1
|
Barreiro Carpio M, Valdes-Pena MA, Molina DA, Espinoza Cabello SEJ, Sialer Guerrero CA, Cribillero G, Vargas Coca KF, Icochea E. Evaluation of commercial doses of a feed additive and silymarin on broiler performance with and without CCl 4-induced liver damage. Poult Sci 2024; 103:103567. [PMID: 38417302 PMCID: PMC10909905 DOI: 10.1016/j.psj.2024.103567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/01/2024] Open
Abstract
Improving productive performance is a daily challenge in the poultry industry. Developing cost-effective additives and strategies that improve performance in antibiotic-free poultry production is critical to maintaining productivity and efficiency. This study evaluates the influence of a commercially available phytogenic feed additive (CA-PFA, that comprises silymarin, betaine and curcumin extracts as main ingredients) and silymarin on commercial broilers' productive performance and liver function with and without carbon tetrachloride (CCl4)-induced liver damage. The experiment was conducted in a completely randomized design, with six treatments, eight replicates, and eight birds per replicate in 18 one-day-old male broilers (Cobb Vantress 500) each; under a 3 × 2 factorial arrangement (3 diets x 2 levels of CCl4, 0 and 1 mL/kg body weight orally). The experimental treatments included 3 diets, commercially recommended doses of CA-PFA (500 mg/kg of feed; this dose provides 70 mg/kg of silymarin, besides the other active ingredients included in the formulation), silymarin (250 mg/kg of feed, containing 28% of active ingredient; this dose provides 70 mg/kg of silymarin as active ingredient) and an additive-free basal diet as a control. A standard commercial silymarin was used as a reference due to its well-known and extensively studied hepatoprotective properties that can mitigate the negative effects of CCl4 in the liver. The data were analyzed as a 2-way ANOVA, and the means showing significant (P ≤ 0.05) differences were then compared using the Post-Hoc Tukey HSD test. No interaction was detected between factors. Exposure to CCl4 had a noticeable detrimental effect on alertness, productive performance, and liver function of broilers without a significant increase in mortality. Including CA-PFA in the diet improved productive performance compared to the basal diet from day 21 to the end of the trial, on day 42. While no influence in feed intake was detected for any treatment, CA-PFA improved body weight gain (BWG) and feed conversion ratio (FCR) significantly (P < 0.05) from day 21 to the end of the trial in healthy and CCl4-exposed birds. The results show that CA-PFA supplementation improves performance parameters in broilers with and without CCl4-induced liver damage, when compared to a basal diet and the addition of a standard commercial silymarin product.
Collapse
Affiliation(s)
- Mabel Barreiro Carpio
- R&D Department, Ilender Perú S.A., Lima, Peru; Department of Chemistry, NC State University, Raleigh, NC.
| | - M Alejandro Valdes-Pena
- R&D Department, Ilender Perú S.A., Lima, Peru; Department of Chemistry, NC State University, Raleigh, NC
| | | | | | | | - Giovanna Cribillero
- School of Veterinary Medicine, Universidad Nacional Mayor de San Marcos, San Borja, Lima, Peru; Department of Poultry Science, Mississippi State University, Mississippi State, MS
| | - Katherine F Vargas Coca
- School of Veterinary Medicine, Universidad Nacional Mayor de San Marcos, San Borja, Lima, Peru
| | - Eliana Icochea
- School of Veterinary Medicine, Universidad Nacional Mayor de San Marcos, San Borja, Lima, Peru
| |
Collapse
|
2
|
Apte U, Bhushan B, Dadhania V. Hepatic Defenses Against Toxicity: Liver Regeneration and Tissue Repair. COMPREHENSIVE TOXICOLOGY 2018:368-396. [DOI: 10.1016/b978-0-12-801238-3.64918-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Dadhania VP, Bhushan B, Apte U, Mehendale HM. Wnt/β-Catenin Signaling Drives Thioacetamide-Mediated Heteroprotection Against Acetaminophen-Induced Lethal Liver Injury. Dose Response 2017; 15:1559325817690287. [PMID: 28210203 PMCID: PMC5302098 DOI: 10.1177/1559325817690287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Preplacement of compensatory tissue repair (CTR) by exposure to a nonlethal dose of a toxicant protects animals against a lethal dose of another toxicant. Although CTR is known to heteroprotect, the underlying molecular mechanisms are not completely known. Here, we investigated the mechanisms of heteroprotection using thioacetamide (TA): acetaminophen (APAP) heteroprotection model. Male Swiss Webster mice received a low dose of TA or distilled water (DW) vehicle 24 hours prior to a lethal dose of APAP. Liver injury, tissue repair, and promitogenic signaling were studied over a time course of 24 hours after APAP overdose to the TA- and DW-primed mice (TA + APAP and DW + APAP, respectively). Thioacetamide pretreatment afforded 100% protection against APAP overdose compared to 100% lethality in the DW + APAP-treated mice. Although hepatic Cyp2e1 was similar at the time of APAP administration, immediate activation of hepatic c-Jun N-terminal kinases (JNK) was observed in the TA + APAP-treated mice compared to its delayed activation in the DW + APAP group. In contrast to the DW + APAP group, the TA + APAP-treated mice exhibited extensive CTR, which was secondary to the timely activation of Wnt/β-catenin pathway. Our data indicate that rapid activation and appropriate termination of Wnt/β-catenin signaling and modulation of JNK activity underlie TA + APAP heteroprotection.
