Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 448
Filter
1.
FEBS J ; 291(8): 1780-1794, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38317509

ABSTRACT

Colorectal cancer (CRC) has emerged as the third most prevalent and second deadliest cancer worldwide. Metabolic reprogramming is a key hallmark of cancer cells. Phosphoglycerate dehydrogenase (PHGDH) is over-expressed in multiple cancers, including CRC. Although the role of PHGDH in metabolism has been extensively investigated, its effects on CRC development remains to be elucidated. In the present study, it was demonstrated that PHGDH expression was significantly up-regulated in colorectal cancer. PHGDH expression was positively correlated with that of the aryl hydrocarbon receptor (AhR) and its target genes, CYP1A1 and CYP1B1, in CRC cells. Knockdown of PHGDH reduced AhR levels and activity, as well as the ratio of reduced to oxidized glutathione. The selective AhR antagonist stemregenin 1 induced cell death through reactive oxygen species-dependent autophagy in CRC cells. PHGDH knockdown induced CRC cell sensitivity to stemregenin 1 via the autophagy pathway. Our findings suggest that PHGDH modulates AhR signaling and the redox-dependent autophagy pathway in CRC, and that the combination of inhibition of both PHGDH and AhR may be a novel therapeutic strategy for CRC.


Subject(s)
Colorectal Neoplasms , Receptors, Aryl Hydrocarbon , Humans , Autophagy/genetics , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Phosphoglycerate Dehydrogenase/deficiency , Phosphoglycerate Dehydrogenase/genetics , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
2.
Int J Mol Sci ; 24(21)2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37958638

ABSTRACT

Herbal extracts represent a wide spectrum of biologically active ingredients with potential medical applications. By screening minor constituents of jasmine essential oil towards aryl hydrocarbon receptor (AhR) activity using a gene reporter assay (GRA), we found the antagonist effects of jasmone (3-methyl-2-[(2Z)-pent-2-en-1-yl]cyclopent-2-en-1-one). It inhibited 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-, benzo[a]pyrene (BaP)-, and 6-formylindolo[3,2-b]carbazole (FICZ)-triggered AhR-dependent luciferase activity in a concentration-dependent manner. However, the inhibition differed markedly between TCDD, BaP, and FICZ, with the latter being significantly less inhibited. The dose-response analysis confirmed an allosteric type of AhR antagonism. Furthermore, jasmone efficiently inhibited AhR activation by AhR agonists and microbial catabolites of tryptophan (MICTs). TCDD- and FICZ-inducible CYP1A1 expression in primary human hepatocytes was inhibited by jasmone, whereas in the human HepG2 and LS180 cells, jasmone antagonized only TCDD-activated AhR. Jasmone only partially displaced radiolabeled TCDD from its binding to mouse Ahr, suggesting it is not a typical orthosteric ligand of AhR. TCDD-elicited AhR nuclear translocation was not affected by jasmone, whereas downstream signaling events, including the formation of the AhR:ARNT complex and enrichment of the CYP1A1 promoter, were inhibited by jasmone. In conclusion, we show that jasmone is a potent allosteric antagonist of AhR. Such discovery may help to find and/or clarify the use of jasmone in pharmaco- and phytotherapy for conditions where AhR plays a key role.


Subject(s)
Polychlorinated Dibenzodioxins , Receptors, Aryl Hydrocarbon , Animals , Humans , Mice , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Ligands , Polychlorinated Dibenzodioxins/adverse effects , Receptors, Aryl Hydrocarbon/antagonists & inhibitors
3.
Toxins (Basel) ; 14(2)2022 01 26.
Article in English | MEDLINE | ID: mdl-35202128

