Aop:131

From AOP-Wiki
Revision as of 14:40, 6 January 2016 by Afarhat (Talk | contribs)

Jump to: navigation, search

AOP Title

Aryl hydrocarbon receptor activation leading to uroporphyria
Short name: AHR activation-uroporphyria

Authors

Amani Farhat and Gillian Manning

Major contributor

Sean W. Kennedy

Status

Please follow the link to snapshots page to view and create Snapshots of this AOP.

Under development: Do not distribute or cite.

This AOP page was last modified on 1/6/2016.

Click here to show/hide revision dates for related pages

Page Revision Date/Time


Abstract

Hepatic uroporphyria is a disorder where the disturbance of heme biosynthesis results in accumulation and excretion of uroporphyrin, heptacarboxylic acid and hexacarboxylic acid: collectively referred to as highly carboxylated porphyrins (HCPs)[1][2][3]. The disorder can be genetically acquired, due to a dysfunction in any of the 7 enzymes involved in the heme biosynthesis pathway [4], or may be chemically induced, which involves the inhibition of uroporphyrinogen decarboxylase (UROD). This adverse outcome pathway (AOP) describes the linkages leading to chemically induced porphyria through the activation of the aryl hydrocarbon receptor (AHR), a transcription factor that plays important endogenous roles in reproduction, liver and heart development, cardiovascular function, immune function and cell cycle regulation [5][6][7][8][9][10][11][12][13][14]. This AOP was developed in accordance with OECD guidelines and demonstrates a high degree of confidence as a qualitative AOP. The quantitative understanding of this AOP however is not yet complete, preventing the accurate prediction of uroporphyria from lower level key events.

Background (optional)

Summary of the AOP

Please follow link to widget page to edit this section. If you manually enter text in this section, it will get automatically altered or deleted in subsequent edits using the widgets.

Molecular Initiating Event

Molecular Initiating Event Support for Essentiality
AHR, Activation Strong

Key Events

Event Support for Essentiality
Uroporphyrinogen, Oxidation Strong
UROD, Inhibition Strong
Highly carboxylated porphyrins, Accumulation Strong
CYP1A2/CYP1A5, Induction Strong

Adverse Outcome

Adverse Outcome
Uroporphyria, N/A

Relationships Among Key Events and the Adverse Outcome

Event Description Triggers Weight of Evidence Quantitative Understanding
Highly carboxylated porphyrins, Accumulation Directly Leads to Uroporphyria, N/A Strong Strong
Uroporphyrinogen, Oxidation Directly Leads to Highly carboxylated porphyrins, Accumulation Strong Weak
Uroporphyrinogen, Oxidation Indirectly Leads to UROD, Inhibition Moderate Weak
CYP1A2/CYP1A5, Induction Directly Leads to Uroporphyrinogen, Oxidation Moderate Weak
AHR, Activation Directly Leads to CYP1A2/CYP1A5, Induction Strong Strong
UROD, Inhibition Indirectly Leads to Uroporphyrinogen, Oxidation Strong Moderate

Network View

   Cytoscape Web will replace the contents of this div with your graph.

Click nodes or edges.

Life Stage Applicability

Life Stage Evidence Links
Adult Strong
Juvenile Strong

Taxonomic Applicability

Name Scientific Name Evidence Links
mouse Mus musculus Strong NCBI
rat Rattus norvegicus Strong NCBI
human Homo sapiens Strong NCBI
chicken Gallus gallus Strong NCBI
herring gull Larus argentatus Strong NCBI
Japanese quail Coturnix japonica Strong NCBI
Common Starling Moderate NCBI

Sex Applicability

Sex Evidence Links

Graphical Representation

Click to upload graphical representation

Overall Assessment of the AOP

Domain of Applicability

Life Stage Applicability, Taxonomic Applicability, Sex Applicability
Elaborate on the domains of applicability listed in the summary section above. Specifically, provide the literature supporting, or excluding, certain domains.

