Find information on thousands of medical conditions and prescription drugs.

Deferoxamine

Deferoxamine, otherwise known as desferrioxamine or desferal, is a chelating agent used to remove excess iron from the body. It acts by binding free iron in the bloodstream and enhancing its elimination in the urine. By removing excess iron, the agent reduces the damage done to various organs and tissues, such as the liver. more...

Home
Diseases
Medicines
A
B
C
D
Dacarbazine
Dactinomycin
Dalmane
Danazol
Dantrolene
Dapoxetine
Dapsone
Daptomycin
Daraprim
Darvocet
Darvon
Daunorubicin
Daunorubicin
Daypro
DDAVP
Deca-Durabolin
Deferoxamine
Delsym
Demeclocycline
Demeclocycline
Demerol
Demulen
Denatonium
Depakene
Depakote
Depo-Provera
Desferal
Desflurane
Desipramine
Desmopressin
Desogen
Desogestrel
Desonide
Desoxyn
Desyrel
Detrol
Dexacort
Dexamethasone
Dexamfetamine
Dexedrine
Dexpanthenol
Dextran
Dextromethorphan
Dextromoramide
Dextropropoxyphene
Dextrorphan
Diabeta
Diacerein
Diacetolol
Dial
Diamox
Diazepam
Diazoxide
Dibenzepin
Diclofenac
Diclohexal
Didanosine
Dieldrin
Diethylcarbamazine
Diethylstilbestrol
Diethyltoluamide
Differin
Diflucan
Diflunisal
Digitoxin
Digoxin
Dihydrocodeine
Dihydroergotamine
Dihydrotachysterol
Dilantin
Dilaudid
Diltahexal
Diltiazem
Dimenhydrinate
Dimercaprol
Dimetapp
Dimethyl sulfoxide
Dimethyltryptamine
Dimetridazole
Diminazene
Diovan
Dioxybenzone
Diphenhydramine
Diphenoxylate
Dipipanone
Dipivefrine
Diprivan
Diprolene
Diproteverine
Dipyridamole
Disulfiram
Disulfiram
Dizocilpine
Dobutamine
Docetaxel
Docusate sodium
Dofetilide
Dolasetron
Dolobid
Dolophine
Domperidone
Donepezil
Dopamine
Dopram
Doral
Doramectin
Doriden
Dornase alfa
Doryx
Dostinex
Doxapram
Doxazosin
Doxepin
Doxil
Doxil
Doxorubicin
Doxy
Doxycycline
Doxyhexal
Doxylamine
Drisdol
Drixoral
Dronabinol
Droperidol
Drospirenone
Duloxetine
Durabolin
Duragesic
Duraphyl
Duraquin
Dutasteride
Dv
Dyclonine
E
F
G
H
I
J
K
L
M
N
O
P
Q
R
S
T
U
V
W
X
Y
Z

Deferoxamine is used to treat acute iron poisoning, especially in small children. Treatment with this agent is also frequently necessary in patients with certain types of chronic anemia (e.g. thalassemia and myelodysplastic syndrome) who require many blood transfusions, which can greatly increase the amount of iron in the body. Administration for chronic conditions is generally accomplished by subcutaneous injection (SQ) over a period of 8-12 hours daily. Administation of deferoxamine after acute intoxication may color the urine a pinkish red, a phenomenon termed "'vin rose urine".

Apart from in iron toxicity, deferoxamine is also used to treat aluminum toxicity (an excess of aluminum in the body) in certain patients.

Read more at Wikipedia.org


[List your site here Free!]