ABSTRACT

The uremic toxin indoxyl sulfate (IS), elevated in chronic kidney disease (CKD), is known to contribute towards progressive cardiovascular disease. IS activates the aryl hydrocarbon receptor (AhR) mediating oxidative stress and endothelial dysfunction via activation of the CYP1A1 pathway. The present study examines AhR inhibition with the antagonist, CH223191, on IS-mediated impairment of vascular endothelial function and disruption of redox balance. The acute effects of IS on endothelium-dependent relaxation were assessed in aortic rings from Sprague Dawley rats exposed to the following conditions: (1) control; (2) IS (300 µM); (3) IS + CH223191 (1 µM); (4) IS + CH223191 (10 µM). Thereafter, tissues were assessed for changes in expression of redox markers. IS reduced the maximum level of endothelium-dependent relaxation (Rmax) by 42% (p < 0.001) compared to control, this was restored in the presence of increasing concentrations of CH223191 (p < 0.05). Rings exposed to IS increased expression of CYP1A1, nitro-tyrosine, NADPH oxidase 4 (NOX4), superoxide, and reduced eNOS expression (p < 0.05). CH223191 (10 µM) restored expression of these markers back to control levels (p < 0.05). These findings demonstrate the adverse impact of IS-mediated AhR activation on the vascular endothelium, where oxidative stress may play a critical role in inducing endothelial dysfunction in the vasculature of the heart and kidneys. AhR inhibition could provide an exciting novel therapy for CVD in the CKD setting.


Subject(s)
Aorta, Thoracic/drug effects , Azo Compounds/pharmacology , Endothelium, Vascular/drug effects , Indican/pharmacology , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiology , Cytochrome P-450 CYP1A1/genetics , Endothelium, Vascular/physiology , Gene Expression/drug effects , Male , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic , Vasodilation/drug effects
4.
Immunity ; 55(2): 324-340.e8, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35139353

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a sensor of products of tryptophan metabolism and a potent modulator of immunity. Here, we examined the impact of AhR in tumor-associated macrophage (TAM) function in pancreatic ductal adenocarcinoma (PDAC). TAMs exhibited high AhR activity and Ahr-deficient macrophages developed an inflammatory phenotype. Deletion of Ahr in myeloid cells or pharmacologic inhibition of AhR reduced PDAC growth, improved efficacy of immune checkpoint blockade, and increased intra-tumoral frequencies of IFNγ+CD8+ T cells. Macrophage tryptophan metabolism was not required for this effect. Rather, macrophage AhR activity was dependent on Lactobacillus metabolization of dietary tryptophan to indoles. Removal of dietary tryptophan reduced TAM AhR activity and promoted intra-tumoral accumulation of TNFα+IFNγ+CD8+ T cells; provision of dietary indoles blocked this effect. In patients with PDAC, high AHR expression associated with rapid disease progression and mortality, as well as with an immune-suppressive TAM phenotype, suggesting conservation of this regulatory axis in human disease.


Subject(s)
Immune Tolerance/immunology , Receptors, Aryl Hydrocarbon/immunology , Tryptophan/immunology , Tumor-Associated Macrophages/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Humans , Indoles/immunology , Indoles/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Microbiota/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/metabolism
5.
J Mol Med (Berl) ; 100(2): 215-243, 2022 02.
Article in English | MEDLINE | ID: mdl-34800164

ABSTRACT

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that can be activated by structurally diverse compounds arising from the environment and the microbiota and host metabolism. Expanding evidence has been shown that the modulation of the canonical pathway of AHR occurs during several chronic diseases and that its abrogation might be of clinical interest for metabolic and inflammatory pathological processes. However, most of the evidence on the pharmacological abrogation of the AHR-CYP1A1 axis has been reported in vitro, and therefore, guidance for in vivo studies is needed. In this review, we cover the state-of-the-art of the pharmacodynamic and pharmacokinetic properties of AHR antagonists and CYP1A1 inhibitors in different in vivo rodent (mouse or rat) models of disease. This review will serve as a road map for those researchers embracing this emerging therapeutic area targeting the AHR. Moreover, it is a timely opportunity as the first AHR antagonists have recently entered the clinical stage of drug development.


Subject(s)
Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Animals , Humans , Receptors, Aryl Hydrocarbon/metabolism
6.
Int J Mol Sci ; 22(22)2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34830207

