Direct Oxidation and Covalent Binding of Isoniazid to Rodent Liver and Human Hepatic Microsomes: Humans Are More Like Mice than Rats
Abstract

Isoniazid (INH) is associated with serious liver injury and autoimmunity. Classic studies in rats indicated that a reactive metabolite of acetylhydrazine is responsible for the covalent binding and toxicity of INH. Studies in rabbits suggested that hydrazine might be the toxic species. However, these models involved acute toxicity with high doses of INH, and INH-induced liver injury in humans has very different features than such animal models. In this study, we demonstrated that a reactive metabolite of INH itself can covalently bind in the liver of mice and also to human liver microsomes. Covalent binding also occurred in rats, but it was much less than that in mice. We were able to trap the reactive metabolite of INH with N-α-acetyl-l-lysine in incubations with human liver microsomes. This suggests that the reactive intermediate of INH that leads to covalent binding is a diazohydroxide rather than a radical or carbocation because those reactive metabolites would be too reactive to trap in this way. Treatment of mice or rats with INH for up to 5 weeks did not produce severe liver injury. The alanine transaminase assay (ALT) is inhibited by INH, and other assays such as glutamate and sorbitol dehydrogenase (SDH) were better biomarkers of INH-induced liver injury. High doses of INH (200 and 400 mg/kg/day) for one week produced steatosis in rats and an increase in SDH, which suggests that it can cause mitochondrial injury. However, steatosis was not observed when INH was given at lower doses for longer periods of time to either mice or rats. We propose that covalent binding of the parent drug can contribute to INH-induced hepatotoxicity and autoimmunity. We also propose that these are immune-mediated reactions, and there are clinical data to support these hypotheses.
Introduction
Materials and Methods
Trapping of the Reactive Metabolite
INA Activated Ester, INH Dimer, and Isonicotinic Acid-N-α-acetyl-l-lysine (INA-NAL) Adduct
Conjugation of INH to BSA/Blue Carrier Protein and Antibody Production
Animal Treatment
INH, AcHz, and Hz Blood Level Measurements
Western Blotting
Histopathology and Immunohistochemistry
Statistical Analysis
Results
HLM | NADPH | INH | NAL | adducts observed | tr (min) |
---|---|---|---|---|---|
+ | – | – | – | no peak | – |
+ | + | – | – | no peak | – |
+ | + | + | – | INA-INH | 9.9 |
+ | + | + | + | INA-NAL | 9.0 |
INA-INH = the INH dimer and INA-NAL = the N-α-acetyl-l-lysine adduct.
Figure 1

Figure 1. INH adduct formation in human liver microsomes. INH = isoniazid, NAL = N-α-acetyl-l-lysine, HLM = human liver microsomes, INA-INH = INH dimer, and INA-NAL = N-α-acetyl-l-lysine adduct.
Figure 2

Figure 2. Specificity of the anti-INH antibody. (A) Antibody specificity was tested by ELISA. The plate was either coated with BSA modified with INA (BSA-INA) or BSA alone. Preimmune serum (SPre) or serum after immunization with Blue Carrier Protein modified with INA (SAft) diluted at 1:100,000 was used as the primary antibody. In the third and fifth column, the primary antibody was preincubated with INH or NAL, respectively, at a concentration of 200 μM for 30 min at room temperature. (B) Antibody was tested for cross-reactivity with binding due to AcHz. Female C57BL/6 mice were treated with either INH or AcHz (Fisher Scientific) by gavage for 7 days at 50 mg/kg/day (n = 2 for each group). There was no binding to hepatic proteins from untreated control and AcHz-treated mice, whereas hepatic proteins from INH-treated mice showed a large number of bands modified with INH. (C) Antiserum was tested for specificity on Western blots by preincubation with INH at 200 μM or 2 mM for 1 h at 4 °C, which prevented binding to the INH-modified hepatic proteins. (D) Binding of the anti-INH serum to INH-modified liver proteins on Western blots was compared to that of the preimmune serum from the same animal.
Figure 3

Figure 3. Covalent binding of INH to hepatic proteins in mice. (A) Male C57BL/6 (n = 4) untreated controls or treated with INH (0.2% of INH by weight in food) for 5 weeks. (B) Male vs female C57BL/6 (n = 4) treated with INH (0.2% of INH by weight in food) for 5 weeks. (C) Female C57BL/6 mice (n = 3) treated for 5 weeks vs Balb C mice (n = 3) treated for 3 weeks, both with 0.2% INH by weight in food. (D) Female Balb C mice treated with INH; either by gavage at 100 mg/kg/day (n = 2) for a period of 1, 3, or 7 days or with 0.2% INH by weight in food for 3 weeks. (E,F) Immunohistochemical staining in the livers of female C57BL/6 mice in untreated control vs those treated with 0.2% INH by weight in food for 5 weeks. Magnification: 5×.
Figure 4

Figure 4. Covalent binding of INH to hepatic proteins in rats. (A) BN rats (n = 3) in untreated controls or treated with INH either by gavage at a dose of 150 mg/kg/day or with 0.2% INH by weight in food for 5 weeks. (B) Wistar rats (n = 4) in untreated controls or treated with INH by gavage at 150 mg/kg/day for 4 weeks. (C) Comparison of covalent binding between male Wistar rats and male BN rats (n = 4) treated with INH by gavage at a dose of 150 mg/kg/day for 4 and 5 weeks, respectively. (D) Comparison of covalent binding between male C57BL/6 mice (n = 4) treated with 0.2% INH by weight in food for 5 weeks vs male Wistar rats (n = 4) treated with INH by gavage at a dose of 150 mg/kg/day for 4 weeks.
In Vitro Binding of INH to Human, Mouse, and Rat Liver Microsomes
Figure 5

Figure 5. In vitro covalent binding of INH to hepatic microsomes (A) to HLM with and without an NADPH-generating system; (B) to MLM with and without a NADPH-generating system; (C) to HLM as a function of time and INH concentration; (D) to MLM as a function of time and INH concentration; (E,F) direct comparison of binding between HLM and MLM at INH concentrations of 10 and 100 μM, respectively.
Figure 6

Figure 6. Comparison of INH covalent binding to rat, mouse, and human liver microsomes at an INH concentration of 50 μM in the presence or absence of a NADPH generating system.
Treatment of Wistar and BN Rats with INH
Figure 7

Figure 7. Serum SDH activities and body weights in INH-treated BN rats. (A) SDH activity in BN rats treated with INH by gavage at 150 mg/kg/day, 50 mg/kg twice daily, or given at 0.2% INH by weight in food. (B) Body weight of BN rats given INH as in panel A. Values represent the mean ± SE. Analyzed for statistical significance by two-way ANOVA. Significantly different from the control group (*p < 0.05; **p < 0.01; ***p < 0.001).
food consumption (g/day) | avg animal weight range (g) | avg INH intake in mg/kg/day | ||
---|---|---|---|---|
BN rats | control | 15.6 ± 1.1 | 240–300 | |
gavaged at 150 mg/kg/day | 4.7 ± 1.0*** | 220–240 | ||
gavaged at 50 mg/kg twice daily | 3.5 ± 2.0*** | 180–240 | ||
INH given in food | 9.1 ± 0.6*** | 210–240 | 79–87 | |
mice | male C57BL/6 | |||
control | 5.0 ± 0.3 | 22–26 | ||
INH | 4.6 ± 0.3 | 22–24 | 386–420 | |
female C57BL/6 | ||||
control | 5.1 ± 0.3 | 16–20 | ||
INH | 4.2 ± 0.3** | 16–20 | 420–525 | |
male Balb C | ||||
control | 4.6 ± 0.3 | 22–23 | ||
INH | 2.9 ± 0.2*** | 18–20 | 290–322 | |
female Balb C | ||||
control | 4.6 ± 0.1 | 17–19 | ||
INH | 3.1 ± 0.3*** | 15–17 | 367–416 |
INH was given to BN rats (n = 4) by gavage as specified or at 0.2% INH by weight in food for 5 weeks maximum. C57BL/6 (n = 4) mice were treated with INH at 0.2% by weight in food for 5 weeks, and Balb C (n = 4) were treated at the same dose for 3 weeks. Animal body weight range was estimated from Figures 5 and 6, and the amount of food consumed was calculated on the basis of weight range and by assuming homogenous mixing of drug with food. Values represent the mean ± standard error of the mean (SE) with 4 animals per group. The data were analyzed for statistical significance by the Mann-Whitney U test. Significantly different from control group (*p < 0.05; **p < 0.01; ***p < 0.001).
Treatment of C57BL/6 and Balb C Mice with INH
Figure 8

Figure 8. GLDH activities and body weights in mice. (A–D) Male and female C57BL/6 mice were treated at 0.2% INH by weight in food for 5 weeks. (E–H) Male and female Balb C mice were treated at 0.2% INH by weight in food for 3 weeks. Values represent the mean ± SE from 4 animals per group. The data were analyzed for statistical significance by two-way ANOVA. Significantly different from the control group (*p < 0.05; **p < 0.01; ***p < 0.001).
Figure 9

Figure 9. Serum concentrations of INH and INH/AcHz ratio in mice and rats. INH was given at a dose of 0.2% by weight in food for up to one week. Values represent the mean ± SE for mice (n = 5) and for rats (n = 4). The data were analyzed for statistical significance by the Mann–Whitney U test. Significantly different groups (*p < 0.05).
Discussion
Supporting Information
Covalent binding of INH to hepatic proteins in mice; liver histology of mice and rats treated with INH; percentage of BSA and blue carrier protein lysine residues modified by INA-NHS; GLDH and ALT activities after 5 weeks of treatment of BN rats with INH; and SDH and ALT activities in Balb C and C57BL/6 mice. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
AcHz | acetylhydrazine |
AcINH | acetylisoniazid |
ALT | alanine aminotransferase |
BSA | bovine serum albumin |
BN | Brown Norway |
CNS | central nervous system |
GLDH | glutamate dehydrogenase |
H&E | hematoxylin and eosin |
HLM | human liver microsomes |
Hz | hydrazine |
INA | isonicotinic acid |
INH | isoniazid |
INA-BSA | isonicotinic acid coupled to bovine serum albumin |
INA-INH | isonicotinic acid coupled to isoniazid |
INA-NAL | isonicotinic acid coupled to N-α-acetyl-l-lysine |
INA-NHS | reactive N-hydroxysuccinimide ester of isonicotinic acid |
LC-MS | liquid chromatography coupled to mass spectrometry |
MLM | mouse liver microsomes |
NAL | N-α-acetyl-l-lysine |
NHS | N-hydroxysuccinimide |
RLM | rat liver microsomes |
SDH | sorbitol dehydrogenase |
References
This article references 26 other publications.
- 1Center for Disease Control and Prevention (2010) Severe isoniazid-associated liver injuries among persons being treated for latent tuberculosis infection: United States, 2004–2008 MMWR 59, 224– 229
- 2Mitchell, J. R., Zimmerman, H. J., Ishak, K. G., Thorgeirsson, U. P., Timbrell, J. A., Snodgrass, W. R., and Nelson, S. D. (1976) Isoniazid liver injury: clinical spectrum, pathology, and probable pathogenesis Ann. Intern. Med. 84, 181– 192[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE287jsVGgsw%253D%253D&md5=3ba6aa4dfbaca727dac947da5c80b913Isoniazid liver injury: clinical spectrum, pathology, and probable pathogenesisMitchell J R; Zimmerman H J; Ishak K G; Thorgeirsson U P; Timbrell J A; Snodgrass W R; Nelson S DAnnals of internal medicine (1976), 84 (2), 181-92 ISSN:0003-4819.The clinical spectrum of isoniazid-induced liver injury seems to be clinically, biochemically, and histologically indistinguishable from viral hepatitis, except that the injury occurs primarily in persons older than 35 years. A possible relation between susceptibility of patients to isoniazid liver injury and rapid metabolism (acetylation) of the drug has been found. Examination of isoniazid metabolites showed that patients with rapid acetylator phenotype hydrolyze much more isoniazid to isonicotinic acid and the free hydrazine moiety than do slow acetylators. The hydrazine moiety liberated from isoniazid is primarily acetylhydrazine, and studies in animals show this metabolite to be converted to a potent acylating agent that produces liver necrosis. It seems likely that formation of chemically reactive metabolites is also the biochemical event initiating isoniazid liver injury in man. Recognition of the seriousness of isoniazid hepatic injury, not readily accepted at first, has led to revisions in the uses of isoniazid prophylaxis.
- 3Snodgrass, W., Potter, W. Z., T., J., Jollow, D. J., and Mitchell, J. R. (1974) Possible mechanism of isoniazid-related hepatic injury Clin. Res. 22, 323AGoogle ScholarThere is no corresponding record for this reference.