Life Stage Applicability: Uroporphyria occurs following chemical exposure in juvenile or adult individuals. Fetal exposure to dioxin-like compounds causes developmental abnormalities and embryolethality rather than HCP accumulation[15][16][17][18][19]. Turkish children under the age of two that were exposed to HCB through breastmilk passed away from a condition called "pink sore”[20].

Taxonomic Applicability: Although the AHR is highly conserved in evolution[21], chemical-induced uroporphyria has only been detected in birds[1][2][3] and mammals[22] , including an accidental outbreak in humans due to hexachlorobenzen-contaminated grain in the 1950s[20]. Fish are less susceptible to chemical-induced uroporphyria, but elevated levels of HCP have been documented in highly contaminated environments[23].

Sex Applicability: Although this AOP applies broadly to both males and females, sexual dimorphism for uroporphyria has been observed in rats exposed to hexachlorobenzene (HCB). Hepatic uroporphyrin III was markedly increased in female rats exposed to HCB whereas exposed males showed levels of hepatic porphyrins similar to controls[24].

Essentiality of the Key Events

Molecular Initiating Event Summary, Key Event Summary
Provide an overall assessment of the essentiality for the key events in the AOP. Support calls for individual key events can be included in the molecular initiating event, key event, and adverse outcome tables above.

Every Key event in this AOP is absolutely essential for downstream events to occur. A summary of evidence for essrntiality of each key event is given below.

Molecular Initiating Event: AHR activation (Essentiality=strong)

  • Mice with a high-affinity Ahr allele (C57BL/6J ) are much more sensitive to uroporphyria than mice with low-affinity Ahr allele (DBA/2)[25][26][27][28][29];
  • The Ah locus influences the susceptibility of C57BL/6J mice to HCB-induced porphyria[30];
  • Ahr knockout mice (C57BL/6) are resistant to development of porphyria, even in the presence of iron loading[25];
  • Primary hepatocytes of avian species indicate that species that are highly sensitive to AHR activation are more sensitive to uroporphyrin accumulation than species with lower sensitivity to AHR activation[31].

Key Event 1: CYP1A2/Cyp1A5 induction (Essentiality=strong)

  • CYP1A2 knockout in mice prevents chemical-induced uroporphyria[32][33][34];
  • CYP1A2 knockout prevents porphyria in genetically predisposed mice (Hfe-/-, Urod-/+) that normally develop porphyria in absence of external stimuli[35];
  • CYP1A2 levels are correlated with the extent of urophorphyrin accumulation in mice[36];
  • 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and non-ortho substituted PCBs that are potent inducers of CYP1A4/5 cause accumulation of only HCPs in chicken embryonic hepatocytes cultures, whereas PCBs that do not induce CYP1A4/5 cause a porphyrin pattern that is not consistent with inhibition of UROD[37];
  • Common tern (Sterna hirundo) embryonic hepatocyte cultures, which are ~50 to > 1600 times less sensitive than chicken embryonic hepatocyte cultures to CYP1A5 induction by TCDD and PCBs, do not accumulate HCPs upon chemical exposure[31].

It should be noted that a recent study by Davies et al.[25] found that both C57BL/6J mice (susceptible to chemical-induced porphyria) and DBA/2 mice (resistant to porphyria due to polymorphism in AHR gene) showed increased expression of CYP1A2 when exposed to TCDD, even though the DBA/2 strain did not develop porphyria. Furthermore AHR-/- mice showed a mild uroporphyric response in the presence of iron loading and 5-aminolevulinic acid (a heme precursor). These findings suggest that the induction of CYP1A2 is not crucial for chemical-induced porphyria, but a basal level of expression is absolutely essential.

Key Event 2: Uroporphyrinogen oxidation (UROX) (Essentiality=strong)

  • Uroporphyria is characterized biochemically by increased formation of HCPs derived by oxidation of the porphyrinogen substrates of uroporphyrinogen decarboxylase (UROD); secondary to decreased activity of this enzyme in the liver[22];
  • Uroporphomethane, derived from oxidizing a single carbon bridge in uroporphyrinogen, has been identified as the UROD inhibitor that leads to chemically- and genetically-induced uroporphyria in mice[38];
  • UROX activity is positively correlated with uroporphyrin levels in mice[36].

