Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Sci Total Environ ; 927: 172069, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38582117

ABSTRACT

Ferroptosis is a newly recognized type of programmed cell death that is implicated in the pathophysiological process of neurological disorders. Our previous studies have revealed that exposure to high concentrations of fluoride for long periods of time induces hippocampal neural injury and cognitive deficits. However, whether ferroptosis is involved in fluoride-induced neuronal death and the underlying mechanism remain unknown. In this study, the results indicated that exposure to high fluoride triggered ferroptosis in SH-SY5Y cells and in the hippocampus of mice. Fluoride exposure accelerated the lysosomal degradation of GPX4 and led to neuronal ferroptosis, while GPX4 overexpression protected SH-SY5Y cells against fluoride-induced neurotoxicity. Intriguingly, the enhanced chaperone-mediated autophagy (CMA) induced by fluoride stimulation was responsible for GPX4 degradation because the inhibition of CMA activity by LAMP2A knockdown effectively prevented fluoride-induced GPX4 loss. Furthermore, mitochondrial ROS (mtROS) accumulation caused by fluoride contributed to CMA activation-mediated GPX4 degradation and subsequent neuronal ferroptosis. Notably, the ferroptosis-specific inhibitor ferrostatin-1 (Fer-1) or the ROS scavenger N-acetyl-L-cysteine (NAC) alleviated fluoride-evoked hippocampal neuronal death and synaptic injury as well as cognitive deficits in mice. The present studies indicates that ferroptosis is a novel mechanism of fluoride-induced neurotoxicity and that chronic fluoride exposure facilitates GPX4 degradation via mtROS chaperone-mediated autophagy, leading to neuronal ferroptosis and cognitive impairment.


Subject(s)
Chaperone-Mediated Autophagy , Cognitive Dysfunction , Ferroptosis , Fluorides , Neurons , Phospholipid Hydroperoxide Glutathione Peroxidase , Reactive Oxygen Species , Animals , Humans , Mice , Autophagy/drug effects , Chaperone-Mediated Autophagy/physiology , Chaperone-Mediated Autophagy/drug effects , Cognitive Dysfunction/chemically induced , Ferroptosis/drug effects , Ferroptosis/physiology , Fluorides/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Reactive Oxygen Species/metabolism
2.
Environ Pollut ; 349: 123958, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38621452

ABSTRACT

Exposure to environmental cadmium (Cd) is known to cause neuronal death and cognitive decline in humans. Ferroptosis, a novel iron-dependent type of regulated cell death, is involved in various neurological disorders. In the present study, Cd exposure triggered ferroptosis in the mouse hippocampus and in the HT22 murine hippocampal neuronal cell line, as indicated by significant increases in ferroptotic marker expression, intracellular iron levels, and lipid peroxidation. Interestingly, ferroptosis of hippocampal neurons in response to Cd exposure relied on the induction of autophagy since the suppression of autophagy by 3-methyladenine (3-MA) and chloroquine (CQ) substantially ameliorated Cd-induced ferroptosis. Furthermore, nuclear receptor coactivator 4 (NCOA4)-mediated degradation of ferritin was required for the Cd-induced ferroptosis of hippocampal neurons, demonstrating that NCOA4 knockdown decreased intracellular iron levels and lipid peroxidation and increased cell survival, following Cd exposure. Moreover, Cd-induced mitochondrial reactive oxygen species (mtROS) generation was essential for the ferritinophagy-mediated ferroptosis of hippocampal neurons. Importantly, pretreatment with the ferroptosis inhibitor ferrostatin-1 (Fer-1) effectively attenuated Cd-induced hippocampal neuronal death and cognitive impairment in mice. Taken together, these findings indicate that ferroptosis is a novel mechanism underlying Cd-induced neurotoxicity and cognitive impairment and that the mtROS-ferritinophagy axis modulates Cd-induced neuronal ferroptosis.


Subject(s)
Cadmium , Cognitive Dysfunction , Ferroptosis , Hippocampus , Neurons , Nuclear Receptor Coactivators , Reactive Oxygen Species , Ferroptosis/drug effects , Animals , Mice , Cadmium/toxicity , Neurons/drug effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Reactive Oxygen Species/metabolism , Nuclear Receptor Coactivators/metabolism , Nuclear Receptor Coactivators/genetics , Ferritins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Male , Autophagy/drug effects , Iron/metabolism , Lipid Peroxidation/drug effects , Cell Line , Mice, Inbred C57BL
3.
ACS Biomater Sci Eng ; 9(11): 6225-6240, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37906514

