal epithelial cells [22, 23]. Such cultures are being used to study intestinal ab- sorption and metabolism of ions and other chemicals [24]. To aid studies on iron and heme uptake and metab- olism by enterocytes, we tested whether cultures of Ca- co-2 cells could take up and metabolize heine, iron, or cobalt, and whether HO and biliverdin reductase could be detected in such cultures.

Materials

Caco-2 cells were a generous gift from Drs. Susan and Robert Baker, Department of Pediatrics, University of Massachusetts Medical Center, Worcester, MA. They may also be obtained from the American Type Culture Collection (Rockvi]lle, MD). Dulbecco's modified Ea- gle's medium (DMEM) was obtained from Fisher (Springfield, IL). Penicillin/streptomycin, L-glutamine, and gentamicin were from Flow ICN (Costa Mesa, CA). Bovine calf serum was from Hyclone Laboratories (Lo- gan, UT). William's E medium was from Gibco BRL (Grand Island, NY). Dimethyl sulfoxide (DMSO), non- essential amino acids, trypsin, cobalt chloride, cadmium chloride, iron chloride, sodium nitrolotriaceate, NADPH, deferoxaraine, bovine serum albumin (BSA), dexamethasone, soybean trypsin inhibitor, triiodothy- ronine, and insulin were from Sigma (St. Louis, MO). Heine was from Porphyrin Products (Logan, UT). RNAzol | was from Biotex (Houston, TX). All other chemicals were of the highest purity commercially avail- able.

Methods

The human colon adenocarcinoma cell line Caco-2 was obtained at the 71 ~t passage and was studied through the 84 th passage. Cells were routinely grown in DMEM with 20% bovine calf serum, 1% non-essential amino acids, 1.74mM sodium bicarbonate, 2mM L-glutamine, 100 U/ml penicillin, 100 mg/ml streptomycin and 50 rag/ ml gentamicin. The cells were grown and treated in 6 cm or 10 cm tissue culture plates in a 37 ~ C incubator with

5% CO z [24]. The medium was changed every 2-3 days. After about 10 days of growth and the cells reached con- fluence, monolayers were gently rinsed with, and ex- posed to serum-free William's E medium containing dexamethasone (300pg/ml), triiodothyronine (1gg/ ml), and insulin (1 gg/ml) [16]. Chemicals were prepared and added at the indicated times and concentrations af- ter which cells were harvested into 100 mM potassium phosphate pH7.50, 20% glycerol (v/v), containing I mM EDTA and sonicated for 3 sec at a setting of 1.5 using a Branson sonicator equipped with a microtip. Heine was administered as a BSA complex (ca. 1.66 moles heine/1 mole albumin). The complex was prepared by adding 100 gl of 10 mM heme, dissolved in dimethyl sulfoxide (DMSO), to 3.9 ml of a solution of BSA, ll.lmg/ml, dissolved in 40mM Tris.HCL (pH 7.4). Heme oxygenase was assayed as described previously [8, 16], except that a 14,000 x g I rain spin su- pernatant was used [25] as the source of the enzyme. 250 units (pmole bilirubin formed~ -1) of exogenous bili- verdin reductase (BVR) was added to all HO assays ex- cept for those in Fig. 4, in which the exogenous BVR activity was as indicated. One unit of heme oxygenase or biliverdin reductase activity is defined as one pmole bili- rubin formed per minute.

Total RNA was isolated by using RNAzol | according to the manufacturer's instructions. 20 Bg of RNA in a final volume of 40 gl were treated with 24 pl of 20 x SSC and 16 gl of 37% formaldehyde (by volume) and incu- bated at 65 ~ C for 15 min. The samples were diluted to 600 gl with 10 x 0.015 M sodium citrate, 0.15 M sodium chloride (SSC) and 10 lxg of RNA were loaded per dot. The blots wre crosslinked by exposure to 2.4 gjoules of UV radiation using a UV-Stratalinker (Stratagene, La Jolla, CA) and prehybridized at 42 ~ C in 15 ml of a buffer containing 50% formamide, 5 x SSC, 100 gg/ml salmon sperm DNA, 5 x Denhardt's solution and 1% SDS. After 2 hr the solution was removed and replaced with a small- er volume (5-7 mls) of the same buffer and a boiled ra- dioactive probe for human heme oxygenase-1 [26] was added to 30 x 106 counts per blot. After hybridization for 18 hr at 42 ~ C, the blots were washed with 0.1 x SSC at room temperature for 30 rain with 3 buffer changes and counted on a beta-counter (Beta-gen, Millvale, CA). To- tal mRNA loaded onto dot-blots was normalized by probing duplicate blots with a poly-T probe, as previous- ly described [27, 28]. Protein concentrations were as- sayed by the bicinchoninic method [29] using bovine se- rum albumin as a standard.