- 4Timbrell, J. A., Mitchell, J. R., Snodgrass, W. R., and Nelson, S. D. (1980) Isoniazid hepatoxicity: the relationship between covalent binding and metabolism in vivo J. Pharmacol. Exp. Ther. 213, 364– 369[PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3cXktlOis7s%253D&md5=22987ead13a85569c3b9156212ef5d21Isoniazid hepatotoxicity: the relationship between covalent binding and metabolism in vivoTimbrell, John A.; Mitchell, Jerry R.; Snodgrass, Wayne R.; Nelson, Sidney D.Journal of Pharmacology and Experimental Therapeutics (1980), 213 (2), 364-9CODEN: JPETAB; ISSN:0022-3565.The relation between the hepatotoxicity and metab. of isoniazid (I) [54-85-3] and its metabolites, acetylisoniazid [1078-38-2] and acetylhydrazine [1068-57-1], was investigated. Toxic doses of acetylisoniazid and acetylhydrazine, radiolabeled in the Ac group, were found to bind covalently to liver protein in vivo. This binding was mediated by the microsomal enzyme system as indicated by the effects of pretreatments altering the activity of these enzymes. Metabolic studies revealed that the pretreatments increased the metab. of the acetylhydrazine moiety of Ac-labeled acetylisoniazid and of acetylhydrazine itself by the microsomal enzyme system. Pretreatment with the acyl amidase inhibit r bis-p-nitrophenyl phosphate inhibited the hydrolysis of acetylisoniazid to isonicotinic acid plus acetylhydrazine and concomitantly decreased the covalent binding of Ac-labeled acetylisoniazid. The changes in the metab. of I, acetylisoniazid, and acetylhydrazine effected by various pretreatments paralleled changes in the severity of the hepatic necrosis caused by the compds. Apparently, acetylhydrazine is the metabolite responsible for the hepatic necrosis caused by I, and microsomal metab. of acetylhydrazine in vivo leads to the prodn. of a reactive acylating species capable of reacting covalently with tissue macromols.
- 5Nelson, S. D., Mitchell, J. R., Timbrell, J. A., Snodgrass, W. R., and Corcoran, G. B. (1976) Isoniazid and iproniazid: activation of metabolites to toxic intermediates in man and rat Science 193, 901– 903Google ScholarThere is no corresponding record for this reference.
- 6Noda, A., Hsu, K. Y., Noda, H., Yamamoto, Y., and Kurozumi, T. (1983) Is isoniazid-hepatotoxicity induced by the metabolite, hydrazine? J. UOEH 5, 183– 190[PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3sXltlegsr8%253D&md5=b6d11194b2a7b901544e4a012b8f330eIs isoniazid-hepatotoxicity induced by the metabolite, hydrazine?Noda, Atsuko; Hsu, Kuang Yang; Noda, Hiroshi; Yamamoto, Yuzo; Kurozumi, TakeshiJournal of UOEH (1983), 5 (2), 183-90CODEN: JOUOD4; ISSN:0387-821X.Treatment of tuberculosis with a combination of isoniazid (I) [54-85-3] and rifampicin (II) [13292-46-1] has been previously shown to cause more severe hepatitis than treatment with I alone. Pretreatment of rabbits with II had no effect on subsequent blood concns. of I or its metabolites acetylisoniazid [1078-38-2], acetylhydrazine [1068-57-1], or diacetylhydrazine [3148-73-0], but it significantly decreased hydrazine [302-01-2] levels. Furthermore, none of the metabolites, except hydrazine, themselves caused hepatic necrosis in rabbits. Since II increased hepatic cytochrome P 450 levels, a mechanism is proposed whereby the II-induced increase in the hepatic enzyme is responsible for the oxidative degrdn. of hydrazine to hepatotoxic products. Hydrazine is thus suggested as the I metabolite responsible for I-induced hepatitis.
- 7Sarich, T. C., Youssefi, M., Zhou, T., Adams, S. P., Wall, R. A., and Wright, J. M. (1996) Role of hydrazine in the mechanism of isoniazid hepatotoxicity in rabbits Arch. Toxicol. 70, 835– 840[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XntVWgs7o%253D&md5=3c75759c797c5170391113797a55c78eRole of hydrazine in the mechanism of isoniazid hepatotoxicity in rabbitsSarich, Troy C.; Youssefi, Mohammed; Zhou, Ting; Adams, Stephen P.; Wall, Richard A.; Wright James M.Archives of Toxicology (1996), 70 (12), 835-840CODEN: ARTODN; ISSN:0340-5761. (Springer)By using a model of isoniazid (INH)-induced hepatotoxicity in rabbits, in which INH-induced hepatotoxicity manifests as hepatic necrosis, hepatic steatosis (hepatic fat accumulation), and hypertriglyceridemia (elevated plasma triglycerides), the severity of these measures of toxicity were compared with plasma levels of INH, acetylhydrazine, and hydrazine. Plasma INH and acetylhydrazine were not correlated with markers of INH-induced hepatic necrosis or fatty changes. Plasma hydrazine at 32 h was correlated with plasma argininosuccinic acid lyase (ASAL) activity as area under the curve (r2 = 0.54, P < 0.002) and log plasma ASAL activity at 48 h after the 1st dose of INH (r2 = 0.53, p < 0.005), but not with fatty changes. Hydrazine, and not INH or acetylhydrazine, is most likely involved in the pathogenic mechanism of hepatic necrosis.
- 8Hofstra, A. H., Li-Muller, S. M., and Uetrecht, J. P. (1992) Metabolism of isoniazid by activated leukocytes. Possible role in drug-induced lupus Drug. Metab. Dispos. 20, 205– 210Google ScholarThere is no corresponding record for this reference.
- 9Metushi, I. G., Cai, P., Zhu, X., Nakagawa, T., and Uetrecht, J. P. (2011) A fresh look at the mechanism of isoniazid-induced hepatotoxicity Clin. Pharmacol. Ther. 89, 911– 914[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmtlagtbc%253D&md5=e444a505f3b86ff0d51408447de8509eA Fresh Look at the Mechanism of Isoniazid-Induced HepatotoxicityMetushi, I. G.; Cai, P.; Zhu, X.; Nakagawa, T.; Uetrecht, J. P.Clinical Pharmacology & Therapeutics (New York, NY, United States) (2011), 89 (6), 911-914CODEN: CLPTAT; ISSN:0009-9236. (Nature Publishing Group)A review. Isoniazid (INH)-induced hepatotoxicity remains a significant clin. problem, and the current mechanistic hypothesis is incomplete; it is simply referred to as metabolic idiosyncrasy, which is believed to involve cytotoxicity caused by bioactivation of acetylhydrazine, a metabolite of INH. However, this hypothesis is based on animal studies, involving characteristics that are very different from those that pertain to hepatotoxicity in humans, such as delayed onset. This article provides a fresh look at this issue.
- 10Bernhardt, P. V., Chin, P., and Richardson, D. R. (2001) Unprecedented oxidation of a biologically active aroylhydrazone chelator catalysed by iron(III): serendipitous identification of diacylhydrazine ligands with high iron chelation efficacy J. Biol. Inorg. Chem. 6, 801– 809[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXptVKns7k%253D&md5=aa8610e965785caee57ef79dd935bd75Unprecedented oxidation of a biologically active aroylhydrazone chelator catalysed by iron(III): serendipitous identification of diacylhydrazine ligands with high iron chelation efficacyBernhardt, Paul V.; Chin, Piao; Richardson, Des R.JBIC, Journal of Biological Inorganic Chemistry (2001), 6 (8), 801-809CODEN: JJBCFA; ISSN:0949-8257. (Springer-Verlag)Ligands of the 2-pyridylcarbaldehyde isonicotinoylhydrazone class show high iron (Fe) sequestering efficacy and have potential as agents for the treatment of Fe overload disease. We have investigated the mechanisms responsible for their high activity. X-ray crystallog. studies show that the tridentate chelate 2-pyridylcarbaldehyde isonicotinoylhydrazone undergoes an unexpected oxidn. to isonicotinoyl(picolinoyl)hydrazine when complexed with FeIII. In contrast, in the absence of FeIII, the parent hydrazone is not oxidized in aerobic aq. soln. To examine whether the diacylhydrazine could be responsible for the biol. effects of 2-pyridylcarbaldehyde isonicotinoylhydrazone, their Fe chelation efficacy was compared. In contrast to its parent hydrazone, the diacylhydrazine showed little Fe chelation activity. Potentiometric titrns. suggested that this might be because the diacylhydrazine was charged at physiol. pH, hindering its access across membranes to intracellular Fe pools. In contrast, the Fe complex of this diacylhydrazine was charge neutral, which may allow facile movement through membranes. These data allow a model of Fe chelation for this compd. to be proposed: the parent aroylhydrazone diffuses through cell membranes to bind Fe and is subsequently oxidized to the diacylhydrazine complex which then diffuses from the cell. Other diacylhydrazine analogs that were charge neutral at physiol. pH demonstrated high Fe chelation efficacy. Thus, for this class of ligands, the charge of the chelator appears to be an important factor for detg. their ability to access intracellular Fe. The results of this study are significant for understanding the biol. activity of 2-pyridylcarbaldehyde isonicotinoylhydrazone and for the design of novel diacylhydrazine chelators for clin. use.
- 11Christensen, J. B. (2001) A simple method for synthesis of active esters of isonicotinic and picolinic acids Molecules 6, 47– 51Google ScholarThere is no corresponding record for this reference.
- 12Zhu, X., Li, J., Liu, F., and Uetrecht, J. P. (2011) Involvement of T helper 17 cells in D-penicillamine-induced autoimmune disease in Brown Norway rats J. Toxicol. Sci. 120, 331– 338[Crossref], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3M3otFaqsA%253D%253D&md5=a6bc54b2bcf093d828caca7239327f7dInvolvement of T helper 17 cells in D-penicillamine-induced autoimmune disease in Brown Norway ratsZhu Xu; Li Jinze; Liu Feng; Uetrecht Jack PToxicological sciences : an official journal of the Society of Toxicology (2011), 120 (2), 331-8 ISSN:.Idiosyncratic drug reactions (IDRs) are poorly understood, but their clinical characteristics suggest that they are immune mediated. Penicillamine-induced autoimmunity in Brown Norway rats has been utilized as an animal model for mechanistic studies of one type of IDR because it closely mimics the autoimmune syndromes that it causes in humans. Our previous work suggested that it is T-cell mediated. It has been shown that T helper 17 (Th17) cells play a central role in many types of autoimmune diseases. This study was designed to test whether Th17 cells are involved in the pathogenesis of penicillamine-induced autoimmunity and to establish an overall serum cytokine/chemokine profile for this IDR. In total, 24 serum cytokines/chemokines were determined and revealed a dynamic process. In sick animals, interleukin (IL) 6 and transforming growth factor-β1, known to be driving forces of Th17 differentiation, were consistently increased at both early and late stages of penicillamine treatment; however, no significant changes in these cytokines were observed in animals that did not develop autoimmunity. IL-17, a characteristic cytokine produced by Th17 cells, was increased in sick animals at both the messenger RNA and serum protein level. In addition, serum concentrations of IL-22, another characteristic cytokine produced by Th17 cells, were found to be elevated. Furthermore, the percentage of IL-17-producing CD4 T cells was significantly increased but only in sick animals. These data strongly suggest that Th17 cells are involved in penicillamine-induced autoimmunity. Such data provide important mechanistic clues that may help to predict which drug candidates will cause a relatively high incidence of such autoimmune IDRs.
- 13O’Brien, P. J., Slaughter, M. R., Polley, S. R., and Kramer, K. (2002) Advantages of glutamate dehydrogenase as a blood biomarker of acute hepatic injury in rats Lab. Anim. 36, 313– 321[Crossref], [PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XlslWis74%253D&md5=1c01835b9a443a27354674278028024eAdvantages of glutamate dehydrogenase as a blood biomarker of acute hepatic injury in ratsO'Brien, P. J.; Slaughter, M. R.; Polley, S. R.; Kramer, K.Laboratory Animals (2002), 36 (3), 313-321CODEN: LBANAX; ISSN:0023-6772. (Royal Society of Medicine Press Ltd.)In a recent study in rats, alanine aminotransferase (ALT), the preferred plasma biomarker of hepatocellular injury in rats, was ineffective at detecting marked hepatic necrosis produced by acetaminophen. In contrast, glutamate dehydrogenase (GLDH) was markedly elevated. Accordingly, these enzymes were comprehensively evaluated as plasma biomarkers of hepatocellular injury in rats using several other models of hepatic injury, including partial hepatectomy and exposure to methapyrilene, dexamethasone, cyproterone, isoniazid, lead nitrate, and Wyeth-14643. Other enzymes also evaluated were aspartate aminotransferase (AST), sorbitol dehydrogenase (SDH), and the hepatobiliary marker alk. phosphatase (ALP). Compared to plasma ALT increases, plasma GLDH increases were up to 10-fold greater, up to 3-fold more persistent, and occurred at times following hepatocellular injury when plasma ALT was not increased. Plasma GLDH activity was not inhibited by the test compds., whereas ALT was substantially inhibited by both isoniazid and lead nitrate. While plasma GLDH activity was unaffected by induction, ALT was induced by cyproterone and dexamethasone, and ALP was induced by Wyeth-14643 and partial hepatectomy. GLDH was concluded to be a more effective biomarker of acute hepatic injury than ALT, AST, SDH or ALP in the rat, based primarily on the large increase following hepatocellular injury, prolonged persistence in the blood following injury, high sensitivity for detection of injury (including pre-necrotic injury), high tissue specificity, and lower susceptibility to inhibition or induction.