ABSTRACT

There is an urgent demand for antibacterial bone grafts in clinics. Worryingly, the misuse and overuse of antibiotics accelerate the emergence of drug-resistant bacteria. Therefore, this study prepared a novel injectable bioceramic cement without antibiotics (FS-BCS), which showed good antibacterial properties by loading iron and strontium onto a matrix composed of brushite and calcium sulfate. The setting time, injectability, microstructure, antibacterial properties, anti-biofilm properties, and cytocompatibility of the novel bioceramic cement were evaluated thoroughly. The results showed that the material was highly injectable and antiwashout. The antibacterial tests revealed that FS-BCS inhibited the growth of 99.9% E. coli and S. aureus separately in the broth due to the synergistic effect of strontium and iron. Simultaneously, crystal violet and fluorescent staining tests revealed that the material could significantly inhibit the formation of E. coli and S. aureus biofilms. In addition, the co-incorporation of iron and strontium promoted the proliferation and migration of osteoblasts. Therefore, FS-BCS has good application potential in antibiotic-free anti-infection bone grafting using minimally invasive surgery.


Subject(s)
Escherichia coli , Staphylococcus aureus , Bone Cements/chemistry , Bone Cements/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Biofilms , Iron/pharmacology , Strontium/pharmacology , Minimally Invasive Surgical Procedures
4.
Sci Total Environ ; 903: 166478, 2023 Dec 10.
Article in English | MEDLINE | ID: mdl-37625726

ABSTRACT

Cadmium (Cd), a toxic heavy metal, exerts deleterious effects on neuronal survival and cognitive function. NOD-like receptor 3 (NLRP3) inflammasome-dependent pyroptosis has been linked to Cd-induced cytotoxicity. The current research intended to elucidate the role of NLRP3 inflammasome-mediated pyroptosis in Cd-evoked neuronal death and cognitive impairments and the underlying mechanisms. Exposure to 1 mg/kg Cd for 8 weeks led to hippocampal-dependent cognitive deficits and neural/synaptic damage in mice. NLRP3 inflammasome-related protein expression (NLRP3, ASC, and caspase1 p20) and neuronal pyroptosis were significantly upregulated in Cd-treated hippocampi and SH-SY5Y cells. Moreover, pretreatment with the NLRP3 inhibitor MCC950 mitigated Cd-elicited NLRP3 inflammasome activation and subsequent neuronal pyroptosis in SH-SY5Y cells. Furthermore, exposure to Cd downregulated Sirt3 expression, suppressed SOD2 activity by hyperacetylation, and enhanced mtROS accumulation in vivo and in vitro. Notably, Cd-induced NLRP3 inflammasome-dependent neuronal pyroptosis was attenuated by a mtROS scavenger or Sirt3 overexpression in SH-SY5Y cells. In addition, Cd failed to further suppress SOD activity and activate NLRP3 inflammasome-dependent neuronal pyroptosis in Sirt3 shRNA-treated SH-SY5Y cells. Collectively, our findings indicate that Cd exposure induces neuronal injury and cognitive deficits by activating NLRP3 inflammasome-dependent neuronal pyroptosis and that activation of the NLRP3 inflammasome is partially mediated by the Sirt3-mtROS axis.

5.
Environ Pollut ; 334: 122158, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37429494

ABSTRACT

The brominated flame retardant 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) is widely distributed in the environment and poses a certain risk to human health. Studies have reported that oxidative stress is a key mechanism underlying BDE-47-induced neurotoxicity. Mitochondrial reactive oxygen species (mtROS) is a crucial mediator of NLRP3 inflammasome activation, which is involved in cognitive dysfunction induced by environmental toxins. However, the function of the mtROS-NLRP3 inflammasome pathway in BDE-47-elicited cognitive deficits and the underlying mechanisms remain elusive. Our data illustrated that eight weeks of BDE-47 (20 mg/kg) gavage led to cognitive deficits and hippocampal neuronal injury in mice. BDE-47 exposure downregulated Sirt3 expression and decreased the activity and expression level of SOD2, thereby inhibiting mtROS scavenging and activating NLRP3 inflammasome-mediated pyroptosis in the mouse hippocampus and BV-2 cells. In vitro, BDE-47-evoked microglial pyroptosis relied on NLRP3 inflammasome activation. Moreover, a mtROS scavenger (TEMPO) attenuated NLRP3 inflammasome activation and subsequent microglial pyroptosis under BDE-47 stress. Furthermore, Sirt3 overexpression restored the activity and expression of SOD2 and enhanced mtROS scavenging, thereby suppressing NLRP3 inflammasome activation and ameliorating microglial pyroptosis. Notably, honokiol (HKL), a pharmacological agonist of Sirt3, mitigated BDE-47-evoked hippocampal neuronal injury and cognitive impairment by inhibiting mtROS-NLRP3 axis-mediated pyroptosis via Sirt3 upregulation.