Extracellular superoxide dismutase protects lung development in hyperoxia-exposed newborn mice
From American Journal of Respiratory and Critical Care Medicine, 2/1/03 by Ahmed, Mohamed N

We tested the hypothesis that targeted transgenic overexpression of human extracellular superoxide dismutase (EC-SOD) would preserve alveolar development in hyperoxia-exposed newborn mice. We exposed newborn transgenic and wild-type mice to 95% oxygen (O^sub 2^) or air x 7 days and measured bronchoalveolar lavage cell counts, and lung homogenate EC-SOD, oxidized and reduced glutathione, and myeloperoxidase. We found that total EC-SOD activity in transgenic newborn mice was approximately 2.5x the wild-type activity. Hyperoxia-exposed transgenic mice had less pulmonary neutrophil influx and oxidized glutathione than wild-type littermates at 7 days. We measured alveolar surface and volume density in animals exposed to 14 days more of air or 60% O^sub 2^. Hyperoxia-exposed transgenic EC-SOD mice had significant preservation of alveolar surface and volume density compared with wild-type littermates. After 7 days 95% O^sub 2^ + 14 days 60% O^sub 2^, lung inflammation measured as myeloperoxidase activity was reduced to control levels in all treatment groups.

Keywords: bronchopulmonary dysplasia; superoxide dismutase; hyperoxia; newborn infant; transgenic mice

In extreme prematurity, respiratory failure due to deficient alveolar development and surfactant production could be complicated by diminished antioxidant stores and enzymatic antioxidant inducibility (1). Inflammatory responses to perinatal infection or lung injury cause further cellular damage by increasing oxidant and proteolytic reactions. Bronchopulmonary dysplasia (BPD) remains a common complication of extreme prematurity, despite advances in surfactant therapy and intensive care. It has recently been characterized as a failure of complete alveolar development (2). Proposed mechanisms include oxidant damage to biomolecules that lead to indirect effects on cell proliferation. Hyperoxia impairs alveolar formation (3), and the relative deficiency of antioxidant defenses may render 21% Fio^sub 2^"hyperoxic" in premature newborns (4). Inflammation contributes by effects of leukocyte-mediated proteolytic enzymatic cellular damage and by releasing oxygen radicals produced during the respiratory burst that further oxidize biomolecules (5). Oxidant injury and inflammation are thought to be dominant mechanisms in the pathogenesis of BPD (6).

Premature newborns are relatively deficient in stores of antioxidant vitamins A and E (7). Although relative pulmonary antioxidant enzyme immaturity has not been corroborated in humans, cord blood antioxidant levels decline with decreasing gestational age (8). There is ample evidence that clinically encountered oxidant stress in premature newborns is accompanied by lung protein and lipid oxidation (9, 10). The rationale for supplementing antioxidant capacity is also supported by studies that supplied either iron chelators (deferoxamine) or iron-binding proteins (transferrin) and demonstrated reduced oxidant stress and improved lung growth, as recently reviewed by Frank and Sosenko (7). At present, antioxidant vitamin prophylaxis does not prevent BPD in a baboon model, (11) and clinical trials of antioxidant vitamin treatment (12) or antioxidant enzyme supplementation (13) have had only modest success or have been ineffective at preventing BPD.

Failure of exogenous antioxidant enzyme prevention of BPD may be due to nonspecific effects or ineffective dose delivery to vulnerable lung compartments (14, 15). Overexpression of manganese superoxide dismutase (SOD) conferred superior survival in hyperoxia-exposed transgenic adult mice (16).

We recently demonstrated protection from hyperoxia in human extracellular SOD (hEC-SOD) transgenic hyperoxia-- exposed adult mice that typically express two- to threefold increases in total EC-SOD activity over wild-type littermates (17). In contrast, EC-SOD knockout mice had increased mortality and lung injury when exposed to hyperoxia (18). ECSOD may confer the particular advantage of targeting the vulnerable extracellular milieu of the lung epithelium, potentially providing superior protection against oxidant-mediated cellular injury, as recently suggested by Berrington and colleagues (14). This may permit normal proliferation and differentiation in newborns, which is known to be inhibited by hyperoxia (19).

To determine whether sustained expression of EC-SOD would promote normal lung development during severe oxidative stress, we evaluated the effect of transgenic overexpression of hEC-SOD in a mouse model of hyperoxia-- induced chronic lung disease. In this model, newborn mice are exposed to Fio^sub 2^ = 0.95 at birth for 1 week and then recovered at Fio^sub 2^ = 0.6 for two more weeks to induce an oxidative and inflammatory insult followed by more indolent oxidative stress.