- 14Ng, W., Lobach, A. R., Zhu, X., Chen, X., Liu, F., Metushi, I. G., Sharma, A., Li, J., Cai, P., Ip, J., Novalen, M., Popovic, M., Zhang, X., Tanino, T., Nakagawa, T., Li, Y., and Uetrecht, J. (2012) Animal models of idiosyncratic drug reactions Adv. Pharmacol. 63, 81– 135Google ScholarThere is no corresponding record for this reference.
- 15Fukino, K., Sasaki, Y., Hirai, S., Nakamura, T., Hashimoto, M., Yamagishi, F., and Ueno, K. (2008) Effects of N-acetyltransferase 2 (NAT2), CYP2E1 and glutathione-S-transferase (GST) genotypes on the serum concentrations of isoniazid and metabolites in tuberculosis patients J. Toxicol. Sci. 33, 187– 195[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXos1aksrc%253D&md5=a715cdb9609b09056fcb3c6589c2dc63Effects of N-acetyltransferase 2 (NAT2), CYP2E1 and glutathione-S-transferase (GST) genotypes on the serum concentrations of isoniazid and metabolites in tuberculosis patientsFukino, Katsumi; Sasaki, Yuka; Hirai, Shigekazu; Nakamura, Takayuki; Hashimoto, Masayo; Yamagishi, Fumio; Ueno, KoichiJournal of Toxicological Sciences (2008), 33 (2), 187-195CODEN: JTSCDR; ISSN:0388-1350. (Japanese Society of Toxicology)For the purpose of a side-effect monitoring of isoniazid (INH), the authors investigated the relationship between the genotypes of drug-metabolizing enzymes involved in INH metab. and the serum concns. of INH and its metabolites in 129 tuberculosis patients hospitalizing in the National Hospital Organization Chiba-East Hospital. Genotype distributions of N-acetyltransferase 2 (NAT2), CYP2E1*5B, CYP2E1*6, glutathione-S-transferase (GST) M1 and GST T1 were similar to those already reported in Japanese populations. Acetylating pathway of INH to acetyl isoniazid (AcINH) tended to shift to the hydrolytic pathway generating hydrazine (Hz) with the increase of mutant alleles in NAT2 gene. Serum concn. of Hz was significantly higher in slow acetylators than in rapid acetylators of NAT2. Also, serum concn. of Hz was significantly higher in the group that showed a high concn. of rifampicin (RFP) than in which RFP was not detected. The effect of CYP2E1 gene polymorphisms on the serum concn. of Hz was rarely obsd., while that of GST gene polymorphism was obsd. in intermediate acetylators of NAT2. Hz tended to accumulate in patients with GST M1 null genotype. Therefore, it is conceivable that the risk factors of Hz accumulation are as follows: NAT2 slow acetylator phenotype, high concn. of serum RFP, and GST M1 null genotype. In these cases, the authors think it's necessary to pay attention to the development of hepatic disorder caused by Hz.
- 16Jencks, B. D. S. a. W. P. (1989) Mechanism of solvolysis of substituted benzoyl halides J. Am. Chem. Soc. 8470– 8479Google ScholarThere is no corresponding record for this reference.
- 17Maddrey, W. C. and Boitnott, J. K. (1973) Isoniazid hepatitis Ann. Intern. Med. 79, 1– 12[Crossref], [PubMed], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE3s3is1ShsQ%253D%253D&md5=a1c3a72b20ea9fe46792a3685bb28f22Isoniazid hepatitisMaddrey W C; Boitnott J KAnnals of internal medicine (1973), 79 (1), 1-12 ISSN:0003-4819.There is no expanded citation for this reference.
- 18Black, M., Mitchell, J. R., Zimmerman, H. J., Ishak, K. G., and Epler, G. R. (1975) Isoniazid-associated hepatitis in 114 patients Gastroenterology 69, 289– 302Google ScholarThere is no corresponding record for this reference.
- 19Chowdhury, A., Santra, A., Bhattacharjee, K., Ghatak, S., Saha, D. R., and Dhali, G. K. (2006) Mitochondrial oxidative stress and permeability transition in isoniazid and rifampicin induced liver injury in mice J. Hepatol. 45, 117– 126[Crossref], [PubMed], [CAS], Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XlsFKkt70%253D&md5=f4fd7ec0bd4832b843330cb2a467da7cMitochondrial oxidative stress and permeability transition in Isoniazid and Rifampicin induced liver injury in miceChowdhury, Abhijit; Santra, Amal; Bhattacharjee, Koutilya; Ghatak, Subhadip; Saha, Dhira Rani; Dhali, Gopal KrishnaJournal of Hepatology (2006), 45 (1), 117-126CODEN: JOHEEC; ISSN:0168-8278. (Elsevier B.V.)Background/Aims: To evaluate the role of mitochondrial oxidative stress and permeability transition (MPT) in isoniazid (INH) and rifampicin (RMP) induced hepatotoxicity in mice. Methods: Liver damage was induced by co-treatment of INH (50 mg/kg) and RMP (100 mg/kg). Pre-treatment with either methionine or phorone was done to modulate hepatic GSH level. Liver cell injury was assessed biochem. and histol.Evidence of apoptosis was sought by TUNEL test, caspase assay and histol. Results: INH and RMP co-treatment caused steatosis and increased apoptosis of the hepatocytes, hepatic oxidative stress, particularly in the mitochondrial fraction with increased mitochondrial permeability transition (MPT). Mitochondrial oxidative stress as well as liver cell injury was increased by prior treatment with phorone. This was attenuated by pretreatment with methionine suggesting a glutathione (GSH) dependent phenomenon. Conclusions: Oxidative stress in the mitochondria and inappropriate MPT are important in the pathogenesis of apoptotic liver cell injury in INH-RMP hepatotoxicity. The phenomenon is GSH dependent and methionine supplementation might have a protective role.
- 20Sodhi, C. P., Rana, S. V., Mehta, S. K., Vaiphei, K., Attari, S., and Mehta, S. (1997) Study of oxidative-stress in isoniazid-rifampicin induced hepatic injury in young rats Drug. Chem. Toxicol. 20, 255– 269[Crossref], [PubMed], [CAS], Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXmt1Crt74%253D&md5=a6a54f98ed783b6eae2a33bb7b43cb85Study of oxidative stress in isoniazid-rifampicin induced hepatic injury in young ratsSodhi, C. P.; Rana, S. V.; Mehta, S. K.; Vaiphei, K.; Attari, S.; Mehta, S.Drug and Chemical Toxicology (1977) (1997), 20 (3), 255-269CODEN: DCTODJ; ISSN:0148-0545. (Dekker)The role of oxidative stress as a mechanism of hepatotoxicity caused by the combination of isoniazid (INH) and rifampicin (RMP) was investigated in young growing rats. A successful model of hepatotoxicity was produced by giving 50 mg/kg/day each of INH and RMP in 2 wk. Liver showed type II hepatocellular changes (microvesicular fat deposition) with mild portal triaditis. The glutathione and related thiols were significantly decreased in both blood and liver tissues with combined INH and RMP treatment. Superoxide dismutase, glutathione peroxidase, catalase, and glutathione S-transferase with CDNB and DCNB as substrates were decreased in the combination treated group. Glutathione reductase, glutathione S-transferase with ethacrynic acid as substrate, and lipid peroxidn. exhibited a significant increase with treatment. The altered profile of antioxidant enzymes with increased lipid peroxidn. indicated the enhanced oxidative stress in the combined INH and RMP treatment. All the findings are faithfully reflected in the blood tissue except superoxide dismutase which showed a significant enhancement in this tissue. INH and RMP hepatotoxicity thus appeared to be mediated through oxidative stress.
- 21Huang, Y. S., Chern, H. D., Su, W. J., Wu, J. C., Lai, S. L., Yang, S. Y., Chang, F. Y., and Lee, S. D. (2002) Polymorphism of the N-acetyltransferase 2 gene as a susceptibility risk factor for antituberculosis drug-induced hepatitis Hepatology 35, 883– 889Google ScholarThere is no corresponding record for this reference.
- 22Peretti, E., Karlaganis, G., and Lauterburg, B. H. (1987) Increased urinary excretion of toxic hydrazino metabolites of isoniazid by slow acetylators. Effect of a slow-release preparation of isoniazid Eur. J. Clin. Pharmacol. 33, 283– 286Google ScholarThere is no corresponding record for this reference.
- 23Lauterburg, B. H., Smith, C. V., Todd, E. L., and Mitchell, J. R. (1985) Pharmacokinetics of the toxic hydrazino metabolites formed from isoniazid in humans J. Pharmacol. Exp. Ther. 235, 566– 570[PubMed], [CAS], Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XotFCrtg%253D%253D&md5=5375eeb30435603e52a44b66184e5a95Pharmacokinetics of the toxic hydrazino metabolites formed from isoniazid in humansLauterburg, B. H.; Smith, C. V.; Todd, E. L.; Mitchell, J. R.Journal of Pharmacology and Experimental Therapeutics (1985), 235 (3), 566-70CODEN: JPETAB; ISSN:0022-3565.The plasma concns. of isoniazid [54-85-3] and its hydrazino metabolites, acetylisoniazid [1078-38-2], acetylhydrazine [1068-57-1], and diacetylhydrazine [3148-73-0], were measured in healthy subjects after the ingestion of 300 mg of isoniazid. The area under the plasma concn.-time curve (AUC) of acetylisoniazid and diacetylhydrazine increased with increasing rate of acetylation of isoniazid. In contrast, the AUC of acetylhydrazine, the postulated precursor of a toxic metabolite formed from isoniazid, was greater in slow acetylators. This occurred even though rapid acetylators from isoniazid, because the rapid acetylators also acetylated acetylhydrazine faster to diacetylhydrazine than did the slow acetylators. Due to this complex relation between the AUC of acetylhydrazine and the rate of isoniazid acetylation (i.e., a faster rate of formation of acetylhydrazine is accompanied by a faster clearance to diacetylhydrazine), the rate of acetylation of isoniazid minimally influences the exposure of most patients to acetylhydrazine. The apparent plasma half-life of acetylhydrazine was about 5 times longer than the plasma half-life of isoniazid, and thus repeated doses of isoniazid should lead to an accumulation of acetylhydrazine in the slowest acetylators in which the plasma half-life of acetylhydrazine is 20-plus h. Furthermore, a large dose (10 mg/kg) of isoniazid led to satn. of acetylation, which may be esp. contributory to an accumulation of acetylhydrazine in slow acetylators by decreasing clearance to diacetylhydrazine. The remarkably high incidence (21.4%) of liver injury reported in slow-acetylator patients receiving high therapeutic doses of isoniazid (10 mg/kg/day) and rifampin may be due metabolic activation of acetylhydrazine to a toxic metabolite which contributes to the pathogenesis of isoniazid hepatitis.
- 24Warrington, R. J., Tse, K. S., Gorski, B. A., Schwenk, R., and Sehon, A. H. (1978) Evaluation of isoniazid-associated hepatitis by immunological tests Clin. Exp. Immunol. 32, 97– 104[PubMed], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE1c3htlGruw%253D%253D&md5=7f3919ec2c35d189fbb271db8bc5f371Evaluation of isoniazid-associated hepatitis by immunological testsWarrington R J; Tse K S; Gorski B A; Schwenk R; Sehon A HClinical and experimental immunology (1978), 32 (1), 97-104 ISSN:0009-9104.In a retrospective study of patients developing hepatitis or persistent serum glutamic oxaloacetic transaminase (SGOT) elevations while receiving isoniazid, it was found that the lymphocyte transformation test (LTT) was positive in nineteen cases (95%) in response to stimulation by isoniazid, isonicotinic acid and conjugates of these compounds with human serum albumin. However, no significant amount of antibody against isoniazid was detected in the sera of these patients by a sensitive radioimmunoassay. By contrast, no positive LTT was seen in normal controls or in patients receiving isoniazid without evidence of liver damage, while in patients with transient SGOT abnormalities, the LTT was positive only at the time of liver dysfunction. There was no correlation between the degree of lymphocyte transformation and the severity of liver damage. However, there were differences in the patterns of response to the four stimulatory preparations used. Thus patients with overt hepatitis most frequently responded to isoniazid, while individuals with only SGOT abnormalities showed stimulation in the LTT more often with a conjugate of isonicotinic acid and human serum albumin. It appears, therefore, that the presence of isoniazid-induced liver damage is associated with the presence of cellular hypersensitivity to the drug. The differences in lymphocyte reactivity in the two groups might indicate a potential means of predicting which individuals are at increased risk of developing overt hepatitis when exhibiting evidence of minor liver dysfunction while receiving isoniazid.
