Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int Immunopharmacol ; 125(Pt A): 111182, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37944217

ABSTRACT

This study sought to determine whether lactoferrin supplementation could counteract the harm that cadmium (Cd) induced to the rats. The effect of Cd and lactoferrin were investigated in hematological, biochemical, histological, immunohistochemical expression and ultrastructural studies. After 30 days of treatment, rats exposed to Cd had significantly higher levels of Cd in their blood, more oxidized lipids, and less antioxidant capacity overall. Supplemental lactoferrin also significantly undoes that effect. Hematological and biochemical parameters changed along with the increase in blood Cd levels. The histological integrity of the liver, kidney, spleen, and (axillary, cervical, mesenteric and popliteal) lymph nodes that had been damaged by Cd exposure was also restored by lactoferrin supplementation. Moreover, the liver and spleen ultrastructure showed the same improvement. In addition, the spleen of Lf/Cd group showed less immunohistochemical expression of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in comparison to the Cd group. In conclusion, the current study showed that supplementing with lactoferrin improved immune response and restored biochemical and oxidative stability induced by Cd.


Subject(s)
Cadmium Chloride , Oxidative Stress , Animals , Male , Rats , Cadmium/toxicity , Cadmium Chloride/antagonists & inhibitors , Cadmium Chloride/toxicity , Dietary Supplements , Lactoferrin/pharmacology
2.
Molecules ; 24(22)2019 Nov 18.
Article in English | MEDLINE | ID: mdl-31752142

ABSTRACT

Cadmium (Cd) imparts nephrotoxicity via triggering oxidative stress and pathological signal transductions in renal cells. The present study was performed to explore the protective mechanism of carnosic acid (CA), a naturally occurring antioxidant compound, against cadmium chloride (CdCl2)-provoked nephrotoxicity employing suitable in vitro and in vivo assays. CA (5 µM) exhibited an anti-apoptotic effect against CdCl2 (40 µM) in normal kidney epithelial (NKE) cells evidenced from cell viability, image, and flow cytometry assays. In this study, CdCl2 treatment enhanced oxidative stress by triggering free radical production, suppressing the endogenous redox defence system, and inhibiting nuclear factor erythroid 2-related factor 2 (Nrf2) activation in NKE cells and mouse kidneys. Moreover, CdCl2 treatment significantly endorsed apoptosis and fibrosis via activation of apoptotic and transforming growth factor (TGF)-ß1/mothers against decapentaplegic homolog (Smad)/collagen IV signalling pathways, respectively. In contrast, CA treatment significantly attenuated Cd-provoked nephrotoxicity via inhibiting free radicals, endorsing redox defence, suppressing apoptosis, and inhibiting fibrosis in renal cells in both in vitro and in vivo systems. In addition, CA treatment significantly (p < 0.05-0.01) restored blood and urine parameters to near-normal levels in mice. Histological findings further confirmed the protective role of CA against Cd-mediated nephrotoxicity. Molecular docking predicted possible interactions between CA and Nrf2/TGF-ß1/Smad/collagen IV. Hence, CA was found to be a potential therapeutic agent to treat Cd-mediated nephrotoxicity.


Subject(s)
Abietanes/pharmacology , Cadmium Chloride/antagonists & inhibitors , Cadmium Chloride/toxicity , Kidney/drug effects , Animals , Antioxidants/pharmacology , Cadmium/pharmacology , Cell Line , Collagen Type IV/metabolism , Heme Oxygenase-1/metabolism , Kidney/metabolism , Kidney/pathology , Mice , Molecular Docking Simulation , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism
3.
Mol Med Rep ; 20(2): 1499-1508, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31257486

ABSTRACT

Geniposide, as a type of iridoid glycoside, has antioxidative capacity. However, the mechanism underlying the effect of geniposide in cadmium (Cd)­induced osteoblast injury remains only partly elucidated. In the present study, Cell Counting Kit­8 (CCK­8) was used to determine MC­3T3­E1 cell viability. Flow cytometry was used to determine the rate of apoptosis and levels of reactive oxygen species (ROS). Oxidative stress­related factors were assessed using enzyme­linked immunosorbent method (ELISA). Quantitative real­time polymerase chain reaction (qPCR) and western blotting were used to evaluate apoptosis­ and bone formation­related genes and nuclear factor erythroid 2­related factor (Nrf2) signaling. It was demonstrated that geniposide increased the viability of the Cd­treated MC­3T3­E1 cells. Geniposide decreased apoptosis and ROS accumulation compared to these parameters in the Cd group. Geniposide attenuated oxidative stress­related factors, malondialdehyde and lactate dehydrogenase and increased antioxidant key enzyme superoxidase dismutase (SOD). The expression levels of Bax, Bcl­2 and survivin were modulated by geniposide. Additionally, the mRNA and protein expression of the receptor activator of NF­κB ligand (RANKL) and osterix were significantly increased, while osteoprotegerin was decreased by geniposide treatment compared to the Cd groups. Geniposide also enhanced Nrf2, heme oxygenase­1 (HO­1) and NAD(P)H quinone dehydrogenase 1 (NQO1) expression. The present study identified a potential agent for the treatment of Cd­induced osteoblast injury.


