1
|
Wiggins BG, Wang YF, Burke A, Grunberg N, Vlachaki Walker JM, Dore M, Chahrour C, Pennycook BR, Sanchez-Garrido J, Vernia S, Barr AR, Frankel G, Birdsey GM, Randi AM, Schiering C. Endothelial sensing of AHR ligands regulates intestinal homeostasis. Nature 2023; 621:821-829. [PMID: 37586410 PMCID: PMC10533400 DOI: 10.1038/s41586-023-06508-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
Endothelial cells line the blood and lymphatic vasculature, and act as an essential physical barrier, control nutrient transport, facilitate tissue immunosurveillance and coordinate angiogenesis and lymphangiogenesis1,2. In the intestine, dietary and microbial cues are particularly important in the regulation of organ homeostasis. However, whether enteric endothelial cells actively sense and integrate such signals is currently unknown. Here we show that the aryl hydrocarbon receptor (AHR) acts as a critical node for endothelial cell sensing of dietary metabolites in adult mice and human primary endothelial cells. We first established a comprehensive single-cell endothelial atlas of the mouse small intestine, uncovering the cellular complexity and functional heterogeneity of blood and lymphatic endothelial cells. Analyses of AHR-mediated responses at single-cell resolution identified tissue-protective transcriptional signatures and regulatory networks promoting cellular quiescence and vascular normalcy at steady state. Endothelial AHR deficiency in adult mice resulted in dysregulated inflammatory responses and the initiation of proliferative pathways. Furthermore, endothelial sensing of dietary AHR ligands was required for optimal protection against enteric infection. In human endothelial cells, AHR signalling promoted quiescence and restrained activation by inflammatory mediators. Together, our data provide a comprehensive dissection of the effect of environmental sensing across the spectrum of enteric endothelia, demonstrating that endothelial AHR signalling integrates dietary cues to maintain tissue homeostasis by promoting endothelial cell quiescence and vascular normalcy.
Collapse
Affiliation(s)
- Benjamin G Wiggins
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
| | - Yi-Fang Wang
- MRC London Institute of Medical Sciences, London, UK
| | - Alice Burke
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Nil Grunberg
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Julia M Vlachaki Walker
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Marian Dore
- MRC London Institute of Medical Sciences, London, UK
| | | | - Betheney R Pennycook
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | | | - Santiago Vernia
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Alexis R Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Gad Frankel
- Department of Life Sciences, Imperial College London, London, UK
| | - Graeme M Birdsey
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Anna M Randi
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Chris Schiering
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
| |
Collapse
|
2
|
Palzkill VR, Tan J, Yang Q, Morcos J, Laitano O, Ryan TE. Activation of the Aryl Hydrocarbon Receptor in Endothelial Cells Impairs Ischemic Angiogenesis in Chronic Kidney Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550410. [PMID: 37546909 PMCID: PMC10401998 DOI: 10.1101/2023.07.24.550410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Rationale Chronic kidney disease (CKD) is a strong risk factor for peripheral artery disease (PAD) that is associated with worsened clinical outcomes. CKD leads to accumulation of tryptophan metabolites that associate with adverse limb events in PAD and are ligands of the aryl hydrocarbon receptor (AHR) which may regulate ischemic angiogenesis. Objectives To test if endothelial cell-specific deletion of the AHR (AHRecKO) alters ischemic angiogenesis and limb function in mice with CKD subjected to femoral artery ligation. Findings Male AHRecKO mice with CKD displayed better limb perfusion recovery and enhanced ischemic angiogenesis compared to wildtype mice with CKD. However, the improved limb perfusion did not result in better muscle performance. In contrast to male mice, deletion of the AHR in female mice with CKD had no impact on perfusion recovery or angiogenesis. Using primary endothelial cells from male and female mice, treatment with indoxyl sulfate uncovered sex-dependent differences in AHR activating potential and RNA sequencing revealed wide ranging sex-differences in angiogenic signaling pathways. Conclusion Endothelium-specific deletion of the AHR improved ischemic angiogenesis in male, but not female, mice with CKD. There are sex-dependent differences in Ahr activating potential within endothelial cells that are independent of sex hormones.
Collapse
Affiliation(s)
- Victoria R. Palzkill
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Qingping Yang
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Juliana Morcos
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
| |
Collapse
|
3
|
Inhibitory Effects of 3-Methylcholanthrene Exposure on Porcine Oocyte Maturation. Int J Mol Sci 2023; 24:ijms24065567. [PMID: 36982641 PMCID: PMC10058619 DOI: 10.3390/ijms24065567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/05/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
3-methylcholanthrene (3-MC) is a highly toxic environmental pollutant that impairs animal health. 3-MC exposure can cause abnormal spermatogenesis and ovarian dysfunction. However, the effects of 3-MC exposure on oocyte maturation and embryo development remain unclear. This study revealed the toxic effects of 3-MC exposure on oocyte maturation and embryo development. 3-MC with different concentrations of 0, 25, 50, and 100 μM was applied for in vitro maturation of porcine oocytes. The results showed that 100 μM 3-MC significantly inhibited cumulus expansion and the first polar body extrusion. The rates of cleavage and blastocyst of embryos derived from 3-MC-exposed oocytes were significantly lower than those in the control group. Additionally, the rates of spindle abnormalities and chromosomal misalignments were higher than those in the control group. Furthermore, 3-MC exposure not only decreased the levels of mitochondria, cortical granules (CGs), and acetylated α-Tubulin, but also increased the levels of reactive oxygen species (ROS), DNA damage, and apoptosis. The expression of cumulus expansion and apoptosis-related genes was abnormal in 3-MC-exposed oocytes. In conclusion, 3-MC exposure disrupted the nuclear and cytoplasmic maturation of porcine oocytes through oxidative stress.
Collapse
|
4
|
Correia MJ, Pimpão AB, Lopes-Coelho F, Sequeira CO, Coelho NR, Gonçalves-Dias C, Barouki R, Coumoul X, Serpa J, Morello J, Monteiro EC, Pereira SA. Aryl Hydrocarbon Receptor and Cysteine Redox Dynamics Underlie (Mal)adaptive Mechanisms to Chronic Intermittent Hypoxia in Kidney Cortex. Antioxidants (Basel) 2021; 10:antiox10091484. [PMID: 34573115 PMCID: PMC8469308 DOI: 10.3390/antiox10091484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/31/2022] Open
Abstract
We hypothesized that an interplay between aryl hydrocarbon receptor (AhR) and cysteine-related thiolome at the kidney cortex underlies the mechanisms of (mal)adaptation to chronic intermittent hypoxia (CIH), promoting arterial hypertension (HTN). Using a rat model of CIH-HTN, we investigated the impact of short-term (1 and 7 days), mid-term (14 and 21 days, pre-HTN), and long-term intermittent hypoxia (IH) (up to 60 days, established HTN) on CYP1A1 protein level (a sensitive hallmark of AhR activation) and cysteine-related thiol pools. We found that acute and chronic IH had opposite effects on CYP1A1 and the thiolome. While short-term IH decreased CYP1A1 and increased protein-S-thiolation, long-term IH increased CYP1A1 and free oxidized cysteine. In addition, an in vitro administration of cystine, but not cysteine, to human endothelial cells increased Cyp1a1 expression, supporting cystine as a putative AhR activator. This study supports CYP1A1 as a biomarker of obstructive sleep apnea (OSA) severity and oxidized pools of cysteine as risk indicator of OSA-HTN. This work contributes to a better understanding of the mechanisms underlying the phenotype of OSA-HTN, mimicked by this model, which is in line with precision medicine challenges in OSA.