Subject(s)
Cognitive Dysfunction , Flame Retardants , Sirtuin 3 , Humans , Animals , Mice , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pyroptosis , Sirtuin 3/genetics , Sirtuin 3/metabolism , Flame Retardants/toxicity , Microglia/metabolism , Down-Regulation , Cognitive Dysfunction/chemically induced , Reactive Oxygen Species/metabolism , Cognition
6.
Front Oncol ; 12: 1045797, 2022.
Article in English | MEDLINE | ID: mdl-36452487

ABSTRACT

Poly(rC)-binding proteins (PCBPs), a defined subfamily of RNA binding proteins, are characterized by their high affinity and sequence-specific interaction with poly-cytosine (poly-C). The PCBP family comprises five members, including hnRNP K and PCBP1-4. These proteins share a relatively similar structure motif, with triple hnRNP K homology (KH) domains responsible for recognizing and combining C-rich regions of mRNA and single- and double-stranded DNA. Numerous studies have indicated that PCBPs play a prominent role in hematopoietic cell growth, differentiation, and tumorigenesis at multiple levels of regulation. Herein, we summarized the currently available literature regarding the structural and functional divergence of various PCBP family members. Furthermore, we focused on their roles in normal hematopoiesis, particularly in erythropoiesis. More importantly, we also discussed and highlighted their involvement in carcinogenesis, including leukemia and lymphoma, aiming to clarify the pleiotropic roles and molecular mechanisms in the hematopoietic compartment.

7.
ACS Appl Mater Interfaces ; 14(42): 47369-47384, 2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36228174

ABSTRACT

Bone implants with the photothermal effect are promising for the treatment of bone tumor defects. Noble metal-based photothermal nanoagents are widely studied for their stable photothermal effect, but they are expensive and difficult to directly grow on implant surfaces. In contrast, non-noble metal photothermal nanoagents are economical but unstable. Herein, to develop a stable and economical photothermal film on bone implants, a Ni nanoparticle-doped oxide semiconductor film was grown in situ on Nitinol via the reduction of Ni-Ti-layered double hydroxides. Ni nanoparticles remained stable in the NiTiO3 structure even when immersed in fluid for 1 month, and thus, the film presented a reliable photothermal effect under near-infrared light irradiation. The film also showed excellent in vitro and in vivo antitumor performance. Moreover, the nanostructure on the film allowed bone differentiation of mouse embryo cells (C3H10T1/2), and the released Ni ions supported the angiogenesis behavior of human vein endothelial cells. Bone implantation experiments further showed the enhancement of osteointegration of the modified Nitinol implant in vivo. This novel multifunctional Nitinol bone implant design offers a promising strategy for the therapy of bone tumor-related defects.


Subject(s)
Bone Neoplasms , Metal Nanoparticles , Nanoparticles , Humans , Mice , Animals , Oxides , Endothelial Cells , Bone Regeneration , Bone Neoplasms/drug therapy , Metal Nanoparticles/therapeutic use , Nanoparticles/chemistry , Hydroxides , Semiconductors
8.
J Hazard Mater ; 437: 129381, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35752048

ABSTRACT

Oxidative stress and mitochondrial dysfunction contribute greatly to fluoride-induced cognitive impairment and behavioural disorders. Honokiol, a natural biphenolic compound, possesses antioxidant and mitochondrial protective properties. The present study investigated the protective actions of honokiol on NaF-elicited cognitive deficits and elucidated the possible mechanism of honokiol-mediated protection. The results demonstrated that honokiol administration markedly attenuated fluoride-induced cognitive impairments and neural/synaptic injury in mice. Moreover, honokiol elevated the activity and expression of SOD2 and promoted mtROS scavenging through Sirt3 activation in NaF-treated mice and SH-SY5Y cell lines. Meanwhile, honokiol substantially lowered mtROS production by enhancing Sirt3-mediated mitochondrial DNA (mtDNA) transcription, thereby leading to significant increases in ATP synthesis and complex I activity. Further studies revealed that honokiol activated AMPK and upregulated the PGC-1α and Sirt3 protein expression in vivo and in vitro. Intriguingly, the protective actions of honokiol on oxidative stress and mitochondrial dysfunction were abolished by AMPK shRNA or Sirt3 shRNA. Notably, AMPK knockdown prevented the increase in PGC-1α and Sirt3 expression induced by honokiol, while Sirt3 shRNA suppressed Sirt3 signaling without significant effects on p-AMPK and PGC-1α expression. In conclusion, our findings indicate that honokiol mitigates NaF-induced oxidative stress and mitochondrial dysfunction by regulating mtROS homeostasis, partly via the AMPK/PGC-1α/Sirt3 pathway, which ultimately contributes to neuronal/synaptic injury and cognitive deficits.