- 25Warrington, R. J., McPhilips-Feener, S., and Rutherford, W. J. (1982) The predictive value of the lymphocyte transformation test in isoniazid-associated hepatitis Clin. Allergy 12, 217– 222[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL383lvFOqsA%253D%253D&md5=e0ab1605cc324b65254281b408836433The predictive value of the lymphocyte transformation test in isoniazid-associated hepatitisWarrington R J; McPhilips-Feener S; Rutherford W JClinical allergy (1982), 12 (3), 217-22 ISSN:0009-9090.Sixty-one patients receiving isoniazid (INH) for chemotherapy or chemoprophylaxis were assessed by the lymphocyte-transformation test (LTT) shortly after starting treatment. Thirty-eight per cent exhibited stimulation with INH, isonicotinic acid (INA) or human-serum albumin conjugates of these haptens. In the LTT-positive group, liver dysfunction subsequently developed in 58.8%, as compared to 22.7% in the LTT-negative group (P less than 0.01). The difference was not accountable on the basis of age, ethnic background, sex or chemotherapy vs chemoprophylaxis. Although there was an excess of alcohol abusers in the LTT-positive group, the probability of developing liver dysfunction amongst alcohol abusers in that group was twice as great as for LTT-negative alcohol abusers. The specificity of the LTT in predicting liver damage was 83-90% (depending upon the criteria used for determining positivity), while the sensitivity of the test was only 50%.
- 26Sharma, S. K., Balamurugan, A., Saha, P. K., Pandey, R. M., and Mehra, N. K. (2002) Evaluation of clinical and immunogenetic risk factors for the development of hepatotoxicity during antituberculosis treatment Am. J. Respir. Crit. Care Med. 166, 916– 919[Crossref], [PubMed], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD38vpslCrtQ%253D%253D&md5=ad1c7f20ef9dcbeb2d77b97ddc483501Evaluation of clinical and immunogenetic risk factors for the development of hepatotoxicity during antituberculosis treatmentSharma Surendra K; Balamurugan Arumugam; Saha Pradip Kumar; Pandey Ravindra M; Mehra Narinder KAmerican journal of respiratory and critical care medicine (2002), 166 (7), 916-9 ISSN:1073-449X.Though several risk factors for the development of hepatotoxicity due to antituberculosis drugs have been suggested, involvement of genetic factors is not fully established. We have studied the major histocompatibility complex (MHC) class II alleles and clinical risk factors for the development of hepatotoxicity in 346 North Indian patients with tuberculosis undergoing antituberculosis treatment. Of these, 56 patients developed drug-induced hepatotoxicity (DIH group), whereas the remaining 290 patients did not (non-DIH group). The DIH group was comparatively older, had lower pretreatment serum albumin, and a higher frequency of moderately/far advanced disease radiographically than the latter. Further, patients with high alcohol intake had threefold higher odds of developing hepatotoxicity. In multivariate logistic regression analysis, older age (odds ratio [OR] 1.2), moderately/far advanced disease (OR 2.0), serum albumin < 3.5 g/dl (OR 2.3), absence of HLA-DQA1*0102 (OR 4.0), and presence of HLA-DQB1*0201 (OR 1.9) were independent risk factors for DIH. Our results suggest that the risk of hepatotoxicity from antituberculosis drugs is influenced by clinical and genetic factors.
Cited By
This article is cited by 74 publications.
- M. Saeed Mirzaei, Maxim V. Ivanov, Avat Arman Taherpour, Saber Mirzaei. Mechanism-Based Inactivation of Cytochrome P450 Enzymes: Computational Insights. Chemical Research in Toxicology 2021, 34 (4) , 959-987. https://doi.org/10.1021/acs.chemrestox.0c00483
- Tiffany Cho, Xijin Wang, Jack Uetrecht. Rotenone Increases Isoniazid Toxicity but Does Not Cause Significant Liver Injury: Implications for the Hypothesis that Inhibition of the Mitochondrial Electron Transport Chain Is a Common Mechanism of Idiosyncratic Drug-Induced Liver Injury. Chemical Research in Toxicology 2019, 32 (7) , 1423-1431. https://doi.org/10.1021/acs.chemrestox.9b00116
- Chaitanya K. Jaladanki, Akbar Shaikh, and Prasad V. Bharatam . Biotransformation of Isoniazid by Cytochromes P450: Analyzing the Molecular Mechanism using Density Functional Theory. Chemical Research in Toxicology 2017, 30 (11) , 2060-2073. https://doi.org/10.1021/acs.chemrestox.7b00129
- Yakov M. Koen, Nadezhda A. Galeva, Imir G. Metushi, Jack Uetrecht, and Robert P. Hanzlik . Protein Targets of Isoniazid-Reactive Metabolites in Mouse Liver in Vivo. Chemical Research in Toxicology 2016, 29 (6) , 1064-1072. https://doi.org/10.1021/acs.chemrestox.6b00098
- Alastair Mak and Jack Uetrecht . The Combination of Anti-CTLA-4 and PD1–/– Mice Unmasks the Potential of Isoniazid and Nevirapine To Cause Liver Injury. Chemical Research in Toxicology 2015, 28 (12) , 2287-2291. https://doi.org/10.1021/acs.chemrestox.5b00305
- Xiaoli Meng, James L. Maggs, Toru Usui, Paul Whitaker, Neil S. French, Dean J. Naisbitt, and B. Kevin Park . Auto-oxidation of Isoniazid Leads to Isonicotinic-Lysine Adducts on Human Serum Albumin. Chemical Research in Toxicology 2015, 28 (1) , 51-58. https://doi.org/10.1021/tx500285k
- Imir G. Metushi, William M. Lee, and Jack Uetrecht . IgG3 Is the Dominant Subtype of Anti-isoniazid Antibodies in Patients with Isoniazid-Induced Liver Failure. Chemical Research in Toxicology 2014, 27 (5) , 738-740. https://doi.org/10.1021/tx500108u
- Imir G. Metushi, Xu Zhu, Xin Chen, Michael A. Gardam, and Jack Uetrecht . Mild Isoniazid-Induced Liver Injury in Humans Is Associated with an Increase in Th17 Cells and T Cells Producing IL-10. Chemical Research in Toxicology 2014, 27 (4) , 683-689. https://doi.org/10.1021/tx500013z
- Violina T. Angelova, Rumyana Simeonova. Effects of a new 1,2,3-thiadiazole containing hydrazone antimycobacterial agent on serum and liver biochemical parameters in female mice. Drug and Chemical Toxicology 2022, 45 (1) , 113-119. https://doi.org/10.1080/01480545.2019.1660671
- Jack Uetrecht, Denis M. Grant, Peter G. Wells. Biochemical mechanisms of drug toxicity. 2022,,, 267-302. https://doi.org/10.1016/B978-0-12-819869-8.00015-X
- Youhao Chen, Shaoxing Guan, Yanping Guan, Siyuan Tang, Yanying Zhou, Xueding Wang, Huichang Bi, Min Huang. Novel clinical biomarkers for drug-induced liver injury. Drug Metabolism and Disposition 2021, , DMD-MR-2021-000732. https://doi.org/10.1124/dmd.121.000732
- Antonio Segovia-Zafra, Daniel E. Di Zeo-Sánchez, Carlos López-Gómez, Zeus Pérez-Valdés, Eduardo García-Fuentes, Raúl J. Andrade, M. Isabel Lucena, Marina Villanueva-Paz. Preclinical models of idiosyncratic drug-induced liver injury (iDILI): Moving towards prediction. Acta Pharmaceutica Sinica B 2021, 11 (12) , 3685-3726. https://doi.org/10.1016/j.apsb.2021.11.013
- Sangeetha Nithiyanandam, Sabina Evan Prince. Toxins mechanism in instigating hepatotoxicity. Toxin Reviews 2021, 40 (4) , 616-631. https://doi.org/10.1080/15569543.2020.1818105
- Xuan Cheng, Jia‐lian Zhu, Yun Li, Wen‐wen Luo, Huai‐rong Xiang, Qi‐zhi Zhang, Wen‐xing Peng. Serum biomarkers of isoniazid‐induced liver injury: Aminotransferases are insufficient, and OPN, L‐FABP and HMGB1 can be promising novel biomarkers. Journal of Applied Toxicology 2021, 48 https://doi.org/10.1002/jat.4236
- Saifei Lei, Ruizhi Gu, Xiaochao Ma. Clinical perspectives of isoniazid-induced liver injury. Liver Research 2021, 5 (2) , 45-52. https://doi.org/10.1016/j.livres.2021.02.001
- Courtney C. Aldrich, Robert S. Wallis, Rory P. Remmel, Pooja Hegde, Christina L. Stallings. Antitubercular Agents. 2021,,, 1-110. https://doi.org/10.1002/0471266949.bmc298
- David S. Umbaugh, Hartmut Jaeschke. Biomarkers of drug-induced liver injury: a mechanistic perspective through acetaminophen hepatotoxicity. Expert Review of Gastroenterology & Hepatology 2021, 15 (4) , 363-375. https://doi.org/10.1080/17474124.2021.1857238
- Alison Jee, Samantha Christine Sernoskie, Jack Uetrecht. Idiosyncratic Drug-Induced Liver Injury: Mechanistic and Clinical Challenges. International Journal of Molecular Sciences 2021, 22 (6) , 2954. https://doi.org/10.3390/ijms22062954
- Kamel Mokhnache, Ahlem Karbab, El-Khamsa Soltani, Walid Bououden, Soraya Ouhida, Lekhmici Arrar, Maria Angeles Esteban, Noureddine Charef, Mohammad S. Mubarak. Synthesis, characterization, toxic substructure prediction, hepatotoxicity evaluation, marine pathogenic bacteria inhibition, and DFT calculations of a new hydrazone derived from isoniazid. Journal of Molecular Structure 2020, 1221 , 128817. https://doi.org/10.1016/j.molstruc.2020.128817
- L.V. Natrus, L.V. Gayova, O.O. Gorkunenko, P.A. Chernovol, M.V. Zelinska. CYP2E1-DEPENDENT VARIATIONS IN HEPATOCYTES DAMAGE DURING TREATMENT OF TUBERCULOSIS. Medical Science of Ukraine (MSU) 2020, 16 (3) , 20-26. https://doi.org/10.32345/2664-4738.3.2020.4
- Guoqiang Zhang, Lin Chen, Yuanjie Wen, Zhi Rao, Yuhui Wei, Xin’an Wu. Pyridoxal isonicotinoyl hydrazone inhibition of FXR is involved in the pathogenesis of isoniazid-induced liver injury. Toxicology and Applied Pharmacology 2020, 402 , 115134. https://doi.org/10.1016/j.taap.2020.115134
- Siyu Fu, Dongbo Wu, Wei Jiang, Juan Li, Jiang Long, Chengyao Jia, Taoyou Zhou. Molecular Biomarkers in Drug-Induced Liver Injury: Challenges and Future Perspectives. Frontiers in Pharmacology 2020, 10 https://doi.org/10.3389/fphar.2019.01667
- Lubna Danish, Riffat Siddiq, Sarwat Jahan, Mehwish Taneez, Manzoor Khan, Marva Sandhu. Comparative Study of Protective Effect of Cimetidine and Verapamil on Paracetamol-Induced Hepatotoxicity in Mice. International Journal of Hepatology 2020, 2020 , 1-9. https://doi.org/10.1155/2020/9185361
- Xiao-hua Wang, Yong-wen Jin, Zhi Rao, Guo-qiang Zhang, Kai-hong Zang, Hong-yan Qin. Curcumin Enhances the Systemic Exposure of Isoniazid in Rats: Role of NAT2 in the Liver and Intestine. International Journal of Pharmacology 2019, 16 (1) , 10-17. https://doi.org/10.3923/ijp.2020.10.17
- Osama Abdel-Ghaf, Amany Mohammed Hegab, Eiman Ismael Rayan. Evaluation of Antioxidative Effect of Green Tea Catechins Against Isoniazid-induced Biochemical Alterations in Rats. International Journal of Pharmacology 2019, 15 (7) , 777-789. https://doi.org/10.3923/ijp.2019.777.789
- Yingying Pan, Mingzhu Cao, Danming You, Genggeng Qin, Zhi Liu. Research Progress on the Animal Models of Drug-Induced Liver Injury: Current Status and Further Perspectives. BioMed Research International 2019, 2019 , 1-12. https://doi.org/10.1155/2019/1283824
- Yinglong Wu, Lihe Sun, Fang Zeng, Shuizhu Wu. A conjugated-polymer-based ratiometric nanoprobe for evaluating in-vivo hepatotoxicity induced by herbal medicine via MSOT imaging. Photoacoustics 2019, 13 , 6-17. https://doi.org/10.1016/j.pacs.2018.11.002
- Jack Uetrecht. Mechanisms of idiosyncratic drug-induced liver injury. 2019,,, 133-163. https://doi.org/10.1016/bs.apha.2018.12.001
- James P. McEvoy, Nhan V. Pham, Hong T. Le, Micah M. Fernandez, Ryan P. Farmer, Surendra N. Mahapatro. Two-electron redox chemistry of p -nitro- and p -cyanobenzene diazohydroxides. RSC Advances 2018, 8 (73) , 41762-41766. https://doi.org/10.1039/C8RA05948A
- Ling-Yu Ruan, Jun-Ting Fan, Wei Hong, He Zhao, Ming-Hui Li, Lei Jiang, Yong-Hong Fu, Yue-Xiao Xing, Cheng Chen, Jun-Song Wang. Isoniazid-induced hepatotoxicity and neurotoxicity in rats investigated by 1H NMR based metabolomics approach. Toxicology Letters 2018, 295 , 256-269. https://doi.org/10.1016/j.toxlet.2018.05.032
- Sayed Ab. Reshad Ghafouri, Alexander Brun, Rohit Bhalla, Craig Margulies, Kevin Skole. Disseminated Granulomatous Disease from Intravesical Instillation of Bacillus Calmette-Guerin. Case Reports in Oncological Medicine 2018, 2018 , 1-4. https://doi.org/10.1155/2018/8280527
- Yifan Bao, Xiaochao Ma, Theodore P. Rasmussen, Xiao-bo Zhong. Genetic Variations Associated with Anti-Tuberculosis Drug-Induced Liver Injury. Current Pharmacology Reports 2018, 4 (3) , 171-181. https://doi.org/10.1007/s40495-018-0131-8