Collapse
Affiliation(s)
- Maria João Correia
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - António B. Pimpão
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Filipa Lopes-Coelho
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Catarina O. Sequeira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Nuno R. Coelho
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Clara Gonçalves-Dias
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Robert Barouki
- INSERM UMR-S 1124, 3TS, Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, Université de Paris, 45 rue des Saints-Pères, 75006 Paris, France; (R.B.); (X.C.)
| | - Xavier Coumoul
- INSERM UMR-S 1124, 3TS, Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, Université de Paris, 45 rue des Saints-Pères, 75006 Paris, France; (R.B.); (X.C.)
| | - Jacinta Serpa
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Judit Morello
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Emília C. Monteiro
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Sofia A. Pereira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
- Correspondence:
| |
Collapse
|
5
|
Ravid JD, Kamel MH, Chitalia VC. Uraemic solutes as therapeutic targets in CKD-associated cardiovascular disease. Nat Rev Nephrol 2021; 17:402-416. [PMID: 33758363 DOI: 10.1038/s41581-021-00408-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2021] [Indexed: 02/01/2023]
Abstract
Chronic kidney disease (CKD) is characterized by the retention of a myriad of solutes termed uraemic (or uremic) toxins, which inflict damage to several organs, including the cardiovascular system. Uraemic toxins can induce hallmarks of cardiovascular disease (CVD), such as atherothrombosis, heart failure, dysrhythmias, vessel calcification and dysregulated angiogenesis. CVD is an important driver of mortality in patients with CKD; however, reliance on conventional approaches to managing CVD risk is insufficient in these patients, underscoring a need to target risk factors that are specific to CKD. Mounting evidence suggests that targeting uraemic toxins and/or pathways induced by uraemic toxins, including tryptophan metabolites and trimethylamine N-oxide (TMAO), can lower the risk of CVD in patients with CKD. Although tangible therapies resulting from our growing knowledge of uraemic toxicity are yet to materialize, a number of pharmacological and non-pharmacological approaches have the potential to abrogate the effects of uraemic toxins, for example, by decreasing the production of uraemic toxins, by modifying metabolic pathways induced by uraemic toxins such as those controlled by aryl hydrocarbon receptor signalling and by augmenting the clearance of uraemic toxins.
Collapse
Affiliation(s)
- Jonathan D Ravid
- School of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Mohamed Hassan Kamel
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Vipul C Chitalia
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA. .,Boston Veterans Affairs Healthcare System, Boston, MA, USA. .,Global Co-creation Lab, Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
6
|
Salyers ZR, Coleman M, Balestrieri NP, Ryan TE. Indoxyl sulfate impairs angiogenesis via chronic aryl hydrocarbon receptor activation. Am J Physiol Cell Physiol 2021; 320:C240-C249. [PMID: 33406025 DOI: 10.1152/ajpcell.00262.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic kidney disease (CKD) is associated with a substantial increased risk of cardiovascular disease. There is growing evidence that uremic metabolites, which accumulate in the blood with CKD, have detrimental impacts on endothelial cell health and function. However, the molecular mechanisms by which uremic metabolites negatively impact endothelial cell biology are not fully understood. In this study, activation of the aryl hydrocarbon receptor (AHR) via indoxyl sulfate, a known uremic metabolite, was found to impair endothelial cell tube formation and proliferation but not migratory function. Moreover, aortic ring cultures treated with indoxyl sulfate also exhibited decreased sprouting and high AHR activation. Next, genetic knockdown of the AHR using shRNA was found to rescue endothelial cell tube formation, proliferation, and aortic ring sprouting. Similarly, pharmacological AHR antagonism using resveratrol and CH223191 were also found to rescue angiogenesis in cell and aortic ring cultures. Finally, a constitutively active AHR (CAAHR) vector was generated and used to confirm AHR-specific effects. Expression of the CAAHR recapitulated the impaired tube formation and proliferation in cultured endothelial cells and decreased sprouting in aortic ring cultures. Taken together, these data define the impact of AHR activation on angiogenesis and highlight the potential for therapeutic AHR antagonists, which may improve angiogenesis in the context of CKD and cardiovascular disease.
Collapse
Affiliation(s)
- Zachary R Salyers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Madeline Coleman
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Nicholas P Balestrieri
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida.,Center for Exercise Science, University of Florida, Gainesville, Florida
| |
Collapse
|
7
|
Li Y, Zhou C, Lei W, Wang K, Zheng J. Roles of aryl hydrocarbon receptor in endothelial angiogenic responses†. Biol Reprod 2020; 103:927-937. [PMID: 32716482 PMCID: PMC7731988 DOI: 10.1093/biolre/ioaa128] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 12/23/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR) is a transcription factor, which can be activated by a plethora of structure-diverse ligands. Historically, AhR is known for its involvements in regulation of metabolism of xenobiotics. However, normal physiological roles of AhR have been defined in other essential biological processes, including vascular growth and function, reproduction, and immunoresponses. In contrast, aberrant expression and activation of the AhR signaling pathway occur in a variety of human diseases, many of which (e.g., preeclampsia, atherosclerosis, and hypertension) could be associated with endothelial dysfunction. Indeed, emerging evidence has shown that either exogenous or endogenous AhR ligands can induce endothelial dysfunction in either an AhR-dependent or AhR-independent manner, possibly reliant on the blood vessel origin (artery and vein) of endothelial cells. Given that the AhR signaling pathway has broad impacts on endothelial and cardiovascular function, AhR ligands, AhR, and their downstream genes could be considered novel therapeutic targets for those endothelial-related diseases. This review will discuss the current knowledge of AhR's mediation on endothelial function and potential mechanisms underlying these actions with a focus on placental endothelial cells.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chi Zhou
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Wei Lei
- Department of Cardiovascular Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Kai Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Cardiovascular Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
8
|
Yamaguchi M, Hankinson O. An aryl hydrocarbon receptor agonist suppresses the growth of human umbilical vein endothelial cells in vitro: Potent effect with polyunsaturated fatty acids. Int J Exp Pathol 2020; 101:248-263. [PMID: 32985761 DOI: 10.1111/iep.12373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/27/2020] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
Human umbilical vein endothelial cells (HUVECs) are a pivotal component of the hematopoietic microenvironment linked to the modulation of the immune response, inflammation and carcinogenesis. HUVEC expresses the aryl hydrocarbon receptor (AHR), which regulates gene expression by binding to the xenobiotic-responsive element. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent agonist for AHR signalling. Treatment with TCDD (0.1-100 nmol/L) was found to suppress the proliferation and to stimulate the death of HUVEC. TCDD's effects were abolished by culturing with CH223191, an inhibitor of AHR signalling. Mechanistically, TCDD treatment increased the protein levels of cell growth suppressors, including p53, Rb, p21 and regucalcin, and caspase-3 implicated in apoptotic cell death, and decreased the levels of Stat3, mitogen-activated protein kinase (MAPK/Erk1/2) and phospho-MAPK/Erk1/2. Treatment with polyunsaturated fatty acids (PUFAs), including docosahexaenoic acid, eicosapentaenoic acid and arachidonic acid, suppressed the proliferation and stimulated the death of HUVEC in vitro, and decreased the levels of Stat3, MAPK/Erk1/2 and phospho-MAPK/Erk1/2 and increased caspase-3. Notably, the effects of TCDD in suppressing proliferation and stimulating death of HUVEC were modulated by coculturing with PUFAs. These effects were reversed by treatment with CH223191, an inhibitor of AHR. Treatment with both TCDD and PUFAs collaboratively enhanced the levels of AHR, CYP1A1, p53, p21, Rb and regucalcin. Moreover, TCDD suppressed migration with wound healing of HUVEC. Notably, the combination of TCDD and PUFAs revealed potent suppressive effects on angiogenesis of HUVEC, potentially related to disorders of the stromal microenvironment.