Subject(s)
Antioxidants/pharmacology , Cadmium Chloride/antagonists & inhibitors , Iridoids/pharmacology , NF-E2-Related Factor 2/genetics , Oxidants/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Cadmium Chloride/pharmacology , Cell Line , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Malondialdehyde/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Oxidants/pharmacology , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Survivin/genetics , Survivin/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
4.
Neurol Res ; 41(7): 594-608, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30973085

ABSTRACT

Recent evidence has suggested that cadmium (Cd) ions-induced neurotoxicity is associated with increased oxidative stress and mitochondrial-dependent and endoplasmic reticulum (ER) stress-induced apoptosis. This study aimed to investigate if rutin hydrate (RH), a well-reported neuroprotective and an antioxidant flavonoid, can ameliorate cadmium chloride (CdCl2)-induced neurotoxicity by inhibiting the resultant ER stress. Rats were divided into 4 groups (n = 16/group) of control, control + RH (100 mg/kg), CdCl2 (5 mg/kg), and CdCl2 + RH. All treatments were administered orally for 30 days, on daily basis. Brain homogenates from CdCl2-treated rats showed increased oxidative stress and induced activation of ER stress characterized by increasing mRNA and protein levels of GRP78, ATF-6, CHOP and Xbp-1 and protein levels of p-elF2α, p-JNK1/2 and cleaved caspase-12. Also, CdCl2 significantly reduced Bcl-2, enhanced Bax translocation to the mitochondrial membrane, increased cytoplasmic levels of cytochrome-C and caspase-3, and reduced mitochondrial membrane potential (Δψm) (increased Vmax and reduced time to Vmax). In contrast, RH significantly enhanced levels GSH and activities of SOD, GSH-Px, decreased levels of MDA and inhibited mitochondrial permeability transition pore (mtPTP) in the brains of both control and CdCl2-treated rats. Interestingly, in brain homogenates of CdCl2-treated rats only, RH reduced all markers of ER stress, increased Bcl-2, reduced mitochondrial Bax translocation and improved mitochondrial coupling. It also reduced cytosolic levels of cytochrome-C, cleaved caspase-3, and cleaved caspase-12. Overall, these findings support the efficiency of RH to inhibit ER stress in brains CdCl2-treated rats which is added to its existing mechanisms of neuroprotection. Abbreviations: ATF-6: activating transcription factor-6; Bax: Bcl-associated x; BBB: blood-brain barrier; Bcl-2: B-cell lymphoma 2; BiP: immunoglobulin heavy-chain-binding protein; [Ca2+]i: intracellular free Ca2+ concentration; Cd: cadmium; CdCl2: cadmium chloride; CHOP: CCAAT/enhancer-binding protein-homologous protein; CMC: carboxymethyl cellulose; Δψm: mitochondrial membrane potential; elF2α: phospho-eukaryotic translation initiation factor 2-alpha; ER: endoplasmic reticulum; ERAD: ER-associated protein degradation; ERK1/2: extracellular signal-regulated kinases 1 and 2; GADD 153: growth arrest and DNA damage-inducible protein 153; GRP78, 78 kDa glucose-regulated protein; GSH: reduced glutathione; GSH: reduced glutathione; GSH-Px: glutathione peroxidase; GSSG: glutathione disulfide (oxidized glutathione); IRE-1: inositol-requiring enzyme-1; JNK: c-Jun N-terminal kinase; MAPK: mitogen-activated protein kinase; MDA: malondialdehyde; mTOR: Akt/mammalian target of rapamycin; mtPTP: mitochondrial permeability transition pore; ONOO-: peroxynitrite; PCR: polymerase chain reaction; PERK: protein kinase RNA-like ER kinase; p-JNK: phospho-JNK; qPCR: quantitative PCR; RCR: respiratory control ratio; RH: rutin hydrate; RHoGDI: Rho-GDP-dissociation inhibitor; ROS: reactive oxygen species; SOD: superoxide dismutase; UPR: unfolded protein response; VDAC: voltage-dependent anion channel; Vmax: maximal rate of pore opening; Xbp-1: X-box binding protein 1.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Cadmium Chloride/toxicity , Endoplasmic Reticulum Stress/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondria/drug effects , Rutin/pharmacology , Animals , Brain/metabolism , Cadmium Chloride/antagonists & inhibitors , Caspase 3/metabolism , Cytochromes c/metabolism , Male , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , bcl-2-Associated X Protein/metabolism
5.
Int J Mol Sci ; 20(3)2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30717178

ABSTRACT

Cadmium (Cd) is harmful for humans and animals, especially for the reproductive system. However, the mechanism of its toxicity has not been elucidated, and how to alleviate its toxicity is very important. This study aimed to explore the role and mechanism of action of sulforaphane (SFN) in protecting mouse Leydigs (TM3) cells from cadmium (Cd)-induced damage. The half-maximal inhibitory concentration (IC50) of Cd and the safe doses of SFN were determined using a methyl thiazolyl tetrazolium (MTT) assay. The testosterone secretion from TM3 cells was measured using the enzyme-linked immunosorbent assay. The intracellular oxidative stress was evaluated using corresponding kits. The cell apoptosis was detected using flow cytometry. The mRNA expression of genes associated with NF-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) signaling was detected using reverse transcription⁻polymerase chain reaction, including Nrf2, heme oxygenase I (HO-1), glutathione peroxidase (GSH-Px), NAD(P)H:quinone acceptor oxidoreductase 1 (NQO1), and γ-glutamylcysteine synthetase (γ-GCS). The protein expression of Nrf2, GSH-Px, HO-1, γ-GCS, and NQO1 was detected using Western blot analysis. The results showed that the IC50 of Cd to TM3 cells was 51.4 µmol/L. SFN reduced the release of lactate dehydrogenase from Cd-exposed cells. Cd + SFN 2.5 treatment significantly elevated testosterone concentration compared with the Cd group (p < 0.05). SFN significantly increased total superoxide dismutase (T-SOD) and GSH-Px activity and GSH content in Cd-treated cells (p < 0.05; p < 0.01), inhibited the production of malondialdehyde or reactive oxygen species caused by Cd (p < 0.05; p < 0.01), and reduced the apoptotic rate of Cd-induced TM3 cells (p < 0.01). SFN upregulated the mRNA expression of Nrf2, GSH-Px, HO-1, NQO1, and γ-GCS in Cd-treated cells, indicating the protective effect of SFN against Cd-induced oxidative stress or cell apoptosis by activating the Nrf2/ARE signaling pathway.