METHODS

We used B6C3 transgenic mice, (17) with the human SP-C promoter targeting human EC-SOD expression mainly in alveolar epithelium (20). Experiments were performed according to institutional guidelines for animal care and use at Duke University Medical Center.

Acute Hyperoxia Exposure

Newborn mice (wild-type and transgenic) were randomly assigned to air and oxygen exposure (Fio^sub 2^= 95% for 7 days) beginning at birth. Nursing dams were alternated between air and oxygen-exposed litters (21). Animals were killed with 150 mg/kg intraperitoneal sodium pentobarbital after exposure. Genotype analysis was performed using polymerase chain reaction (17). Body weights, survival, and lung weights were recorded.

EC-SOD Expression by Activity

Tissues from three or four mice in each treatment group were weighed and homogenized in a buffer containing an anti-protease cocktail (see details in the online supplement). Supernatants obtained after centrifugation were pooled and passed over a concanavalin-A Sepharose column to isolate EC-SOD, and activity was measured three or four times by inhibition of xanthine/xanthine oxidase-mediated cytochrome c reduction, as described previously (17).

EC-SOD Expression by Immunoblot

Lungs were homogenized, and 20 jig total lung protein from each animal was analyzed by western blotting, detected with anti-mouse ECSOD or anti-human EC-SOD as described in the online supplement. EC-SOD Expression by Immunohistochemistry

Lungs from five mice in each treatment group were fixed as described previously (22). Sections from inflation-fixed lung were immunostained for hEC-SOD. We evaluated the histochemical signal as described previously using rabbit anti-human EC-SOD 1:3,000 and detected them with biotinylated goat anti-rabbit 1:4,000 (23). Preimmune rabbit serum was used as a control for the EC-SOD immunostaining.

Total and Oxidized Glutathione

Total and oxidized glutathione was measured in lung homogenates from a minimum of four pups/treatment group by high-performance liquid chromatography according to a procedure slightly modified from Paroni and coworkers (24).

Bronchoalveolar Lavage

Five wild-type and five trangenic pups/treatment group underwent bronchoalveolar lavage (BAL) to determine cell counts using 0.5 ml 0.9% sodium chloride, I mM ethylenediamine tetraacetic acid, and repeated four times before pooling (25). Total cell counts from BAL fluid were counted in a hemacytometer, then cells were cytocentrifuged, and Wright stained. At least 300 cells were counted to obtain a differential leukocyte cell count.

Myeloperoxidase

Myeloperoxidase activity was measured spectrophotometrically in whole lung homogenates from five animals/treatment group by reaction with o-dianisidine dye using a microplate assay described previously (21, 22).

Chronic Hyperoxia Exposure

In other animals, after 7 days air or hyperoxia, the FIo^sub 2^ in the hyperoxia groups was reduced from 95 to 60% until age 21 days, at which time all pups were killed and lungs inflation-fixed and sectioned as described previously. Survival, body weights, and lung weights were measured. Five animals/treatment group were evaluated morphometrically to determine alveolar surface density and alveolar volume density as estimates of surface area and alveolar number, as described previously and in the online supplement (26). Activities of EC-SOD were measured in lung homogenates pooled from four animals/treatment group to achieve adequate detection. Whole lung myeloperoxidase measurements were made in five animals/treatment group as in the acute hyperoxia studies referred to previously.

Statistical Analysis

Groups were compared using analysis of variance, and a post hoc Tukey-Kramer test was performed to determine statistical differences. We accepted p values less than 0.05 as significant, assuming an alpha error = 0.05 and Beta error = 0.10. Analysis was performed using JMP Software version 3.2 (SAS, Cary, NC).

RESULTS

Effects of Acute Hyperoxia on Newborn Mouse Body and Lung Weights

There was no mortality in the air-exposed groups. At 7 days, 95% oxygen exposure resulted in 17% mortality for wild-type and 15% for transgenic mice (p = 0.50, not significant) and decreased body weight in both genotypes: wild-type 2.9+/- 0.6 g, n = 56, versus transgenic 2.8+/- 0.3 g, n = 62 (mean +/- SEM). Hyperoxia exposure decreased lung weight: air, 76.7 8.0 versus hyperoxia: 62.0 + 1.4, p value less than 0.05, and n = 9/group. Genotype had no effect on lung weight.