ABSTRACT

Nearly 18 million people died from cardiovascular diseases in 2019, of these 85% were due to heart attack and stroke. The available therapies although efficacious, have narrow therapeutic window and long list of contraindications. Therefore, there is still an urgent need to find novel molecular targets that could protect the brain and heart against ischemia without evoking major side effects. Nuclear receptors are one of the promising targets for anti-ischemic drugs. Modulation of estrogen receptors (ERs) and peroxisome proliferator-activated receptors (PPARs) by their ligands is known to exert neuro-, and cardioprotective effects through anti-apoptotic, anti-inflammatory or anti-oxidant action. Recently, it has been shown that the expression of aryl hydrocarbon receptor (AhR) is strongly increased after brain or heart ischemia and evokes an activation of apoptosis or inflammation in injury site. We hypothesize that activation of ERs and PPARs and inhibition of AhR signaling pathways could be a promising strategy to protect the heart and the brain against ischemia. In this Review, we will discuss currently available knowledge on the mechanisms of action of ERs, PPARs and AhR in experimental models of stroke and myocardial infarction and future perspectives to use them as novel targets in cardiovascular diseases.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Ischemia/metabolism , Myocardial Ischemia/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Estrogen/metabolism , Stroke/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Brain Ischemia/drug therapy , Disease Models, Animal , Humans , Ligands , Mice , Molecular Targeted Therapy/methods , Myocardial Ischemia/drug therapy , Peroxisome Proliferator-Activated Receptors/agonists , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Selective Estrogen Receptor Modulators/pharmacology , Selective Estrogen Receptor Modulators/therapeutic use , Signal Transduction/drug effects , Stroke/drug therapy , Treatment Outcome
7.
Biomolecules ; 11(10)2021 10 15.
Article in English | MEDLINE | ID: mdl-34680153

ABSTRACT

Ischemia-reperfusion injury is the commonest form of acute kidney injury (AKI). Tubular epithelial cell senescence contributes to incomplete recovery from AKI and predisposes to subsequent chronic kidney disease. In cultures of primary proximal renal tubular epithelial cells (RPTECs) subjected to anoxia or reoxygenation, we evaluated the role of indoleamine 2,3-dioxygenase 1 (IDO) in cellular senescence. Proteins of interest were assessed with Western blotting or enzyme-linked immunosorbent assay or histochemically. Under anoxia or reoxygenation, IDO expression and activity were increased. Moreover, the two IDO-derived pathways, the general control nonderepressible 2 kinase (GCN2K) pathway and the aryl-hydrocarbon receptor (AhR) pathway, were also activated. A DNA damage response (DDR) took place and led to increased levels of the cell-cycle inhibitors p21 and p16, and senescence-associated ß-galactosidase (SA-ß-Gal) activity. Cell proliferation was inhibited, and more IL-6 was produced. The IDO inhibitor 1-DL-methyl-tryptophan ameliorated the DDR; decreased p21, p16, and SA-ß-Gal activity; restored cell proliferation; and decreased IL-6 production. The AhR inhibitor CH223191 did not affect the above parameters. In conclusion, anoxia and the subsequent reoxygenation upregulate IDO. IDO depletes tryptophan and activates GCN2K. The latter enhances the anoxia- or reoxygenation-induced DDR, resulting in increased p21 and p16 expression and eventually leading to RPTEC senescence. Since cellular senescence affects AKI outcome, the role of IDO in cellular senescence and the possible therapeutic role of IDO inhibitors deserve further investigation.


Subject(s)
Cellular Senescence/genetics , Hypoxia/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interleukin-6/genetics , Protein Serine-Threonine Kinases/genetics , Tryptophan/analogs & derivatives , Acute Kidney Injury/drug therapy , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Azo Compounds/pharmacology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Damage/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Humans , Hypoxia/drug therapy , Hypoxia/pathology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mice , Oxygen/metabolism , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Tryptophan/pharmacology , rho GTP-Binding Proteins/genetics
8.
J Am Heart Assoc ; 10(20): e022701, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34622690

ABSTRACT

Background Hyperglycemia is associated with greater hematoma expansion (HE) and worse clinical prognosis after intracerebral hemorrhage (ICH). However, the clinical benefits of intensive glucose normalization remain controversial, and there are no approved therapies for reducing HE. The aryl hydrocarbon receptor (AHR) has been shown to participate in hyperglycemia-induced blood-brain barrier (BBB) dysfunction and brain injury after stroke. Herein, we investigated the role of AHR in hyperglycemia-induced HE in a male mouse model of ICH. Methods and Results CD1 mice (n=387) were used in this study. Mice were subjected to ICH by collagenase injection. Fifty percent dextrose was injected intraperitoneally 3 hours after ICH. AHR knockout clustered regularly interspaced short palindromic repeat was administered intracerebroventricularly to evaluate the role of AHR after ICH. A selective AHR inhibitor, 6,2',4'-trimethoxyflavone, was administered intraperitoneally 2 hours or 6 hours after ICH for outcome study. To evaluate the effect of AHR on HE, 3-methylcholanthrene, an AHR agonist, was injected intraperitoneally 2 hours after ICH. The results showed hyperglycemic ICH upregulated AHR accompanied by greater HE. AHR inhibition provided neurological benefits by restricting HE and preserving BBB function after hyperglycemic ICH. In vivo knockdown of AHR further limited HE and enhanced the BBB integrity. Hyperglycemia directly activated AHR as a physiological stimulus in vivo. The thrombospondin-1/transforming growth factor-ß/vascular endothelial growth factor axis partly participated in AHR signaling after ICH, which inhibited the expressions of BBB-related proteins, ZO-1 and Claudin-5. Conclusions AHR may serve as a potential therapeutic target to attenuate hyperglycemia-induced hematoma expansion and to preserve the BBB in patients with ICH.