Subject(s)
Antioxidants/pharmacology , Cadmium Chloride/antagonists & inhibitors , Isothiocyanates/pharmacology , Leydig Cells/drug effects , NF-E2-Related Factor 2/genetics , Signal Transduction/drug effects , Animals , Antioxidant Response Elements/drug effects , Apoptosis/drug effects , Cadmium Chloride/toxicity , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Leydig Cells/cytology , Leydig Cells/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Sulfoxides , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Testosterone/biosynthesis
6.
J Cell Mol Med ; 22(9): 4221-4235, 2018 09.
Article in English | MEDLINE | ID: mdl-29993192

ABSTRACT

Cadmium, a highly ubiquitous toxic heavy metal, has been widely recognized as an environmental and industrial pollutant, which confers serious threats to human health. The molecular mechanisms of the cadmium-induced cardiotoxicity (CIC) have not been studied in human cardiomyocytes at the cellular level. Here we showed that human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) can recapitulate the CIC at the cellular level. The cadmium-treated hPSC-CMs exhibited cellular phenotype including reduced cell viability, increased apoptosis, cardiac sarcomeric disorganization, elevated reactive oxygen species, altered action potential profile and cardiac arrhythmias. RNA-sequencing analysis revealed a differential transcriptome profile and activated MAPK signalling pathway in cadmium-treated hPSC-CMs, and suppression of P38 MAPK but not ERK MAPK or JNK MAPK rescued CIC phenotype. We further identified that suppression of PI3K/Akt signalling pathway is sufficient to reverse the CIC phenotype, which may play an important role in CIC. Taken together, our data indicate that hPSC-CMs can serve as a suitable model for the exploration of molecular mechanisms underlying CIC and for the discovery of CIC cardioprotective drugs.


Subject(s)
Cadmium Chloride/toxicity , Gene Expression Regulation/drug effects , Models, Biological , Myocytes, Cardiac/drug effects , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Cadmium Chloride/antagonists & inhibitors , Cardiotoxicity/genetics , Cardiotoxicity/metabolism , Cardiotoxicity/prevention & control , Cell Differentiation/drug effects , Cell Line , Chromones/pharmacology , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Imides/pharmacology , Insulin/pharmacology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Morpholines/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Toxicol Lett ; 288: 35-43, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29444456

ABSTRACT

Cadmium (Cd), a toxic heavy metal, is known to induce renal toxicity by primarily targeting at renal proximal tubule. Endoplasmic reticulum (ER) stress and gap junctional intercellular communication (GJIC) regulate many pathophysiological processes. Yet, how ER stress and GJIC regulate Cd-induced nephrotoxicity remain elusive. In this study, we treated human proximal tubule (HK-2) cells with 1 µM CdCl2 every other day for 12 days and found that Cd significantly increased cell apoptosis at 10 and 12 days. This cytotoxicity correlated with activation of ER stress and apoptotic signaling evidenced by upregulation of inositol-requiring enzyme 1 (IRE1α), splice X-box binding protein-1 (XBP-1s), and apoptosis signal-regulating kinase 1 (ASK1) proteins. Interestingly, the AKT signaling was activated at 2- and 4-day and then inhibited at 10- and 12-day of Cd treatment; by contrast, Cd decreased GJIC levels at 2- and 4-day followed by a significant increase at 10- and 12-day treatment. Activation of AKT by SC79 or inhibition of GJIC by 18α-glycyrrhetinic acid (18α-GA) completely abolished Cd-induced AKT inhibition and IRE1α-ASK1 activation. Importantly, pretreatment with ER stress inhibitor or 18α-GA significantly mitigated Cd-induced apoptosis. These results suggest that GJIC collaborates with AKT signaling and ER stress in regulating prolonged Cd-treatment-induced apoptosis in HK-2 cells.


Subject(s)
Apoptosis/drug effects , Cadmium Chloride/toxicity , Cell Communication/drug effects , Endoplasmic Reticulum Stress/drug effects , Gap Junctions/drug effects , Cadmium Chloride/antagonists & inhibitors , Cell Line , Cell Survival/drug effects , Extracellular Space , Gene Expression Regulation/drug effects , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Oncogene Protein v-akt , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
8.
Biol Trace Elem Res ; 185(2): 404-413, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29427035