Collapse
Affiliation(s)
- Masayoshi Yamaguchi
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Oliver Hankinson
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.,Molecular Toxicology Program, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| |
Collapse
|
9
|
Bock KW. Human AHR functions in vascular tissue: Pro- and anti-inflammatory responses of AHR agonists in atherosclerosis. Biochem Pharmacol 2018; 159:116-120. [PMID: 30508524 DOI: 10.1016/j.bcp.2018.11.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023]
Abstract
Despite decades of intense research physiologic aryl hydrocarbon receptor (AHR) functions have not been elucidated. Challenges include marked species differences and dependence on cell type and cellular context. A previous commentary on human AHR functions in skin and intestine has been extended to vascular tissue. Similar functions appear to be operating in vascular tissue including microbial defense, modulation of stem/progenitor cells as well as control of immunity and inflammation. However, AHR functions are Janus faced: Detrimental AHR functions in vascular tissue are well documented, e.g., upon exposure to polycyclic aromatic hydrocarbons in cigarette smoke leading to oxidative stress and generation of oxidized LDL. Modified LDL particles accumulate in macrophages and smooth muscle-derived pro-inflammatory foam cells, the hallmark of atherosclerosis. On the other hand, numerous anti-inflammatory AHR agonists have been identified including bilirubin and quercetin. Mechanisms as to how AHR produces pro- and anti-inflammatory responses in the vascular system need further investigation.
Collapse
Affiliation(s)
- Karl Walter Bock
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstrasse 56, D-72074 Tübingen, Germany.
| |
Collapse
|
10
|
Pang LP, Li Y, Zou QY, Zhou C, Lei W, Zheng J, Huang SA. ITE inhibits growth of human pulmonary artery endothelial cells. Exp Lung Res 2018; 43:283-292. [PMID: 29140133 DOI: 10.1080/01902148.2017.1367868] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIM Pulmonary arterial hypertension (PAH), a deadly disorder is associated with excessive growth of human pulmonary artery endothelial (HPAECs) and smooth muscle (HPASMCs) cells. Current therapies primarily aim at promoting vasodilation, which only ameliorates clinical symptoms without a cure. 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) is an endogenous aryl hydrocarbon receptor (AhR) ligand, and mediates many cellular function including cell growth. However, the roles of ITE in human lung endothelial cells remain elusive. Herein, we tested a hypothesis that ITE inhibits growth of human pulmonary artery endothelial cells via AhR. MATERIALS AND METHODS Immunohistochemistry was performed to localize AhR expression in human lung tissues. The crystal violet method and MTT assay were used to determine ITE's effects on growth of HPAECs. The AhR activation in HPAECs was confirmed using Western blotting and RT-qPCR. The role of AhR in ITE-affected proliferation of HPAECs was assessed using siRNA knockdown method followed by the crystal violet method. RESULTS Immunohistochemistry revealed that AhR was present in human lung tissues, primarily in endothelial and smooth muscle cells of pulmonary veins and arteries, as well as in bronchial and alveolar sac epithelia. We also found that ITE dose- and time-dependently inhibited proliferation of HPAECs with a maximum inhibition of 83% at 20 µM after 6 days of treatment. ITE rapidly decreased AhR protein levels, while it increased mRNA levels of cytochrome P450 (CYP), family 1, member A1 (CYP1A1) and B1 (CYP1B1), indicating activation of the AhR/CYP1A1 and AhR/CYP1B1 pathways in HPAECs. The AhR siRNA significantly suppressed AhR protein expression, whereas it did not significantly alter ITE-inhibited growth of HPAECs. CONCLUSIONS ITE suppresses growth of HPAECs independent of AhR, suggesting that ITE may play an important role in preventing excessive growth of lung endothelial cells.
Collapse
Affiliation(s)
- Ling-Pin Pang
- a Cardiovascular Medicine Center , Affiliated Hospital of Guangdong Medical University , Zhanjiang , Guangdong , P.R. China.,b Department of Obstetrics and Gynecology , University of Wisconsin-Madison , Madison , WI , USA
| | - Yan Li
- b Department of Obstetrics and Gynecology , University of Wisconsin-Madison , Madison , WI , USA
| | - Qing-Yun Zou
- b Department of Obstetrics and Gynecology , University of Wisconsin-Madison , Madison , WI , USA
| | - Chi Zhou
- b Department of Obstetrics and Gynecology , University of Wisconsin-Madison , Madison , WI , USA
| | - Wei Lei
- a Cardiovascular Medicine Center , Affiliated Hospital of Guangdong Medical University , Zhanjiang , Guangdong , P.R. China
| | - Jing Zheng
- a Cardiovascular Medicine Center , Affiliated Hospital of Guangdong Medical University , Zhanjiang , Guangdong , P.R. China.,b Department of Obstetrics and Gynecology , University of Wisconsin-Madison , Madison , WI , USA
| | - Shi-An Huang
- a Cardiovascular Medicine Center , Affiliated Hospital of Guangdong Medical University , Zhanjiang , Guangdong , P.R. China
| |
Collapse
|
11
|
Aryl Hydrocarbon Receptor: A New Player of Pathogenesis and Therapy in Cardiovascular Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6058784. [PMID: 29984241 PMCID: PMC6015699 DOI: 10.1155/2018/6058784] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 03/14/2018] [Accepted: 04/05/2018] [Indexed: 01/04/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a DNA binding protein that acts as a nuclear receptor mediating xenobiotic metabolism and environmental responses. Owing to the evolutionary conservation of this gene and its widespread expression in the immune and circulatory systems, AhR has for many years been almost exclusively studied by the pharmacological/toxicological field for its role in contaminant toxicity. More recently, the functions of AhR in environmental adaption have been examined in the context of the occurrence, development, and therapy of cardiovascular diseases. Increasing evidence suggests that AhR is involved in maintaining homeostasis or in triggering pathogenesis by modulating the biological responses of critical cell types in the cardiovascular system. Here, we describe the structure, distribution, and ligands of AhR and the AhR signaling pathway and review the impact of AhR on cardiovascular physiology. We also discuss the potential contribution of AhR as a new potential factor in the targeted treatment of cardiovascular diseases.
Collapse
|
12
|
Li Y, Wang K, Zou QY, Jiang YZ, Zhou C, Zheng J. ITE Suppresses Angiogenic Responses in Human Artery and Vein Endothelial Cells: Differential Roles of AhR. Reprod Toxicol 2017; 74:181-188. [PMID: 28986273 DOI: 10.1016/j.reprotox.2017.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/22/2017] [Accepted: 09/27/2017] [Indexed: 12/26/2022]
Abstract
Aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor is involved in regulation of many essential biological processes including vascular development and angiogenesis. 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) is an AhR ligand, which regulates immune responses and cancer cell growth. However, the roles of the ITE/AhR pathway in mediating placental angiogenesis remains elusive. Here, we determined if ITE affected placental angiogenic responses via AhR in human umbilical vein (HUVECs) and artery endothelial (HUAECs) cells in vitro. We observed that ITE dose- and time-dependently inhibited proliferation and viability of HUAECs and HUVECs, whereas it inhibited migration of HUAECs, but not HUVECs. While AhR siRNA significantly suppressed AhR protein expression in HUVECs and HUAECs, it attenuated the ITE-inhibited angiogenic responses of HUAECs, but not HUVECs. Collectively, ITE suppressed angiogenic responses of HUAECs and HUVECs, dependent and independent of AhR, respectively. These data suggest that ITE may regulate placental angiogenesis.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Kai Wang
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Qing-Yun Zou
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Yi-Zhou Jiang
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Chi Zhou
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States; Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China.