All results are shown as mean + SEM, n = 3. Statistical

Page 3: Induction of heme oxygenase in intestinal epithelial cells: studies in Caco-2 cell cultures

E

x o

cl') E E - r ' . ~

o m

._~ X::}

< o E l:)..

150

100

- A. Heme

50

0 I I I I 0 40 80 120

[Heme] (I.tM)

200- B. Cadmium

150

100

50 -

. . . . . . . . . . . . 1 . . . . . . . . . . . . . . . . 1 . . . . . . . . . . . . 1 . . . . . . . . . . . . 1 . . . . . . . . . . . . . 1 . . . . . . . . . 1 . . . . . . . . . . .

0 5 10 15 20 25 30 [Cadmium Chloride] (t.tM)

Fig. i. Effect of heme (A) and cadmium chloride (B) on activity of heme oxygenase in Caco-2 cells. Cells were cultured, harvested, and heme oxygenase activity measured as described in Materials and Me- thods. Heme was added as an albumin complex at the indicated con- centrations 18 hr prior to harvest. Cadmium chloride was added at the indicated concentrations 18 hr prior to harvest. Data are mean _+ SEM, n = 3; when no error bars are shown the SEM lies within the size of the symbol.

analyses were done using Student ' s t-tests, Dunne t t ' s test, or analysis of variance, with Scheff6 's correc t ion for

multiple comparisons, as appropriate . With all tests, a 95% conf idence level was the lower limit for signifi-

cance.

Results

In initial studies, the possible effect of selected culture

condit ions on activity of H O in Caco-2 cells were as- sessed. Since the cells were initially cul tured with D M E M and 20% bovine serum, various doses of h e m e

were added in this m e d i u m and in Wil iam's E m e d i u m with added insulin, dexamethasone and t r i iodothyro- nine. N o H O induct ion was observed in cells exposed to heine in D M E M with 20% serum (data not shown). In

95

2.5-

Z

2.0- I - -

-6

~ 1 . 5 - ~ 7 ~ e r

1.0-

0

o~ 0.5-

0.0 ~

, , , , , , ' , , , , ' , , , ,

,;,;,;,;,;,;,;

Control I

50 gM Heme

% g k l % g % g % g % 1 % I ,

% g % l % l % g % l k l k g ,

. , . , . . . . . . . . >i 1 10 IxM Cd

Fig. 2. Effects of heme and cadmium on heme oxygenase mRNA levels in Caco-2 cells. Cells were cultured, harvested, and total RNA isolated and probed as described in Materials and Methods. Heme (50 gM), added as the albumin complex, or cadmium chloride (10 gM) was ad- ded to some cultures for 10 hr prior to harvest. RNA was isolated and assayed as described in Materials and Methods. Data are mean + SEM, n = 3. The counts per minute for the blots hybridized to human heme oxygenase were normalized by dividing by the counts per minu- te from duplicate blots probed with radioactive poly-T. An asterisk (*) denotes a significant increase above control (Dunnett's test).

contrast , H O activity was induced in the cells exposed to

heme in Will iam's E m e d i u m containing dexametha-

sone, t r i iodothyronine and insulin (Fig. 1). The hormon- es that were added to the Will iam's E med ium were no t

essential for induction, but their inclusion increased the

activity of H O by 20% (data not shown).