- Hozeifa M. Hassan, Bashir A. Yousef, Hongli Guo, Liu Xiaoxin, Luyong Zhang, Zhenzhou Jiang. Investigating the CYP2E1 Potential Role in the Mechanisms Behind INH/LPS-Induced Hepatotoxicity. Frontiers in Pharmacology 2018, 9 https://doi.org/10.3389/fphar.2018.00198
- A. Mak, J. Uetrecht. Idiosyncratic Adverse Drug Reactions. 2018,,, 681-716. https://doi.org/10.1016/B978-0-12-801238-3.64242-3
- Kuppusamy Rajesh, Jayadevan Santhanalakshmi. Fabrication of a SnO 2 –graphene nanocomposite based electrode for sensitive monitoring of an anti-tuberculosis agent in human fluids. New Journal of Chemistry 2018, 42 (4) , 2903-2915. https://doi.org/10.1039/C7NJ03411C
- Pengcheng Wang, Amina I. Shehu, Jie Lu, Rujuta H. Joshi, Raman Venkataramanan, Kim S. Sugamori, Denis M. Grant, Xiao-bo Zhong, Xiaochao Ma. Deficiency of N -acetyltransferase increases the interactions of isoniazid with endobiotics in mouse liver. Biochemical Pharmacology 2017, 145 , 218-225. https://doi.org/10.1016/j.bcp.2017.09.001
- Osama Abdel-Ghaf, Salwa Thabet Mahmoud, Azza Ali Said, Fatma Abdel-Azee. Hepatoprotective Effect of Rutin Against Oxidative Stress of Isoniazid in Albino Rats. International Journal of Pharmacology 2017, 13 (6) , 516-528. https://doi.org/10.3923/ijp.2017.516.528
- Hozeifa M. Hassan, Hongli Guo, Bashir A. Yousef, Ding Ping-Ping, Luyong Zhang, Zhenzhou Jiang. Dexamethasone Pretreatment Alleviates Isoniazid/Lipopolysaccharide Hepatotoxicity: Inhibition of Inflammatory and Oxidative Stress. Frontiers in Pharmacology 2017, 8 https://doi.org/10.3389/fphar.2017.00133
- Erica J. Fratz-Berilla, Leonid Breydo, Laurent Gouya, Hervé Puy, Vladimir N. Uversky, Gloria C. Ferreira. Isoniazid inhibits human erythroid 5-aminolevulinate synthase: Molecular mechanism and tolerance study with four X-linked protoporphyria patients. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 2017, 1863 (2) , 428-439. https://doi.org/10.1016/j.bbadis.2016.11.011
- Amina Ibrahim Shehu, Xiaochao Ma, Raman Venkataramanan. Mechanisms of Drug-Induced Hepatotoxicity. Clinics in Liver Disease 2017, 21 (1) , 35-54. https://doi.org/10.1016/j.cld.2016.08.002
- Toru Usui, Xiaoli Meng, Katy Saide, John Farrell, Paul Thomson, Paul Whitaker, John Watson, Neil S. French, B. Kevin Park, Dean J. Naisbitt. From the Cover: Characterization of Isoniazid-Specific T-Cell Clones in Patients with anti-Tuberculosis Drug-Related Liver and Skin Injury. Toxicological Sciences 2017, 155 (2) , 420-431. https://doi.org/10.1093/toxsci/kfw218
- Jingran Dong, Yanli Ren, Sufang Sun, Jiao Yang, Chunxia Nan, Hongmei Shi, Jianzhong Xu, Jie Duan, Tiesheng Shi, Lars I. Elding. Kinetics and mechanism of oxidation of the anti-tubercular prodrug isoniazid and its analog by iridium( iv ) as models for biological redox systems. Dalton Transactions 2017, 46 (26) , 8377-8386. https://doi.org/10.1039/C7DT00885F
- F. Li, P. Wang, K. Liu, M. G. Tarrago, J. Lu, E. N. Chini, X. Ma. A High Dose of Isoniazid Disturbs Endobiotic Homeostasis in Mouse Liver. Drug Metabolism and Disposition 2016, 44 (11) , 1742-1751. https://doi.org/10.1124/dmd.116.070920
- Jinping Gan, Shuguang Ma, Donglu Zhang. Non-cytochrome P450-mediated bioactivation and its toxicological relevance. Drug Metabolism Reviews 2016, 48 (4) , 473-501. https://doi.org/10.1080/03602532.2016.1225756
- Pengcheng Wang, Komal Pradhan, Xiao-bo Zhong, Xiaochao Ma. Isoniazid metabolism and hepatotoxicity. Acta Pharmaceutica Sinica B 2016, 6 (5) , 384-392. https://doi.org/10.1016/j.apsb.2016.07.014
- Hozeifa Mohamed Hassan, Hongli Guo, Bashir Alsiddig Yousef, Mounia Guerram, Aida Mejda Hamdi, Luyong Zhang, Zhenzhou Jiang. Role of Inflammatory and Oxidative Stress, Cytochrome P450 2E1, and Bile Acid Disturbance in Rat Liver Injury Induced by Isoniazid and Lipopolysaccharide Cotreatment. Antimicrobial Agents and Chemotherapy 2016, 60 (9) , 5285-5293. https://doi.org/10.1128/AAC.00854-16
- Imir Metushi, Jack Uetrecht, Elizabeth Phillips. Mechanism of isoniazid-induced hepatotoxicity: then and now. British Journal of Clinical Pharmacology 2016, 81 (6) , 1030-1036. https://doi.org/10.1111/bcp.12885
- John Mach, Aniko Huizer - Pajkos, Sarah J. Mitchell, Catriona McKenzie, Leo Phillips, Alice Kane, Brett Jones, Rafael de Cabo, Victoria Cogger, David G. Le Couteur, Sarah N. Hilmer. The effect of ageing on isoniazid pharmacokinetics and hepatotoxicity in Fischer 344 rats. Fundamental & Clinical Pharmacology 2016, 30 (1) , 23-34. https://doi.org/10.1111/fcp.12157
- Saifur R. Khan, Naif Aljuhani, Andrew G.M. Morgan, Argishti Baghdasarian, Richard P. Fahlman, Arno G. Siraki. Cytoprotective effect of isoniazid against H2O2 derived injury in HL-60 cells. Chemico-Biological Interactions 2016, 244 , 37-48. https://doi.org/10.1016/j.cbi.2015.11.026
- Yilin Yang, Lulu Jiang, Shuo Wang, Tao Zeng, Keqin Xie. Diallyl trisulfide protects the liver against hepatotoxicity induced by isoniazid and rifampin in mice by reducing oxidative stress and activating Kupffer cells. Toxicology Research 2016, 5 (3) , 954-962. https://doi.org/10.1039/C5TX00440C
- Tian-Guang Zhang, Yi-Mei Wang, Jun Zhao, Ming-Yu Xia, Shuang-Qing Peng, Takashi Ikejima. Induction of protective autophagy against apoptosis in HepG2 cells by isoniazid independent of the p38 signaling pathway. Toxicology Research 2016, 5 (3) , 963-972. https://doi.org/10.1039/C5TX00470E
- Hozeifa M. Hassan, Hong-li Guo, Bashir A. Yousef, Zhang Luyong, Jiang Zhenzhou. Hepatotoxicity mechanisms of isoniazid: A mini-review. Journal of Applied Toxicology 2015, 35 (12) , 1427-1432. https://doi.org/10.1002/jat.3175
- Kok-Yong Seng, Kim-Hor Hee, Gaik-Hong Soon, Nicholas Chew, Saye H. Khoo, Lawrence Soon-U Lee. Population Pharmacokinetic Analysis of Isoniazid, Acetylisoniazid, and Isonicotinic Acid in Healthy Volunteers. Antimicrobial Agents and Chemotherapy 2015, 59 (11) , 6791-6799. https://doi.org/10.1128/AAC.01244-15
- Imir G. Metushi, Ping Cai, Dzana Dervovic, Feng Liu, Alexandra Lobach, Tetsuya Nakagawa, Jack Uetrecht. Development of a novel mouse model of amodiaquine-induced liver injury with a delayed onset. Journal of Immunotoxicology 2015, 12 (3) , 247-260. https://doi.org/10.3109/1547691X.2014.934977
- Imir G. Metushi, M. Anthony Hayes, Jack Uetrecht. Treatment of PD-1 −/− mice with amodiaquine and anti-CTLA4 leads to liver injury similar to idiosyncratic liver injury in patients. Hepatology 2015, 61 (4) , 1332-1342. https://doi.org/10.1002/hep.27549
- Winnie Ng, Imir G. Metushi, Jack Uetrecht. Hepatic effects of aminoglutethimide: A model aromatic amine. Journal of Immunotoxicology 2015, 12 (1) , 24-32. https://doi.org/10.3109/1547691X.2013.867912
- Imir G. Metushi, Jack Uetrecht. Isoniazid-induced liver injury and immune response in mice. Journal of Immunotoxicology 2014, 11 (4) , 383-392. https://doi.org/10.3109/1547691X.2013.860644
- Imir G. Metushi, Xu Zhu, Jack Uetrecht. D-penicillamine-induced granulomatous hepatitis in brown Norway rats. Molecular and Cellular Biochemistry 2014, 393 (1-2) , 229-235. https://doi.org/10.1007/s11010-014-2065-8
- Rachel J. Church, Hong Wu, Merrie Mosedale, Susan J. Sumner, Wimal Pathmasiri, Catherine L. Kurtz, Mathew T. Pletcher, John S. Eaddy, Karamjeet Pandher, Monica Singer, Ameesha Batheja, Paul B. Watkins, Karissa Adkins, Alison H. Harrill. A Systems Biology Approach Utilizing a Mouse Diversity Panel Identifies Genetic Differences Influencing Isoniazid-Induced Microvesicular Steatosis. Toxicological Sciences 2014, 140 (2) , 481-492. https://doi.org/10.1093/toxsci/kfu094
- Jayaprakasam Bolleddula, Kevin DeMent, James P. Driscoll, Philip Worboys, Patrick J. Brassil, David L. Bourdet. Biotransformation and bioactivation reactions of alicyclic amines in drug molecules. Drug Metabolism Reviews 2014, 46 (3) , 379-419. https://doi.org/10.3109/03602532.2014.924962
- Imir G. Metushi, Ping Cai, Libia Vega, Denis M. Grant, Jack Uetrecht. Paradoxical Attenuation of Autoimmune Hepatitis by Oral Isoniazid in Wild-Type and N -Acetyltransferase–Deficient Mice. Drug Metabolism and Disposition 2014, 42 (6) , 963-973. https://doi.org/10.1124/dmd.113.056622
- Adam J Shuhendler, Kanyi Pu, Lina Cui, Jack P Uetrecht, Jianghong Rao. Real-time imaging of oxidative and nitrosative stress in the liver of live animals for drug-toxicity testing. Nature Biotechnology 2014, 32 (4) , 373-380. https://doi.org/10.1038/nbt.2838
- Urs A Boelsterli, Kang Kwang Lee. Mechanisms of isoniazid-induced idiosyncratic liver injury: Emerging role of mitochondrial stress. Journal of Gastroenterology and Hepatology 2014, 29 (4) , 678-687. https://doi.org/10.1111/jgh.12516
- Imir G. Metushi, Corron Sanders, , William M. Lee, Jack Uetrecht. Detection of anti-isoniazid and anti-cytochrome P450 antibodies in patients with isoniazid-induced liver failure. Hepatology 2014, 59 (3) , 1084-1093. https://doi.org/10.1002/hep.26564
- Laura James, Dean Roberts. Isoniazid hepatotoxicity: Progress in understanding the immunologic component. Hepatology 2014, 59 (3) , 746-748. https://doi.org/10.1002/hep.26707
- Angelique Leone, Alex Nie, J. Brandon Parker, Sharmilee Sawant, Leigh-Anne Piechta, Michael F. Kelley, L. Mark Kao, S. Jim Proctor, Geert Verheyen, Mark D. Johnson, Peter G. Lord, Michael K. McMillian. Oxidative stress/reactive metabolite gene expression signature in rat liver detects idiosyncratic hepatotoxicants. Toxicology and Applied Pharmacology 2014, 275 (3) , 189-197. https://doi.org/10.1016/j.taap.2014.01.017
- Imir G. Metushi, Jack Uetrecht. Lack of liver injury in Wistar rats treated with the combination of isoniazid and rifampicin. Molecular and Cellular Biochemistry 2014, 387 (1-2) , 9-17. https://doi.org/10.1007/s11010-013-1864-7
- Dennie GA Hebels, Marlon JA Jetten, Hugo JW Aerts, Ralf Herwig, Daniël HJ Theunissen, Stan Gaj, Joost H van Delft, Jos CS Kleinjans. Evaluation of database-derived pathway development for enabling biomarker discovery for hepatotoxicity. Biomarkers in Medicine 2014, 8 (2) , 185-200. https://doi.org/10.2217/bmm.13.154
- Ke Liu, Feng Li, Jie Lu, Zhiwei Gao, Curtis D. Klaassen, Xiaochao Ma. Role of CYP3A in Isoniazid Metabolism In Vivo. Drug Metabolism and Pharmacokinetics 2014, 29 (2) , 219-222. https://doi.org/10.2133/dmpk.DMPK-13-NT-089
- . ABSTRACTS FROM THE 10 TH INTERNATIONAL ISSX MEETING. Drug Metabolism Reviews 2014,,, 1-286. https://doi.org/10.3109/03602532.2013.868114
- Kang Kwang Lee, Kazunori Fujimoto, Carmen Zhang, Christine T. Schwall, Nathan N. Alder, Carl A. Pinkert, Winfried Krueger, Theodore Rasmussen, Urs A. Boelsterli. Isoniazid-induced cell death is precipitated by underlying mitochondrial complex I dysfunction in mouse hepatocytes. Free Radical Biology and Medicine 2013, 65 , 584-594. https://doi.org/10.1016/j.freeradbiomed.2013.07.038
- Yanyan Chen, Peng Xue, Yongyong Hou, Hao Zhang, Hongzhi Zheng, Tong Zhou, Weidong Qu, Weiping Teng, Qiang Zhang, Melvin E. Andersen, Jingbo Pi. Isoniazid suppresses antioxidant response element activities and impairs adipogenesis in mouse and human preadipocytes. Toxicology and Applied Pharmacology 2013, 273 (3) , 435-441. https://doi.org/10.1016/j.taap.2013.10.005
- Yong Lian, Jing Zhao, Peiyu Xu, Yimei Wang, Jun Zhao, Li Jia, Ze Fu, Li Jing, Gang Liu, Shuangqing Peng, . Protective Effects of Metallothionein on Isoniazid and Rifampicin-Induced Hepatotoxicity in Mice. PLoS ONE 2013, 8 (8) , e72058. https://doi.org/10.1371/journal.pone.0072058
- Feng Li, Jie Lu, Jie Cheng, Laiyou Wang, Tsutomu Matsubara, Iván L Csanaky, Curtis D Klaassen, Frank J Gonzalez, Xiaochao Ma. Human PXR modulates hepatotoxicity associated with rifampicin and isoniazid co-therapy. Nature Medicine 2013, 19 (4) , 418-420. https://doi.org/10.1038/nm.3104
Abstract
Figure 1
Figure 1. INH adduct formation in human liver microsomes. INH = isoniazid, NAL = N-α-acetyl-l-lysine, HLM = human liver microsomes, INA-INH = INH dimer, and INA-NAL = N-α-acetyl-l-lysine adduct.