| |
Collapse
|
13
|
Hexachlorobenzene promotes angiogenesis in vivo, in a breast cancer model and neovasculogenesis in vitro, in the human microvascular endothelial cell line HMEC-1. Toxicol Lett 2015; 239:53-64. [DOI: 10.1016/j.toxlet.2015.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/31/2015] [Accepted: 09/03/2015] [Indexed: 12/13/2022]
|
14
|
The aryl hydrocarbon receptor links integrin signaling to the TGF-β pathway. Oncogene 2015; 35:3260-71. [PMID: 26500056 DOI: 10.1038/onc.2015.387] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 08/31/2015] [Accepted: 09/08/2015] [Indexed: 01/02/2023]
Abstract
Glioblastoma is the most common and aggressive form of intrinsic brain tumor. Transforming growth factor (TGF)-β represents a central mediator of the malignant phenotype of these tumors by promoting invasiveness and angiogenesis, maintaining tumor cell stemness and inducing profound immunosuppression. Integrins, which are highly expressed in glioma cells, interact with the TGF-β pathway. Furthermore, a link has been described between activity of the transcription factor aryl hydrocarbon receptor (AhR) and TGF-β expression. Here we demonstrate that integrin inhibition, using αv, β3 or β5 neutralizing antibodies, RNA interference-mediated integrin gene silencing or pharmacological inhibition by the cyclic RGD peptide EMD 121974 (cilengitide) or the non-peptidic molecule GLPG0187, inhibits AhR activity. These effects are independent of cell detachment or cell density. While AhR mRNA expression was not affected by integrin inhibition, AhR total and nuclear protein levels were reduced, suggesting that integrin inhibition-mediated regulation of AhR may occur at a post-transcriptional level. AhR-null astrocytes, AhR-null hepatocytes or glioblastoma cells with a transiently silenced AhR gene showed reduced sensitivity to integrin inhibition-mediated alterations in TGF-β signaling, indicating that AhR mediates integrin control of the TGF-β pathway. Accordingly, there was a significant correlation of αv integrin levels with nuclear AhR and pSmad2 levels as determined by immunohistochemistry in human glioblastoma in vivo. In summary, this study identifies a signaling network comprising integrins, AhR and TGF-β and validates integrin inhibition as a promising strategy not only to inhibit angiogenesis, but also to block AhR- and TGF-β-controlled features of malignancy in human glioblastoma.
Collapse
|
15
|
Li Y, Wang K, Zou QY, Magness RR, Zheng J. 2,3,7,8-Tetrachlorodibenzo-p-dioxin differentially suppresses angiogenic responses in human placental vein and artery endothelial cells. Toxicology 2015; 336:70-8. [PMID: 26275813 DOI: 10.1016/j.tox.2015.08.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/06/2015] [Accepted: 08/09/2015] [Indexed: 10/23/2022]
Abstract
Placental angiogenesis is dramatically increased during pregnancy in association with the elevated placental blood flows to support the rapidly growing fetus. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is an environmental toxicant and a ligand of aryl hydrocarbon receptor (AhR). Herein, we investigated the effects of TCDD on proliferation, migration, and viability of fetoplacental endothelial cells in response to a complete growth medium which contained serum and growth supplement using human umbilical cord vein (HUVECs) and artery (HUAECs) cells as models. We found that TCDD dose- and time-dependently inhibited (p < 0.05) proliferation of HUVECs and HUAECs. Treatment with TCDD at 10 nM for 6 days inhibited (p < 0.05) migration (by ∼ 30%) of HUAECs, but not HUVECs. TCDD at 10nM also decreased (p < 0.05) viability of HUVECs and HUAECs. Interestingly, specific AhR siRNA blocked (p < 0.05) the TCDD-inhibited cellular responses in HUAECs, but not HUVECs. Nonetheless, TCDD at 10nM neither affected the cell cycle progression, nor did it induce cell apoptosis in HUVECs and HUAECs. In addition, TCDD at 10 nM also did not alter activation of ERK1/2 and AKT1 in HUVECs and HUAECs. Collectively, TCDD suppresses proliferation and/or migration (two key steps of angiogenesis) of HUVECs and HUAECs independent and dependent of AhR, respectively. These data suggest that TCDD inhibited growth of HUVECs and HUAECs via decreasing cell viability. Thus, TCDD may inhibit fetoplacental angiogenesis, leading to negative pregnancy outcomes.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Kai Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, PR China
| | - Qing-Yun Zou
- Department of Obstetrics and Gynecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Ronald R Magness
- Department of Obstetrics and Gynecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI 53715, USA; Departments of Pediatrics, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jing Zheng
- Department of Obstetrics and Gynecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Cardiovascular Medicine, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, Guangdong, PR China.
| |
Collapse
|
16
|
Park HR, Lee SE, Yang H, Son GW, Park YS. Functional screening of altered microRNA expression in 3-methylcholanthrene-treated human umbilical vein endothelial cells. BIOCHIP JOURNAL 2014. [DOI: 10.1007/s13206-014-8403-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
17
|
Fukasawa K, Kagaya S, Maruyama S, Kuroiwa S, Masuda K, Kameyama Y, Satoh Y, Akatsu Y, Tomura A, Nishikawa K, Horie S, Ichikawa YI. A novel compound, NK150460, exhibits selective antitumor activity against breast cancer cell lines through activation of aryl hydrocarbon receptor. Mol Cancer Ther 2014; 14:343-54. [PMID: 25522763 DOI: 10.1158/1535-7163.mct-14-0158] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antiestrogen agents are commonly used to treat patients with estrogen receptor (ER)-positive breast cancer. Tamoxifen has been the mainstay of endocrine treatment for patients with early and advanced breast cancer for many years. Following tamoxifen treatment failure, however, there are still limited options for subsequent hormonal therapy. We discovered a novel compound, NK150460, that inhibits 17β-estradiol (E2)-dependent transcription without affecting binding of E2 to ER. Against our expectations, NK150460 inhibited growth of not only most ER-positive, but also some ER-negative breast cancer cell lines, while never inhibiting growth of non-breast cancer cell lines. Cell-based screening using a random shRNA library, identified aryl hydrocarbon receptor nuclear translocator (ARNT) as a key gene involved in NK150460's antitumor mechanism. siRNAs against not only ARNT but also its counterpart aryl hydrocarbon receptor (AhR) and their target protein, CYP1A1, dramatically abrogated NK150460's growth-inhibitory activity. This suggests that the molecular cascade of AhR/ARNT plays an essential role in NK150460's antitumor mechanism. Expression of ERα was decreased by NK150460 treatment, and this was inhibited by an AhR antagonist. Unlike two other AhR agonists now undergoing clinical developmental stage, NK150460 did not induce histone H2AX phosphorylation or p53 expression, suggesting that it did not induce a DNA damage response in treated cells. Cell lines expressing epithelial markers were more sensitive to NK150460 than mesenchymal marker-expressing cells. These data indicate that NK150460 is a novel AhR agonist with selective antitumor activity against breast cancer cell lines, and its features differ from those of the other two AhR agonists.