Subject(s)
Cognitive Dysfunction , Neuroblastoma , Sirtuin 3 , AMP-Activated Protein Kinases/metabolism , Animals , Biphenyl Compounds , Cognition , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , DNA, Mitochondrial , Fluorides/pharmacology , Humans , Lignans , Mice , Mitochondria , Neuroblastoma/metabolism , Oxidative Stress , RNA, Small Interfering , Sirtuin 3/genetics , Sirtuin 3/metabolism , Sirtuin 3/pharmacology
9.
Antioxidants (Basel) ; 11(5)2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35624708

ABSTRACT

Nuclear factor erythroid factor E2-related factor 2 (Nrf2) transcribes antioxidant genes that reduce the blood pressure (BP), yet its activation with tert-butylhydroquinone (tBHQ) in mice infused with angiotensin II (Ang II) increased mean arterial pressure (MAP) over the first 4 days of the infusion. Since tBHQ enhanced cyclooxygenase (COX) 2 expression in vascular smooth muscle cells (VSMCs), we tested the hypothesis that tBHQ administration during an ongoing Ang II infusion causes an early increase in microvascular COX-dependent reactive oxygen species (ROS) and contractility. Mesenteric microarteriolar contractility was assessed on a myograph, and ROS by RatioMaster™. Three days of oral tBHQ administration during the infusion of Ang II increased the mesenteric microarteriolar mRNA for p47phox, the endothelin type A receptor and thromboxane A2 synthase, and increased the excretion of 8-isoprostane F2α and the microarteriolar ROS and contractions to a thromboxane A2 (TxA2) agonist (U-46,619) and endothelin 1 (ET1). These were all prevented in Nrf2 knockout mice. Moreover, the increases in ROS and contractility were prevented in COX1 knockout mice with blockade of COX2 and by blockade of thromboxane prostanoid receptors (TPRs). In conclusion, the activation of Nrf2 over 3 days of Ang II infusion enhances microarteriolar ROS and contractility, which are dependent on COX1, COX2 and TPRs. Therefore, the blockade of these pathways may diminish the early adverse cardiovascular disease events that have been recorded during the initiation of Nrf2 therapy.

10.
Front Oncol ; 12: 1132485, 2022.
Article in English | MEDLINE | ID: mdl-36866022

ABSTRACT

[This corrects the article DOI: 10.3389/fonc.2022.1045797.].

11.
Food Chem Toxicol ; 158: 112665, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34780879

ABSTRACT

Excessive fluoride is capable of inducing cognitive deficits, but the mechanisms remain elusive. This study aimed to investigate the effects and underlying mechanisms of fluoride on mitochondrial dysfunction and neurobiological alterations, as well as cognitive impairment. C57BL/6 mice were orally administered 25, 50, and 100 mg/L NaF for 90 days. Cultured human neuroblastoma SH-SY5Y cells were exposed to NaF (110 mg/L) for 24 h in the presence or absence of Sirt3 overexpression. The results demonstrated that chronic exposure to high fluoride induced cognitive deficits and neural/synaptic injury in mice. Fluoride reduced mitochondrial antioxidant enzyme activities and elevated SOD2 acetylation by downregulating Sirt3 expression in the brains of mice and NaF-treated SH-SY5Y cells. Moreover, fluoride lowered mtDNA transcription and induced mitochondrial dysfunction along with increased FoxO3A acetylation in the brains of mice and NaF-treated SH-SY5Y cells. Subsequent experiments revealed that overexpression of Sirt3 significantly attenuated the adverse effects of fluoride on radical scavenging capabilities and mtDNA transcription, as well as mitochondrial function in SH-SY5Y cells. These results suggest that chronic long-term fluoride exposure evokes neural/synaptic injury and cognitive impairment through mitochondrial dysfunction and its associated oxidative stress, which is, at least partly, mediated by Sirt3 inhibition in the mouse brain.