H e m e oxygenase activity in Caco-2 cells, grown in

Will iam's E medium, was inducible by some metal ions,

as well as by heme (Fig. 1). H O activity was greatest in

Caco-2 cells t rea ted for 18 hr with 50--100 ktM heme or

10 gM cadmium. 25 g M cadmium chloride decreased

H O activity, while even higher concent ra t ions (50-

100 gM) resulted in no detectable activity, apparent ly

due to toxicity to the cell cultures. 200 ~tM cobalt chlo- ride or ferric nitr i lotr iacetate (prepared as in [16]) pro-

duced slight (ca. 1.5 to 2-fold), but significant increases

in H O activity (data not shown). Glu te th imide (50 gM),

a phenobarbi ta l - l ike drug that induces H O in cul tured

hepatocytes [8, 16], had no detectable effect on activity

of the enzyme in Caco-2 cells (data not shown). A t 10 hr, 50 g M heme or 10 gM cadmium produced significant in-

creases in H O m R N A levels (Fig. 2). Induc t ion of heme oxygenase activity by heme oc-

curred rapidly and was maximal at 12 hr. The first signif-

icant increase in activity occurred at 8 hr. This heme-de- penden t induct ion was short-lived, with activities signif- icantly decreased f rom m a x i m u m at 16 hr (Fig. 3).

Act ivi ty of H O after heme t rea tment (50 gM) was

Page 4: Induction of heme oxygenase in intestinal epithelial cells: studies in Caco-2 cell cultures

96

'.o

c O55

E 7- �9

o

E t ~

150

100

50

0 4 8 12 16

Hours after Treatment

Fig. 3. Time course of induction of heine oxygenase by heme in Caco-2

cells. Cells were cultured, harvested, and heme oxygenase activity

measured as described in Materials and Methods. Heme (50/aM) was

added as an a lbumin complex and cells harvested at the t imes indica-

ted (solid symbols). Controls treated with a DMSO/a lbumin solution

identical to that used to carry the heme had no change in activity (open

symbols). Data are mean + SEM, n = 3; when no error bars are shown,

the SEM lies within the size of the symbol.

measured with and without added BVR (Fig. 4). When no BVR was added, the measured HO activity was 42 +_ 4 units per mg protein. However, the apparent activity of HO increased to 93 units per mg protein when 500 units of exogenous BVR was added to the assay. The maximum amount of added BVR yielded a 5/1 ratio of BVR/HO activities. Thus, the endogenous BVR activity was rate limiting for bilirubin formation when heme ox- ygenase was induced.

Discussion

Our results show that Caco-2 cells contain the enzymes required for catabolism of heme to bilirubin, namely, HO and biliverdin reductase. Indeed, basal levels of ac- tivity of HO are equal those of the reductase. Further- more, HO activities are induced by heme or by metal ions, such as cadmium (Fig. 1), but not by glutethimide, a phenobarbital-like drug that induces the enzyme in liver cultures (data not shown, [8,16]). After induction of HO, biliverdin reductase activity becomes rate-limiting for bilirubin formation (Fig. 4), in contrast to liver, for ex- ample, in which the reductase is present in large excess [5-7].

The increase in activities of HO produced by heine or cadmium treatments were accompanied by, and prob- ably due to, increased amounts of mRNA for HO (Fig.

100

~ i 80 ~-.~ O ~ 60

"6~, 40 ._Z, ,=

"6"~ 20 < O E

0

# r

I . p 4

10 20 30 40 50

BVR in Assay (pl)

Fig. 4. Effects of exogenous biliverdin reductase (BVR) on apparent

activity of heme oxygenase in extracts of Caco-2 cells. Cells were cultu-

red, harvested, and heine oxygenase activity measured as described in

Materials and Methods. Heine (50 gM) was added as an albumin com-

plex 18 hr prior to harvest. 40 gl equals 500 units of biliverdin reducta-

se activity. Data are mean _+ SEM; n = 3. w represents a significant in-

crease over 0 l-tI BVR, and * represents a significant increase over tl3 gl

BVR, Scheffe's test for multiple comparisons. The difference between

20 and 40/_tl BVR was not statistically significant.

2). Thus, as for other types of cells studied by us [16, 28] and others [12, 13], induction of HO in Caco-2 cells by heme or metals probably occurs chiefly as the result of an increase in gene transcription rate. The lack of strict correspondence between the increases in HO mRNA and activity is not surprising, since the mRNA amounts have been measured at only one time point (10 hr). Re- cent results from our laboratory [28] established that berne-dependent induction of HO mRNA in liver cells was transient, with a peak at 5-7 hr, and short-lived. Fur- thermore, the half-life of the message (3 hr) was much shorter than that of the enzyme protein (15 hr).