ABSTRACT

Cadmium chloride (CdCl2) is a ubiquitous environmental toxicant that causes a variety of disturbances in biological systems, including brain dysfunction and testicular tissue degeneration. On the other hand, it is supposed that beneficial properties of probiotic bacteria (Lactobacillus and Acidobacillus) are related to their capacity to adhere or bind different targets, thus leading to improved intestinal microbial balance and other benefits to the host. Bearing aforementioned in mind, the present study was undertaken to investigate the protective effect of probiotic supplementation against cadmium chloride-induced brain and testis toxicity in mice model. Animals received Lactobacillus and Acidobacillus either alone or added to folic acid for 1 week before CdCl2 intoxication in a dose of 20 mg/kg BW followed by probiotics (5 × 109) and/or folic acid (12 mg/kg) treatment for 3 weeks. The levels of malondialdehyde (MDA), butyrl choline esterase (BCHE), reduced glutathione (GSH), and total superoxide dismutase (SOD) activities were investigated. Finally, cadmium neurotoxicity was determined by estimating the gene expression of ß-catenin and brain-derived neurotrophic factor (BDNF), as well as estimating the alterations in testicular function by determining acid phosphatase level in addition to steroidogenic acute regulatory protein (StAR) and 17-hydroxy steroid dehydrogenase (17-ß HSD) gene expression. Based on our results, we can conclude that exposure of mice to cadmium chloride resulted in a significant elevation in MDA, BCHE levels accompanied with a significant reduction in GSH and SOD activities compared to the control value. CdCl2 also downregulated the gene expression of ß-catenin and BDNF, as well as acid phosphatase level, in addition to StAR and 17-ß HSD gene expression. These deviated parameters were significantly modulated in the co-treated animals with probiotics compared with the cadmium-treated group. In conclusion, Lactobacillus and folic acid in a mixture with cadmium acted beneficially to an organism, increasing the cadmium excretion in feces, and consequently increasing ß-catenin and BDNF in brain tissue and StAR and 17-ß HSD in testis and improving their functions. Histoarchitecture analysis confirmed these results.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cadmium Chloride/antagonists & inhibitors , Cadmium Chloride/toxicity , Folic Acid/pharmacology , Lactobacillus/isolation & purification , Phosphoproteins/metabolism , Probiotics , beta Catenin/metabolism , Animals , Brain Injuries/chemically induced , Brain Injuries/pathology , Brain-Derived Neurotrophic Factor/genetics , Cadmium Chloride/administration & dosage , Complementary Therapies , Disease Models, Animal , Folic Acid/administration & dosage , Male , Mice , Phosphoproteins/genetics , Signal Transduction/drug effects , beta Catenin/genetics
9.
Biol Trace Elem Res ; 185(2): 344-355, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29397540

ABSTRACT

Quercetin, a plant-derived flavonoid, is frequently used as an antioxidant for efficient anti-oxidative capacity. However, whether quercetin has protective effects on goat sperm and preimplantation embryos against Cd2+-induced oxidative injury is still unclear. So, we researched the influence of quercetin on goat sperm and zygotes respectively under the oxidative stress induced by Cd2+. In our study, quercetin decreased the malonaldehyde (MDA) and reactive oxygen species (ROS) levels caused by Cd2+ in goat sperm (p < 0.05), which facilitated sperm characteristics including motility, survival rates, membrane integrity, and mitochondria activity during storage in vitro and subsequent embryo development (p < 0.05). Moreover, in goat zygotes, quercetin decreased peroxidation products including ROS, MDA, and carbonyl through preserving or maintaining mitochondrial function, gene expression, and anti-oxidative products such as glutathione peroxidase, superoxide dismutase, and catalase, which ameliorated subsequent embryo development and embryo quality (p < 0.05). Taken together, these results suggest that quercetin protects both goat sperm and preimplantation embryos from Cd2+-induced oxidative stress.


Subject(s)
Cadmium Chloride/antagonists & inhibitors , Oxidative Stress/drug effects , Quercetin/pharmacology , Spermatozoa/drug effects , Zygote/drug effects , Animals , Antioxidants/metabolism , Cadmium Chloride/pharmacology , Goats , Male , Sperm Motility/drug effects , Spermatozoa/metabolism , Spermatozoa/pathology , Zygote/metabolism , Zygote/pathology
10.
Biol Trace Elem Res ; 181(2): 378-387, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28567583

ABSTRACT

For experiments of cadmium toxicity in animal models, cadmium (II) chloride is often used due to its solubility in water and its ability to produce high concentrations of cadmium at the target site. The present study was designed to investigate the potential inhibitory effect of the Fragaria ananassa fruit extract on cadmium (II) chloride-induced renal toxicity in rats. Tested animals were pretreated with the extract of F. ananassa and injected with cadmium (II) chloride (6.5-mg/kg body weight) for 5 days. Cadmium (II) chloride significantly increased kidney cadmium concentration, kidney weight, lipid peroxidation, and nitric oxide production. Plasma uric acid, urea, and creatinine levels also increased significantly, indicative of kidney dysfunction. These effects were accompanied by significantly decreased levels of nonenzymatic and enzymatic antioxidant molecules (i.e., glutathione content and the activities of superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase). Moreover, messenger RNA (mRNA) expression of the antiapoptotic protein, Bcl-2, and the antioxidant proteins, superoxide dismutase 2 and glutathione reductase, were downregulated markedly, whereas mRNA expression of tumor necrosis factor-α was upregulated significantly in kidney tissues of cadmium-treated rats. Histology of kidney tissue demonstrated severe, adverse changes that reflected cadmium-induced tissue damage. Pretreatment of rats with the extract of F. ananassa ameliorated all aforementioned cadmium (II) chloride-induced changes. In conclusion, the present study showed acute renal toxicity in rats treated with cadmium (II) chloride. The study also revealed that pretreatment with the extract of F. ananassa could protect the kidney against cadmium (II) chloride-induced acute renal toxicity.