Collapse
Affiliation(s)
- Vivekkumar P Dadhania
- Department of Toxicology, College of Health & Pharmaceutical Sciences, The University of Louisiana at Monroe (ULM), Monroe, LA, USA
| | - Bharat Bhushan
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center (KUMC), Kansas City, KS, USA
| | - Udayan Apte
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center (KUMC), Kansas City, KS, USA
| | - Harihara M Mehendale
- Department of Toxicology, College of Health & Pharmaceutical Sciences, The University of Louisiana at Monroe (ULM), Monroe, LA, USA
| |
Collapse
|
4
|
Calabrese EJ, Baldwin LA. The Dose Determines the Stimulation (and Poison): Development of A Chemical Hormesis Database. Int J Toxicol 2016. [DOI: 10.1080/109158197226874] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
A comprehensive effort was undertaken to identify articles demonstrating chemical hormesis. Nearly 4000 potentially relevant articles were retrieved from preliminary computer searches utilizing various keyword descriptors and extensive cross-referencing. A priori evaluation criteria were established including study design features (e.g., number of doses, dose range), statistical analysis, and reproducibility of results. Evidence of chemical hormesis was judged to have occurred in approximately 350 of the 4000 studies evaluated. Chemical hormesis was observed in a wide range of taxonomic groups and involved agents representing highly diverse chemical classes, many of potential environmental relevance. Numerous biologic endpoints were assessed, with growth responses the most prevalent, followed by metabolic effects, longevity, reproductive responses, and survival. Hormetic responses were generally observed to be of limited magnitude with the average low-dose maximum stimulation approximately 50% greater than controls. The hormetic dose-response range was generally limited to about one order of magnitude with the upper end of the hormetic curve approaching the estimated no-observed-effect level (NOEL) for the particular endpoint. Based on the evaluation criteria, high to moderate evidence of hormesis was observed in studies comprised of ≥ doses with <3 doses in the hormetic zone. The present analysis suggests that chem ical hormesis is a reproducible and generalizable biologic phenomenon. Over the last decade advances have been made providing mechanistic insight helpful in explaining the phenomenon of chemical hormesis in multiple biologic systems with various endpoints. The reason for the uncertainty surrounding the existence of hormesis as a “real phenomenon” is believed to be the result of its relatively infrequent observation in the literature due to experimental design considerations, especially with respect to the number of doses, range of doses, and endpoint selection.
Collapse
Affiliation(s)
- Edward J. Calabrese
- Department of Environmental Health Sciences, School of Public Health, University of Massachusetts, Amherst, MA 01003, USA
| | - Linda A. Baldwin
- Department of Environmental Health Sciences, School of Public Health, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
5
|
Ramaiah SK, Apte U, Mehendale HM. Diet Restriction as a Protective Mechanism in Noncancer Toxicity Outcomes: A Review. Int J Toxicol 2016. [DOI: 10.1080/109158100750058776] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
It is well documented that diet restriction (DR) increases life expectancy, slows aging, and decreases the incidence of a variety of age-associated diseases including cancer and chemical-induced carcinogenesis. With regard to chemical toxicity, very few studies have attempted to investigate the effects of DR on noncancer toxicity outcomes. This review summarizes the findings of how DR influences acute toxicity outcomes and mechanisms. DR-induced protection in ozone lung inflammation, acute toxicity of isoproterenol, ganciclovir-, aspirin-and acidified ethanol-induced gastric injury are discussed. Because similar physiologic mechanisms exist in humans, DR, if practiced as a fife-style option, may improve quality of life in addition to accruing savings in burgeoning health care costs. Finally, these studies may be of value in boosting research in the area of pharmacology and therapeutics in developing potential therapeutic and safety assessment tools in human and veterinary medicine.
Collapse
Affiliation(s)
- Shashi K. Ramaiah
- Department of Toxicology and Louisiana Institute of Toxicology, College of Pharmacy, The University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - Udayan Apte
- Department of Toxicology and Louisiana Institute of Toxicology, College of Pharmacy, The University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - Harihara M. Mehendale
- Department of Toxicology and Louisiana Institute of Toxicology, College of Pharmacy, The University of Louisiana at Monroe, Monroe, Louisiana, USA
| |
Collapse
|
6
|
Abstract
Dietary restriction (DR) also called dietary control or calorie restriction is reported to have many advantages with regard to human health. It leads to suppression of obesity, mitigates free radicals and increases available antioxidants which are accounted for extending the life span of individuals. DR is also reported to induce synthesis of heat shock proteins in animals as a control mechanism against stress. Further, it is known to play a significant role in decreasing toxicity and lethality due to a variety of toxic chemicals and drugs by stimulating tissue repair damaged by the toxicants leading to restoration of intact organ and its functions. Moreover, extensive work done on animals indicate DR has an important role in suppressing certain types of cancer. In this review an effort is made to highlight the various advantages of DR from the point of human health perspective.
Collapse
Affiliation(s)
- M N Astagimath
- Department of Biochemistry, Kasturba Medical College, 575 004 Bejai, Mangalore
| | | |
Collapse
|
7
|
Lyon P, Cohen M, Quintner J. An Evolutionary Stress-Response Hypothesis for Chronic Widespread Pain (Fibromyalgia Syndrome). PAIN MEDICINE 2011; 12:1167-78. [DOI: 10.1111/j.1526-4637.2011.01168.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
8
|
Moyer AM, Fridley BL, Jenkins GD, Batzler AJ, Pelleymounter LL, Kalari KR, Ji Y, Chai Y, Nordgren KKS, Weinshilboum RM. Acetaminophen-NAPQI hepatotoxicity: a cell line model system genome-wide association study. Toxicol Sci 2010; 120:33-41. [PMID: 21177773 DOI: 10.1093/toxsci/kfq375] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acetaminophen is the leading cause of acute hepatic failure in many developed nations. Acetaminophen hepatotoxicity is mediated by the reactive metabolite N-acetyl-p-benzoquinonimine (NAPQI). We performed a "discovery" genome-wide association study using a cell line-based model system to study the possible contribution of genomics to NAPQI-induced cytotoxicity. A total of 176 lymphoblastoid cell lines from healthy subjects were treated with increasing concentrations of NAPQI. Inhibiting concentration 50 values were determined and were associated with "glutathione pathway" gene single nucleotide polymorphisms (SNPs) and genome-wide basal messenger RNA expression, as well as with 1.3 million genome-wide SNPs. A group of SNPs in linkage disequilibrium on chromosome 3 was highly associated with NAPQI toxicity. The p value for rs2880961, the SNP with the lowest p value, was 1.88 × 10(-7). This group of SNPs mapped to a "gene desert," but chromatin immunoprecipitation assays demonstrated binding of several transcription factor proteins including heat shock factor 1 (HSF1) and HSF2, at or near rs2880961. These chromosome 3 SNPs were not significantly associated with variation in basal expression for any of the genome-wide genes represented on the Affymetrix U133 Plus 2.0 GeneChip. We have used a cell line-based model system to identify a SNP signal associated with NAPQI cytotoxicity. If these observations are validated in future clinical studies, this SNP signal might represent a potential biomarker for risk of acetaminophen hepatotoxicity. The mechanisms responsible for this association remain unclear.