Subject(s)
Cognitive Dysfunction/chemically induced , Mitochondria , Sirtuin 3 , Sodium Fluoride/toxicity , Animals , Brain Chemistry/drug effects , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Morris Water Maze Test/drug effects , Neurotoxins/toxicity , Sirtuin 3/genetics , Sirtuin 3/metabolism
12.
J Biomed Nanotechnol ; 15(7): 1482-1491, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31196352

ABSTRACT

As a widely used first-line chemotherapy drug for tumor, Doxorubicin (DOX) can induce various side effects on normal tissues because of its non-specific distribution in the body. Emerging evidence has shown that platelets have the capability to recognize and interact with tumor cells. Inspired by this, the platelet-based drug delivery system was constructed by loading of DOX in platelet cytoplasm and modification of transferrin on the surface of platelet (Tf-P-DOX). The encapsulation efficiency of DOX in platelet was the highest at the DOX concentration of 0.05 mM, and reached to 64.9%. Fluorescence microscopy showed that the Tf-P-DOX facilitated cell uptakes and enhanced intracellular drug accumulation in B16F10 cells. Compared with free DOX, Tf-P-DOX exhibited an enhanced effect on cell apoptosis at the same concentration of DOX. In vivo imaging system showed that the near-infrared fluorescence of B16F10 tumor-bearing mice was mainly accumulated in the tumor site, which caused the inhibition of tumor growth in mice. The morphological changes of tumor tissue in Tf-P-DOX group was significant in comparison with those of the control group, including the small nucleus, the insufficiency of cancerous nest, and the infiltration of inflammatory cells, while Tf-P-DOX did not show significant adverse effects on normal tissues. Compared with the control group, the levels of caspase 9 and caspase 3 protein expressions were increased significantly in Tf-P-DOX group. Our studies suggest platelets can be repurposed as promising carriers for efficient targeting and treatment of solid tumors.


Subject(s)
Melanoma , Animals , Cell Line, Tumor , Doxorubicin , Drug Carriers , Drug Delivery Systems , Mice
13.
Oncotarget ; 9(10): 9468-9479, 2018 Feb 06.
Article in English | MEDLINE | ID: mdl-29507703

ABSTRACT

Polymorphism (rs3918242) in the MMP9 gene has been reported to be associated with coronary artery disease (CAD). This study aims to investigate a more accurate estimation of the relationship between CAD and rs3918242 polymorphism by a meta-analysis method. We systematically searched studies on the association of rs3918242 polymorphism and CAD in PubMed, Web of Science, the Cochrane Library, Wanfang Data and CNKI. We used Stata 12.0 and RevMan 5.3 software to perform the meta-analyses. A total of 37 case-control studies involving 13,035 CAD patients and 11,372 non-CAD controls were included. A statistically significant association between rs3918242 polymorphism and CAD was observed in allelic model (Odds ratio (OR) 1.34; 95% confidence interval (CI) 1.20-1.50; p < 0.00001), recessive model (OR 1.43; 95% CI 1.17-1.75; p = 0.0004), and in dominant model ( OR 1.36; 95% CI 1.20-1.53; p < 0.00001). Moreover, we also found that there is a statistically significant association between rs3918242 polymorphism and myocardial infarction (MI) in Asians with allelic model (OR 1.66; 95% CI 1.29-2.14; p < 0.0001), recessive model (OR 2.29; 95% CI 1.44-3.63; p = 0.004), and dominant (OR 1.74; 95% CI 1.29-2.35; p = 0.0003) model. A similar result in Caucasians with allelic model (OR 1.14; 95% CI 1.02-1.27; p = 0.02), and in dominant (OR 1.17; 95% CI 1.04-1.32; p = 0.01) model. Our meta-analysis suggested that the MMP9 T allele is a risk factor for CAD and MI.

14.
Biol Trace Elem Res ; 185(1): 124-134, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29294227

ABSTRACT

Nanometer zinc oxide (nano-ZnO) is widely used in diverse industrial and agricultural fields. Due to the extensive contact humans have with these particles, it is crucial to understand the potential effects that nano-ZnO have on human health. Currently, information related to the toxicity and mechanisms of nano-ZnO is limited. The aim of the present study was to investigate acute and cumulative toxic effects of 50-nm unmodified ZnO in mice. This investigation will seek to establish median lethal dose (LD50), a cumulative coefficient, and target organs. The acute and cumulative toxicity was investigated by Karber's method and via a dose-increasing method, respectively. During the experiment, clinical signs, mortality, body weights, hematology, serum biochemistry, gross pathology, organ weight, and histopathology were examined. The LD50 was 5177-mg/kg·bw; the 95% confidence limits for the LD50 were 5116-5238-mg/kg·bw. It could be concluded that the liver, kidney, lung, and gastrointestinal tract were target organs for the 50-nm nano-ZnO acute oral treatment. The cumulative coefficient (K) was 1.9 which indicated that the cumulative toxicity was apparent. The results also indicated that the liver, kidney, lung, and pancrea were target organs for 50-nm nano-ZnO cumulative oral exposure and might be target organs for subchronic and chronic toxicity of oral administered 50-nm ZnO.