Key Event 3: Uroporphyrinogen decarboxylase (UROD) inhibition (Essentiality=strong)

  • Mutations in the UROD gene that reduce or eliminate UROD activity lead to porphyria in mammals; a decrease in hepatic UROD activity of at least 70% is necessary to observe symptoms from overproduction of porphyrins[22];
  • A marked progressive decrease in UROD enzyme activity is a common feature in animal models of chemical-induced porphyria[22][34][39][40][41];
  • Liver cytosol UROD activity in female rats exposed to HCB was decreased more than 70% and correlated with elevated hepatic uroporphyrin levels, whereas male rats, which did not develop porphyria, showed UROD activity similar to controls[24];
  • UROD activity is inversely proportional to uroporphyrin levels in mice[36];
  • In chicken hepatocytes, the strongest inducers of porphyrin accumulation were also the strongest inhibitors of UROD activity[41];
  • Reduced UROD enzyme activity, not protein levels, is characteristic of uroporphyria in humans and rats[24][42][43].

Key Event 4: Highly carboxylated porphyrin (HCP) accumulation (Essentiality=strong)

  • Under normal heme biosynthesis, porphyrins are only present in trace amounts in the liver; however, in the absence of UROD activity, the oxidation of Uroporphorynogen to uroporphyrins dominates, leading to an accumulation of HCPs;
  • Porphyrins are strongly fluorescent compounds resulting in a characteristic red fluorescence of hepatic tissue under UV light that is proportional to the level of porphyrins[44][45]. Increased urinary excretion of porphyrins is also indicative of their accumulation and can lead to dark red/brown urine[22]. HCPs also accumulate in the skin causing solar hypersensitivity and increased skin fragility[46];
  • HCP accumulation was observed in avian embryo hepatocyte cultures following exposure potent AHR agonists (dioxin-like compounds)[37][47][48][49] and in the livers of Japanese quails and chickens exposed to PCBs[50][51][52];
  • HCP accumulation was evident in mice treated with polyhalogenated aromatic compounds[36] and TCDD-treated rats[25].

Weight of Evidence Summary

Summary Table
Provide an overall summary of the weight of evidence based on the evaluations of the individual linkages from the Key Event Relationship pages.

Dose concordance

Table 1 demonstrates that upstream KEs (monooxygenase activity/quantity) are significantly affected at lower doses than downstream KEs (porphyrin levels). After a 6 month recovery period, CYP450 and hepatic porphyrin levels were dramatically reduced, however, they did not return to normal. Furthermore, urinary porphyrin excretion remained maximally elevatedCite error: Invalid <ref> tag; invalid names, e.g. too many

Uroporphyria Table 1 TCDD recovery.png

Temporal Concordance

Table 2 demonstrates that upstream KEs (CYP1A2 expression and UROD inhibition) are significantly affected at earlier time-points than downstream KEs (porphyrin levels). These studies also show that upstream KEs are more sensitive to change than downstream KEs; ddY mice showed a 44% reduction in UROD activity but did not develop uroporphyria[25][53].

Uroporphyria Table 2 sensitive vs resistant.png

Key Events Relationships

Table 3 shows a sampling of the literature that demonstrates changes in KEs at multiple levels of organization leading to uroporphyria. The use of animal models resistant to porphyria (low AHR affinity of AHR/CYP1A2 knockout) illustrates the essentiality of these KEs in for downstream effects.

Uroporphyria Table 3 KER Summary.png

Quantitative Considerations

Summary Table
Provide an overall discussion of the quantitative information available for this AOP. Support calls for the individual relationships can be included in the Key Event Relationship table above.