Collapse
Affiliation(s)
- Ann M Moyer
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Liu Y, Chen X, Duan S, Feng Y, An M. Mathematical modeling of plant allelopathic hormesis based on ecological-limiting-factor models. Dose Response 2010; 9:117-29. [PMID: 21431081 PMCID: PMC3057639 DOI: 10.2203/dose-response.09-050.liu] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Allelopathy arises from the release of chemicals by one plant species that affect other species in its vicinity, usually to their detriment. Allelopathic effects have been demonstrated to be limiting factors for species distributions and ecological processes in some natural or agricultural communities. Based on the biphasic hormetic responses of plants to allelochemicals, ecological-limiting-factor models were introduced into the An-Johnson-Lovett hormesis model to improve modelling the phenomenon of allelopathic hormesis and to better reflect the nature of allelopathy as a limiting factor in ecological processes. Outcomes of the models have been compared for several sets of experimental data from the literature and good agreement between the models and data was observed, which indicates that the new models give some insight into the ecological mechanisms involved and may provide more options for modelling the allelopathic phenomenon as well as platforms for further research on plant allelopathic hormesis.
Collapse
Affiliation(s)
- Yinghu Liu
- Key Laboratory of Ecological Agriculture of Ministry of Agriculture, College of Science, South China Agricultural University, Wushan, Guangzhou 510642, China
| | | | | | | | | |
Collapse
|
10
|
|
11
|
Liu J, Zhou ZX, Zhang W, Bell MW, Waalkes MP. Changes in hepatic gene expression in response to hepatoprotective levels of zinc. Liver Int 2009; 29:1222-9. [PMID: 19490425 PMCID: PMC3500762 DOI: 10.1111/j.1478-3231.2009.02007.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Zinc (Zn) administration at non-toxic doses protects against the hepatotoxicity produced by many agents, but the underlying mechanisms remain elusive. AIM To examine the basis of Zn-induced generalised hepatoprotective effects. METHODS Rats and mice were given Zn at known hepatoprotective levels (100 mumol ZnCl2/kg/day, s.c., for 4 days) and molecular responses were assessed. RESULTS Zn treatment produced changes in 5% of the genes on custom-designed mouse liver array and Rat Toxicology-II array. Changes in gene expression were further confirmed and extended by real-time reverse transcriptase-polymerase chain reaction. Zn treatment dramatically increased the expression of the metallothionein (Mt), and modestly increased the expression of acute-phase protein genes (ceruloplasmin, Stat3, egr1, Cxc chemokines and heat-shock proteins). For genes encoding for antioxidant enzymes, some were increased (Nrf2 and Nqo1), while others remained unaltered (Cu, Zn SOD and glutathione S-transferases). Expressions of cytokine and pro-inflammatory genes were not affected, while genes related to cell proliferation (cyclin D1) were modestly upregulated. Some metabolic enzyme genes, including cytochrome P450s and UDP-glucuronosyltransferase, were modestly suppressed, perhaps to switch cellular metabolic energy to acute-phase responses. Liver Zn content was increased between 1.6- and 2.1-fold, while hepatic MT protein was increased between 50 and 200-fold. Mice typically showed greater responses than rats. CONCLUSION Such gene expression changes, particularly the dramatic induction of MT and Nrf2 antioxidant pathway, occur in the absence of overt liver injury, and are probably important in the hepatoprotective effects of Zn against toxic insults.
Collapse
Affiliation(s)
- Jie Liu
- Inorganic Carcinogenesis Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at NIEHS, Research Triangle Park, NC
| | - Zhan-Xiang Zhou
- Department of Medicine, University of Louisville, Louisville, KY
| | - Wei Zhang
- Laboratory of Pharmacology, NIEHS, Research Triangle Park, NC
| | - Matthew W. Bell
- Inorganic Carcinogenesis Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at NIEHS, Research Triangle Park, NC
| | - Michael P. Waalkes
- Inorganic Carcinogenesis Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at NIEHS, Research Triangle Park, NC
| |
Collapse
|
12
|
Liu J, Wu Q, Lu YF, Pi J. New insights into generalized hepatoprotective effects of oleanolic acid: key roles of metallothionein and Nrf2 induction. Biochem Pharmacol 2008; 76:922-8. [PMID: 18706400 DOI: 10.1016/j.bcp.2008.07.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 07/14/2008] [Accepted: 07/15/2008] [Indexed: 12/15/2022]
Abstract
Oleanolic acid (OA) is a natural triperpenoid that protects against a variety of hepatotoxicants such as carbon tetrachloride, cadmium, acetaminophen, and bromobenzene. To gain insight into the molecular mechanisms of this generalized hepatoprotection, genomic analysis was performed on mouse and rat livers after OA treatment. Mice and rats were given OA at a hepatoprotective dose (50 micromol/kg, s.c., daily for 4 days) and hepatic RNA was isolated, purified, and subjected to gene expression analysis. OA treatment produced changes in 5% of the genes on custom-designed mouse liver array and rat toxicology-II array. Changes in key gene expressions were further analyzed by real-time RT-PCR. OA treatment dramatically increased expression of hepatic metallothionein (Mt), and increased the expression of the nuclear factor E2-related factor 2 (Nrf2), NAD(P)H:quinone oxidoreductase 1 (Nqo1), heme oxygenase-1 (Hmox1), and glutamate-cysteine ligases (Gclc and Gclm). OA treatment also increased the expression of genes related to cell proliferation and suppressed the expression of several cytochrome P450 genes possibly to switch cellular metabolic energy to an acute-phase response. Hepatic MT protein was increased 60- and 15-fold in mice and rats, respectively, together with a 30% increase in mouse liver zinc. These gene expression changes, particularly the dramatic induction of MT and the Nrf2 signaling, occur with hepatoprotection doses of OA, and likely are important in the generalized protective effects of OA against hepatotoxicants.