Collapse
Affiliation(s)
- Kazuteru Fukasawa
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan. Department of Urology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan.
| | - Shigehide Kagaya
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| | - Sakiko Maruyama
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| | - Shunsuke Kuroiwa
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| | - Kuniko Masuda
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| | - Yoshio Kameyama
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| | - Yoshitaka Satoh
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| | - Yuichi Akatsu
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| | - Arihiro Tomura
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| | - Kiyohiro Nishikawa
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| | - Shigeo Horie
- Department of Urology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Yuh-ichiro Ichikawa
- Pharmaceutical Research Laboratories, Research and Development Group, Nippon Kayaku Co., Ltd., Kita-ku, Tokyo, Japan
| |
Collapse
|
18
|
Li Y, Wang K, Jiang YZ, Chang XW, Dai CF, Zheng J. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) inhibits human ovarian cancer cell proliferation. Cell Oncol (Dordr) 2014; 37:429-37. [PMID: 25404385 DOI: 10.1007/s13402-014-0206-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2014] [Indexed: 01/06/2023] Open
Abstract
PURPOSE The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, mediates a broad spectrum of biological processes, including ovarian growth and ovulation. Recently, we found that an endogenous AhR ligand (ITE) can inhibit ovarian cancer proliferation and migration via the AhR. Here, we tested whether 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, an exogenous AhR ligand) may exert similar anti-ovarian cancer activities using human ovarian cancer and non-cancerous human ovarian surface epithelial cells. METHODS Two human ovarian cancer cell lines (SKOV-3 and OVCAR-3) and one human ovarian surface epithelial cell line (IOSE-385) were used. Cell proliferation and migration activities were determined using crystal violet and FluoroBlok insert system assays, respectively. AhR protein expression was assessed by Western blotting. Expression of cytochrome P450, family 1, member A1 (CYP1A1) and member B1 (CYP1B1) mRNA was assessed by qPCR. Small interfering RNAs (siRNAs) were used to knock down AhR expression. RESULTS We found that TCDD dose-dependently suppressed OVCAR-3 cell proliferation, with a maximum effect (~70% reduction) at 100 nM. However, TCDD did not affect SKOV-3 and IOSE-385 cell proliferation and migration. The estimated IC50 of TCDD for inhibiting OVCAR-3 cell proliferation was 4.6 nM. At 10 nM, TCDD time-dependently decreased AhR protein levels, while it significantly increased CYP1A1 and CYP1B1 mRNA levels in SKOV-3, OVCAR-3 and IOSE-385 cells, indicating activation of AhR signaling. siRNA-mediated AhR knockdown readily blocked TCDD-mediated suppression of OVCAR-3 cell proliferation. CONCLUSION Our data indicate that TCDD can suppress human ovarian cancer cell proliferation via the AhR signaling pathway and that TCDD exhibits an anti-proliferative activity in at least a subset of human ovarian cancer cells.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, University of Wisconsin, 202 S. Park St., Madison, WI, 53715, USA
| | | | | | | | | | | |
Collapse
|
19
|
Hsu YH, Chang CC, Yang NJ, Lee YH, Juan SH. RhoA-Mediated Inhibition of Vascular Endothelial Cell Mobility: Positive Feedback Through Reduced Cytosolic p21 and p27. J Cell Physiol 2014; 229:1455-65. [DOI: 10.1002/jcp.24583] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/14/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Yung-Ho Hsu
- Department of Nephrology; Taipei Medical University-Shuan-Ho Hospital; Taipei Taiwan
| | - Chih-Cheng Chang
- Graduate Institute of Medical Sciences; Taipei Medical University; Taipei Taiwan
- Department of Physiology, School of Medicine, College of Medicine; Taipei Medical University; Taipei Taiwan
| | - Nian-Jie Yang
- Graduate Institute of Medical Sciences; Taipei Medical University; Taipei Taiwan
- Department of Physiology, School of Medicine, College of Medicine; Taipei Medical University; Taipei Taiwan
| | - Yi-Hsuan Lee
- Institute of Physiology; National Yang-Ming University; Taipei Taiwan
| | - Shu-Hui Juan
- Graduate Institute of Medical Sciences; Taipei Medical University; Taipei Taiwan
- Department of Physiology, School of Medicine, College of Medicine; Taipei Medical University; Taipei Taiwan
| |
Collapse
|
20
|
Microarray analysis of gene expression in 3-methylcholanthrene-treated human endothelial cells. Mol Cell Toxicol 2014. [DOI: 10.1007/s13273-014-0003-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
21
|
Chang CC, Sue YM, Yang NJ, Lee YH, Juan SH. 3-Methylcholanthrene, an AhR agonist, caused cell-cycle arrest by histone deacetylation through a RhoA-dependent recruitment of HDAC1 and pRb2 to E2F1 complex. PLoS One 2014; 9:e92793. [PMID: 24658119 PMCID: PMC3962457 DOI: 10.1371/journal.pone.0092793] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 02/25/2014] [Indexed: 12/15/2022] Open
Abstract
We previously showed that treating vascular endothelial cells with 3-methylcholanthrene (3MC) caused cell-cycle arrest in the Go/G1 phase; this resulted from the induction of p21 and p27 and a decreased level and activity of the cyclin-dependent kinase, Cdk2. We further investigated the molecular mechanisms that modulate cell-cycle regulatory proteins through the aryl-hydrocarbon receptor (AhR)/Ras homolog gene family, member A (RhoA) dependent epigenetic modification of histone. AhR/RhoA activation mediated by 3MC was essential for the upregulation of retinoblastoma 2 (pRb2) and histone deacetylase 1 (HDAC1), whereas their nuclear translocation was primarily modulated by RhoA activation. The combination of increased phosphatase and tensin homolog (PTEN) activity and decreased phosphatidylinositide 3-kinase (PI3K) activation by 3MC led to the inactivation of the Ras-cRaf pathway, which contributed to pRb2 hypophosphorylation. Increased HDAC1/pRb2 recruitment to the E2F1 complex decreased E2F1-transactivational activity and H3/H4 deacetylation, resulting in the downregulation of cell-cycle regulatory proteins (Cdk2/4 and Cyclin D3/E). Co-immunoprecipitation and electrophoretic mobility shift assay (EMSA) results showed that simvastatin prevented the 3MC-increased binding activities of E2F1 proteins in their promoter regions. Additionally, RhoA inhibitors (statins) reversed the effect of 3MC in inhibiting DNA synthesis by decreasing the nuclear translocation of pRb2/HDAC1, leading to a recovery of the levels of cell-cycle regulatory proteins. In summary, 3MC decreased cell proliferation by the epigenetic modification of histone through an AhR/RhoA-dependent mechanism that can be rescued by statins.
Collapse
Affiliation(s)
- Chih-Cheng Chang
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Yuh-Mou Sue
- Department of Nephrology, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| | - Nian-Jie Yang
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Institute of Physiology, National Yang-Ming University, Taipei, Taiwan
| | - Shu-Hui Juan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
22
|
Sallée M, Dou L, Cerini C, Poitevin S, Brunet P, Burtey S. The aryl hydrocarbon receptor-activating effect of uremic toxins from tryptophan metabolism: a new concept to understand cardiovascular complications of chronic kidney disease. Toxins (Basel) 2014; 6:934-49. [PMID: 24599232 PMCID: PMC3968369 DOI: 10.3390/toxins6030934] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 12/13/2022] Open
Abstract
Patients with chronic kidney disease (CKD) have a higher risk of cardiovascular diseases and suffer from accelerated atherosclerosis. CKD patients are permanently exposed to uremic toxins, making them good candidates as pathogenic agents. We focus here on uremic toxins from tryptophan metabolism because of their potential involvement in cardiovascular toxicity: indolic uremic toxins (indoxyl sulfate, indole-3 acetic acid, and indoxyl-β-d-glucuronide) and uremic toxins from the kynurenine pathway (kynurenine, kynurenic acid, anthranilic acid, 3-hydroxykynurenine, 3-hydroxyanthranilic acid, and quinolinic acid). Uremic toxins derived from tryptophan are endogenous ligands of the transcription factor aryl hydrocarbon receptor (AhR). AhR, also known as the dioxin receptor, interacts with various regulatory and signaling proteins, including protein kinases and phosphatases, and Nuclear Factor-Kappa-B. AhR activation by 2,3,7,8-tetrachlorodibenzo-p-dioxin and some polychlorinated biphenyls is associated with an increase in cardiovascular disease in humans and in mice. In addition, this AhR activation mediates cardiotoxicity, vascular inflammation, and a procoagulant and prooxidant phenotype of vascular cells. Uremic toxins derived from tryptophan have prooxidant, proinflammatory, procoagulant, and pro-apoptotic effects on cells involved in the cardiovascular system, and some of them are related with cardiovascular complications in CKD. We discuss here how the cardiovascular effects of these uremic toxins could be mediated by AhR activation, in a “dioxin-like” effect.