EC-SOD expression. We found that EC-SOD activity in air-- exposed transgenic mice was more than double the activity compared with wild-type littermates, as shown in Figure 1. This increase was further induced by hyperoxia at 7 days in transgenic mice. Immunoblotting showed that human EC-SOD was detected only in the transgenic mice, but mouse EC-SOD was detected in both wild-type and transgenic mice. Hyperoxia had no effect on either human or mouse EC-SOD protein expression measured by immunoblot (Figure 1). In the transgenic newborn mice, hEC-SOD immunohistochemical expression was primarily located in alveolar epithelium and also in bronchiolar epithelium (Figure 2). No hEC-SOD immunostaining was seen in wild-type littermates.

Glutathione. Total glutathione in lung homogenates was increased by hyperoxia exposure at 7 days but less so in EC-SOD transgenic mice. Lung homogenates from mice exposed to hyperoxia for 7 days had elevated oxidized glutathione compared with air-exposed mice. The elevation in the EC-SOD transgenic mice was slightly less than in the wild-type mice. Hyperoxia-exposed transgenic mice had significantly lower levels of oxidized and reduced glutathione than hyperoxia-exposed wild-type littermates (Figure 3).

Inflammation. Transgenic mice exposed to hyperoxia for 7 days showed a qualitative reduction in the inflammatory cell infiltrate and preservation of septal integrity (Figure 4). Hyperoxia increased BAL leukocyte accumulation in both wild-type and transgenic mice, but transgenic mice had significantly reduced BAL leukocytes compared with wild-type littermates. Transgenic hEC-SOD expression also prevented accumulation of MPO activity in whole lung homogenates from hyperoxia-exposed mice compared with wild-type littermates.

Chronic Hyperoxia

Survival, body and lung weights. Chronic hyperoxia impaired growth: hyperoxia, 8.9 +/- 0.4 g, n = 15, versus air, 11.4 +/- 0.7 g, n = 22, p value less than 0.001. Genotype had no effect on body weight or lung weight at 21 days in either air or hyperoxia exposure. After 21 days, lung wet weights were similar in air and hyperoxia exposed groups, hyperoxia, 97 +/- 3 mg, versus air 89 +/- 5mg, n = 14/group p = 0.16. There were no differences in late mortality in wild-type versus transgenic hyperoxia-exposed mice.

Alveolar development. There were no differences in surface density or volume density between 21-day air-exposed wild-type and transgenic mice. Hyperoxia exposure for 21 days significantly reduced alveolar surface density and alveolar volume density in wild-type mice but not in EC-SOD transgenic mice (Figure 5).

Inflammation. Tissue myeloperoxidase accumulation was similar among all groups at Day 21 (Figure 6). There was a trend toward decreased myeloperoxidase accumulation in the hEC-- SOD transgenic animals in both treatment groups, but this did not reach statistical significance.

EC-SOD activity. EC-SOD transgenic mice maintained elevated total lung EC-SOD activity at 21 days. Transgenic animals had more than threefold the activity levels of those observed in wild-type littermates, as in the 7-day exposed newborns (Figure 7). Hyperoxia-exposure induced an additional increase in total EC-SOD activity compared with air-exposed transgenic animals. Hyperoxia had no effect on EC-SOD activity in wild-- type littermates.

DISCUSSION

Overexpression of human EC-SOD in hyperoxia-exposed adult mice confers improved survival and decreased inflammation (17). In our model of chronic lung disease, we sought to use a sublethal injury that shares some features of human chronic lung disease, namely oxidant stress, inflammation, and impaired alveolar development (16). We found that overexpression of human EC-SOD targeted to pulmonary epithelium partly prevented glutathione oxidation, and reduced hyperoxia-induced interstitial and airway inflammation at 7 days. Later alveolar development-as determined by alveolar surface and volume density-was preserved in hEC-SOD transgenic mice exposed to hyperoxia for 21 days.