The ready inducibility of HO makes it unlikely that heme could pass through the Caco-2 monolayer intact, escaping breakdown by the oxygenase. Therefore, heine must apparently be administered parenterally in order to gain access to the circulation or internal organs with the iron-protoporphyrin IX macrocycle intact. Further- more, administration of large doses of heme enterally, in an effort to exceed the capacity of enterocyte HO, is not likely to be feasible, since induction of HO will occur. Thus, despite the difficulties that may attend the admin- istration of heme parenterally [30, 31], the parenteral route is the only one that currently is known to be effica- cious [32-34]. Some of the potentially adverse side-ef- fects of parenteral heme may be obviated by its adminis- tration complexed to albumin [34] or arginine [33].

Page 5: Induction of heme oxygenase in intestinal epithelial cells: studies in Caco-2 cell cultures

It is possible that heme could pass through entero- cytes intact if the HO of these cells were strongly inhib- ited. Some non-iron metalloporphyrins, especially tin or zinc-diiododeuteroporphyrin, -mesoporphyrin, or -pro- toporphyrin, are potent inhibitors of HO [35-39], in- cluding HO of the intestinal mucosa [40-43]. However, recent preliminary results indicated that tin protopor- phyrin blocked transport of heme iron across Caco-2 cells, suggesting that catabolism of heme by HO may be required for iron from heme to be transported through the cell [44].

The mechanism of intestinal iron absorption and de- livery of iron to the basolateral side of the intestinal mu- cosa is still uncharacterized. The ability of heme to in- duce heme oxygenase, and the recent demonstration of directional transport of radioactive iron in Caco-2 cells [44], shows that such cells can be used as a model to in- vestigate heme and iron metabolism of the intestinal ep- ithelium. These cells could also be used to test possible enteral heine delivery systems, as an alternative to pa- rentheral heme administration, for example, in patients with acute porphyria [32-34].

In summary, Caco-2 cells contain HO and biliverdin reductase activities, and, as in other tissues, the mRNA levels of HO are increased by heme or by transition met- als. Further studies to establish the molecular mecha- nisms responsible for this induction are in progress in our laboratory.

Acknowledgements

We thank R. and S. Baker for their generous gift of the Caco-2 cells, S. Shibahara for the generous gift of the plasmid (pHHO1) containing human heme oxygenase cDNA, K. Laruso for assistance with tissue culture, and R. Lambrecht for helpful discussion. Supported by grants from NIH (DK 38825) and from the American Porphyria Foundation.

References

1. Tenhunen R, Marver HS, Schmid R: The enzymatic catabolism of hemoglobin: Stimulation of microsomal heme oxygenase by he- min. J Lab Clin Med 75: 410M21,1970

2. Maines MD: Heme oxygenase: [unction, multiplicity, regulatory mechanisms, and clinical applications. FASEB J 2: 2557-2568, 1988

3. Kikuchi G, Yoshida T: Function and induction of the microsomal heme oxygenase. Mol Cell Biochem 53/54: 163-183, 1983

97

4. Lincoln BC, Aw TY, Bonkovsky HL: Heme catabolism in cultur- ed hepatocytes: evidence that heine oxygenase is the predominant pathway and that a proportion of synthesized heine is converted rapidly to biliverdin. Biochim Biophys Acta 992: 49-58, 1989

5. Tenhunen R, Ross ME, Marver HS, Schmid R: Reduced nicotin- amide-adenine dinucleotide phosphate dependent biliverdin re- ductase: partial purification and characterization. Biochem 9: 298-303, 1970

6. Tenhunen R, Marver HS, Schmid R: The enzymatic conversion of heme to bilirubin by microsomal heine oxygenase, Proc Natl Acad Sci USA 61: 748-755, 1968

7. Tenhunen R, Marver HS, Schmid R: Microsomal heme oxyge- nase: characterization of the enzyme. J Biol Chem 244: 6388-6394, 1969