Considerations for Potential Applications of the AOP (optional)

References

  1. 1.0 1.1 Fox, G. A., Norstrom, R. J., Wigfield, D. C., and Kennedy, S. W. (1988) Porphyria in herring gulls: A biochemical response to chemical contamination of great lakes food chains. ‘’Environmental Toxicology and Chemistry’’ ‘’’7’’’ (10), 831-839
  2. 2.0 2.1 Kennedy, S. W., and Fox, G. A. (1990) Highly carboxylated porphyrins as a biomarker of polyhalogenated aromatic hydrocarbon exposure in wildlife: Confirmation of their presence in Great Lakes herring gull chicks in the early 1970s and important methodological details. Chemosphere 21 (3), 407-415.
  3. 3.0 3.1 Kennedy, S. W., Fox, G. A., Trudeau, S. F., Bastien, L. J., and Jones, S. P. (1998) Highly carboxylated porphyrin concentration: A biochemical marker of PCB exposure in herring gulls. Marine Environmental Research 46 (1-5), 65-69.
  4. Thunell, S. (2000) Porphyrins, porphyrin metabolism and porphyrias. I. Update. Scand. J. Clin. Lab Invest 60 (7), 509-540.
  5. Baba, T., Mimura, J., Nakamura, N., Harada, N., Yamamoto, M., Morohashi, K., and Fujii-Kuriyama, Y. (2005) Intrinsic function of the aryl hydrocarbon (dioxin) receptor as a key factor in female reproduction. Mol. Cell Biol. 25 (22), 10040-10051.
  6. Denison, M. S., Soshilov, A. A., He, G., DeGroot, D. E., and Zhao, B. (2011) Exactly the same but different: promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol. Sci. 124 (1), 1-22.
  7. Fernandez-Salguero, P. M., Pineau, T., Hilbert, D. M., McPhail, T., Lee, S. S., Kimura, S., Nebert, D. W., Rudikoff, S., Ward, J. M., and Gonzalez, F. J. (1995) Immune system impairment and hepatic fibrosis in mice lacking the dioxin-binding Ah receptor. Science 268 (5211), 722-726.
  8. Ichihara, S., Yamada, Y., Ichihara, G., Nakajima, T., Li, P., Kondo, T., Gonzalez, F. J., and Murohara, T. (2007) A role for the aryl hydrocarbon receptor in regulation of ischemia-induced angiogenesis. Arterioscler. Thromb. Vasc. Biol. 27 (6), 1297-1304.
  9. Lahvis, G. P., Lindell, S. L., Thomas, R. S., McCuskey, R. S., Murphy, C., Glover, E., Bentz, M., Southard, J., and Bradfield, C. A. (2000) Portosystemic shunting and persistent fetal vascular structures in aryl hydrocarbon receptor-deficient mice. Proc. Natl. Acad. Sci U. S. A 97 (19), 10442-10447.
  10. Mimura, J., Yamashita, K., Nakamura, K., Morita, M., Takagi, T. N., Nakao, K., Ema, M., Sogawa, K., Yasuda, M., Katsuki, M., and Fujii-Kuriyama, Y. (1997) Loss of teratogenic response to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in mice lacking the Ah (dioxin) receptor. Genes Cells 2 (10), 645-654.
  11. Omiecinski, C. J., Vanden Heuvel, J. P., Perdew, G. H., and Peters, J. M. (2011) Xenobiotic metabolism, disposition, and regulation by receptors: from biochemical phenomenon to predictors of major toxicities. Toxicol. Sci. 120 Suppl 1, S49-S75.
  12. Schmidt, J. V., Su, G. H., Reddy, J. K., Simon, M. C., and Bradfield, C. A. (1996) Characterization of a murine Ahr null allele: involvement of the Ah receptor in hepatic growth and development. Proc. Natl. Acad. Sci U. S. A 93 (13), 6731-6736.