Subject(s)
Cadmium Chloride/antagonists & inhibitors , Fragaria/chemistry , Plant Extracts/pharmacology , Protective Agents/pharmacology , Renal Insufficiency/prevention & control , Animals , Cadmium Chloride/toxicity , Immunohistochemistry , Male , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Protective Agents/chemistry , Protective Agents/isolation & purification , Rats , Renal Insufficiency/chemically induced , Renal Insufficiency/pathology
11.
Arch Physiol Biochem ; 124(4): 367-377, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29214892

ABSTRACT

This study aimed at studying the potential neuroprotective effect of Rutin hydrate (RH) alone or in conjugation with α-tocopherol against cadmium chloride (CdCl2)-induced neurotoxicity and cognitive impairment in rats and to investigate the mechanisms of action. Rats intoxicated with CdCl2 were treated with the vehicle, RH, α-tocopherol or combined treatment were examined, and compared to control rats received vehicle or individual doses of either drug. Data confirmed that RH improves spatial memory function by increasing acetylcholine availability, boosting endogenous antioxidant potential, activating cell survival and inhibiting apoptotic pathways, an effect that is more effective when RH was conjugated with α-tocopherol. Mechanism of RH action includes activation of PP2A mediated inhibiting of ERK1/2 and JNK apoptotic pathways and inhibition of PTEN mediated activation of mTOR survival pathway. In conclusion, RH affords a potent neuroprotection against CdCl2-induced brain damage and memory dysfunction and co-administration of α-tocopherol enhances its activity.


Subject(s)
Apoptosis , Brain/metabolism , Dietary Supplements , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/prevention & control , Rutin/therapeutic use , Acetylcholine/agonists , Acetylcholine/metabolism , Animals , Antioxidants/therapeutic use , Apoptosis/drug effects , Brain/drug effects , Brain/enzymology , Cadmium Chloride/antagonists & inhibitors , Cadmium Chloride/toxicity , Cadmium Poisoning/physiopathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Enzyme Activation/drug effects , MAP Kinase Signaling System/drug effects , Male , Maze Learning/drug effects , Memory Disorders/etiology , Memory Disorders/prevention & control , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/enzymology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Rats, Wistar , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/metabolism , alpha-Tocopherol/therapeutic use
12.
Free Radic Biol Med ; 112: 12-23, 2017 11.
Article in English | MEDLINE | ID: mdl-28690196

ABSTRACT

Mismatch repair (MMR) corrects replicative errors and minimizes DNA damage that occurs frequently in microsatellites. MMR deficiency is manifested as microsatellite instability (MSI), which contributes to hypermutability and cancer pathogenesis. Genomic instability, including MSI and chromosomal instability, appears to be responsible for the carcinogenesis of arsenic and cadmium, common contaminants in our environment. However, few studies have addressed arsenic- or cadmium-induced MSI, especially its potential link with arsenic- or cadmium-generated oxidative stress, due to the lack of quantifiable MSI assays and cost-effective animal models. Here, using a dual-fluorescent reporter, we demonstrate that sub-lethal doses of cadmium or arsenite, but not arsenate, increased the MSI frequency in human colorectal cancer cells. Arsenite- and cadmium-induced MSI occurred concomitantly with increased levels of reactive species and oxidative DNA damage, and with decreased levels of MMR proteins. However, N-acetyl-l-cysteine (NAC) suppressed arsenite- and cadmium-induced MSI and oxidative stress while restoring the levels of MMR proteins in the cells. Similarly, MSI was induced separately by arsenite and cadmium, and suppressed by NAC, in zebrafish in a fluorescinated PCR-based assay with newly-developed microsatellite markers and inter-segmental comparisons. Of five selected antioxidants examined, differential effects were exerted on the MSI induction and cytotoxicity of both arsenite and cadmium. Compared to MMR-proficient cells, MMR-deficient cells were more resistant to arsenic-mediated and cadmium-mediated cytotoxicity. Our findings demonstrate a novel linkage between arsenite-generated and cadmium-generated oxidative stress and MSI induction. Our findings also caution that antioxidants must be individually validated before being used for preventing arsenite- and cadmium-induced MSI that is associated with cancer development.


Subject(s)
Arsenites/toxicity , Cadmium Chloride/toxicity , DNA Mismatch Repair/drug effects , DNA/genetics , Microsatellite Instability/drug effects , Sodium Compounds/toxicity , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Arsenites/antagonists & inhibitors , Cadmium Chloride/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , DNA/metabolism , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genes, Reporter , HCT116 Cells , Humans , Microsatellite Repeats/drug effects , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Oxidative Stress , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Sodium Compounds/antagonists & inhibitors , Zebrafish
13.
Colloids Surf B Biointerfaces ; 154: 357-364, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28365425

ABSTRACT

Microbial extracellular polymeric substances (EPS) play an important role in resisting the shock load of toxic contaminants to microbial aggregates. In order to investigate the surface interaction process of bacteria with heavy metals, in this work, the kinetics and affinity of heavy metal (CdCl2 and PbCl2) binding on Bacillus subtilis with EPS and without EPS were determined using in situ surface plasmon resonance. The binding mechanism between bacteria (with EPS and without EPS) and heavy metals was probed using Fourier-transform infrared spectra. The effect of heavy metals on aggregations of microbial cells with EPS and without EPS was investigated. The results showed that both the binding of Pb2+ and Cd2+ to bacteria with EPS had a similar kinetics process, however Pb2+ bound to bacterial surface without EPS more firmly compared with Cd2+. From our results we theorized that heavy metals changed the protein secondary structures of bacteria without EPS protection, that EPS reduced the influence of heavy metals on microbial aggregation, and that Pb2+ inhibited cell aggregation more easily compared with Cd2+ in the absence of EPS.