Collapse
Affiliation(s)
- Marion Sallée
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Laetitia Dou
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Claire Cerini
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Stéphane Poitevin
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Philippe Brunet
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Stéphane Burtey
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| |
Collapse
|
23
|
Wang K, Li Y, Jiang YZ, Dai CF, Patankar MS, Song JS, Zheng J. An endogenous aryl hydrocarbon receptor ligand inhibits proliferation and migration of human ovarian cancer cells. Cancer Lett 2013; 340:63-71. [PMID: 23851185 DOI: 10.1016/j.canlet.2013.06.026] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 06/20/2013] [Accepted: 06/23/2013] [Indexed: 11/28/2022]
Abstract
The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor mediates many biological processes. Herein, we investigated if 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE, an endogenous AhR ligand) regulated proliferation and migration of human ovarian cancer cells via AhR. We found that AhR was widely present in many histotypes of ovarian cancer tissues. ITE suppressed OVCAR-3 cell proliferation and SKOV-3 cell migration in vitro, which were blocked by AhR knockdown. ITE also suppressed OVCAR-3 cell growth in mice. These data suggest that the ITE might potentially be used for therapeutic intervention for at least a subset of human ovarian cancer.
Collapse
Affiliation(s)
- Kai Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, PR China.,Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, United States
| | - Yan Li
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, United States
| | - Yi-Zhou Jiang
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, United States
| | - Cai-Feng Dai
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, United States.,Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China
| | - Manish S Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, United States
| | - Jia-Sheng Song
- AhR Pharmaceuticals, Inc., Madison, WI 53719, United States
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, United States.,Department of Cardiovascular Medicine, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, Guangdong, PR China
| |
Collapse
|
24
|
Feng S, Cao Z, Wang X. Role of aryl hydrocarbon receptor in cancer. Biochim Biophys Acta Rev Cancer 2013; 1836:197-210. [PMID: 23711559 DOI: 10.1016/j.bbcan.2013.05.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/14/2013] [Accepted: 05/17/2013] [Indexed: 01/01/2023]
Abstract
Aryl hydrocarbon receptor (AHR), a cytosolic ligand-activated transcription factor, belongs to the member of bHLH/PAS family of heterodimeric transcriptional regulators and is widely expressed in a variety of animal species and humans. Recent animal and human data suggested that AHR is involved in various signaling pathways critical to cell normal homeostasis, which covers multiple aspects of physiology, such as cell proliferation and differentiation, gene regulation, cell motility and migration, inflammation and others. Dysregulation of these physiological processes is known to contribute to events such as tumor initiation, promotion, and progression. Increasing epidemiological and experimental animal data provided substantial support for an association between abnormal AHR function and cancer, implicating AHR may be a novel drug-interfering target for cancers. The proposed underlying mechanisms of its actions in cancer involved multiple aspects, (a) inhibiting the functional expression of the key anti-oncogenes (such as p53 and BRCA1), (b) promoting stem cells transforming and angiogenesis, (c) altering cell survival, proliferation and differentiation by influencing the physiologic processes of cell-cycle, apoptosis, cell contact-inhibition, metabolism and remodel of extracellular matrix, and cell-matrix interaction, (d) cross-talking with the signaling pathways of estrogen receptor and inflammation. This review aims to provide a brief overview of recent investigations into the role of AHR and the underlying mechanisms of its actions in cancer, which were explored by the new technologies emerging in recent years.
Collapse
Affiliation(s)
- Shaolong Feng
- The School of Public Health, University of South China, Hengyang 421001, China.
| | | | | |
Collapse
|
25
|
Chang CC, Lee PS, Chou Y, Hwang LL, Juan SH. Mediating effects of aryl-hydrocarbon receptor and RhoA in altering brain vascular integrity: the therapeutic potential of statins. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:211-21. [PMID: 22720799 DOI: 10.1016/j.ajpath.2012.03.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 03/14/2012] [Accepted: 03/19/2012] [Indexed: 12/13/2022]
Abstract
We have demonstrated previously that focal adhesion kinase (FAK)/RhoA alteration by the aryl-hydrocarbon receptor (AhR) agonist 3-methylcholanthrene (3MC) is involved in the antimigratory effects of 3MC in human umbilical vascular endothelial cells. Here, we identified that signaling properties and molecular mechanisms of RhoA/β-catenin were both implicated in alterations to blood-brain barrier integrity. The mechanisms of action were the down-regulation of integrin, the extracellular matrix, and adherens junction stability. PTEN phosphorylation by 3MC-mediated AhR/RhoA activation increased the proteasomal degradation of β-catenin through PKCδ/pGSK3β-mediated β-catenin phosphorylation; the crucial roles of AhR/RhoA in this process were verified by using gain- or loss-of-function experiments. The decrease in β-catenin led to decreased expression of fibronectin and α5β1 integrin. Additionally, protein interactions among FAK, VE-cadherin, vinculin, and β-actin were simultaneously decreased, resulting in adherens junction instability. Novel functional TCF/LEF1 binding sites in the promoter regions of fibronectin and α5/β1 integrin were identified by electrophoretic mobility shift and chromatin immunoprecipitation assays. The results indicate that the binding activities of β-catenin decreased in mouse cerebrovascular endothelial cells treated with 3MC. In addition, simvastatin and pravastatin treatment reversed 3MC-mediated alterations in mouse cerebrovascular endothelial cells by RhoA inactivation, and the in vitro findings were substantiated by an in vivo blood-brain barrier assay. Thus, endothelial barrier dysfunction due to 3MC occurs through AhR/RhoA-mediated β-catenin down-regulation, which is reversed by simvastatin treatment in vivo.
Collapse
Affiliation(s)
- Chih-Cheng Chang
- Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
26
|
Wang ML, Shih CK, Chang HP, Chen YH. Antiangiogenic activity of indole-3-carbinol in endothelial cells stimulated with activated macrophages. Food Chem 2012; 134:811-20. [PMID: 23107695 DOI: 10.1016/j.foodchem.2012.02.185] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 09/07/2011] [Accepted: 02/28/2012] [Indexed: 01/18/2023]
Abstract
The effect of indole-3-carbinol (I3C), a major indolic metabolite in cruciferous vegetables, on lipopolysaccharide (LPS)-activated macrophage-induced tube formation and its associated factors in endothelial EA hy926 cells was investigated. LPS significantly enhanced the capillary-like structure of endothelial cells (ECs) co-cultured with macrophages, but no such effect was observed in single-cultured ECs. I3C, on the other hand, suppressed such enhancement in concert with decreased secretions of vascular endothelial growth factor (VEGF), nitric oxide (NO), interleukin-6 (IL-6), and matrix metalloproteinases (MMPs). The results obtained from cultivating ECs with conditioned medium (CM) collected from macrophages suggested that both ECs and macrophages were inactivated by I3C. These results indicate that I3C from cruciferous vegetables may possess potential roles in preventing inflammation-associated angiogenic diseases.