EC-SOD expression may confer particular protection against oxidant stress. Mutant mice lacking EC-SOD are more susceptible to hyperoxia-induced injury and inflammation (18). Transgenic mice overexpressing mitochondrial (manganese) SOD showed improved survival and attenuation of acute injury (16). Hyperoxia-exposure studies in transgenic mice overexpressing cytosolic (copper-zinc) SOD conferred similar benefits (27). Because EC-SOD is a secreted protein, it may dismutate superoxide in vulnerable lung compartments relatively inaccessible to intracellular antioxidants, such as the surface of alveolar epithelium or the alveolar hypophase (28).

Inflammation may exacerbate oxidant stress and thus contribute to propagation of lung injury, and it may impede repair and cell proliferation. Our previous studies in hyperoxia-exposed adult EC-SOD overexpressing mice showed attenuation of inflammatory cytokines and adhesion molecules (17). We have recently shown that blockade of neutrophil influx protects against adverse hyperoxia effects on lung development in newborn rats (26). The preservation of alveolar development we observed at 21 days in the present studies may be partly attributable to the early reduction of inflammation we observed as reduced BAL fluid leukocyte counts and tissue myeloperoxidase in transgenic mice at 7 days.

Overexpression of EC-SOD preserved lung development during oxidative stress, evident as partly preserved alveolar surface and volume density. This would indicate that secondary septation is likely preserved because decreased septation would result in lower surface density. There were no differences in surface density between air-exposed newborn wild-type and transgenic mice. This suggests that EC-SOD transgenic animals do not have constitutively superior alveolar development in the absence of oxidative stress.

The beneficial effects of EC-SOD overexpression may be mediated indirectly by alterations in ROS signaling of cellular processes. Hydrogen peroxide, produced spontaneously or by SOD by single electron reduction of superoxide, has been increasingly recognized as a mediator of in vitro apoptosis and may alter cell proliferation (29). Reduction of hydrogen peroxide by exogenous catalase in vitro can abrogate the effect of cellular mitogens, as recently reviewed by Jankov and coworkers(30). Potential effects of SOD on hydrogen peroxide-mediated signaling would likely depend greatly on the timing, concentration, and location of hydrogen peroxide.

Hyperoxia induced even higher levels of total EC-SOD activity in the transgenic animals at 7 and 21 days. The transferred hEC-SOD gene is under control of the human SP-C promoter in the transgenic mice. It is known that hyperoxia induces increased SP-C messenger RNA in mice, and we speculate that similar mechanisms may be enhancing the human SP-C promoter-controlled transgenic EC-SOD overexpression (31, 32). In contrast, human EC-SOD protein levels measured by immunoblot did not appear to be induced by hyperoxia, but this was not strictly quantified. Cellular protein processing in the transgenic mice may alter the EC-SOD activity without changing the total lung EC-SOD accumulation. The wild-type littermates showed no hyperoxia-induced changes in either EC-SOD activity or mouse EC-SOD accumulation in whole lung.

We conclude that overexpression of hEC-SOD in neonatal mice protects against hyperoxia-induced inflammation at 7 days and impairment of lung development at 21 days. Although the specific mechanism is still unclear, it likely includes preservation of reduced glutathione-important for lung oxidant-antioxidant equilibrium-and attenuation of inflammation that may cause cell death and alveolar destruction. Newborn mice exposed to hyperoxia for 21 days showed maintenance of both alveolar volume and surface density. We speculate that correctly targeted SOD overexpression will prevent adverse effects of oxidant stress on lung development and may protect lung development in newborns at risk to develop BPD.

Acknowledgment: The authors gratefully acknowledge the technical assistance of Mary Whorton and Kathryn Auten.

References

1. Jobe AH, Ikegami M. Mechanisms initiating lung injury in the preterm. Early Hum Dev 1998;53:81-94.

2. Jobe A. The new BPD: an arrest of lung development. Pediatr Res 1999; 46:641-643.

3. Randell SH, Mercer RR, Young SL. Neonatal hyperoxia alters the pulmonary alveolar and capillary structure of 40-day-old rats. Am J Pathol 1990;136:1259-1266.