Figure 2
Figure 2. Specificity of the anti-INH antibody. (A) Antibody specificity was tested by ELISA. The plate was either coated with BSA modified with INA (BSA-INA) or BSA alone. Preimmune serum (SPre) or serum after immunization with Blue Carrier Protein modified with INA (SAft) diluted at 1:100,000 was used as the primary antibody. In the third and fifth column, the primary antibody was preincubated with INH or NAL, respectively, at a concentration of 200 μM for 30 min at room temperature. (B) Antibody was tested for cross-reactivity with binding due to AcHz. Female C57BL/6 mice were treated with either INH or AcHz (Fisher Scientific) by gavage for 7 days at 50 mg/kg/day (n = 2 for each group). There was no binding to hepatic proteins from untreated control and AcHz-treated mice, whereas hepatic proteins from INH-treated mice showed a large number of bands modified with INH. (C) Antiserum was tested for specificity on Western blots by preincubation with INH at 200 μM or 2 mM for 1 h at 4 °C, which prevented binding to the INH-modified hepatic proteins. (D) Binding of the anti-INH serum to INH-modified liver proteins on Western blots was compared to that of the preimmune serum from the same animal.
Figure 3
Figure 3. Covalent binding of INH to hepatic proteins in mice. (A) Male C57BL/6 (n = 4) untreated controls or treated with INH (0.2% of INH by weight in food) for 5 weeks. (B) Male vs female C57BL/6 (n = 4) treated with INH (0.2% of INH by weight in food) for 5 weeks. (C) Female C57BL/6 mice (n = 3) treated for 5 weeks vs Balb C mice (n = 3) treated for 3 weeks, both with 0.2% INH by weight in food. (D) Female Balb C mice treated with INH; either by gavage at 100 mg/kg/day (n = 2) for a period of 1, 3, or 7 days or with 0.2% INH by weight in food for 3 weeks. (E,F) Immunohistochemical staining in the livers of female C57BL/6 mice in untreated control vs those treated with 0.2% INH by weight in food for 5 weeks. Magnification: 5×.
Figure 4
Figure 4. Covalent binding of INH to hepatic proteins in rats. (A) BN rats (n = 3) in untreated controls or treated with INH either by gavage at a dose of 150 mg/kg/day or with 0.2% INH by weight in food for 5 weeks. (B) Wistar rats (n = 4) in untreated controls or treated with INH by gavage at 150 mg/kg/day for 4 weeks. (C) Comparison of covalent binding between male Wistar rats and male BN rats (n = 4) treated with INH by gavage at a dose of 150 mg/kg/day for 4 and 5 weeks, respectively. (D) Comparison of covalent binding between male C57BL/6 mice (n = 4) treated with 0.2% INH by weight in food for 5 weeks vs male Wistar rats (n = 4) treated with INH by gavage at a dose of 150 mg/kg/day for 4 weeks.
Figure 5
Figure 5. In vitro covalent binding of INH to hepatic microsomes (A) to HLM with and without an NADPH-generating system; (B) to MLM with and without a NADPH-generating system; (C) to HLM as a function of time and INH concentration; (D) to MLM as a function of time and INH concentration; (E,F) direct comparison of binding between HLM and MLM at INH concentrations of 10 and 100 μM, respectively.
Figure 6
Figure 6. Comparison of INH covalent binding to rat, mouse, and human liver microsomes at an INH concentration of 50 μM in the presence or absence of a NADPH generating system.
Figure 7
Figure 7. Serum SDH activities and body weights in INH-treated BN rats. (A) SDH activity in BN rats treated with INH by gavage at 150 mg/kg/day, 50 mg/kg twice daily, or given at 0.2% INH by weight in food. (B) Body weight of BN rats given INH as in panel A. Values represent the mean ± SE. Analyzed for statistical significance by two-way ANOVA. Significantly different from the control group (*p < 0.05; **p < 0.01; ***p < 0.001).
Figure 8
Figure 8. GLDH activities and body weights in mice. (A–D) Male and female C57BL/6 mice were treated at 0.2% INH by weight in food for 5 weeks. (E–H) Male and female Balb C mice were treated at 0.2% INH by weight in food for 3 weeks. Values represent the mean ± SE from 4 animals per group. The data were analyzed for statistical significance by two-way ANOVA. Significantly different from the control group (*p < 0.05; **p < 0.01; ***p < 0.001).
Figure 9
Figure 9. Serum concentrations of INH and INH/AcHz ratio in mice and rats. INH was given at a dose of 0.2% by weight in food for up to one week. Values represent the mean ± SE for mice (n = 5) and for rats (n = 4). The data were analyzed for statistical significance by the Mann–Whitney U test. Significantly different groups (*p < 0.05).
References
ARTICLE SECTIONSThis article references 26 other publications.
- 1Center for Disease Control and Prevention (2010) Severe isoniazid-associated liver injuries among persons being treated for latent tuberculosis infection: United States, 2004–2008 MMWR 59, 224– 229
- 2Mitchell, J. R., Zimmerman, H. J., Ishak, K. G., Thorgeirsson, U. P., Timbrell, J. A., Snodgrass, W. R., and Nelson, S. D. (1976) Isoniazid liver injury: clinical spectrum, pathology, and probable pathogenesis Ann. Intern. Med. 84, 181– 192[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE287jsVGgsw%253D%253D&md5=3ba6aa4dfbaca727dac947da5c80b913Isoniazid liver injury: clinical spectrum, pathology, and probable pathogenesisMitchell J R; Zimmerman H J; Ishak K G; Thorgeirsson U P; Timbrell J A; Snodgrass W R; Nelson S DAnnals of internal medicine (1976), 84 (2), 181-92 ISSN:0003-4819.The clinical spectrum of isoniazid-induced liver injury seems to be clinically, biochemically, and histologically indistinguishable from viral hepatitis, except that the injury occurs primarily in persons older than 35 years. A possible relation between susceptibility of patients to isoniazid liver injury and rapid metabolism (acetylation) of the drug has been found. Examination of isoniazid metabolites showed that patients with rapid acetylator phenotype hydrolyze much more isoniazid to isonicotinic acid and the free hydrazine moiety than do slow acetylators. The hydrazine moiety liberated from isoniazid is primarily acetylhydrazine, and studies in animals show this metabolite to be converted to a potent acylating agent that produces liver necrosis. It seems likely that formation of chemically reactive metabolites is also the biochemical event initiating isoniazid liver injury in man. Recognition of the seriousness of isoniazid hepatic injury, not readily accepted at first, has led to revisions in the uses of isoniazid prophylaxis.
- 3Snodgrass, W., Potter, W. Z., T., J., Jollow, D. J., and Mitchell, J. R. (1974) Possible mechanism of isoniazid-related hepatic injury Clin. Res. 22, 323AGoogle ScholarThere is no corresponding record for this reference.
- 4Timbrell, J. A., Mitchell, J. R., Snodgrass, W. R., and Nelson, S. D. (1980) Isoniazid hepatoxicity: the relationship between covalent binding and metabolism in vivo J. Pharmacol. Exp. Ther. 213, 364– 369[PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3cXktlOis7s%253D&md5=22987ead13a85569c3b9156212ef5d21Isoniazid hepatotoxicity: the relationship between covalent binding and metabolism in vivoTimbrell, John A.; Mitchell, Jerry R.; Snodgrass, Wayne R.; Nelson, Sidney D.Journal of Pharmacology and Experimental Therapeutics (1980), 213 (2), 364-9CODEN: JPETAB; ISSN:0022-3565.The relation between the hepatotoxicity and metab. of isoniazid (I) [54-85-3] and its metabolites, acetylisoniazid [1078-38-2] and acetylhydrazine [1068-57-1], was investigated. Toxic doses of acetylisoniazid and acetylhydrazine, radiolabeled in the Ac group, were found to bind covalently to liver protein in vivo. This binding was mediated by the microsomal enzyme system as indicated by the effects of pretreatments altering the activity of these enzymes. Metabolic studies revealed that the pretreatments increased the metab. of the acetylhydrazine moiety of Ac-labeled acetylisoniazid and of acetylhydrazine itself by the microsomal enzyme system. Pretreatment with the acyl amidase inhibit r bis-p-nitrophenyl phosphate inhibited the hydrolysis of acetylisoniazid to isonicotinic acid plus acetylhydrazine and concomitantly decreased the covalent binding of Ac-labeled acetylisoniazid. The changes in the metab. of I, acetylisoniazid, and acetylhydrazine effected by various pretreatments paralleled changes in the severity of the hepatic necrosis caused by the compds. Apparently, acetylhydrazine is the metabolite responsible for the hepatic necrosis caused by I, and microsomal metab. of acetylhydrazine in vivo leads to the prodn. of a reactive acylating species capable of reacting covalently with tissue macromols.
- 5Nelson, S. D., Mitchell, J. R., Timbrell, J. A., Snodgrass, W. R., and Corcoran, G. B. (1976) Isoniazid and iproniazid: activation of metabolites to toxic intermediates in man and rat Science 193, 901– 903Google ScholarThere is no corresponding record for this reference.
- 6Noda, A., Hsu, K. Y., Noda, H., Yamamoto, Y., and Kurozumi, T. (1983) Is isoniazid-hepatotoxicity induced by the metabolite, hydrazine? J. UOEH 5, 183– 190[PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3sXltlegsr8%253D&md5=b6d11194b2a7b901544e4a012b8f330eIs isoniazid-hepatotoxicity induced by the metabolite, hydrazine?Noda, Atsuko; Hsu, Kuang Yang; Noda, Hiroshi; Yamamoto, Yuzo; Kurozumi, TakeshiJournal of UOEH (1983), 5 (2), 183-90CODEN: JOUOD4; ISSN:0387-821X.Treatment of tuberculosis with a combination of isoniazid (I) [54-85-3] and rifampicin (II) [13292-46-1] has been previously shown to cause more severe hepatitis than treatment with I alone. Pretreatment of rabbits with II had no effect on subsequent blood concns. of I or its metabolites acetylisoniazid [1078-38-2], acetylhydrazine [1068-57-1], or diacetylhydrazine [3148-73-0], but it significantly decreased hydrazine [302-01-2] levels. Furthermore, none of the metabolites, except hydrazine, themselves caused hepatic necrosis in rabbits. Since II increased hepatic cytochrome P 450 levels, a mechanism is proposed whereby the II-induced increase in the hepatic enzyme is responsible for the oxidative degrdn. of hydrazine to hepatotoxic products. Hydrazine is thus suggested as the I metabolite responsible for I-induced hepatitis.