Collapse
Affiliation(s)
- Jie Liu
- Zunyi Medical College, Key Laboratory of Pharmacology, 201 Dalian Road, Zunyi 563000, China.
| | | | | | | |
Collapse
|
13
|
Goldani HAS, Matte US, Ramos ARL, Costa TG, Winkelmann LV, Meurer L, Vieira SMG, Kieling CO, Silveira TR. The role of food restriction on CCl4-induced cirrhosis model in rats. ACTA ACUST UNITED AC 2007; 58:331-7. [PMID: 17275271 DOI: 10.1016/j.etp.2006.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Accepted: 11/11/2006] [Indexed: 12/14/2022]
Abstract
Effects of food restriction on susceptibility to the toxic effect of some chemicals are controversial. In order to identify an exposure model that could maximize cirrhosis and minimize mortality rate, this study aimed to evaluate the effect of food restriction on tetrachloride carbon (CCl(4))-induced cirrhosis model in rats. Fifty-three male Wistar rats received CCl(4) 0.25 ml/kg weekly intragastrically once a week. Thirty-three had 44% food restriction (group 1); 10 rats had 25% food restriction (group 2); and 10 rats received ad libitum food (group 3). After 10 weeks, the animals were sacrificed and liver sections were collected for histology. Of the 53 animals enrolled for the study, 22 (41.5%) died before completing 10-week CCl(4). Mortality rate was significantly higher in group 1 compared to other groups (p<0.05). Cirrhosis was significantly more prevalent in group 1 than in group 3 (p<0.01), but without significant difference between groups 1 and 2 (p=0.624). We concluded that food restriction is an important issue to be considered when establishing a CCl(4)-induced cirrhosis model in rats. Moreover, there is an ideal range of food intake that predisposes to liver damage without increasing mortality leading to a more effective model.
Collapse
Affiliation(s)
- Helena A S Goldani
- Laboratório de Hepatologia Experimental, Centro de Pesquisas do Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, 90035-003 Porto Alegre-RS, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Sawant SP, Dnyanmote AV, Mehendale HM. Mechanisms of inhibited liver tissue repair in toxicant challenged type 2 diabetic rats. Toxicology 2007; 232:200-15. [PMID: 17298859 DOI: 10.1016/j.tox.2007.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 01/01/2007] [Accepted: 01/05/2007] [Indexed: 01/08/2023]
Abstract
Liver injury initiated by non-lethal doses of CCl(4) and thioacetamide (TA) progresses to hepatic failure and death of type 2 diabetic (DB) rats due to failed advance of liver cells from G(0)/G(1) to S-phase and inhibited tissue repair. Objective of the present study was to investigate cellular signaling mechanisms of failed cell division in DB rats upon hepatotoxicant challenge. In CCl(4)-treated non-diabetic (non-DB) rats, increased IL-6 levels, sustained activation of extracellular regulated kinases 1/2 (ERK1/2) MAPK, and sustained phosphorylation of retinoblastoma protein (p-pRB) via cyclin D1/cyclin-dependent kinase (cdk) 4 and cyclin D1/cdk6 complexes stimulated G(0)/G(1) to S-phase transition of liver cells. In contrast to the non-DB rats, CCl(4) administration led to lower plasma IL-6, decreased ERK1/2 activation, lower cyclin D1, and cdk 4/6 expression resulting in decreased p-pRB and inhibition of liver cell division in the DB rats. Furthermore, higher TGFbeta1 expression and p21 activation may also contribute to decreased p-pRB in DB rats compared to non-DB rats. Similarly, after TA administration to DB rats, down-regulation of cyclin D1 and p-pRB leads to markedly decreased advance of liver cells from G(0)/G(1) to S-phase and tissue repair compared to the non-DB rats. Hepatic ATP levels did not differ between the DB and non-DB rats obviating its role in failed tissue repair in the DB rats. In conclusion, decreased p-pRB may contribute to blocked advance of cells from G(0)/G(1) to S-phase and failed cell division in DB rats exposed to CCl(4) or TA, leading to progression of liver injury and hepatic failure.
Collapse
Affiliation(s)
- Sharmilee P Sawant
- Department of Toxicology, College of Pharmacy, The University of Louisiana at Monroe, 700 University Avenue, Sugar Hall #306, Monroe, LA 71209-0470, USA
| | | | | |
Collapse
|
15
|
Abstract
The mechanism of carbon tetrachloride (CCl4)-induced hepatotoxicity, especially necrosis and fatty liver, has long been a challenging subject of many researchers from various fields over the past 50 years. Even though the mechanisms of tissue damages are different among chemicals and affected tissues, CCl4 has played a role as a key substance of tissue injury. A number of studies have been conducted and various hypotheses have been raised. As a result, several important basic mechanisms of tissue damages have emerged, involving metabolic activation, reactive free radical metabolites, lipid peroxidation, covalent binding and disturbance of calcium homeostasis. Recent studies also revealed inflammation and regeneration as important modification factors in the tissue injury. The author attempted to summarize the history of CCl4 research with some emphasis on the experiments done by the author and his colleagues. Their studies with isolated perfused rat liver suggest that covalent binding of CCl4 metabolites rather than lipid peroxidation has a significant role in the production of centrilobular necrosis following CCl4 administration. Further studies are necessary to unveil detailed mechanisms of hepatocyte necrosis induced by CCl4.
Collapse
Affiliation(s)
- Yasusuke Masuda
- Faculty of Pharmacy, Niigata University of Pharmacy and Applied Life Sciences, Niigata City, Japan
| |
Collapse
|
16
|
McCarty LS, Borgert CJ. Review of the toxicity of chemical mixtures: Theory, policy, and regulatory practice. Regul Toxicol Pharmacol 2006; 45:119-43. [PMID: 16701933 DOI: 10.1016/j.yrtph.2006.03.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Indexed: 10/24/2022]
Abstract
An analysis of current mixture theory, policy, and practice was conducted by examining standard reference texts, regulatory guidance documents, and journal articles. Although this literature contains useful theoretical concepts, clear definitions of most terminology, and well developed protocols for study design and statistical analysis, no general theoretical basis for the mechanisms and interactions of mixture toxicity could be discerned. There is also a poor understanding of the relationship between exposure-based and internal received dose metrics. This confounds data interpretation and limits reliable determinations of the nature and extent of additivity. The absence of any generally accepted classification scheme for either modes/mechanisms of toxic action or of mechanisms of toxicity interactions is problematic as it produces a cycle in which research and policy are interdependent and mutually limiting. Current regulatory guidance depends heavily on determination of toxicological similarity concluded from the presence of a few prominent constituents, assumed from a common toxicological effect, or presumed from an alleged similar toxic mode/mechanism. Additivity, or the lack of it, is largely based on extrapolation of existing knowledge for single chemicals in this context. Thus, regulatory risk assessment protocols lack authoritative theoretical underpinnings, creating substantial uncertainty. Development of comprehensive classification schemes for modes/mechanisms of toxic action and mechanisms of interaction is needed to ensure a sound theoretical foundation for mixture-related regulatory activity and provide a firm basis for iterative hypothesis development and experimental testing.