Subject(s)
Cerebral Hemorrhage , Hematoma , Hyperglycemia , Receptors, Aryl Hydrocarbon , Animals , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/pathology , Disease Models, Animal , Hematoma/etiology , Hyperglycemia/complications , Male , Mice , Receptors, Aryl Hydrocarbon/antagonists & inhibitors
9.
Acta Biochim Biophys Sin (Shanghai) ; 53(11): 1428-1439, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34508548

ABSTRACT

Bone marrow mesenchymal stromal cells (BMMSCs) are widely sourced and easily amplified in vitro; thus, they have a great potential in the treatment of hemopathies. Recent findings suggested that BMMSCs express the aryl hydrocarbon receptor (AHR). However, few studies have reported on the regulation of proliferative behaviors and metabolism by AHR in BMMSCs. In the present study, we found that activating AHR reduced the proliferation of BMMSCs and enhanced their mitochondrial function, whereas inhibiting AHR exerted the opposite effects. This study may provide the basis for further unveiling the molecular mechanisms and therapeutic potential of AHR in BMMSCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Marrow Cells/metabolism , Cell Cycle/genetics , Mesenchymal Stem Cells/metabolism , Mitochondria/genetics , Receptors, Aryl Hydrocarbon/genetics , Adult , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Cells/cytology , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Proliferation , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation , Healthy Volunteers , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Membrane Potential, Mitochondrial , Mesenchymal Stem Cells/cytology , Middle Aged , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Oxid Med Cell Longev ; 2021: 5530390, 2021.
Article in English | MEDLINE | ID: mdl-34394826

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are toxicants in particulate matter (PM). The vocal fold, part of the larynx and a key structure for voicing, is always in contact with air. In recent epidemic studies, PM was shown to cause laryngitis; however, the basic mechanism has not been evaluated. In the present study, intracellular reactive oxygen species (ROS) and proinflammatory cytokine levels were analyzed after exposing human vocal fold fibroblasts (hVFFs) to PM standard reference material (SRM 2786). Expression levels of the aryl hydrocarbon receptor (AhR) and Cytochrome P450 Family 1 Subfamily A Member 1 (CYP1A1) were also evaluated. PM induced ROS formation and proinflammatory cytokines via the AhR CYP1A1 pathway and caused lipid peroxidation and DNA damage. Blocking AhR or CYP1A1 production using siRNAs significantly decreased ROS production and IL-6 and IL-9 expression in PM-exposed hVFFs, thus protecting the cells against oxidative stress. These results confirm that PAHs in PM play an important role in cell damage and inflammation, confirming a basic pathophysiologic relationship between PM exposure and laryngitis.


Subject(s)
Oxidative Stress/drug effects , Particulate Matter/chemistry , Polycyclic Aromatic Hydrocarbons/toxicity , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Polycyclic Aromatic Hydrocarbons/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Vocal Cords/cytology
11.
J Immunol ; 207(6): 1672-1682, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34417259

ABSTRACT

NK cells are known to be developmentally blocked and functionally inhibited in patients with acute myeloid leukemia (AML), resulting in poor clinical outcomes. In this study, we demonstrate that whereas NK cells are inhibited, closely related type 1 innate lymphoid cells (ILC1s) are enriched in the bone marrow of leukemic mice and in patients with AML. Because NK cells and ILC1s share a common precursor (ILCP), we asked if AML acts on the ILCP to alter developmental potential. A combination of ex vivo and in vivo studies revealed that AML skewing of the ILCP toward ILC1s and away from NK cells represented a major mechanism of ILC1 generation. This process was driven by AML-mediated activation of the aryl hydrocarbon receptor (AHR), a key transcription factor in ILCs, as inhibition of AHR led to decreased numbers of ILC1s and increased NK cells in the presence of AML. These results demonstrate a mechanism of ILC developmental skewing in AML and support further preclinical study of AHR inhibition in restoring normal NK cell development and function in the setting of AML.