- 7Sarich, T. C., Youssefi, M., Zhou, T., Adams, S. P., Wall, R. A., and Wright, J. M. (1996) Role of hydrazine in the mechanism of isoniazid hepatotoxicity in rabbits Arch. Toxicol. 70, 835– 840[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XntVWgs7o%253D&md5=3c75759c797c5170391113797a55c78eRole of hydrazine in the mechanism of isoniazid hepatotoxicity in rabbitsSarich, Troy C.; Youssefi, Mohammed; Zhou, Ting; Adams, Stephen P.; Wall, Richard A.; Wright James M.Archives of Toxicology (1996), 70 (12), 835-840CODEN: ARTODN; ISSN:0340-5761. (Springer)By using a model of isoniazid (INH)-induced hepatotoxicity in rabbits, in which INH-induced hepatotoxicity manifests as hepatic necrosis, hepatic steatosis (hepatic fat accumulation), and hypertriglyceridemia (elevated plasma triglycerides), the severity of these measures of toxicity were compared with plasma levels of INH, acetylhydrazine, and hydrazine. Plasma INH and acetylhydrazine were not correlated with markers of INH-induced hepatic necrosis or fatty changes. Plasma hydrazine at 32 h was correlated with plasma argininosuccinic acid lyase (ASAL) activity as area under the curve (r2 = 0.54, P < 0.002) and log plasma ASAL activity at 48 h after the 1st dose of INH (r2 = 0.53, p < 0.005), but not with fatty changes. Hydrazine, and not INH or acetylhydrazine, is most likely involved in the pathogenic mechanism of hepatic necrosis.
- 8Hofstra, A. H., Li-Muller, S. M., and Uetrecht, J. P. (1992) Metabolism of isoniazid by activated leukocytes. Possible role in drug-induced lupus Drug. Metab. Dispos. 20, 205– 210Google ScholarThere is no corresponding record for this reference.
- 9Metushi, I. G., Cai, P., Zhu, X., Nakagawa, T., and Uetrecht, J. P. (2011) A fresh look at the mechanism of isoniazid-induced hepatotoxicity Clin. Pharmacol. Ther. 89, 911– 914[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmtlagtbc%253D&md5=e444a505f3b86ff0d51408447de8509eA Fresh Look at the Mechanism of Isoniazid-Induced HepatotoxicityMetushi, I. G.; Cai, P.; Zhu, X.; Nakagawa, T.; Uetrecht, J. P.Clinical Pharmacology & Therapeutics (New York, NY, United States) (2011), 89 (6), 911-914CODEN: CLPTAT; ISSN:0009-9236. (Nature Publishing Group)A review. Isoniazid (INH)-induced hepatotoxicity remains a significant clin. problem, and the current mechanistic hypothesis is incomplete; it is simply referred to as metabolic idiosyncrasy, which is believed to involve cytotoxicity caused by bioactivation of acetylhydrazine, a metabolite of INH. However, this hypothesis is based on animal studies, involving characteristics that are very different from those that pertain to hepatotoxicity in humans, such as delayed onset. This article provides a fresh look at this issue.
- 10Bernhardt, P. V., Chin, P., and Richardson, D. R. (2001) Unprecedented oxidation of a biologically active aroylhydrazone chelator catalysed by iron(III): serendipitous identification of diacylhydrazine ligands with high iron chelation efficacy J. Biol. Inorg. Chem. 6, 801– 809[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXptVKns7k%253D&md5=aa8610e965785caee57ef79dd935bd75Unprecedented oxidation of a biologically active aroylhydrazone chelator catalysed by iron(III): serendipitous identification of diacylhydrazine ligands with high iron chelation efficacyBernhardt, Paul V.; Chin, Piao; Richardson, Des R.JBIC, Journal of Biological Inorganic Chemistry (2001), 6 (8), 801-809CODEN: JJBCFA; ISSN:0949-8257. (Springer-Verlag)Ligands of the 2-pyridylcarbaldehyde isonicotinoylhydrazone class show high iron (Fe) sequestering efficacy and have potential as agents for the treatment of Fe overload disease. We have investigated the mechanisms responsible for their high activity. X-ray crystallog. studies show that the tridentate chelate 2-pyridylcarbaldehyde isonicotinoylhydrazone undergoes an unexpected oxidn. to isonicotinoyl(picolinoyl)hydrazine when complexed with FeIII. In contrast, in the absence of FeIII, the parent hydrazone is not oxidized in aerobic aq. soln. To examine whether the diacylhydrazine could be responsible for the biol. effects of 2-pyridylcarbaldehyde isonicotinoylhydrazone, their Fe chelation efficacy was compared. In contrast to its parent hydrazone, the diacylhydrazine showed little Fe chelation activity. Potentiometric titrns. suggested that this might be because the diacylhydrazine was charged at physiol. pH, hindering its access across membranes to intracellular Fe pools. In contrast, the Fe complex of this diacylhydrazine was charge neutral, which may allow facile movement through membranes. These data allow a model of Fe chelation for this compd. to be proposed: the parent aroylhydrazone diffuses through cell membranes to bind Fe and is subsequently oxidized to the diacylhydrazine complex which then diffuses from the cell. Other diacylhydrazine analogs that were charge neutral at physiol. pH demonstrated high Fe chelation efficacy. Thus, for this class of ligands, the charge of the chelator appears to be an important factor for detg. their ability to access intracellular Fe. The results of this study are significant for understanding the biol. activity of 2-pyridylcarbaldehyde isonicotinoylhydrazone and for the design of novel diacylhydrazine chelators for clin. use.
- 11Christensen, J. B. (2001) A simple method for synthesis of active esters of isonicotinic and picolinic acids Molecules 6, 47– 51Google ScholarThere is no corresponding record for this reference.
- 12Zhu, X., Li, J., Liu, F., and Uetrecht, J. P. (2011) Involvement of T helper 17 cells in D-penicillamine-induced autoimmune disease in Brown Norway rats J. Toxicol. Sci. 120, 331– 338[Crossref], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3M3otFaqsA%253D%253D&md5=a6bc54b2bcf093d828caca7239327f7dInvolvement of T helper 17 cells in D-penicillamine-induced autoimmune disease in Brown Norway ratsZhu Xu; Li Jinze; Liu Feng; Uetrecht Jack PToxicological sciences : an official journal of the Society of Toxicology (2011), 120 (2), 331-8 ISSN:.Idiosyncratic drug reactions (IDRs) are poorly understood, but their clinical characteristics suggest that they are immune mediated. Penicillamine-induced autoimmunity in Brown Norway rats has been utilized as an animal model for mechanistic studies of one type of IDR because it closely mimics the autoimmune syndromes that it causes in humans. Our previous work suggested that it is T-cell mediated. It has been shown that T helper 17 (Th17) cells play a central role in many types of autoimmune diseases. This study was designed to test whether Th17 cells are involved in the pathogenesis of penicillamine-induced autoimmunity and to establish an overall serum cytokine/chemokine profile for this IDR. In total, 24 serum cytokines/chemokines were determined and revealed a dynamic process. In sick animals, interleukin (IL) 6 and transforming growth factor-β1, known to be driving forces of Th17 differentiation, were consistently increased at both early and late stages of penicillamine treatment; however, no significant changes in these cytokines were observed in animals that did not develop autoimmunity. IL-17, a characteristic cytokine produced by Th17 cells, was increased in sick animals at both the messenger RNA and serum protein level. In addition, serum concentrations of IL-22, another characteristic cytokine produced by Th17 cells, were found to be elevated. Furthermore, the percentage of IL-17-producing CD4 T cells was significantly increased but only in sick animals. These data strongly suggest that Th17 cells are involved in penicillamine-induced autoimmunity. Such data provide important mechanistic clues that may help to predict which drug candidates will cause a relatively high incidence of such autoimmune IDRs.
- 13O’Brien, P. J., Slaughter, M. R., Polley, S. R., and Kramer, K. (2002) Advantages of glutamate dehydrogenase as a blood biomarker of acute hepatic injury in rats Lab. Anim. 36, 313– 321[Crossref], [PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XlslWis74%253D&md5=1c01835b9a443a27354674278028024eAdvantages of glutamate dehydrogenase as a blood biomarker of acute hepatic injury in ratsO'Brien, P. J.; Slaughter, M. R.; Polley, S. R.; Kramer, K.Laboratory Animals (2002), 36 (3), 313-321CODEN: LBANAX; ISSN:0023-6772. (Royal Society of Medicine Press Ltd.)In a recent study in rats, alanine aminotransferase (ALT), the preferred plasma biomarker of hepatocellular injury in rats, was ineffective at detecting marked hepatic necrosis produced by acetaminophen. In contrast, glutamate dehydrogenase (GLDH) was markedly elevated. Accordingly, these enzymes were comprehensively evaluated as plasma biomarkers of hepatocellular injury in rats using several other models of hepatic injury, including partial hepatectomy and exposure to methapyrilene, dexamethasone, cyproterone, isoniazid, lead nitrate, and Wyeth-14643. Other enzymes also evaluated were aspartate aminotransferase (AST), sorbitol dehydrogenase (SDH), and the hepatobiliary marker alk. phosphatase (ALP). Compared to plasma ALT increases, plasma GLDH increases were up to 10-fold greater, up to 3-fold more persistent, and occurred at times following hepatocellular injury when plasma ALT was not increased. Plasma GLDH activity was not inhibited by the test compds., whereas ALT was substantially inhibited by both isoniazid and lead nitrate. While plasma GLDH activity was unaffected by induction, ALT was induced by cyproterone and dexamethasone, and ALP was induced by Wyeth-14643 and partial hepatectomy. GLDH was concluded to be a more effective biomarker of acute hepatic injury than ALT, AST, SDH or ALP in the rat, based primarily on the large increase following hepatocellular injury, prolonged persistence in the blood following injury, high sensitivity for detection of injury (including pre-necrotic injury), high tissue specificity, and lower susceptibility to inhibition or induction.
- 14Ng, W., Lobach, A. R., Zhu, X., Chen, X., Liu, F., Metushi, I. G., Sharma, A., Li, J., Cai, P., Ip, J., Novalen, M., Popovic, M., Zhang, X., Tanino, T., Nakagawa, T., Li, Y., and Uetrecht, J. (2012) Animal models of idiosyncratic drug reactions Adv. Pharmacol. 63, 81– 135Google ScholarThere is no corresponding record for this reference.
- 15Fukino, K., Sasaki, Y., Hirai, S., Nakamura, T., Hashimoto, M., Yamagishi, F., and Ueno, K. (2008) Effects of N-acetyltransferase 2 (NAT2), CYP2E1 and glutathione-S-transferase (GST) genotypes on the serum concentrations of isoniazid and metabolites in tuberculosis patients J. Toxicol. Sci. 33, 187– 195[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXos1aksrc%253D&md5=a715cdb9609b09056fcb3c6589c2dc63Effects of N-acetyltransferase 2 (NAT2), CYP2E1 and glutathione-S-transferase (GST) genotypes on the serum concentrations of isoniazid and metabolites in tuberculosis patientsFukino, Katsumi; Sasaki, Yuka; Hirai, Shigekazu; Nakamura, Takayuki; Hashimoto, Masayo; Yamagishi, Fumio; Ueno, KoichiJournal of Toxicological Sciences (2008), 33 (2), 187-195CODEN: JTSCDR; ISSN:0388-1350. (Japanese Society of Toxicology)For the purpose of a side-effect monitoring of isoniazid (INH), the authors investigated the relationship between the genotypes of drug-metabolizing enzymes involved in INH metab. and the serum concns. of INH and its metabolites in 129 tuberculosis patients hospitalizing in the National Hospital Organization Chiba-East Hospital. Genotype distributions of N-acetyltransferase 2 (NAT2), CYP2E1*5B, CYP2E1*6, glutathione-S-transferase (GST) M1 and GST T1 were similar to those already reported in Japanese populations. Acetylating pathway of INH to acetyl isoniazid (AcINH) tended to shift to the hydrolytic pathway generating hydrazine (Hz) with the increase of mutant alleles in NAT2 gene. Serum concn. of Hz was significantly higher in slow acetylators than in rapid acetylators of NAT2. Also, serum concn. of Hz was significantly higher in the group that showed a high concn. of rifampicin (RFP) than in which RFP was not detected. The effect of CYP2E1 gene polymorphisms on the serum concn. of Hz was rarely obsd., while that of GST gene polymorphism was obsd. in intermediate acetylators of NAT2. Hz tended to accumulate in patients with GST M1 null genotype. Therefore, it is conceivable that the risk factors of Hz accumulation are as follows: NAT2 slow acetylator phenotype, high concn. of serum RFP, and GST M1 null genotype. In these cases, the authors think it's necessary to pay attention to the development of hepatic disorder caused by Hz.
- 16Jencks, B. D. S. a. W. P. (1989) Mechanism of solvolysis of substituted benzoyl halides J. Am. Chem. Soc. 8470– 8479Google ScholarThere is no corresponding record for this reference.