Collapse
Affiliation(s)
- L S McCarty
- L.S. McCarty Scientific Research & Consulting, 94 Oakhaven Drive, Markham, Ont., Canada L6C 1X8.
| | | |
Collapse
|
17
|
Mehendale HM. Tissue repair: an important determinant of final outcome of toxicant-induced injury. Toxicol Pathol 2005; 33:41-51. [PMID: 15805055 DOI: 10.1080/01926230590881808] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Tissue repair is a dynamic compensatory cell proliferation and tissue regeneration response stimulated in order to overcome acute toxicity and recover organ/tissue structure and function. Extensive evidence in rodent models using structurally and mechanistically diverse hepatotoxicants such as acetaminophen (APAP), carbon tetrachloride (CCl4), chloroform (CHCl3), thioacetamide (TA), trichloroethylene (TCE), and allyl alcohol (AA) have demonstrated that tissue repair plays a critical role in determining the final outcome of toxicity, i.e., recovery from injury and survival or progression of injury leading to liver failure and death. Tissue repair is a complex process governed by intricate cellular signaling involving a number of chemokines, cytokines, growth factors, and nuclear receptors leading to promitogenic gene expression and cell division. Tissue repair also encompasses regeneration of hepatic extracellular matrix and angiogenesis, the processes necessary to completely restore the structure and function of the liver tissue lost to toxicant-induced initiation followed by progression of injury. New insights have emerged over the last quarter century indicating that tissue repair follows a dose response. Tissue repair increases with dose until a threshold dose, beyond which it is delayed and impaired due to inhibition of cellular signaling resulting in runaway secondary events causing tissue destruction, organ failure, and death. Prompt and adequately stimulated tissue repair response to toxic injury is critical for recovery from toxic injury. Tissue repair is modulated by a variety of factors including species, strain, age, nutrition, and disease condition causing marked changes in susceptibility and toxic outcome. This review focuses on the properties of tissue repair, different factors affecting tissue repair, and the mechanisms that govern tissue repair and progression of injury. It also highlights the significance of tissue repair as a target for drug development strategies and an important consideration in the assessment of risk from exposure to toxicants.
Collapse
Affiliation(s)
- Harihara M Mehendale
- Department of Toxicology College of Health Sciences, The University of Louisiana at Monroe, Monroe, Louisiana 71209, USA. mehendale @ulm.edu
| |
Collapse
|
18
|
Anand SS, Mehendale HM. Liver regeneration: a critical toxicodynamic response in predictive toxicology. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2004; 18:149-160. [PMID: 21782744 DOI: 10.1016/j.etap.2004.02.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2003] [Accepted: 02/19/2004] [Indexed: 05/31/2023]
Abstract
The objective of the present review is to discuss the importance tissue repair in the mixture risk assessment. Studies have revealed the existence of two stages of toxicity: an inflictive stage (stage I) and progressive or regressive stage (stage II). While much is known about mechanisms by which injury is inflicted (stage I), very little is known about the mechanisms that lead to progression or regression of injury. A wide variety of additional experimental evidence suggests that tissue repair impacts decisively on the final toxic outcome and any modulation in this response has profound impact in the final outcome of toxicity. We designed the present research to investigate the importance of tissue repair in the final acute hepatotoxic outcome upon exposures to mixture of toxicants comprising thioacetamide (TA), allyl alcohol (AA), chloroform (CHCl(3)) and trichloroethylene (TCE). Dose response studies with individual compounds, binary mixtures (BM), ternary (TM) and quaternary mixtures (QM) have been conducted. Results of CHCl(3) + AA BM [Anand, S.S., Murthy, S.N., Vishal, V.S., Mumtaz, M.M., Mehendale, H.M., 2003. Tissue repair plays pivotal role in final outcome of supra-additive liver injury after chloroform and allyl alcohol binary mixture. Food Chem. Toxicol. 41, 1123] and CHCl(3) + AA + TA +TCE QM [Soni, M.G., Ramaiah, S.K., Mumtaz, M.M., Clewell, H., Mehendale, H.M., 1999. Toxicant-inflicted injury and stimulated tissue repair are opposing toxicodynamic forces in predictive toxicology. Regul. Phramcol. Toxicol. 19, 165], and two representative individual compounds (TA and AA) [Mangipudy, R.S., Chanda, S., Mehendale, H.M., 1995a. Tissue repair response as a function of dose in thioacetamide hepatotoxicity. Environ. Health Perspect. 103, 260; Soni, M.G., Ramaiah, S.K., Mumtaz, M.M., Clewell, H., Mehendale, H.M., 1999. Toxicant-inflicted injury and stimulated tissue repair are opposing toxicodynamic forces in predictive toxicology. Regul. Phramcol. Toxicol. 19, 165] are described in this review. In addition, modulation of tissue repair in the outcome of hepatotoxicity and its implications in the risk assessment have been discussed. Male Sprague-Dawley (S-D) rats (250-300g) received a single i.p. injection of individual toxicants as well as mixtures. Liver injury was assessed by plasma alanine amino transferase (ALT) and histopathology. Tissue regeneration response was measured by [(3)H]-thymidine ((3)H-T) incorporation into hepatocellular nuclear DNA and PCNA. Only ALT and (3)H-T data have been presented in this review for the sake of simplicity. Studies with individual hepatotoxicants showed a dose-related increase in injury as well as tissue repair up to a threshold dose. Beyond this threshold, tissue repair was inhibited, and liver injury progressed leading to mortality. Since the highest dose of individual compounds resulted in mortality, this dose was not employed for mixture studies. While CHCl(3) + AA BM caused supra-additive liver injury, QM caused additive liver injury. Due to the prompt and robust compensatory tissue repair, all the rats exposed to BM survived. With QM, the rats receiving the highest dose combination experienced some mortality consequent to the progression of liver injury attendant to suppressed tissue repair. These findings suggest that liver tissue repair, the opposing biological response that restores tissue lost to injury, may play a critical and determining role in the outcome of liver injury regardless of the number of toxicants in the mixture or the mechanism of initiation of injury. These data suggest that inclusion of this response in risk assessment might help in fine-tuning the prediction of toxic outcomes.