4. Massaro DJ, Massaro GD. Regulation of pulmonary alveoli formation. In: Bland RD, Coalson J, editors. Chronic lung disease in early infancy, lung biology in health and disease. New York: Marcel Dekker; 2000. p. 479-492.

5. Lorant DE, Albertine K, Bohnsack JF. Chronic lung disease of early infancy: role of neutrophils. In: Bland RD, Coalson J, editors. Chronic lung disease in early infancy, lung biology in health and disease. New York: Marcel Dekker; 2000. p. 793-812.

6. Coalson J. Pathology of chronic lung disease of early infancy. In: Bland RD, Coalson J, editors. Chronic lung disease in early infancy, lung biology in health and disease. New York: Marcel Dekker; 2000. p. 85-124.

7. Frank L, Sosenko IR. Oxidants and antioxidants: what role do they play in chronic lung disease? In: Bland RD, Coalson J. editors. Chronic lung disease in early infancy, lung biology in health and disease. New York: Marcel Dekker; 2000. p. 257-284.

8. Rogers S, Witz G. Anwar M, Hiatt M, Hegyi T. Antioxidant capacity and oxygen radical diseases in the preterm newborn. Arch Pediatr Adolesc Med 2000;154:544-548.

9. Schock BC, Sweet DG, Ennis M, Warner JA, Young IS, Halliday HL. Oxidative stress and increased type-IV collagenase levels in bronchoalveolar lavage fluid from newborn babies. Pediatr Res 2001;50:29-33.

10. Singh SK, Tandon A, Kumari S, Ravi RN, Ray GN, Batra S. Changes in anti-oxidant enzymes and lipid peroxidation in hyaline membrane disease. Indian J Pediatr 1998;65:609-614.

11. Berger TM, Frei B, Rifai N, Avery ME, Suh J, Yoder BA, Coalson JJ. Early high dose antioxidant vitamins do not prevent bronchopulmo

nary dysplasia in premature baboons exposed to prolonged hyperoxia: a pilot study. Pediatr Res 1998;43:719-726.

12. Tyson JE, Wright LL, Oh W, Kennedy KA, Mele L, Ehrenkranz RA, Stoll BJ. Lemons JA, Stevenson DK, Bauer CR, et aL Vitamin A supplementation for extremely-low-birth-weight infants: National Institute of Child Health and Human Development Neonatal Research Network. N Engl J Med 1999;340:1962-1968.

13. Davis JM, Rosenfeld WN, Richter SE, Parad MR, Gewolb IH, Spitzer AR. Carlo WA, Couser RJ, Price A, Flaster E. et al. Safety and pharmacokinetics of multiple doses of recombinant human CuZn superoxide dismutase administered intratracheally to premature neonates with respiratory distress syndrome. Pediatrics 1997:100:24-30.

14. Berrington WR, Tarpey MM, Freeman BA, Pitt BR. Site- and mechanism-targeted interventions for tissue free radical injury. In: Bland RD, Coalson J, editors. Chronic lung disease in early infancy, lung biology in health and disease. New York: Marcel Dekker; 2000. p. 883-909.

15. Welty SE, Smith CV. Rationale for antioxidant therapy in premature infants to prevent bronchopulmonary dysplasia. Nutr Rev 2001;59: 10-17.

16. Wispe JR, Warner BB, Clark JC, Dey CR, Neuman J, Glasser SW, Crapo JD, Chang LY, Whitsett JA. Human Mn-superoxide dismutase in pulmonary epithelial cells of transgenic mice confers protection from oxygen injury. J Biol Chem 1992:267:23937-23941.

17. Folz RJ, Abushamaa AM, Suliman HB. Extracellular superoxide dismutase in the airways of transgenic mice reduces inflammation and attenuates lung toxicity following hyperoxia. J Clin Invest 1999:103:1055-- 1066.

18. Carlsson LM, Johnsson J, Edlung T, Marklund SL. Mice lacking extracellular superoxide dismutase are more sensitive to hyperoxia. Proc Natl Acad Sci USA 1995;92:6246-6268.