Subject(s)
Zinc Oxide/toxicity , Animals , Female , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Male , Mice , Nanoparticles/toxicity , Pancreas/drug effects , Pancreas/metabolism , Particle Size
15.
Int J Mol Sci ; 16(10): 23572-86, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26437401

ABSTRACT

Aspartic acid semialdehyde dehydrogenase (ASADH) lies at the first branch point in the essential aspartic acid biosynthetic pathway that is found in bacteria and plants but is absent from animals. Mutations in the asadh gene encoding ASADH produce an inactive enzyme, which is lethal. Therefore, in this study, we investigated the hypothesis that ASADH represents a new anti-Mycobacterium tuberculosis (MTB) target. An asadh promoter-replacement mutant MTB, designated MTB::asadh, in which asadh gene expression is regulated by pristinamycin, was constructed to investigate the physiological functions of ASADH in the host bacteria. Bacterial growth was evaluated by monitoring OD600 and ASADH expression was analyzed by Western blotting. The results showed that the growth and survival of MTB::asadh was completely inhibited in the absence of the inducer pristinamycin. Furthermore, the growth of the mutant was rigorously dependent on the presence of the inducer in the medium. The starved mutant exhibited a marked reduction (approximately 80%) in the cell wall materials compared to the wild-type, in addition to obvious morphological differences that were apparent in scanning electron microscopy studies; however, with the addition of pristinamycin, the cell wall contents and morphology similar to those of the wild-type strain were recovered. The starved mutant also exhibited almost no pathogenicity in an in vitro model of infection using mouse macrophage J774A.1 cells. The mutant showed a concentration-dependent recovery of pathogenicity with the addition of the inducer. These findings implicate ASADH as a promising target for the development of novel anti-MTB drugs.


Subject(s)
Antitubercular Agents/pharmacology , Aspartate-Semialdehyde Dehydrogenase/metabolism , Mycobacterium tuberculosis/enzymology , Animals , Blotting, Western , Cell Line , Cell Wall/drug effects , Cell Wall/metabolism , Electrophoresis, Agar Gel , Mice , Mutation/genetics , Mycobacterium tuberculosis/cytology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Promoter Regions, Genetic/genetics , Reproducibility of Results
16.
PLoS One ; 9(2): e88127, 2014.
Article in English | MEDLINE | ID: mdl-24498431

ABSTRACT

BACKGROUND: Adult metabolic syndrome may in part have origins in fetal or early life. This study was designed to explore the effect of prenatal exposure to lipopolysaccharide and high-fat diet on metabolic syndrome in offspring rats. METHODS: 32 pregnant rats were randomly divided into four groups, including Control group; LPS group (pregnant rats were injected with LPS 0.4 mg/kg intraperitoneally on the 8(th), 10(th) and 12(th) day of pregnancy); High-fat group (maternal rats had high-fat diet during pregnancy and lactation period, and their pups also had high-fat diet up to the third month of life); LPS + High-fat group (rats were exposed to the identical experimental scheme with LPS group and High-fat group). RESULTS: Blood pressure elevated in LPS group and High-fat group, reduced in LPS+High-fat group, accompanied by the increase of serum leptin level in LPS and High-fat group and increase of serum IL-6, TNF-a in High-fat group; both serum insulin and cholesterol increased in High-fat and LPS+High-fat group, as well as insulin in LPS group. HOMA-IR value increased in LPS, High-fat and LPS+High-fat group, and QUICKI decreased in these groups; H-E staining showed morphologically pathological changes in thoracic aorta and liver tissue in the three groups. Increased serum alanine and aspartate aminotransferase suggest impaired liver function in LPS+High-fat group. CONCLUSION/SIGNIFICANCE: Prenatal exposure to lipopolysaccharide combined with pre- and postnatal high-fat diet result in lowered blood pressure, insulin resistance and impaired liver function in three-month old offspring rats. The lowered blood pressure might benefit from the predictive adaptive response to prenatal inflammation.