Collapse
Affiliation(s)
- Sathanandam S Anand
- Department of Toxicology, School of Pharmacy, College of Health Sciences, The University of Louisiana at Monroe, 700 University Avenue, Sugar Hall 306, Monroe, LA 71209, USA
| | | |
Collapse
|
19
|
Weber LWD, Boll M, Stampfl A. Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Crit Rev Toxicol 2004; 33:105-36. [PMID: 12708612 DOI: 10.1080/713611034] [Citation(s) in RCA: 1140] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of many halogenated alkanes such as carbon tetrachloride (CCl4), chloroform (CHCl3) or iodoform (CHI3), has been banned or severely restricted because of their distinct toxicity. Yet CCl4 continues to provide an important service today as a model substance to elucidate the mechanisms of action of hepatotoxic effects such as fatty degeneration, fibrosis, hepatocellular death, and carcinogenicity. In a matter of dose,exposure time, presence of potentiating agents, or age of the affected organism, regeneration can take place and lead to full recovery from liver damage. CCl4 is activated by cytochrome (CYP)2E1, CYP2B1 or CYP2B2, and possibly CYP3A, to form the trichloromethyl radical, CCl3*. This radical can bind to cellular molecules (nucleic acid, protein, lipid), impairing crucial cellular processes such as lipid metabolism, with the potential outcome of fatty degeneration (steatosis). Adduct formation between CCl3* and DNA is thought to function as initiator of hepatic cancer. This radical can also react with oxygen to form the trichloromethylperoxy radical CCl3OO*, a highly reactive species. CCl3OO* initiates the chain reaction of lipid peroxidation, which attacks and destroys polyunsaturated fatty acids, in particular those associated with phospholipids. This affects the permeabilities of mitochondrial, endoplasmic reticulum, and plasma membranes, resulting in the loss of cellular calcium sequestration and homeostasis, which can contribute heavily to subsequent cell damage. Among the degradation products of fatty acids are reactive aldehydes, especially 4-hydroxynonenal, which bind easily to functional groups of proteins and inhibit important enzyme activities. CCl4 intoxication also leads to hypomethylation of cellular components; in the case of RNA the outcome is thought to be inhibition of protein synthesis, in the case of phospholipids it plays a role in the inhibition of lipoprotein secretion. None of these processes per se is considered the ultimate cause of CCl4-induced cell death; it is by cooperation that they achieve a fatal outcome, provided the toxicant acts in a high single dose, or over longer periods of time at low doses. At the molecular level CCl4 activates tumor necrosis factor (TNF)alpha, nitric oxide (NO), and transforming growth factors (TGF)-alpha and -beta in the cell, processes that appear to direct the cell primarily toward (self-)destruction or fibrosis. TNFalpha pushes toward apoptosis, whereas the TGFs appear to direct toward fibrosis. Interleukin (IL)-6, although induced by TNFalpha, has a clearly antiapoptotic effect, and IL-10 also counteracts TNFalpha action. Thus, both interleukins have the potential to initiate recovery of the CCl4-damaged hepatocyte. Several of the above-mentioned toxication processes can be specifically interrupted with the use of antioxidants and mitogens, respectively, by restoring cellular methylation, or by preserving calcium sequestration. Chemicals that induce cytochromes that metabolize CCl4, or delay tissue regeneration when co-administered with CCl4 will potentiate its toxicity thoroughly, while appropriate CYP450 inhibitors will alleviate much of the toxicity. Oxygen partial pressure can also direct the course of CCl4 hepatotoxicity. Pressures between 5 and 35 mmHg favor lipid peroxidation, whereas absence of oxygen, as well as a partial pressure above 100 mmHg, both prevent lipid peroxidation entirely. Consequently, the location of CCl4-induced damage mirrors the oxygen gradient across the liver lobule. Mixed halogenated methanes and ethanes, found as so-called disinfection byproducts at low concentration in drinking water, elicit symptoms of toxicity very similar to carbon tetrachloride, including carcinogenicity.
Collapse
Affiliation(s)
- Lutz W D Weber
- Institute of Toxicology, GSF-National Research Center for Environment and Health, Munich, P.O. Box 1129, D-85758 Neuherberg (FRG).
| | | | | |
Collapse
|
20
|
Murali B, Korrapati MC, Warbritton A, Latendresse JR, Mehendale HM. Tolerance of aged Fischer 344 rats against chlordecone-amplified carbon tetrachloride toxicity. Mech Ageing Dev 2004; 125:421-35. [PMID: 15178132 DOI: 10.1016/j.mad.2004.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2003] [Revised: 02/27/2004] [Accepted: 03/01/2004] [Indexed: 01/11/2023]
Abstract
We have investigated the effects of chlordecone 1(CD)+CCl4 combination in adult (3 months), middle aged (14 months), and old aged (24 months) male Fischer 344 (F344) rats. After a non-toxic dietary regimen of CD (10 ppm) or normal powdered diet for 15 days, rats received a single non-toxic dose of CCl4 (100 microl/kg, i.p., 1:4 in corn oil) or corn oil (500 microl/kg, i.p.) alone on day 16. Liver injury was assessed by plasma ALT, AST, and histopathology during a time course of 0-96 h. Liver tissue repair was measured by [3H-CH3]-thymidine (3H-T) incorporation into hepatic nuclear DNA and proliferating cell nuclear antigen (PCNA) immunohistochemistry. Hepatomicrosomal CYP2E1 protein, enzyme activity, and covalent binding of 14CCl4-derived radiolabel were measured in normal and CD fed rats. Exposure to CCl4 alone caused modest liver injury only in 14- and 24-month-old rats but neither progression of injury nor mortality. The CD+CCl4 combination led to 100% mortality in 3-month-old rats by 72 h, whereas none of the 14- and 24-month-old rats died. Both 3- and 14-month-old rats exposed to CD+Cl4 had identical liver injury up to 36 h indicating that bioactivation-mediated CCl4 injury was the same in the two age groups. Thereafter, liver injury escalated only in 3-month-old while it declined in 14-month-old rats. In 24-month-old rats initial liver injury at 6 h was similar to the 3- and 14-month-old rats and thereafter did not develop to the level of the other two age groups, recovering from injury by 96 h as in the 14-month-old rats. Neither hepatomicrosomal CYP2E1 protein nor the associated p-nitrophenol hydroxylase activity or covalent binding of 14CCl4-derived radiolabel to liver tissue differed between the age groups or diet regimens 2 h after the administration of 14CCl4. Compensatory liver tissue repair (3H-T, PCNA) was prompt and robust soon after CCl4 liver injury in the 14- and 24-month-old rats. In stark contrast, in the 3-month-old rats it failed allowing unabated progression of liver injury. These findings suggest that stimulation of early onset and robust liver tissue repair rescue the 14- and 24-month-old F344 rats from the lethal effect of the CD+CCl4 combination.