Collapse
Affiliation(s)
- Mei-Lin Wang
- School of Nutrition and Health Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110, Taiwan, ROC
| | | | | | | |
Collapse
|
27
|
Volkov MS, Bolotina NA, Evteev VA, Koblyakov VA. Ah-receptor-independent stimulation of hepatoma 27 culture cell proliferation by polycyclic aromatic hydrocarbons. BIOCHEMISTRY (MOSCOW) 2012; 77:201-7. [DOI: 10.1134/s0006297912020125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
28
|
Li CH, Cheng YW, Hsu YT, Hsu YJ, Liao PL, Kang JJ. Benzo[a]pyrene inhibits angiogenic factors-induced alphavbeta3 integrin expression, neovasculogenesis, and angiogenesis in human umbilical vein endothelial cells. Toxicol Sci 2010; 118:544-53. [PMID: 20876236 DOI: 10.1093/toxsci/kfq279] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
New blood vessel formation is necessary for the repair of ischemia-damaged tissues. Endothelial cells produce exogenous and endogenous angiogenic factors in the mediation of angiogenesis and neovasculogenesis during neovascularization. Exposure to environmental pollutants may alter proangiogenic capacity or desensitize the responses of endothelial cells to stimulation by basic fibroblast growth factor and vascular endothelial growth factor. Human umbilical vein endothelial cells (HUVECs) were pretreated with benzo[a]pyrene (B[a]P), the major carcinogenic constituent found in tobacco smoke, for 24 h. Neovasculogenesis, migration, and proliferation were evaluated in solvent-treated and B[a]P-treated HUVECs. Endothelial capillary-like tube formation, cell migration, mitogen-activated protein kinase (MAPK) phosphorylation, and integrin expression were reduced in B[a]P-treated HUVECs with angiogenic factor stimulation, in comparison to solvent-treated HUVECs, although cell proliferation and Akt activation remained unaffected. Inhibition of B[a]P-mediated MAPK and neovasculogenesis was significantly rescued by pretreatment with α-naphthoflavone, an aryl hydrocarbon receptor (AhR) antagonist. The B[a]P-mediated inhibition of neovasculogenesis was also rescued in AhR-silenced HUVECs, suggesting the requirement for AhR in B[a]P-associated effects. B[a]P also inhibited angiogenesis in a chorioallantoic membrane assay. We conclude that B[a]P is a potent inhibitor of angiogenesis, and its effects are mediated via AhR-dependent phenotypic changes in B[a]P-treated HUVECs. These findings contribute to an understanding of the involvement of AhR agonists in vasculotoxicity.
Collapse
Affiliation(s)
- Ching-Hao Li
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | | | | | | | | | | |
Collapse
|
29
|
Jiang YZ, Wang K, Fang R, Zheng J. Expression of aryl hydrocarbon receptor in human placentas and fetal tissues. J Histochem Cytochem 2010; 58:679-85. [PMID: 20354149 DOI: 10.1369/jhc.2010.955955] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, mediates many biological processes, including fetal development. In this study, we examined AhR protein expression in human placentas from normal (N) and severe preeclamptic (sPE) pregnancies, as well as human fetal tissues from the second trimester of pregnancy, using immunohistochemistry and/or Western blot analysis. In the placentas, the AhR immunoreactivity was present primarily in syncytiotrophoblasts. The AhR staining was also seen in endothelium of large blood vessels in villi and endothelium of umbilical cord arteries and veins. No difference in AhR protein levels was found between N and sPE placentas. In fetal tissues, the AhR immunoreactivity was localized in lung, kidney, esophagus, pancreas, liver, testicle, thymus gland, retina, and choroid, mainly in epithelial cells, whereas it was absent in heart, brain, sclera, and thoracic aorta. These findings suggest that the AhR plays a critical role in syncytiotrophoblasts of human placentas and epithelium of many fetal organs. These data also imply that human placentas and those fetal organs with high AhR expression (e.g., lung, kidney, liver, pancreas, and thymus gland) during fetal development are highly susceptible to environmental toxicants such as dioxin.
Collapse
Affiliation(s)
- Yi-zhou Jiang
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, USA
| | | | | | | |
Collapse
|
30
|
Chang CC, Tsai SY, Lin H, Li HF, Lee YH, Chou Y, Jen CY, Juan SH. Aryl-hydrocarbon receptor-dependent alteration of FAK/RhoA in the inhibition of HUVEC motility by 3-methylcholanthrene. Cell Mol Life Sci 2009; 66:3193-205. [PMID: 19649566 PMCID: PMC11115561 DOI: 10.1007/s00018-009-0102-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/23/2009] [Accepted: 07/09/2009] [Indexed: 01/01/2023]
Abstract
We previously demonstrated the antiproliferative and antiangiogenic effects of 3-methylcholanthrene (3MC), an aryl-hydrocarbon receptor (AhR) agonist, in human umbilical vascular endothelial cells (HUVECs). Herein, we unraveled its molecular mechanisms in inhibiting HUVEC motility. 3MC down-regulated FAK, but up-regulated RhoA, which was rescued by AhR knockdown. It led us to identify novel AhR binding sites in the FAK/RhoA promoters. Additionally, 3MC increased RhoA activity via suppression of a negative feedback pathway of FAK/p190RhoGAP. With an increase in membrane-bound RhoA, subsequent stress fiber and focal adhesion complex formation was observed in 3MC-treated cells, and this was reversed by a RhoA inhibitor and AhR antagonists. Notably, these compounds significantly reversed 3MC-mediated anti-migration in a transwell assay. The in vitro findings were further confirmed using an animal model of Matrigel formation in Balb/c mice. Collectively, AhR's genomic regulation of FAK/RhoA, together with RhoA activation, is ascribable to the anti-migration effect of 3MC in HUVECs.