Subject(s)
Blood Pressure/drug effects , Diet, High-Fat/adverse effects , Insulin Resistance , Lipopolysaccharides/adverse effects , Metabolic Syndrome/etiology , Prenatal Exposure Delayed Effects/etiology , Animals , Animals, Newborn , Blood Glucose/metabolism , Body Weight , Cholesterol/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Insulin/metabolism , Kidney Function Tests , Liver Function Tests , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Pregnancy , Rats , Rats, Sprague-Dawley , Triglycerides/metabolism
17.
J Renin Angiotensin Aldosterone Syst ; 15(2): 162-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23161146

ABSTRACT

OBJECTIVE: The objective of this article is to explore the role of imidapril on pulmonary hypertension induced by low ambient temperature in broiler chickens. MATERIALS AND METHODS: Ninety chickens were randomly divided into three groups (n = 30): a control group, a low-temperature group and an imidapril group. Chickens in the low-temperature group and imidapril group were exposed to low ambient temperature from 14 days of age until 45 days of age; chickens in the imidapril group were gavaged with imidapril 3 mg/kg once daily for 30 days. The pulmonary arterial pressure, main pulmonary arterial diameter and pulmonary arterial wall thickness were measured, and lung tissue ACE, ACE2 mRNA expression, proliferating cell nuclear antigen (PCNA)-positive cells and Ang II, Ang (1-7) concentration were evaluated. RESULTS: The pulmonary arterial pressure was higher, the main pulmonary arterial diameter was wider and the pulmonary arterial wall was thicker in the low-temperature group than those in the control group and the imidapril group. ACE mRNA and PCNA-positive cells increased significantly in the low-temperature group compared with the control group and imidapril group; lung tissue Ang II concentration in the low-temperature group was higher, but Ang (1-7) content was lower than that in the control group and imidapril group. CONCLUSION: Imidapril provides a protective effect on pulmonary hypertension induced by low ambient temperature in broiler chickens.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cold Temperature , Hypertension, Pulmonary/prevention & control , Hypertension, Pulmonary/veterinary , Imidazolidines/therapeutic use , Poultry Diseases/prevention & control , Angiotensin I/metabolism , Angiotensin II/metabolism , Animals , Arterial Pressure , Chickens , Lung/metabolism , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Pulmonary Artery/pathology
18.
Poult Sci ; 92(6): 1492-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23687144

ABSTRACT

This study explored the effect of imidapril on the right ventricular remodeling induced by low ambient temperature in broiler chickens. Twenty-four broiler chickens were randomly divided into 3 groups (n = 8), including the control group, low temperature group, and imidapril group. Chickens in the control group were raised at normal temperature, whereas chickens in the low temperature group and imidapril group were exposed to low ambient temperature (12 to 18°C) from 14 d of age until 45 d of age. At the same time, chickens in the imidapril group were gavaged with imidapril at 3 mg/kg once daily for 30 d. The thickness of the right ventricular wall was observed with echocardiography. The BW and wet lung weight as well as weight of right and left ventricles and ventricular septum were measured. Both wet lung weight index and right ventricular hypertrophy index were calculated. Pulmonary arterial systolic pressure was assessed according to echocardiography. The expression of ACE and ACE2 mRNA in the right ventricular myocardial tissue was quantified by real-time PCR. Proliferating cell nuclear antigen-positive cells were detected by immunohistostaining. The concentration of angiotensin (Ang) II and Ang (1-7) in the right ventricular myocardial tissue was measured with ELISA. The results showed that right ventricular hypertrophy index, wet lung weight index, pulmonary arterial systolic pressure, expression of ACE mRNA in the right ventricular tissue, Ang II concentration, and the thickness of the right ventricular wall in the low temperature group increased significantly compared with those in the control group and imidapril group. The ACE2 mRNA expression increased 36%, whereas Ang (1-7) concentration decreased significantly in the low temperature group compared with that in the control group and imidapril group. In conclusion, imidapril inhibits right ventricular remodeling induced by low ambient temperature in broiler chickens.