Collapse
Affiliation(s)
- B Murali
- Department of Toxicology, Sugar Hall #306, School of Pharmacy, College of Health Sciences, The University of Louisiana at Monroe, 700 University Avenue, Monroe, LA 71209, USA
| | | | | | | | | |
Collapse
|
21
|
Vaidya VS, Shankar K, Lock EA, Bucci TJ, Mehendale HM. Renal injury and repair following S-1, 2 dichlorovinyl-L-cysteine administration to mice. Toxicol Appl Pharmacol 2003; 188:110-21. [PMID: 12691729 DOI: 10.1016/s0041-008x(02)00080-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
S-(1,2-dichlorovinyl)-L-cysteine (DCVC), a metabolite of a common environmental contaminant, trichloroethylene, is a selective proximal tubular nephrotoxicant. The objective of our study was to examine the dose-response relationship of renal injury and repair following DCVC administration. Male Swiss-Webster mice were injected with DCVC [15, 30, or 75 mg/kg ip in distilled water (10 ml/kg)] and the extent of nephrotoxicity and tissue repair was assessed over a 14-day period. The renal injury due to the low and medium doses of DCVC peaked at 36 and 72 h after dosing, respectively, and then regressed over time due to a timely and adequate tissue repair response. At the highest dose tissue repair was inhibited, thereby causing progression of renal injury, which led to acute renal failure and death of the mice. The possibility that compromised tissue repair was a result of the extensive nephrotoxic injury attendant to the high dose of DCVC was investigated via an equinephrotoxicity study in which separate groups of mice received 40 (LD40) and 75 (LD90) mg DCVC/kg, respectively. Bioactivation-based renal proximal tubular injury measured in these two groups over a time course was identical but there was a marked difference in mortality due to an early and robust tissue repair in the first group relative to the second group. These results support the concept that quantitative evaluation of renal tissue repair in parallel with injury is useful in the assessment of the likely toxic outcome associated with exposure to nephrotoxic drugs and toxicants.
Collapse
Affiliation(s)
- Vishal S Vaidya
- Department of Toxicology, College of Pharmacy, The University of Louisiana at Monroe, Monroe, LA 71209, USA
| | | | | | | | | |
Collapse
|
22
|
Abstract
The chlorinated methanes, particularly carbon tetrachloride and chloroform, are classic models of liver injury and have developed into important experimental hepatoxicants over the past 50 years. Hepatocellular steatosis and necrosis are features of the acute lesion. Mitochondria and the endoplasmic reticulum as target sites are discussed. The sympathetic nervous system, hepatic hemodynamic alterations, and role of free radicals and biotransformation are considered. With carbon tetrachloride, lipid peroxidation and covalent binding to hepatic constituents have been dominant themes over the years. Potentiation of chlorinated methane-induced liver injury by alcohols, aliphatic ketones, ketogenic compounds, and the pesticide chlordecone is discussed. A search for explanations for the potentiation phenomenon has led to the discovery of the role of tissue repair in the overall outcome of liver injury. Some final thoughts about future research are also presented.
Collapse
Affiliation(s)
- G L Plaa
- Département de Pharmacologie, Faculté de Médecine, Université de Montréal, Québec, Canada.
| |
Collapse
|
23
|
Liu J, Sendelbach LE, Parkinson A, Klaassen CD. Endotoxin pretreatment protects against the hepatotoxicity of acetaminophen and carbon tetrachloride: role of cytochrome P450 suppression. Toxicology 2000; 147:167-76. [PMID: 10924799 DOI: 10.1016/s0300-483x(00)00193-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bacterial endotoxin (lipopolysaccharide, LPS) is known to potentiate the toxicity of many hepatotoxicants. However, exposure to a sublethal dose of LPS renders animals tolerant to a lethal dose of LPS, and protects against the toxicity of some chemicals. This study was designed to examine the effects of LPS pretreatment on acetaminophen- and carbon tetrachloride (CCl(4))-induced liver injury in LPS-sensitive C3H/OuJ and LPS-resistant C3H/HeJ mice. Pretreatment of male C3H/OuJ mice with a single injection of LPS (0. 1 mg/kg, ip, for 24 h) protected against the hepatotoxic effects of acetaminophen (400 mg/kg) and carbon tetrachloride (CCl(4), 30 mg/kg), as indicated by serum alanine aminotransferase activity. In contrast, pretreatment of C3H/HeJ mice with 0.1 or 10 mg/kg LPS afforded no protection against the hepatotoxic effects of acetaminophen and CCl(4). In an attempt to determine the mechanism of LPS-induced protection against acetaminophen- and CCl(4)-induced hepatotoxicity in C3H/OuJ mice, liver cytochrome P450 was determined 24 h after LPS injection. LPS treatment caused a 26% decrease in total P450 content in C3H/OuJ but not in C3H/HeJ mice. CYP3A-catalized testosterone 6 beta-, 2 beta-, and 15 beta-hydroxylation was decreased 40% by LPS only in C3H/OuJ mice. To determine whether the differences to LPS-response in the two stains of mice is mediated by a strain-related difference in the release of cytokines, mice were pretreated with interleukin-1 (IL-1 alpha, 5 x 10(5) U/mouse), and the hepatoprotection and hepatic P450 enzymes were examined. IL-1 alpha pretreatment equally protected against the hepatotoxicity of acetaminophen and CCl(4) in both strains, and suppressed the total microsomal P450 and P450 enzyme-catalyzed testosterone hydroxylation to a similar extent. In conclusion, LPS pretreatment suppressed hepatic cytochrome P450 enzymes and protected against the hepatotoxicity of acetaminophen and CCl(4) in LPS-sensitive C3H/OuJ mice, but not in LPS-refractory C3H/HeJ mice. This protective effect of LPS appears to be mediated through the release of cytokines such as IL-1 alpha, which in turn suppresses the cytochrome P450 responsible for the activation of acetaminophen and CCl(4) to reactive metabolites.