Subject(s)
Cell Differentiation/immunology , Immunity, Innate , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , Animals , Azo Compounds/pharmacology , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow/immunology , Carbazoles/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Disease Models, Animal , Female , Humans , Leukemia, Myeloid, Acute/blood , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects
12.
Chem Biodivers ; 18(8): e2100222, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34085382

ABSTRACT

Yinzhihuang oral liquid (YZH) is a traditional Chinese medicine that has been widely used in Asia to prevent and treat neonatal hyperbilirubinemia, but the published preclinical studies on its anti-hyperbilirubinemia effect are conducted in adult animals, partly due to the lack of preclinical neonatal hyperbilirubinemia animal models. In the present study, we tested six reagents to induce hyperbilirubinemia in neonatal rats, and established two appropriate neonatal hyperbilirubinemia rat models by subcutaneous injection of δ-Aminolevulinic acid (ALA, 200 mg/kg) or novobiocin (NOVO, 200 mg/kg). Oral treatment of YZH (80, 160 and 320 mg/kg) significantly decreased serum conjugated bilirubin levels in ALA-treated neonatal rats and serum unconjugated bilirubin levels in NOVO-treated neonatal rats, respectively. Additionally, pre-treatment of YZH also prevented the increase of serum bilirubin levels in both ALA- and NOVO-treated rats. Mechanistically, YZH significantly up-regulated the mRNA expression of genes involved in hepatic bilirubin disposition (organic anion-transporting polypeptide 1b2, Oatp1b2; multidrug resistance-associated protein 2, Mrp2) and bilirubin conjugation (UDP-glucuronosyltransferase 1a1, Ugt1a1). Additionally, YZH up-regulated the mRNA expression of cytochrome P450 1A1 (Cyp1a1), the target gene of aryl hydrocarbon receptor (AhR), and increased the nuclear protein levels of AhR in livers of neonatal rats. YZH and its two active ingredients, namely baicalin (BCL) and 4'-hydroxyacetophenone (4-HT), up-regulated the mRNA expression of AhR target genes (CYP1A1 and UGT1A1) and increased nuclear protein levels of AhR in HepG2 cells. In conclusion, the present study provides two neonatal hyperbilirubinemia animal models and evaluates the anti-hyperbilirubinemia effect and mechanisms of YZH in neonatal animals.


Subject(s)
Drugs, Chinese Herbal/chemistry , Administration, Oral , Aminolevulinic Acid/toxicity , Animals , Animals, Newborn , Bilirubin/blood , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Hep G2 Cells , Humans , Hyperbilirubinemia/chemically induced , Hyperbilirubinemia/drug therapy , Hyperbilirubinemia/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Medicine, Chinese Traditional , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Novobiocin/toxicity , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Up-Regulation/drug effects
13.
Mol Med Rep ; 23(6)2021 06.
Article in English | MEDLINE | ID: mdl-33899121

ABSTRACT

The present study evaluated indoleamine 2,3­dioxygenase 1 (IDO) kinetics and how it affects cell survival during the two distinct phases of ischemia­reperfusion (I­R) injury. Primary renal proximal tubular epithelial cells (RPTECs) were cultured under anoxia or reoxygenation with or without the IDO inhibitor 1­DL­methyltryptophan, the aryl­hydrocarbon receptor (AhR) inhibitor CH223191 or the ferroptosis inhibitor α­tocopherol. Using cell imaging, colorimetric assays, PCR and western blotting, it was demonstrated that IDO was upregulated and induced apoptosis during anoxia. The related molecular pathway entails tryptophan degradation, general control non­derepressible­2 kinase (GCN2K) activation, increased level of phosphorylated eukaryotic translation initiation factor 2α, activating transcription factor (ATF)4, ATF3, C/EBP homologous protein, phosphorylated p53, p53, Bax, death receptor­5 and eventually activated cleaved caspase­3. Reoxygenation also upregulated IDO, which, in this case, induced ferroptosis. The related molecular pathway encompasses kynurenine production, AhR activation, cytochrome p450 enzymes increase, reactive oxygen species generation and eventually ferroptosis. In conclusion, in RPTECs, both anoxia and reoxygenation upregulated IDO, which in turn induced GCN2K­mediated apoptosis and AhR­mediated ferroptosis. Since both phases of I­R injury share IDO upregulation as a common point, its inhibition may prove a useful therapeutic strategy for preventing or attenuating I­R injury.