- 17Maddrey, W. C. and Boitnott, J. K. (1973) Isoniazid hepatitis Ann. Intern. Med. 79, 1– 12[Crossref], [PubMed], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE3s3is1ShsQ%253D%253D&md5=a1c3a72b20ea9fe46792a3685bb28f22Isoniazid hepatitisMaddrey W C; Boitnott J KAnnals of internal medicine (1973), 79 (1), 1-12 ISSN:0003-4819.There is no expanded citation for this reference.
- 18Black, M., Mitchell, J. R., Zimmerman, H. J., Ishak, K. G., and Epler, G. R. (1975) Isoniazid-associated hepatitis in 114 patients Gastroenterology 69, 289– 302Google ScholarThere is no corresponding record for this reference.
- 19Chowdhury, A., Santra, A., Bhattacharjee, K., Ghatak, S., Saha, D. R., and Dhali, G. K. (2006) Mitochondrial oxidative stress and permeability transition in isoniazid and rifampicin induced liver injury in mice J. Hepatol. 45, 117– 126[Crossref], [PubMed], [CAS], Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XlsFKkt70%253D&md5=f4fd7ec0bd4832b843330cb2a467da7cMitochondrial oxidative stress and permeability transition in Isoniazid and Rifampicin induced liver injury in miceChowdhury, Abhijit; Santra, Amal; Bhattacharjee, Koutilya; Ghatak, Subhadip; Saha, Dhira Rani; Dhali, Gopal KrishnaJournal of Hepatology (2006), 45 (1), 117-126CODEN: JOHEEC; ISSN:0168-8278. (Elsevier B.V.)Background/Aims: To evaluate the role of mitochondrial oxidative stress and permeability transition (MPT) in isoniazid (INH) and rifampicin (RMP) induced hepatotoxicity in mice. Methods: Liver damage was induced by co-treatment of INH (50 mg/kg) and RMP (100 mg/kg). Pre-treatment with either methionine or phorone was done to modulate hepatic GSH level. Liver cell injury was assessed biochem. and histol.Evidence of apoptosis was sought by TUNEL test, caspase assay and histol. Results: INH and RMP co-treatment caused steatosis and increased apoptosis of the hepatocytes, hepatic oxidative stress, particularly in the mitochondrial fraction with increased mitochondrial permeability transition (MPT). Mitochondrial oxidative stress as well as liver cell injury was increased by prior treatment with phorone. This was attenuated by pretreatment with methionine suggesting a glutathione (GSH) dependent phenomenon. Conclusions: Oxidative stress in the mitochondria and inappropriate MPT are important in the pathogenesis of apoptotic liver cell injury in INH-RMP hepatotoxicity. The phenomenon is GSH dependent and methionine supplementation might have a protective role.
- 20Sodhi, C. P., Rana, S. V., Mehta, S. K., Vaiphei, K., Attari, S., and Mehta, S. (1997) Study of oxidative-stress in isoniazid-rifampicin induced hepatic injury in young rats Drug. Chem. Toxicol. 20, 255– 269[Crossref], [PubMed], [CAS], Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXmt1Crt74%253D&md5=a6a54f98ed783b6eae2a33bb7b43cb85Study of oxidative stress in isoniazid-rifampicin induced hepatic injury in young ratsSodhi, C. P.; Rana, S. V.; Mehta, S. K.; Vaiphei, K.; Attari, S.; Mehta, S.Drug and Chemical Toxicology (1977) (1997), 20 (3), 255-269CODEN: DCTODJ; ISSN:0148-0545. (Dekker)The role of oxidative stress as a mechanism of hepatotoxicity caused by the combination of isoniazid (INH) and rifampicin (RMP) was investigated in young growing rats. A successful model of hepatotoxicity was produced by giving 50 mg/kg/day each of INH and RMP in 2 wk. Liver showed type II hepatocellular changes (microvesicular fat deposition) with mild portal triaditis. The glutathione and related thiols were significantly decreased in both blood and liver tissues with combined INH and RMP treatment. Superoxide dismutase, glutathione peroxidase, catalase, and glutathione S-transferase with CDNB and DCNB as substrates were decreased in the combination treated group. Glutathione reductase, glutathione S-transferase with ethacrynic acid as substrate, and lipid peroxidn. exhibited a significant increase with treatment. The altered profile of antioxidant enzymes with increased lipid peroxidn. indicated the enhanced oxidative stress in the combined INH and RMP treatment. All the findings are faithfully reflected in the blood tissue except superoxide dismutase which showed a significant enhancement in this tissue. INH and RMP hepatotoxicity thus appeared to be mediated through oxidative stress.
- 21Huang, Y. S., Chern, H. D., Su, W. J., Wu, J. C., Lai, S. L., Yang, S. Y., Chang, F. Y., and Lee, S. D. (2002) Polymorphism of the N-acetyltransferase 2 gene as a susceptibility risk factor for antituberculosis drug-induced hepatitis Hepatology 35, 883– 889Google ScholarThere is no corresponding record for this reference.
- 22Peretti, E., Karlaganis, G., and Lauterburg, B. H. (1987) Increased urinary excretion of toxic hydrazino metabolites of isoniazid by slow acetylators. Effect of a slow-release preparation of isoniazid Eur. J. Clin. Pharmacol. 33, 283– 286Google ScholarThere is no corresponding record for this reference.
- 23Lauterburg, B. H., Smith, C. V., Todd, E. L., and Mitchell, J. R. (1985) Pharmacokinetics of the toxic hydrazino metabolites formed from isoniazid in humans J. Pharmacol. Exp. Ther. 235, 566– 570[PubMed], [CAS], Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XotFCrtg%253D%253D&md5=5375eeb30435603e52a44b66184e5a95Pharmacokinetics of the toxic hydrazino metabolites formed from isoniazid in humansLauterburg, B. H.; Smith, C. V.; Todd, E. L.; Mitchell, J. R.Journal of Pharmacology and Experimental Therapeutics (1985), 235 (3), 566-70CODEN: JPETAB; ISSN:0022-3565.The plasma concns. of isoniazid [54-85-3] and its hydrazino metabolites, acetylisoniazid [1078-38-2], acetylhydrazine [1068-57-1], and diacetylhydrazine [3148-73-0], were measured in healthy subjects after the ingestion of 300 mg of isoniazid. The area under the plasma concn.-time curve (AUC) of acetylisoniazid and diacetylhydrazine increased with increasing rate of acetylation of isoniazid. In contrast, the AUC of acetylhydrazine, the postulated precursor of a toxic metabolite formed from isoniazid, was greater in slow acetylators. This occurred even though rapid acetylators from isoniazid, because the rapid acetylators also acetylated acetylhydrazine faster to diacetylhydrazine than did the slow acetylators. Due to this complex relation between the AUC of acetylhydrazine and the rate of isoniazid acetylation (i.e., a faster rate of formation of acetylhydrazine is accompanied by a faster clearance to diacetylhydrazine), the rate of acetylation of isoniazid minimally influences the exposure of most patients to acetylhydrazine. The apparent plasma half-life of acetylhydrazine was about 5 times longer than the plasma half-life of isoniazid, and thus repeated doses of isoniazid should lead to an accumulation of acetylhydrazine in the slowest acetylators in which the plasma half-life of acetylhydrazine is 20-plus h. Furthermore, a large dose (10 mg/kg) of isoniazid led to satn. of acetylation, which may be esp. contributory to an accumulation of acetylhydrazine in slow acetylators by decreasing clearance to diacetylhydrazine. The remarkably high incidence (21.4%) of liver injury reported in slow-acetylator patients receiving high therapeutic doses of isoniazid (10 mg/kg/day) and rifampin may be due metabolic activation of acetylhydrazine to a toxic metabolite which contributes to the pathogenesis of isoniazid hepatitis.
- 24Warrington, R. J., Tse, K. S., Gorski, B. A., Schwenk, R., and Sehon, A. H. (1978) Evaluation of isoniazid-associated hepatitis by immunological tests Clin. Exp. Immunol. 32, 97– 104[PubMed], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE1c3htlGruw%253D%253D&md5=7f3919ec2c35d189fbb271db8bc5f371Evaluation of isoniazid-associated hepatitis by immunological testsWarrington R J; Tse K S; Gorski B A; Schwenk R; Sehon A HClinical and experimental immunology (1978), 32 (1), 97-104 ISSN:0009-9104.In a retrospective study of patients developing hepatitis or persistent serum glutamic oxaloacetic transaminase (SGOT) elevations while receiving isoniazid, it was found that the lymphocyte transformation test (LTT) was positive in nineteen cases (95%) in response to stimulation by isoniazid, isonicotinic acid and conjugates of these compounds with human serum albumin. However, no significant amount of antibody against isoniazid was detected in the sera of these patients by a sensitive radioimmunoassay. By contrast, no positive LTT was seen in normal controls or in patients receiving isoniazid without evidence of liver damage, while in patients with transient SGOT abnormalities, the LTT was positive only at the time of liver dysfunction. There was no correlation between the degree of lymphocyte transformation and the severity of liver damage. However, there were differences in the patterns of response to the four stimulatory preparations used. Thus patients with overt hepatitis most frequently responded to isoniazid, while individuals with only SGOT abnormalities showed stimulation in the LTT more often with a conjugate of isonicotinic acid and human serum albumin. It appears, therefore, that the presence of isoniazid-induced liver damage is associated with the presence of cellular hypersensitivity to the drug. The differences in lymphocyte reactivity in the two groups might indicate a potential means of predicting which individuals are at increased risk of developing overt hepatitis when exhibiting evidence of minor liver dysfunction while receiving isoniazid.
- 25Warrington, R. J., McPhilips-Feener, S., and Rutherford, W. J. (1982) The predictive value of the lymphocyte transformation test in isoniazid-associated hepatitis Clin. Allergy 12, 217– 222[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL383lvFOqsA%253D%253D&md5=e0ab1605cc324b65254281b408836433The predictive value of the lymphocyte transformation test in isoniazid-associated hepatitisWarrington R J; McPhilips-Feener S; Rutherford W JClinical allergy (1982), 12 (3), 217-22 ISSN:0009-9090.Sixty-one patients receiving isoniazid (INH) for chemotherapy or chemoprophylaxis were assessed by the lymphocyte-transformation test (LTT) shortly after starting treatment. Thirty-eight per cent exhibited stimulation with INH, isonicotinic acid (INA) or human-serum albumin conjugates of these haptens. In the LTT-positive group, liver dysfunction subsequently developed in 58.8%, as compared to 22.7% in the LTT-negative group (P less than 0.01). The difference was not accountable on the basis of age, ethnic background, sex or chemotherapy vs chemoprophylaxis. Although there was an excess of alcohol abusers in the LTT-positive group, the probability of developing liver dysfunction amongst alcohol abusers in that group was twice as great as for LTT-negative alcohol abusers. The specificity of the LTT in predicting liver damage was 83-90% (depending upon the criteria used for determining positivity), while the sensitivity of the test was only 50%.
- 26Sharma, S. K., Balamurugan, A., Saha, P. K., Pandey, R. M., and Mehra, N. K. (2002) Evaluation of clinical and immunogenetic risk factors for the development of hepatotoxicity during antituberculosis treatment Am. J. Respir. Crit. Care Med. 166, 916– 919[Crossref], [PubMed], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD38vpslCrtQ%253D%253D&md5=ad1c7f20ef9dcbeb2d77b97ddc483501Evaluation of clinical and immunogenetic risk factors for the development of hepatotoxicity during antituberculosis treatmentSharma Surendra K; Balamurugan Arumugam; Saha Pradip Kumar; Pandey Ravindra M; Mehra Narinder KAmerican journal of respiratory and critical care medicine (2002), 166 (7), 916-9 ISSN:1073-449X.Though several risk factors for the development of hepatotoxicity due to antituberculosis drugs have been suggested, involvement of genetic factors is not fully established. We have studied the major histocompatibility complex (MHC) class II alleles and clinical risk factors for the development of hepatotoxicity in 346 North Indian patients with tuberculosis undergoing antituberculosis treatment. Of these, 56 patients developed drug-induced hepatotoxicity (DIH group), whereas the remaining 290 patients did not (non-DIH group). The DIH group was comparatively older, had lower pretreatment serum albumin, and a higher frequency of moderately/far advanced disease radiographically than the latter. Further, patients with high alcohol intake had threefold higher odds of developing hepatotoxicity. In multivariate logistic regression analysis, older age (odds ratio [OR] 1.2), moderately/far advanced disease (OR 2.0), serum albumin < 3.5 g/dl (OR 2.3), absence of HLA-DQA1*0102 (OR 4.0), and presence of HLA-DQB1*0201 (OR 1.9) were independent risk factors for DIH. Our results suggest that the risk of hepatotoxicity from antituberculosis drugs is influenced by clinical and genetic factors.
Supporting Information
Supporting Information
ARTICLE SECTIONSCovalent binding of INH to hepatic proteins in mice; liver histology of mice and rats treated with INH; percentage of BSA and blue carrier protein lysine residues modified by INA-NHS; GLDH and ALT activities after 5 weeks of treatment of BN rats with INH; and SDH and ALT activities in Balb C and C57BL/6 mice. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.