Collapse
Affiliation(s)
- J Liu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160-7417, USA
| | | | | | | |
Collapse
|
24
|
Calabreseci EJ. Evidence that hormesis represents an "overcompensation" response to a disruption in homeostasis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 1999; 42:135-137. [PMID: 10051361 DOI: 10.1006/eesa.1998.1729] [Citation(s) in RCA: 137] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The hypothesis that growth hormesis represents an overcompensation to a disruption in homeostasis was supported in experiments assessing the effects of the synthetic plant growth inhibitor phosfon (2,4-dichlorobenzyl tributyl phosphonium chloride) on peppermint. While dose-dependent decreases in growth occurred initially in all treatment groups substantial compensation growth subsequently occurred across treatment groups. The low-dose treatment groups more than fully compensated for the initial growth decrement (20-25%), displaying a net stimulatory response by 5 weeks, whereas the high-dose treatment groups never fully compensated for the initial growth decrement (50-75%). These findings, within the context of other similar reported observations, are believed to be widely generalizable and may have important implications for biomedical fields including the design and interpretation of the bioassay especially within the context of the hazard/risk assessment process.
Collapse
Affiliation(s)
- E J Calabreseci
- School of Public Health, University of Massachusetts, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
25
|
Moghaddam AP, Eggers JS, Calabrese EJ. Evaluation of sex difference in tissue repair following acute carbon tetrachloride toxicity in male and female Sprague-Dawley rats. Toxicology 1998; 130:95-105. [PMID: 9865477 DOI: 10.1016/s0300-483x(98)00095-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cellular regeneration and tissue repair greatly influence the outcome of acute carbon tetrachloride (CCl4) hepatotoxicity. This study examined the temporal kinetics of cellular regeneration and tissue repair processes in male and female Sprague Dawley (SD) rats following an acute CCl4 exposure (0.8 ml/kg, i.p.). In female rats, hepatic damage peaked at 24 h following the treatment and was approximately 2.5-fold (AST 2.7-fold, ALT 2.3 fold) greater than the damage observed in male rats. The hepatic damage in male rats appeared to peak by 3 h post-exposure and did not significantly change through the 36-h time-point. The activity of cytochrome P 4502E1 was 20% greater in male rats and did not correlate with the magnitude of hepatic damage. Morphometric analysis of cell cycle indices revealed that cellular regeneration was significantly greater in female rats as compared to male rats at 48 h and corresponded proportionally to the extent of liver damage. This study demonstrated that female SD rats respond more severely to acute CCl4 hepatotoxicity than male SD rats and the extent of tissue repair and cellular regeneration was greater in female rats. Furthermore, our results suggest that tissue repair is unlikely to result in accounting for the different responses exhibited by male and female SD rats to CCl4 hepatotoxicity.
Collapse
Affiliation(s)
- A P Moghaddam
- Operational Toxicology Branch, Air Force Research Laboratory, Wright-Patterson AFB, OH 84056-5012, USA.
| | | | | |
Collapse
|
26
|
Ramaiah SK, Soni MG, Bucci TJ, Mehendale HM. Diet restriction enhances compensatory liver tissue repair and survival following administration of lethal dose of thioacetamide. Toxicol Appl Pharmacol 1998; 150:12-21. [PMID: 9630448 DOI: 10.1006/taap.1998.8365] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diet restriction is known to prevent a plethora of age-associated diseases including cancer. However, the effects of diet restriction on noncancer end points are not known. The objective of this study was to investigate whether diet restriction protects against hepatotoxicity of thioacetamide (TA), and if so, to investigate the underlying mechanism. Male Sprague-Dawley rats (250-275 g) were maintained on 65% of their ad libitum (AL) food consumption for a period of 3 weeks and then treated with a single low dose of 50 mg TA/kg i.p.. Plasma enzymes (ALT and SDH), hepatic glycogen levels, and 3H-thymidine incorporation into hepatocellular nuclear DNA were measured during a time course (0-120 h) after TA administration. Liver sections were examined for histopathology, and cell-cycle progression was assessed by proliferating cell nuclear antigen (PCNA) immunohistochemistry. In AL rats hepatic necrosis was evident at 12 h, peaked at 36 h, persisted up to 72 h, and was resolved by 96 h. In the diet-restricted (DR) group hepatic necrosis was observed at 12 h, peaked at 24 h, persisted till 72 h, and was resolved by 96 h. Maximal injury indicated by enzyme elevation occurred in DR rats and was approximately sixfold greater than that observed in the AL group. Histopathological examination of the liver sections revealed liver injury concordant with plasma enzyme elevations. There was a higher and sustained S-phase synthesis in the DR rats compared to AL group. S-phase stimulation was evident at 36 h, peaked at 48 h, and persisted until 96 h in the DR rats, whereas in the AL rats peak S-phase stimulation occurred at 36 h and subsided by 72 h. PCNA studies revealed a corresponding stimulation of cell-cycle progression indicating highly stimulated compensatory tissue repair. The 14-day lethality experiments (600 mg TA/kg i.p.) indicated 70% survival in the DR rats compared to 10% survival in the AL group. Although diet restriction increases hepatotoxic injury of TA, it protects from the lethal outcome by enhanced liver tissue repair. Comparison of liver injury and tissue repair employing an equitoxic dose (600 mg TA/kg in AL rats yields similar liver injury as observed with 50 mg TA/kg in DR rats) revealed that in spite of near equal injury up to 36 h, tissue repair response in DR rats is much higher. The compensatory tissue repair allows the DR rats to escape death in contrast to much lower compensation in AL rats leading to progression of liver injury culminating in death.
Collapse
Affiliation(s)
- S K Ramaiah
- Division of Toxicology, College of Pharmacy and Health Sciences, North Louisiana University, Monroe 71209-0470, USA
| | | | | | | |
Collapse
|