Subject(s)
Chickens , Cold Temperature , Heart Ventricles/pathology , Housing, Animal , Imidazolidines/pharmacology , Ventricular Remodeling/drug effects , Angiotensin I/genetics , Angiotensin I/metabolism , Angiotensin II/genetics , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animal Husbandry , Animals , Gene Expression Regulation , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism
19.
Biol Trace Elem Res ; 152(1): 117-24, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23377609

ABSTRACT

The detection of cadmium ions using enzyme-linked immunosorbent assays (ELISA) has been reported by several research groups. Because cadmium ions are too small to stimulate the immune system, high molecular weight immunogens of cadmium are constructed using bifunctional chelators. At present, the most commonly used bifunctional chelator for the preparation of antigens for heavy metal ions is 1-(4-isothiocyanobenzyl) ethylenediamine N,N,N',N'-tetraacetic acid (ITCBE). However, the price of ITCBE is high. So we are interested in a cheaper bifunctional chelator, 1-(4-aminobenzyl) ethylenediamine N,N,N',N'-tetraacetic acid (aminobenzyl-EDTA). Here, cadmium ions were conjugated to carrier proteins using aminobenzyl-EDTA to make artificial antigens. Then, several mice were immunized with the antigen. And monoclonal antibodies (MAbs) against cadmium were produced. Spleen cells of immunized mice were fused with myeloma cells. The resulting hybridomas were screened using protein conjugates which were covalently bound to metal-free EDTA or cadmium. Three hybridoma cell lines (A3, E4 and B5) that produced MAbs with high selectivity and sensitivity were expanded for further study. Cross-reactivities with other metals were below 1 %. These antibodies were used to construct competitive ELISAs. The IC50 for A3 was 8.4 µg/l. The detection range and the lowest detection limit using the antibody A3 was 0.394-64.39 and 0.051 µg/l, respectively. Spike-recovery studies in tap water showed that the antibody A3 could be used for cadmium detection in drinking water.


Subject(s)
Acetates/immunology , Antibodies, Monoclonal/immunology , Cadmium/immunology , Edetic Acid/analogs & derivatives , Ethylenediamines/immunology , Serum Albumin, Bovine/immunology , Acetates/chemistry , Animals , Antibodies, Monoclonal/metabolism , Antigens/chemistry , Antigens/immunology , Cadmium/analysis , Cadmium/chemistry , Cattle , Drinking Water/analysis , Drinking Water/chemistry , Edetic Acid/chemistry , Edetic Acid/immunology , Enzyme-Linked Immunosorbent Assay/methods , Ethylenediamines/chemistry , Hybridomas/cytology , Hybridomas/metabolism , Immunization , Mice , Molecular Structure , Reproducibility of Results , Serum Albumin, Bovine/chemistry , Spectrophotometry, Ultraviolet
20.
Exp Toxicol Pathol ; 65(4): 433-9, 2013 May.
Article in English | MEDLINE | ID: mdl-22342485

ABSTRACT

UNLABELLED: Prenatal exposure to LPS(lipopolysaccharide) results in renal damage in offspring rats, but the mechanism is unknown. The present study was to explore the role of angiotensin II and inflammation in the development of renal damage induced by prenatal exposure to LPS. The pregnant rats were randomly divided into two groups, i.e., control group, LPS group. The rats in the two groups were administered intraperitoneally with vehicle or 0.79 mg/kg LPS on 8th, 10th and 12th day during gestation. The mRNA expression of angiotensinogen, renin, AT(1)-R, AT(2)-R, TNF-α and IL-6 in embryos were assessed. Renal Ang II-positive cells, monocytes/macrophages, lymphocytes, collagen I and TUNEL-positive cells were identified by immunohistochemical staining in newborn and 7-week-old offspring rats. The number of glomeruli and creatinine clearance rate were determined in offspring at 7 weeks of age. The results showed that prenatal LPS decreased AT(2)-R mRNA expression but increased TNF-α and IL-6 mRNA expression in embryos. Prenatal LPS decreased renal angiotensin II-positive cells in newborn offspring rats, while these increased in 7-week-old offspring rats. Prenatal LPS decreased glomerular number and creatinine clearance rate but increased renal infiltrating monocytes/macrophages and lymphocytes at 7 weeks of age. Prenatal LPS also increased TUNEL-positive cells and collagen I expressions in newborn rats and 7-week-old offspring rats. CONCLUSION: Alteration of embryonic AT(2)-R and inflammatory cytokines gene expression induced by prenatal exposure to lipopolysaccharide affects renal development.


Subject(s)
Inflammation/physiopathology , Kidney/embryology , Prenatal Exposure Delayed Effects/physiopathology , Receptor, Angiotensin, Type 2/metabolism , Renin-Angiotensin System/physiology , Angiotensin II , Animals , Apoptosis , Cytokines/metabolism , Embryo, Mammalian , Female , Immunohistochemistry , In Situ Nick-End Labeling , Inflammation/metabolism , Kidney/drug effects , Kidney/physiopathology , Kidney Function Tests , Lipopolysaccharides/toxicity , Oligonucleotide Array Sequence Analysis , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Renin-Angiotensin System/drug effects , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...