  13. Thackaberry, E. A., Gabaldon, D. M., Walker, M. K., and Smith, S. M. (2002) Aryl hydrocarbon receptor null mice develop cardiac hypertrophy and increased hypoxia-inducible factor-1alpha in the absence of cardiac hypoxia. Cardiovasc. Toxicol. 2 (4), 263-274.
  14. Zhang, N., Agbor, L. N., Scott, J. A., Zalobowski, T., Elased, K. M., Trujillo, A., Duke, M. S., Wolf, V., Walsh, M. T., Born, J. L., Felton, L. A., Wang, J., Wang, W., Kanagy, N. L., and Walker, M. K. (2010) An activated renin-angiotensin system maintains normal blood pressure in aryl hydrocarbon receptor heterozygous mice but not in null mice. Biochem. Pharmacol. 80 (2), 197-204.
  15. Brunström, B. (1988) Sensitivity of embryos from duck, goose, herring gull, and various chicken breeds to 3,3',4,4'-tetrachlorobiphenyl. Poultry science 67 (1), 52-57.
  16. Carro, T., Taneyhill, L. A., and Ottinger, M. A. (2013) The effects of an environmentally relevant 58 congener polychlorinated biphenyl (PCB) mixture on cardiac development in the chick embryo. Environ. Toxicol. Chem. 23(6), 1325-31
  17. Gilbertson, M. (1983) Etiology of chick edema disease in herring gulls in the lower Great Lakes. Chemosphere 12 (3), 357-370.
  18. Lavoie, E. T., and Grasman, K. A. (2007) Effects of in ovo exposure to PCBs 126 and 77 on mortality, deformities and post-hatch immune function in chickens. J. Toxicol. Environ. Health A 70 (6), 547-558.
  19. Wells, P. G., Lee, C. J., McCallum, G. P., Perstin, J., and Harper, P. A. (2010) Receptor- and reactive intermediate-mediated mechanisms of teratogenesis. Handb. Exp. Pharmacol. (196), 131-162.
  20. 20.0 20.1 Cripps, D. J., Peters, H. A., Gocmen, A., and Dogramici, I. (1984) Porphyria turcica due to hexachlorobenzene: a 20 to 30 year follow-up study on 204 patients. Br. J Dermatol. 111 (4), 413-422.
  21. Kewley, R. J., Whitelaw, M. L., and Chapman-Smith, A. (2004) The mammalian basic helix-loop-helix/PAS family of transcriptional regulators. Int. J. Biochem. Cell Biol. 36 (2), 189-204.
  22. 22.0 22.1 22.2 22.3 22.4 Smith, A. G., and Elder, G. H. (2010) Complex gene-chemical interactions: hepatic uroporphyria as a paradigm. Chem. Res. Toxicol. 23 (4), 712-723.
  23. Wainwright, J. S., Hopkins, K. M., Bums Jr., T.A., and Di Giulio, R. T. (1995) Investigation of potential biomarkers of exposure to bleached kraft mill effluent in North Carolina rivers. Durham, NC.
  24. 24.0 24.1 24.2 Mylchreest, E., and Charbonneau, M. (1997) Studies on the mechanism of uroporphyrinogen decarboxylase inhibition in hexachlorobenzene-induced porphyria in the female rat. Toxicol. Appl. Pharmacol. 145 (1), 23-33.
  25. 25.0 25.1 25.2 25.3 25.4 Davies, R., Clothier, B., Robinson, S. W., Edwards, R. E., Greaves, P., Luo, J., Gant, T. W., Chernova, T., and Smith, A. G. (2008) Essential role of the AH receptor in the dysfunction of heme metabolism induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Chem. Res. Toxicol. 21 (2), 330-340.
  26. Jones, K. G., and Sweeney, G. D. (1977) Association between induction of aryl hydrocarbon hydroxylase and depression of uroporphyrinogen decarboxylase activity. Res. Commun. Chem. Pathol. Pharmacol. 17 (4), 631-637.
  27. Jones, K. G., and Sweeney, G. D. (1980) Dependence of the porphyrogenic effect of 2,3,7,8-tetrachlorodibenzo(p)dioxin upon inheritance of aryl hydrocarbon hydroxylase responsiveness. Toxicol. Appl. Pharmacol. 53 (1), 42-49.