8. Lincoln BC, Healey JF, Bonkovsky HL: Regulation of hepatic haem metabolism: disparate mechanisms of induction of haem oxygenase by drugs and metals. Biochem J 250:189-196,1988

9. Maines MD, Kappas A: Cobalt induction of hepatic heine oxyge- nase; with evidence that cytochrome P-450 is not essential for this enzyme activity. Proc Natl Acad Sci USA 71: 42034297, 1974

10. Maines MD, Kappas A: The induction of heine oxidation in vari- ous tissues by trace metals: evidence for the catabolism of endoge- nous heine by hepatic heine oxygenase. Ann Clin Res 8 Supp117: 39-46, 1976

11. Sardana MK, Sassa S, Kappas A: Metal ion-mediated regulation of heme oxygenase induction in cultured avian liver cells. J Biol Chem 257: 4806-4811, 1982

12. Shibahara S, Muller RM, Taguchi H: Transcriptional control of rat heme oxygenase by heat shock. J Biol Chem 262: 12889.12892, 1987

13. Alam J, Shibahara S, Smith A: Transcriptional activation of the heme oxygenase gene by heme and cadmium in mouse hepatoma cells. J Biol Chem 264: 6371-6375,1989

14. Maines M, Kappas A: Metals as regulators of heme metabolism. Science 198: 1215-1221, 1977

15. Taketani S, Kohno H, Yoshinaga T, Tokunaga R: The human 32- kDa stress protein induced by exposure to arsenite and cadmium ions is heine oxygenase. FEBS Lett 245: 173.176,1989

16. Cable E, Greene Y, Healey J, Evans C-O, Bonkovsky H: Mecha- nism of synergistic induction of hepatic heine oxygenase by glu- tethimide and iron: Studies in cultured chick embryo liver cells. Biochem Biophys Res Commun 168:176-181,1990

17. Thannoun AM, Mahoney AW, Hendricks DG: Hemoglobin re- generation and iron absorption from meat loaf diets fed to anemic and healthy rats. Nut Reports Int 36: 1273-1284, 1987

18. Thannoun AM, Mahoney AW, Buchowski MS, Hendricks DG: Heme and nonheme iron absorption from meat and meat loaf by anemic and healthy rats. Nut Report Int 37: 487-497, 1988

19. Lynch SR, Dassenko SA, Morck TA, Beard JL, Cook JD: Soy pro- tein product and heme iron absorption in humans. Am J Clin Nutr 41: 13-20, 1985

20. Hallberg L, BjCrn-Rasmussen E, Howard L, Rossander L: Diet- ary heme iron absorption: a discussion of possible mechanisms for the absorption promoting effect of meat and for the regulation of iron absorption. Scand J Gastroent 14: 769-779,1979

21. Bezwoda WR, Bothwell TH, Charlton RW, Torrance JD, Mac- phail AP, Derman DR Mayet F: The relative dietary importance of haem and non-haem iron. S Afr Med J 64: 552-556,1983

22. Kedinger M, Haffen K, Simon-Assmann P: Intestinal tissue and cell culture. Differentiation 36: 71-85,1987

Page 6: Induction of heme oxygenase in intestinal epithelial cells: studies in Caco-2 cell cultures

98

23. Halleux C, Schneider Y-J: Iron absorption by intestinal epithelial cells: 1. Caco-2 cells cultivated in serum-free medium, on poly- ethyleneterephthalate microporous membranes, as an in vitro

model. In Vitro Cell Dev Biol 27A: 293-302, 1991 24. Daniele B, Quaroni A: Effects of epidermal growth factor on dia-

mine oxidase expression and cell growth in Caco-2 cells. Am J Physiot Gastrointest Liver Physiol 261: G669-G676, 1991

25. Sierra EE, Nutter LM: A microassay for heme oxygenase activity using thin-layer chromatography. Anal Biochem 200: 27-30,1992

26. Yoshida T, Biro R Cohen T, Muller RM, Shibahara S: Human heme oxygenase cDNA and induction of its mRNA by hemin. Eu- ropean J Biochem 171: 457461, 1988

27. Hollander MC, Fornace AJ Jr: Estimation of relative mRNA con- tent by filter hybridization to a polythymidylate probe. Biotech- niques 9: 174-179,1990