19. Randell SH, Young SL. Unique features of the immature lung. In: Bland RD, Coalson J, editors. Chronic lung disease in early infancy, lung biology in health and disease. New York: Marcel Dekker; 2000. p. 377-403.

20. Korfhagen TR, Glasser SW, Wert SE, Bruno MD, Daugherty CC, McNeish JD, Stock JL, Potter SS, Whitsett JA. Cis-acting sequences from a human surfactant protein gene confer pulmonary-specific gene expression in transgenic mice. Proc Natl Acad Sci USA 1990;87:6122-- 6126.

21. Deng H, Mason SN, Auten RL Jr. Lung inflammation in hyperoxia can be prevented by antichemokine treatment in newborn rats. Am J Respir Crit Care Med 2000;162:2316-2323.

22. Lundberg C, Arfors KE. Polymorphonuclear leukocyte accumulation in inflammatory dermal sites as measured by "Cr-labeled cells and myeloperoxidase. Inflammation 1983;7:247-255.

23. Aderibigbe AO, Thomas RF, Mercer RR, Auten R. Brief exposure to 95% oxygen alters surfactant protein D and mRNA in adult rat alveolar and bronchiolar epithelium. Am J Respir Cell Mol Biol 1999;20:219-- 227.

24. Paroni R, De Vecchi E, Cighetti G, Arcelloni C, Fermo I. Grossi A, Bonini P. HPLC with o-phthalaldehyde precolumn derivatization to measure total, oxidized, and protein-bound glutathione in blood, plasma, and tissue. Clin Chem 1995:41:448-454.

25. Ahmed MN, Grayck EN, Suliman H, Mason SN, Auten RL. Overexpression of extracellular superoxide dismutase (EC-SOD) protects against hyperoxia-induced airway inflammation in transgenic newborn mice [abstract]. Pediatr Res 2001;49:300a.

26. Auten RL Jr, Mason SN, Tanaka DT, Welty-Wolf K, Whorton MH. Antineutrophil chemokine preserves alveolar development in hyperoxia-- exposed newborn rats. Am J Physiol Lung Cell Mol Physiol 2001; 281:L336-L344.

27. White CW. Avraham KB, Shanley PF, Groner Y. Transgenic mice with expression of elevated levels of copper-zinc superoxide dismutase in the lungs are resistant to pulmonary oxygen toxicity. J Clin Invest 1991;87:2162-2168.

28. Cantin AM, Hubbard RC, Crystal RG. Glutathione deficiency in the epithelial lining fluid of the lower respiratory tract in idiopathic pulmonary fibrosis. Am Rev Respir Dis 1989;139:370-372.

29. Hoidal JR. Hoidal MK. Proteolytic enzymes and their inhibitors in lung health and disease. In: Lenfant C, editor. Chronic lung disease in early infancy. New York: Marcel Dekker; 2000. p. 859-882.

30. Jankov RP, Negus A, Tanswell AK. Antioxidants as therapy in the newborn: some words of caution. Pediatr Res 2001;50:681-687.

31. Minoo P, Segura L, Coalson JJ. King RJ, DeLemos RA. Alterations in surfactant protein gene expression associated with premature birth and exposure to hyperoxia. Am J Physiol 1991;261:L386L392.

32. Allred TF, Mercer RR, Thomas RF, Deng H, Auten RL. Brief 95% O^sub 2^ exposure effects on surfactant protein and mRNA in rat alveolar and bronchiolar epithelium. Am J Physiol 1999;276:1-999-1-1009.

Mohamed N. Ahmed, Hagir B. Suliman, Rodney J. Folz, Eva Nozik-Grayck, Maria L. Golson, S. Nicholas Mason, and Richard L. Auten

Departments of Pediatrics, Medicine, Anesthesiology, Cell Biology, Duke University Medical Center, Durham, North Carolina

Correspondence and requests for reprints should be addressed to Richard L. Auten, M.D., DUMC Box 3373, Duke University Medical Center, Durham, NC 27710. E-mail: auten@duke.edu

Copyright American Thoracic Society Feb 1, 2003
Provided by ProQuest Information and Learning Company. All rights Reserved

Return to Deferoxamine
Home Contact Resources Exchange Links ebay