  28. Smith, A. G., Francis, J. E., Kay, S. J., and Greig, J. B. (1981) Hepatic toxicity and uroporphyrinogen decarboxylase activity following a single dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin to mice. Biochem. Pharmacol. 30 (20), 2825-2830.
  29. Smith, A. G., Clothier, B., Robinson, S., Scullion, M. J., Carthew, P., Edwards, R., Luo, J., Lim, C. K., and Toledano, M. (1998) Interaction between iron metabolism and 2,3,7,8-tetrachlorodibenzo-p-dioxin in mice with variants of the Ahr gene: a hepatic oxidative mechanism. Mol. Pharmacol. 53 (1), 52-61.
  30. Hahn, M. E., Gasiewicz, T. A., Linko, P., and Goldstein, J. A. (1988) The role of the Ah locus in hexachlorobenzene-induced porphyria. Studies in congenic C57BL/6J mice. Biochem. J. 254 (1), 245-254.
  31. 31.0 31.1 Lorenzen, A., Shutt, J. L., and Kennedy, S. W. (1997) Sensitivity of common tern (Sterna hirundo) embryo hepatocyte cultures to CYP1A induction and porphyrin accumulation by halogenated aromatic hydrocarbons and common tern egg extracts. Archives of Environmental Contamination and Toxicology 32 (2), 126-134.
  32. Greaves, P., Clothier, B., Davies, R., Higginson, F. M., Edwards, R. E., Dalton, T. P., Nebert, D. W., and Smith, A. G. (2005) Uroporphyria and hepatic carcinogenesis induced by polychlorinated biphenyls-iron interaction: absence in the Cyp1a2(-/-) knockout mouse. Biochem. Biophys. Res. Commun. 331 (1), 147-152.
  33. Sinclair, P. R., Gorman, N., Dalton, T., Walton, H. S., Bement, W. J., Sinclair, J. F., Smith, A. G., and Nebert, D. W. (1998) Uroporphyria produced in mice by iron and 5-aminolaevulinic acid does not occur in Cyp1a2(-/-) null mutant mice. Biochem. J. 330 ( Pt 1'), 149-153.
  34. 34.0 34.1 Smith, A. G., Clothier, B., Carthew, P., Childs, N. L., Sinclair, P. R., Nebert, D. W., and Dalton, T. P. (2001) Protection of the Cyp1a2(-/-) null mouse against uroporphyria and hepatic injury following exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol. Appl. Pharmacol. 173 (2), 89-98.
  35. Phillips, J. D., Kushner, J. P., Bergonia, H. A., and Franklin, M. R. (2011) Uroporphyria in the Cyp1a2-/- mouse. Blood Cells Mol. Dis. 47 (4), 249-254.
  36. 36.0 36.1 36.2 36.3 Gorman, N., Ross, K. L., Walton, H. S., Bement, W. J., Szakacs, J. G., Gerhard, G. S., Dalton, T. P., Nebert, D. W., Eisenstein, R. S., Sinclair, J. F., and Sinclair, P. R. (2002) Uroporphyria in mice: thresholds for hepatic CYP1A2 and iron. Hepatology 35 (4), 912-921.
  37. 37.0 37.1 Lorenzen, A., Kennedy, S. W., Bastien, L. J., and Hahn, M. E. (1997) Halogenated aromatic hydrocarbon-mediated porphyrin accumulation and induction of cytochrome P4501A in chicken embryo hepatocytes. Biochemical Pharmacology 53 (3), 373-384.
  38. Phillips, J. D., Bergonia, H. A., Reilly, C. A., Franklin, M. R., and Kushner, J. P. (2007) A porphomethene inhibitor of uroporphyrinogen decarboxylase causes porphyria cutanea tarda. Proc. Natl. Acad. Sci. U. S. A 104 (12), 5079-5084.
  39. Kawanishi, S., Mizutani, T., and Sano, S. (1978) Induction of porphyrin synthesis in chick embryo liver cell culture by synthetic polychlorobiphenyl isomers. Biochim. Biophys. Acta 540 (1), 83-92.