Subject(s)
Bacillus subtilis/drug effects , Bacterial Proteins/pharmacology , Cadmium Chloride/antagonists & inhibitors , Humic Substances/analysis , Polysaccharides, Bacterial/pharmacology , Adsorption , Bacillus subtilis/chemistry , Bacillus subtilis/growth & development , Bacterial Proteins/isolation & purification , Cadmium Chloride/toxicity , Cations, Divalent , Culture Media/chemistry , Kinetics , Lead/toxicity , Polysaccharides, Bacterial/isolation & purification , Spectroscopy, Fourier Transform Infrared , Surface Properties
14.
Molecules ; 22(1)2016 Dec 24.
Article in English | MEDLINE | ID: mdl-28029133

ABSTRACT

Cinnamaldehyde (CA) is natural plant-derived compound that has been highly appreciated for its medicinal properties. However, little information is known about the regulation of plant intrinsic physiology by CA. To address these gaps, physiological, histochemical, and biochemical approaches were applied to investigate CA-facilitated cadmium (Cd) tolerance in the roots of tobacco (Nicotiana tabacum) seedlings. Treatment with CdCl2 at 20 µM for 72 h resulted in the significant decrease in root elongation by 40.39% as compared to control. CA alleviated Cd-inhibited root elongation in dose- and time-dependent manners. The addition of CA at 20 µM induced significant increase in root elongation by 42.58% as compared to Cd treatment alone. CA abolished Cd-induced ROS (reactive oxygen species) accumulation, lipid peroxidation, loss of membrane integrity, cell death, and free Cd2+ accumulation in roots. CA blocked the Cd-induced increase in the endogenous H2S level through the down-regulation of d-cysteine desulfhydrase (DCD) expression. H2S scavenger hypotaurine (HT) or potent H2S-biosynthetic inhibitor dl-propargylglicine (PAG) were able mimic the action of CA on the blockade of Cd-induced H2S accumulation, cell death, and growth inhibition. Enhancement of the endogenous H2S level with NaHS (H2S donor) abrogated all the beneficial capabilities of CA, HT, and PAG. Collectively, these results suggest that CA has great potential to confer plant tolerance against Cd stress, which is closely associated with its capability to inhibit Cd-induced H2S production. This study not only provides evidences for the regulation of plant physiology by CA but also sheds new light on the cross-talk between CA and H2S in physiological modulations.


Subject(s)
Cadmium Chloride/antagonists & inhibitors , Cystathionine gamma-Lyase/antagonists & inhibitors , Hydrogen Sulfide/antagonists & inhibitors , Nicotiana/drug effects , Plant Roots/drug effects , Seedlings/drug effects , Acrolein/analogs & derivatives , Acrolein/pharmacology , Alkynes/pharmacology , Antioxidants/pharmacology , Cadmium Chloride/pharmacology , Cell Death/drug effects , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Gene Expression , Glycine/analogs & derivatives , Glycine/pharmacology , Hydrogen Sulfide/metabolism , Lipid Peroxidation/drug effects , Plant Roots/growth & development , Plant Roots/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Seedlings/growth & development , Seedlings/metabolism , Sulfides/pharmacology , Taurine/analogs & derivatives , Taurine/pharmacology , Nicotiana/growth & development , Nicotiana/metabolism
15.
Environ Toxicol ; 30(6): 704-11, 2015.
Article in English | MEDLINE | ID: mdl-24420767

ABSTRACT

Cadmium is commercially profitable element, but it causes toxicity in humans and animals leading to diseases in various organs. The main route of cadmium exposure to humans is through inhalation. Lungs respond to insult through secretion of cytokines. In this study, the chemoprotective effect of monoisoamyl 2, 3-dimercaptosuccinate (MiADMS) was evaluated on viability and cytokines expression in CdCl2 treated human lung A549 cells by cytokine array. Cells were treated with 0, 50, 75, and 100 µM CdCl2 alone, 300 µM MiADMS alone, and co-treated with 300 µM MiADMS and 75 µM CdCl2 for 24 h. The viability was measured by crystal violet dye. The level of cytokines in the cells' lysate and cell culture medium was measured using Ray Biotech's Human Cytokine Array 6 in control cells, 75 µM CdCl2 alone and MiADMS co-treated cells. Array results were validated by ELISA kit. The CdCl2 caused a dose dependent decrease in cell viability, while MiADMS co-treatment resulted in a significant increase in viability of CdCl2 treated cells. Morphology of the cells treated with CdCl2 was destroyed, while MiADMS restored the lost shape in CdCl2 treated cells. In addition, the cells co-treated with MiADMS and CdCl2 showed modulation of cytokines expression in comparison to the CdCl2 alone treated cells. The ELISA results showed the similar pattern of cytokine expression as Human Cytokine Array and validated the array results. These results clearly show the chemoprotective effect of MiADMS and suggest that MiADMS can be used as antidote at moderate dose against CdCl2 toxicity.