Subject(s)
Cell Hypoxia , Epithelial Cells/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kidney Tubules, Proximal/cytology , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 4/metabolism , Animals , Apoptosis , Azo Compounds/pharmacology , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Ferroptosis , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism
14.
Nutrients ; 13(4)2021 Apr 03.
Article in English | MEDLINE | ID: mdl-33916690

ABSTRACT

Tryptophan metabolism, via the kynurenine (Kyn) pathway, and microbial transformation of tryptophan to indolic compounds are fundamental for host health; both of which are altered in colon carcinogenesis. Alterations in tryptophan metabolism begin early in colon carcinogenesis as an adaptive mechanism for the tumor to escape immune surveillance and metastasize. The microbial community is a key part of the tumor microenvironment and influences cancer initiation, promotion and treatment response. A growing awareness of the impact of the microbiome on tryptophan (Trp) metabolism in the context of carcinogenesis has prompted this review. We first compare the different metabolic pathways of Trp under normal cellular physiology to colon carcinogenesis, in both the host cells and the microbiome. Second, we review how the microbiome, specifically indoles, influence host tryptophan pathways under normal and oncogenic metabolism. We conclude by proposing several dietary, microbial and drug therapeutic modalities that can be utilized in combination to abrogate tumorigenesis.


Subject(s)
Carcinogenesis/metabolism , Colonic Neoplasms/therapy , Gastrointestinal Microbiome/drug effects , Tryptophan/metabolism , Tumor Escape/drug effects , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinogenesis/drug effects , Carcinogenesis/immunology , Colon/microbiology , Colon/pathology , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/microbiology , Combined Modality Therapy/methods , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gastrointestinal Microbiome/immunology , Host Microbial Interactions/immunology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoles/administration & dosage , Indoles/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Kynurenine/metabolism , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/immunology , Probiotics/administration & dosage , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Symbiosis/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
15.
Cells ; 10(4)2021 03 24.
Article in English | MEDLINE | ID: mdl-33804845

ABSTRACT

Microglia are activated after ischemic stroke and induce neuroinflammation. The expression of the aryl hydrocarbon receptor (AhR) has recently been reported to elicit cytokine expression. We previously reported that microglial activation mediates ischemic edema progression. Thus, the purpose of this study was to examine the role of AhR in inflammation and edema after ischemia using a mouse middle cerebral artery occlusion (MCAO) model. MCAO upregulated AhR expression in microglia during ischemia. MCAO increased the expression of tumor necrosis factor α (TNFα) and then induced edema progression, and worsened the modified neurological severity scores, with these being suppressed by administration of an AhR antagonist, CH223191. In THP-1 macrophages, the NADPH oxidase (NOX) subunit p47phox was significantly increased by AhR ligands, especially under inflammatory conditions. Suppression of NOX activity by apocynin or elimination of superoxide by superoxide dismutase decreased TNFα expression, which was induced by the AhR ligand. AhR ligands also elicited p47phox expression in mouse primary microglia. Thus, p47phox may be important in oxidative stress and subsequent inflammation. In MCAO model mice, P47phox expression was upregulated in microglia by ischemia. Lipid peroxidation induced by MCAO was suppressed by CH223191. Taken together, these findings suggest that AhR in the microglia is involved in neuroinflammation and subsequent edema, after MCAO via p47phox expression upregulation and oxidative stress.