28. Srivastava KK, Cable lEE, Donohue SE, Bonkovsky HL: Mole- cular basis for heme-dependent induction of heme oxygenase in primary cultures of chick hepatocytes: demonstration of acquired refractoriness to heine. European J Biochem 213: 909-917, 1993

29. Smith PK, Krohn RI, Hermanson GT, Mallia AK, Gartner FH, Provenzano MD, Fujimoto EK, Goeke NM, Olson B J, KLenk DC: Measurement of protein using bicinchoninic acid. Anal Bio- chem 150: 76-85,1985

30. Morris DL, Dudley MD, Pearson RD: Coagulopathy associated with hematin treatment for acute intermittent porphyria. Ann Int Med 95: 700-701, 1981

31. Simionatto CS, Cabal R, Jones RL, Galbraith RA: Thrombo- phlebitis and disturbed hemostasis following administration of in- travenous hematin in normal volunteers. Amer J Med 85: 538- 540, 1988

32. Bonkowsky HL, Tschudy DR Collins A, Doherty J, Bossenmaier I, Cardinal R, Watson C J: Repression of the overproduction of porphyrin precursors in acute intermittent porphyria by intrave- nous infusions of hematin. Proc Natl Acad Sci USA 68: 2725- 2739, 1971

33. Tenhunen R, Tokola O, Lind6n I-B: Haem arginate: a new stable haem compound. J Pharm Pharmacol 39: 780-786, 1987

34. Bonkovsky HL, Healey JF, Lourie AN, Gerron GG: Intravenous heme-albumin in acute intermittent porphyria: evidence for re-

pletion of hepatic hemoproteins and regulatory heme pools. Am J Gastroent 86: 1050-1056, 1991

35. Galbraith RA, Kappas A: Pharmacokinetics of tin-mesoporphy- rin in man and the effects of tin-chelated porphyrins on hyperex- cretion of heme pathway precursors in patients with acute induc- ible porphyria. Hepatology 9: 882-888, 1989

36. Fort FL, Gold J: Phototoxicity of tin protoporphyrin, tin meso- porphyrin, and tin diiododeuteroporphyrin under neonatal pho- totherapy conditions. Pediatrics 84: 1031-1037, 1989

37. Drummond GS, Galbraith RA, Sardana MK, Kappas A: Reduc- tion of the C2 and C4 vinyl groups of Sn-protoporphyrin to form Sn-mesoporphyrin markedly enhances the ability of the metallo- porphyrin to inhibit in vivo heme catabolism. Arch Biochem Bio- phys 255: 64-74, 1987

38. Galbraith RA, Drummond GS, Kappas A: Suppression of biliru- bin production in the Crigler-Najj ar type I syndrome: Studies with the heine oxygenase inhibitor tin-mesoporphyrin. Pediatrics 89: 175-182, 1992

39. Vreman H J, Rodgers PA, Stevenson DK: Zinc protophorphyrin administration for suppression of increased bilirubin production by iatrogenic hemolysis in rhesus neonates. J Pediatr 117: 292-297, 1990

40. Rosenberg DW, Drummond GS, Kappas A: The in vitro and in

vivo inhibition of intestinal heme oxygenase by tin-photop0rphy- rin. Pharmacology 39: 224-229,1989

41. Vreman H J, Gillman M J, Stevenson DK: In vitro inhibition of adult rat intestinal heme oxygenase by metalloporphyrins. Pe- diatr Res 26: 362-365, i989

42. Vreman HJ, Hintz SR, Kim CB, Castillo RO, Stevenson DK: Ef- fects of oral administration of tin and zinc protoporphyrin on neo- natal and adult rat tissue heme oxygenase activity. J Pediatr Gas- troent Nutr 7: 902-906, 1988

43. Rosenberg DW, Kappas A: Actions of orally administered orga- notin compounds on heme metabolism and cytochrome P-450 content and function in intestinal epithelium. Biochem Pharma- col 38: 1155-1161, 1989

44. Khansari MG, Nichols GM, Teaque MD, Alvarez X, Glass J: Ca- co-2 cells: a model for intestinal heme iron uptake and transport. FASEB J 6:A1090 (Abstract), 1992