Subject(s)
Cadmium Chloride/antagonists & inhibitors , Cadmium Chloride/toxicity , Cytokines/metabolism , Lung Diseases/pathology , Lung Diseases/prevention & control , Protective Agents/pharmacology , Succimer/analogs & derivatives , Cadmium/toxicity , Cell Line , Cell Survival/drug effects , Down-Regulation/drug effects , Humans , Lung/pathology , Lung Diseases/metabolism , Succimer/pharmacology
16.
Food Chem Toxicol ; 74: 98-106, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25265456

ABSTRACT

This study aimed to investigate the potential protective role of Physalis peruviana L. (family Solanaceae) against cadmium-induced hepatorenal toxicity in Wistar rats. Herein, cadmium chloride (CdCl2) (6.5 mg/kg bwt/day) was intraperitoneally injected for 5 days, and methanolic extract of physalis (MEPh) was pre-administered to a group of Cd-treated rats by an oral administration at a daily dose of 200 mg/kg bwt for 5 days. The findings revealed that CdCl2 injection induced significant decreases in kidney weight and kidney index. Cadmium intoxication increased the activities of liver enzymes and the bilirubin level, in addition to the levels of uric acid, urea and creatinine were increased in the serum. The pre-administration of MEPh alleviated hepatorenal toxicity in Cd-treated rats. Physalis was noted to play a good hepatorenal protective role, reducing lipid peroxidation, nitric oxide, and enhancing enzymatic activities and non-enzymatic antioxidant molecule, glutathione, in hepatic and renal tissues of Cd-treated rats. Moreover, physalis treatment was able to reverse the histopathological changes in liver and kidney tissues and also increased the expression of Bcl-2 protein in liver and kidney of rats. Overall, the results showed that MEPh can induce antioxidant and anti-apoptotic effects and also exerts beneficial effects for the treatment of Cd-induced hepatorenal toxicity.


Subject(s)
Acute Kidney Injury/chemically induced , Cadmium Chloride/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Fruit , Physalis , Phytotherapy/methods , Acute Kidney Injury/prevention & control , Animals , Cadmium Chloride/antagonists & inhibitors , Chemical and Drug Induced Liver Injury/drug therapy , Kidney/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Male , Oxidative Stress/drug effects , Plant Extracts/therapeutic use , Rats , Rats, Wistar
17.
Toxicol Mech Methods ; 24(8): 567-74, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25133553

ABSTRACT

The Mimosa (Mimosa caesalpiniifolia) is a plant native from South America; it is used in the traditional medicine systems for treating bacterial, fungal, parasitic and inflammatory conditions. The aim of this study was to evaluate the antigenotoxic and antioxidant activities induced by mimosa (M. caesalpiniifolia) in multiple rodent organs subjected to intoxication with cadmium chloride. A total of 40 Wistar rats (8 weeks old, 250 g) were distributed into eight groups (n = 5), as follows: Control group (non-treated group, CTRL); Cadmium exposed group (Cd); cadmium exposure and treated with extract at 62.5 mg/kg/day; cadmium exposure and treated with extract at 125 mg/kg/day; cadmium exposure and treated with extract at 250 mg/kg/day; cadmium exposure and treated with ethyl acetate fraction at 62.5 mg/kg/day. For evaluating the toxicogenetic potential of mimosa, two groups were included in the study being treated with extract at 250 mg/kg/day and acetate fraction of mimosa at 62 mg/kg/day, only. Extract of mimosa at concentrations of 62.5 and 125 mg decreased DNA damage in animals intoxicated with cadmium when compared to cadmium group. In a similar manner, treatment with ethyl acetate fraction of mimosa at 62.5 mg concentration in animals previously exposed to cadmium reduced genetic damage in peripheral blood cells. In a similar manner, the treatment with ethyl acetate fraction reduced DNA damage in liver cells. Oxidative DNA damage was reduced to animals exposed to cadmium and treated with 125 mg of extract as well as those intoxicated to cadmium and treated with 62.5 of acetate fraction of mimosa. Taken together, our results indicate that mimosa prevents genotoxicity induced by cadmium exposure in liver and peripheral blood cells of rats as a result of antioxidant activity.


Subject(s)
Antioxidants/therapeutic use , Cadmium Poisoning/drug therapy , DNA Damage/drug effects , Mimosa/chemistry , Phytotherapy , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Acetates/chemistry , Animals , Antioxidants/administration & dosage , Antioxidants/adverse effects , Antioxidants/isolation & purification , Blood Cells/drug effects , Blood Cells/metabolism , Blood Cells/pathology , Brazil , Cadmium Chloride/antagonists & inhibitors , Cadmium Chloride/toxicity , Cadmium Poisoning/blood , Cadmium Poisoning/metabolism , Cadmium Poisoning/pathology , Dose-Response Relationship, Drug , Ethnopharmacology , Liver/drug effects , Liver/metabolism , Medicine, Traditional , Mutagens/chemistry , Mutagens/toxicity , Oxidative Stress/drug effects , Phytotherapy/adverse effects , Plant Extracts/administration & dosage , Plant Extracts/adverse effects , Plant Extracts/isolation & purification , Rats, Wistar , Solvents/chemistry
18.
J Reprod Dev ; 60(2): 115-27, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24492640