Subject(s)
Brain Edema/etiology , Brain Edema/metabolism , Encephalitis/etiology , Encephalitis/metabolism , Ischemic Stroke/complications , Microglia/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Brain Edema/pathology , Brain Injuries/etiology , Cytochrome P-450 CYP1A1/metabolism , Cytokines/metabolism , Encephalitis/pathology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Infarction, Middle Cerebral Artery/complications , Ligands , Macrophages/metabolism , Male , Mice, Inbred ICR , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidative Stress , Promoter Regions, Genetic/genetics , Protein Subunits/metabolism , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
16.
Anticancer Drugs ; 32(5): 558-566, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33595948

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most prevalent malignant diseases and causes a third of cancer-related death. The prognosis and effective treatment of advanced HCC remains poor in spite of the development of novel therapeutic strategies. In the present study, we investigate anticancer effects of the botanical molecule p-hydroxycinnamic acid (HCA) in the HepG2 liver cancer model in vitro. Culturing with HCA (10-1000 nM) suppressed colony formation and growth of HepG2 cells. Mechanistically, culturing with HCA decreased levels of Ras, PI3K, Akt, MAPK, NF-κB p65 and ß-catenin, which are linked to processes of cell signaling and transcription, and increased levels of retinoblastoma and regucalcin, which are suppressors for carcinogenesis. These alterations may lead to the suppression of cell growth. Furthermore, culturing with HCA (10-1000 nM) stimulated cell death due to increased caspase-3 levels. Interestingly, the effects of HCA on the growth and death of HepG2 cells were inhibited by culturing with CH223191, an antagonist of aryl hydrocarbon receptor (AHR), suggesting that the flavonoid effects are, at least partly, mediated by activation of AHR signaling. Notably, HCA blocked stimulatory effects of Bay K 8644, an agonist of L-type calcium channel, on the growth of HepG2 cells. Thus, our study demonstrates that HCA suppresses the growth and stimulates the death of human liver cancer HepG2 cells in vitro. The botanical molecule HCA may therefore be a useful tool in the treatment of HCC, providing a novel strategy for the therapy of human liver cancers.


Subject(s)
Cell Death/drug effects , Cell Proliferation/drug effects , Coumaric Acids/pharmacology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Azo Compounds/pharmacology , Hep G2 Cells , Humans , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Signal Transduction
17.
EBioMedicine ; 64: 103227, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33530002

ABSTRACT

BACKGROUND: Urolithin A (URA) is an intestinal microbiota metabolic product from ellagitannin-containing foods with multiple biological activities. However, its role in autoimmune diseases is largely unknown. Here, for first time, we demonstrate the therapeutic effect of URA in an experimental autoimmune encephalomyelitis (EAE) animal model. METHODS: Therapeutic effect was evaluated via an active and passive EAE animal model in vivo. The function of URA on bone marrow-derived dendritic cells (BM-DCs), T cells, and microglia were tested in vitro. FINDINGS: Oral URA (25 mg/kg/d) suppressed disease progression at prevention, induction, and effector phases of preclinical EAE. Histological evaluation showed that significantly fewer inflammatory cells, decreased demyelination, lower numbers of M1-type microglia and activated DCs, as well as reduced infiltrating Th1/Th17 cells were present in the central nervous system (CNS) of the URA-treated group. URA treatment at 25 µM inhibited the activation of BM-DCs in vitro, restrained Th17 cell differentiation in T cell polarization conditions, and in a DC-CD4+ T cell co-culture system. Moreover, we confirmed URA inhibited pathogenicity of Th17 cells in adoptive EAE. Mechanism of URA action was directly targeting Aryl Hydrocarbon Receptor (AhR) and modulating the signaling pathways. INTERPRETATION: Collectively, our study offers new evidence that URA, as a human microbial metabolite, is valuable to use as a prospective therapeutic candidate for autoimmune diseases.


Subject(s)
Coumarins/pharmacology , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Biomarkers , Coumarins/chemistry , Disease Models, Animal , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Immunohistochemistry , Mice , Models, Molecular , Molecular Targeted Therapy , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/chemistry , Receptors, Aryl Hydrocarbon/genetics , Structure-Activity Relationship
18.
Neuropharmacology ; 187: 108490, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33607146