  40. Miranda, C. L., Henderson, M. C., Wang, J. L., Nakaue, H. S., and Buhler, D. R. (1992) Comparative effects of the polychlorinated biphenyl mixture, Aroclor 1242, on porphyrin and xenobiotic metabolism in kidney of Japanese quail and rat. Comp Biochem. Physiol C. 103 (1), 149-152.
  41. 41.0 41.1 Sano, S., Kawanishi, S., and Seki, Y. (1985) Toxicity of polychlorinated biphenyl with special reference to porphyrin metabolism. Environ. Health Perspect. 59, 137-143.
  42. Elder, G. H., and Sheppard, D. M. (1982) Immunoreactive uroporphyrinogen decarboxylase is unchanged in porphyria caused by TCDD and hexachlorobenzene. Biochem. Biophys. Res. Commun. 109 (1), 113-120.
  43. Elder, G. H., Urquhart, A. J., De Salamanca, R. E., Munoz, J. J., and Bonkovsky, H. L. (1985) Immunoreactive uroporphyrinogen decarboxylase in the liver in porphyria cutanea tarda. Lancet 2 (8449), 229-233.
  44. Kennedy, S. W. (1988) Studies on Porphyria as an Indicator of Polyhalogenated Aromatic Hydrocarbon Exposure. Carleton University
  45. Lundvall, O., and Enerback, L. (1969) Hepatic fluorescence in porphyria cutanea tarda studied in fine needle aspiration biopsy smears. J Clin Pathol 22 (6), 704-709.
  46. Frank, J., and Poblete-Gutierrez, P. (2010) Porphyria cutanea tarda--when skin meets liver. Best. Pract. Res. Clin Gastroenterol. 24(5), 735-745.
  47. Lambrecht, R. W., Sinclair, P. R., Bement, W. J., Sinclair, J. F., Carpenter, H. M., Buhler, D. R., Urquhart, A. J., and Elder, G. H. (1988) Hepatic uroporphyrin accumulation and uroporphyrinogen decarboxylase activity in cultured chick-embryo hepatocytes and in Japanese quail (Coturnix coturnix japonica) and mice treated with polyhalogenated aromatic compounds. Biochem. J. 253 (1), 131-138.
  48. Marks, G. S., Zelt, D. T., and Cole, S. P. (1982) Alterations in the heme biosynthetic pathway as an index of exposure to toxins. Can. J. Physiol Pharmacol. 60 (7), 1017-1026.
  49. Sassa, S., Sugita, O., Ohnuma, N., Imajo, S., Okumura, T., Noguchi, T., and Kappas, A. (1986) Studies of the influence of chloro-substituent sites and conformational energy in polychlorinated biphenyls on uroporphyrin formation in chick-embryo liver cell cultures. Biochem. J. 235 (1), 291-296.
  50. Goldstein, J. A., McKinney, J. D., Lucier, G. W., Hickman, P., Bergman, H., and Moore, J. A. (1976) Toxicological assessment of hexachlorobiphenyl isomers and 2,3,7,8,-tetrachlorodibenzofuran in chicks. II. Effects on drug metabolism and porphyrin accumulation. Toxicol. Appl. Pharmacol. 36 (1), 81-92.
  51. McKinney, J. D., Chae, K., Gupta, B. N., Moore, J. A., and Goldstein, H. A. (1976) Toxicological assessment of hexachlorobiphenyl isomers and 2,3,7,8 tetrachlorodibenzofuran in chicks. I. Relationship of chemical parameters. Toxicol. Appl. Pharmacol. 36 (1), 65-80.
  52. Miranda, C. L., Henderson, M. C., Wang, J. L., Nakaue, H. S., and Buhler, D. R. (1987) Effects of polychlorinated biphenyls on porphyrin synthesis and cytochrome P-450-dependent monooxygenases in small intestine and liver of Japanese quail. J. Toxicol. Environ. Health 20 (1-2), 27-35.
  53. eki, Y., Kawanishi, S., and Sano, S. (1987). Mechanism of PCB-induced porphyria and yusho disease. Ann. N. Y. Acad. Sci. 514, 222-234.