ABSTRACT

This study was performed to investigate the protective and therapeutic effects of resveratrol (RES) against CdCl2-induced toxicity in rat testes. Seven experimental groups of adult male rats were formulated as follows: A) controls+NS, B) control+vehicle (saline solution of hydroxypropyl cyclodextrin), C) RES treated, D) CdCl2+NS, E) CdCl2+vehicle, F) RES followed by CdCl2 and M) CdCl2 followed by RES. At the end of the protocol, serum levels of FSH, LH and testosterone were measured in all groups, and testicular levels of TBARS and superoxide dismutase (SOD) activity were measured. Epididymal semen analysis was performed, and testicular expression of Bcl-2, p53 and Bax was assessed by RT-PCR. Also, histopathological changes of the testes were examined microscopically. Administration of RES before or after cadmium chloride in rats improved semen parameters including count, motility, daily sperm production and morphology, increased serum concentrations of gonadotropins and testosterone, decreased testicular lipid peroxidation and increased SOD activity. RES not only attenuated cadmium chloride-induced testicular histopathology but was also able to protect against the onset of cadmium chloride testicular toxicity. Cadmium chloride downregulated the anti-apoptotic gene Bcl2 and upregulated the expression of pro-apoptotic genes p53 and Bax. Resveratrol protected against and partially reversed cadmium chloride testicular toxicity via upregulation of Bcl2 and downregulation of p53 and Bax gene expression. The antioxidant activity of RES protects against cadmium chloride testicular toxicity and partially reverses its effect via upregulation of BCl2 and downregulation of p53 and Bax expression.


Subject(s)
Cadmium Chloride/antagonists & inhibitors , Gonadotropins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Stilbenes/pharmacology , Testis/drug effects , Tumor Suppressor Protein p53/biosynthesis , bcl-2-Associated X Protein/biosynthesis , Animals , Antioxidants/pharmacology , Cadmium Chloride/toxicity , Drug Interactions , Follicle Stimulating Hormone/blood , Gene Expression Regulation/drug effects , Gonadotropins/genetics , Histocytochemistry , Infertility, Male/blood , Infertility, Male/chemically induced , Infertility, Male/drug therapy , Luteinizing Hormone/blood , Male , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Rats, Wistar , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/blood , Testis/metabolism , Testosterone/blood , Thiobarbituric Acid Reactive Substances/metabolism , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/genetics
19.
Environ Toxicol Pharmacol ; 36(3): 937-47, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24035924

ABSTRACT

This study evaluates the possible protective potentials of chelidonine and its poly lactide-co-glycolide (PLGA) encapsulated nano-form against cadmium chloride (CdCl2) induced oxidative stress and hepatotoxicity in mice, ex vivo and in vivo. Acute exposure to CdCl2 (1.0 mg/kg b.w; i.p., twice a week for 30 days) generated oxidative stress in mice through accumulation of reactive oxygen species and increased lipid peroxidation, and levels of certain liver marker enzymes (ALT, AST, ALP) with decrease in levels of GSH and certain other antioxidant enzymes (SOD, CAT, GR) in liver. Treatment with nano-chelidonine for 30 days after CdCl2 intoxication significantly reduced oxidative stress and lipid peroxidation and restored levels of GSH, cholesterol, triglyceride and antioxidant enzymes, showing ameliorative changes in histopathology of liver. Expression pattern of certain inflammatory and apoptotic signal proteins also indicated better hepato-protective abilities of nano-chelidonine, making it a more suitable protective drug than chelidonine against cadmium toxicity in mice.


Subject(s)
Benzophenanthridines/chemistry , Cadmium Chloride/antagonists & inhibitors , Cadmium Chloride/toxicity , Cadmium Poisoning/prevention & control , Chelidonium/chemistry , Chemical and Drug Induced Liver Injury/prevention & control , Drug Compounding/methods , Lactic Acid/chemistry , Oxidative Stress/drug effects , Polyglycolic Acid/chemistry , Algorithms , Animals , Benzophenanthridines/administration & dosage , Benzophenanthridines/pharmacokinetics , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/pathology , Excipients , Hepatocytes/drug effects , Immunohistochemistry , Indicators and Reagents , Light , Mice , Nanoparticles , Nanotechnology , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Scattering, Radiation , Spectrophotometry, Atomic
20.
J Trace Elem Med Biol ; 27(4): 355-63, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23993482

ABSTRACT

The protective effect of cannabidiol, the non-psychoactive component of Cannabis sativa, against liver toxicity induced by a single dose of cadmium chloride (6.5 mgkg(-1) i.p.) was investigated in rats. Cannabidiol treatment (5 mgkg(-1)/day, i.p.) was applied for five days starting three days before cadmium administration. Cannabidiol significantly reduced serum alanine aminotransferase, and suppressed hepatic lipid peroxidation, prevented the depletion of reduced glutathione and nitric oxide, and catalase activity, and attenuated the elevation of cadmium level in the liver tissue resulted from cadmium administration. Histopathological examination showed that cadmium-induced liver tissue injury was ameliorated by cannabidiol treatment. Immunohistochemical analysis revealed that cannabidiol significantly decreased the cadmium-induced expression of tumor necrosis factor-α, cyclooxygenase-2, nuclear factor-κB, caspase-3, and caspase-9, and increased the expression of endothelial nitric oxide synthase in liver tissue. It was concluded that cannabidiol may represent a potential option to protect the liver tissue from the detrimental effects of cadmium toxicity.


Subject(s)
Cadmium Chloride/antagonists & inhibitors , Cannabidiol/pharmacology , Liver/drug effects , Animals , Cadmium Chloride/administration & dosage , Cadmium Chloride/toxicity , Cannabidiol/administration & dosage , Injections, Intraperitoneal , Liver/pathology , Male , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...