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA) is an amphetamine derivative that has been shown to produce serotonergic damage in the brains of primates, including humans, and of rats. Tryptophan, the precursor of serotonin, is primarily degraded through the kynurenine (KYN) pathway, producing among others KYN, the main metabolite of this route. KYN has been reported as an endogenous agonist of the aryl hydrocarbon receptor (AhR), a transcription factor involved in several neurological functions. This study aims to determine the effect of MDMA on the KYN pathway and on AhR activity and to establish their role in the long-term serotonergic neurotoxicity induced by the drug in rats. Our results show that MDMA induces the activation of the KYN pathway, mediated by hepatic tryptophan 2,3-dioxygenase (TDO). MDMA also activated AhR as evidenced by increased AhR nuclear translocation and CYP1B1 mRNA expression. Autoradiographic quantification of serotonin transporters showed that both the TDO inhibitor 680C91 and the AhR antagonist CH-223191 potentiated the neurotoxicity induced by MDMA, while administration of exogenous l-kynurenine or of the AhR positive modulator 3,3'-diindolylmethane (DIM) partially prevented the serotonergic damage induced by the drug. The results demonstrate for the first time that MDMA increases KYN levels and AhR activity, and these changes appear to play a role in limiting the neurotoxicity induced by the drug. This work provides a better understanding of the physiological mechanisms that attenuate the brain damage induced by MDMA and identify modulation of the KYN pathway and of AhR as potential therapeutic strategies to limit the negative effects of MDMA.


Subject(s)
Hippocampus/drug effects , Kynurenine/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Receptors, Aryl Hydrocarbon/drug effects , Serotonin Agents/toxicity , Tryptophan Oxygenase/drug effects , Animals , Autoradiography , Hippocampus/metabolism , Kynurenine/pharmacology , Neurotoxicity Syndromes , Rats , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Serotonin , Serotonin Plasma Membrane Transport Proteins/metabolism , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/metabolism
19.
Curr Opin Immunol ; 70: 15-26, 2021 06.
Article in English | MEDLINE | ID: mdl-33429228

ABSTRACT

Sustained transcriptional activation of the aryl hydrocarbon receptor (AhR) promotes tumour growth and impairs the immune defence, at least for cutaneous melanoma and glioma. AhR ligands are produced by the tumour microenvironment (TME) and by the tumour itself (intracrine). The recent identification of interleukin-4-induced-1 (IL4I1), a parallel pathway to indoleamine 2 3-dioxygenase 1 (IDO1)/ tryptophan 2,3-dioxygenase (TDO), and its ability to generate AhR ligands, confirms that a complete inhibition of AhR ligand production might be difficult to reach. Here, we have focused on recent discoveries explaining the large varieties of AhR ligands and the functional consequences in terms of cancer cell plasticity and consecutive therapy resistance. We also examined therapeutic strategies targeting the AhR signalling pathway and their possible adverse effects. Since the end of 2019, two phase I clinical trials have investigated the ability of the AhR antagonist to 'reset' the immune system and re-sensitize the cancer cells to therapies by preventing their dedifferentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Neoplasms/immunology , Receptors, Aryl Hydrocarbon/immunology , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/immunology , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/immunology
20.
Ecotoxicol Environ Saf ; 210: 111892, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33429317

ABSTRACT

Human activities have generated air pollution, with extremely small particles (PM 2.5, particulate matter less than 2.5 µm in diameter) and liquid droplets, which become a menace to human health. Among the pollutants, polycyclic aromatic hydrocarbons (PAHs), which enhance the risks of pulmonary dysfunction and cancer development, have been extensively studied. Numerous studies have addressed the effects of PAHs on the respiratory system, whereas the effects on lung stem/progenitor cells remain unknown. Here, we provide evidence that benzo[a]pyrene (BaP), a major toxic PAH, induces fibrotic changes with a loss of α-1,6-fucosylation in CD54+CD157+CD45- cells (lung stem cells). In studies with aryl hydrocarbon receptor (AHR) antagonist, we found that these effects by BaP are independent of the canonical AHR pathway. In addition, these BaP-induced fibrotic changes are reduced by TGF-ß antagonist, suggesting an alternative pathway of BaP toxicity is different from other PAH/AHR signaling pathways. Finally, it was observed that BaP impairs the spheroid formation and the podoplanin expression of CD54+CD157+CD45- cells, indicating that BaP suppresses the differentiation of lung stem cells. Taken together, our findings reveal specific BaP-induced injuries in CD54+CD157+CD45- cells.


Subject(s)
Air Pollutants/toxicity , Benzo(a)pyrene/toxicity , Lung/cytology , Stem Cells/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Cell Differentiation/drug effects , Cells, Cultured , Fibrosis , Mice , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Stem Cells/pathology , Transforming Growth Factor beta/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...