Collapse
MESH Headings
- Animals
- Binding Sites
- Cell Movement/drug effects
- Cell Movement/physiology
- Cells, Cultured
- Chromatin Immunoprecipitation
- Endothelial Cells/cytology
- Endothelial Cells/drug effects
- Endothelial Cells/enzymology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Feedback, Physiological
- Focal Adhesion Kinase 1/genetics
- Focal Adhesion Kinase 1/metabolism
- Focal Adhesion Kinase 1/physiology
- Gene Expression Regulation
- Humans
- Methylcholanthrene/pharmacology
- Mice
- Mice, Inbred BALB C
- Promoter Regions, Genetic
- RNA Interference
- RNA, Messenger/metabolism
- RNA, Small Interfering
- Receptors, Aryl Hydrocarbon/agonists
- Receptors, Aryl Hydrocarbon/antagonists & inhibitors
- Receptors, Aryl Hydrocarbon/physiology
- Umbilical Cord/cytology
- rhoA GTP-Binding Protein/genetics
- rhoA GTP-Binding Protein/metabolism
- rhoA GTP-Binding Protein/physiology
Collapse
Affiliation(s)
- Chih-Cheng Chang
- Department of Physiology, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110 Taiwan
- Department of Physiology, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ying Tsai
- Department of Physiology, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110 Taiwan
- Department of Physiology, Taipei Medical University, Taipei, Taiwan
| | - Heng Lin
- Institute of Pharmacology and Toxicology, Tzu Chi University, Hualien, Taiwan
| | - Hsiao-Fen Li
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Department of Physiology, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110 Taiwan
- Department of Physiology, Taipei Medical University, Taipei, Taiwan
| | - Ying Chou
- Department of Physiology, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110 Taiwan
- Department of Physiology, Taipei Medical University, Taipei, Taiwan
| | - Chih-Yu Jen
- Department of Physiology, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110 Taiwan
- Department of Physiology, Taipei Medical University, Taipei, Taiwan
| | - Shu-Hui Juan
- Department of Physiology, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110 Taiwan
- Department of Physiology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
31
|
Ishimura R, Kawakami T, Ohsako S, Tohyama C. Dioxin-induced toxicity on vascular remodeling of the placenta. Biochem Pharmacol 2008; 77:660-9. [PMID: 19027717 DOI: 10.1016/j.bcp.2008.10.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 10/14/2008] [Accepted: 10/15/2008] [Indexed: 12/22/2022]
Abstract
Arylhydrocarbon receptor (AhR) activated by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) triggers its downstream signaling pathway to exert adverse effects on vasculature development, which can be initiated by vasculogenesis, followed by angiogenesis, or vascular remodeling, in a variety of animals including avians, piscines and mammals. The placenta, a mammalian organ rich in vasculature, consists of endothelial and trophoblast cells of fetal origin, which proliferate and differentiate under hypoxic condition in the uterine horn. Our studies demonstrated that vascular remodeling occurs prominently in the placenta of the control Holtzman rat strain during the late period of gestation, and induces changes in cell shape and elimination by apoptosis of trophoblasts. As a result, the net volumes of both maternal and fetal blood in the placenta increase to cope with the essential requirements of oxygen and nutrients in the late period of gestation. On the other hand, in utero exposure to TCDD markedly suppressed the development of sinusoids and trophoblast cells and the apoptosis of trophoblast cells with a concomitant increase in the incidence of fetal death under hypoxic condition. A crosstalk between the hypoxia-inducible factor (HIF)-mediated pathway and AhR-mediated pathway is considered to play an important role in this physiological process. No such changes were observed in the Sprague-Dawley rat strain that turned out to have an AhR conformation identical to that of the Holtzman rat strain. In this commentary, we will discuss a possible link of the TCDD toxicities with the AhR signaling pathway and gestation-related diseases.
Collapse
Affiliation(s)
- Ryuta Ishimura
- National Institute for Environmental Studies, Tsukuba, Japan
| | | | | | | |
Collapse
|
32
|
Kung T, Murphy KA, White LA. The aryl hydrocarbon receptor (AhR) pathway as a regulatory pathway for cell adhesion and matrix metabolism. Biochem Pharmacol 2008; 77:536-46. [PMID: 18940186 DOI: 10.1016/j.bcp.2008.09.031] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 09/17/2008] [Accepted: 09/19/2008] [Indexed: 01/07/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is an orphan receptor in the basic helix-loop-helix PAS family of transcriptional regulators. Although the endogenous regulator of this pathway has not been identified, the AhR is known to bind and be activated by a variety of compounds ranging from environmental contaminants to flavanoids. The function of this receptor is still unclear; however, animal models indicate that the AhR is important for normal development. One hypothesis is that the AhR senses cellular stress and initiates the cellular response by altering gene expression and inhibiting cell cycle progression and that activation of the AhR by exogenous environmental chemicals results in the dysregulation of this normal function. In this review we will examine the role of the AhR in the regulation of genes and proteins involved in cell adhesion and matrix remodeling, and discuss the implications of these changes in development and disease. In addition, we will discuss evidence suggesting that the AhR pathway is responsive to changes in matrix composition as well as cell-cell and cell-matrix interactions.
Collapse
Affiliation(s)
- Tiffany Kung
- Department of Biochemistry and Microbiology, Rutgers, The State University of NJ, New Brunswick, NJ 08901, USA
| | | | | |
Collapse
|
33
|
Pang PH, Lin YH, Lee YH, Hou HH, Hsu SP, Juan SH. Molecular mechanisms of p21 and p27 induction by 3-methylcholanthrene, an aryl-hydrocarbon receptor agonist, involved in antiproliferation of human umbilical vascular endothelial cells. J Cell Physiol 2008; 215:161-71. [PMID: 18022818 DOI: 10.1002/jcp.21299] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We previously reported that 3-methylcholanthrene (3MC), an aryl-hydrocarbon receptor (AhR) agonist, inhibits the proliferation of human umbilical vascular endothelial cells (HUVECs; Juan et al., 2006, Eur J Pharmacol 530: 1-8). Herein, pretreatment of HUVECs with p21 or p27 small interfering (si)RNA reduced 3MC-induced elimination of [(3)H]thymidine incorporation, demonstrating their essential roles in the antiproliferation of HUVECs. The molecular mechanisms of p21 and p27 involved in the antiproliferative effects of 3MC were elucidated in this study. 3MC time- and concentration-dependently increased p21 and p27 levels, and decreased the protein level of CDK2 with no apparent alteration of p53. Interestingly, 3MC-mediated p21 and p27 inductions were eliminated by resveratrol, an AhR antagonist, suggesting their AhR dependency, further confirmed by AhR siRNA. Among the relevant pathways, p38MAPK activation sustained the levels of p21 and p27 induced by 3MC, which was eliminated by AhR antagonists and N-acetylcysteine (NAC), an antioxidant. 3MC concentration-dependently enhanced not only the consensus dioxin-responsive element (DRE)-driven luciferase activity, but also the binding activity of the AhR to the putative DRE derived from the p21 and p27 promoters. A deletion of the DRE (-285/-270) in p21 (-2,300/+8) only partially alleviated the 3MC-induced luciferase activity unless NAC was added, suggesting that there may be a DRE-independent mechanism associated with oxidative stress. However, a deletion of the DRE (-660/-645) in p27 (-1,358/-100) almost completely abrogated the activation. Our study demonstrated that both the functional DRE and the phosphorylation of p38MAPK are essential for the induction of p21 and p27, resulting in the antiproliferative action of 3MC in HUVECs.
Collapse
Affiliation(s)
- Pai-Huei Pang
- Department of Ophthalmology, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
34
|
Hillegass JM, Murphy KA, Villano CM, White LA. The impact of aryl hydrocarbon receptor signaling on matrix metabolism: implications for development and disease. Biol Chem 2008; 387:1159-73. [PMID: 16972783 DOI: 10.1515/bc.2006.144] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The aryl hydrocarbon receptor (AhR) was identified as the receptor for polycyclic aromatic hydrocarbons and related compounds. However, novel data indicate that the AhR binds a variety of unrelated endogenous and exogenous compounds. Although AhR knockout mice demonstrate that this receptor has a role in normal development and physiology, the function of this receptor is still unclear. Recent evidence suggests that AhR signaling also alters the expression of genes involved in matrix metabolism, specifically the matrix metalloproteinases (MMPs). MMP expression and activity is critical to normal physiological processes that require tissue remodeling, as well as in mediating the progression of a variety of diseases. MMPs not only degrade structural proteins, but are also important mediators of cell signaling near or at the cell membrane through exposure of cryptic sites, release of growth factors, and cleavage of receptors. Therefore, AhR modulation of MMP expression and activity may be critical, not only in pathogenesis, but also in understanding the endogenous function of the AhR. In this review we will examine the data indicating a role for the AhR-signaling pathway in the regulation of matrix remodeling, and discuss potential molecular mechanisms.
Collapse
Affiliation(s)
- Jedd M Hillegass
- Department of Biochemistry and Microbiology, Rutgers, The State University of New Jersey, 76 Lipman Dr., New Brunswick, NJ 08901, USA
| | | | | | | |
Collapse
|
35
|
Sanyal MK, Li YL. Deleterious effects of polynuclear aromatic hydrocarbon on blood vascular system of the rat fetus. ACTA ACUST UNITED AC 2007; 80:367-73. [PMID: 17615575 DOI: 10.1002/bdrb.20122] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Polynuclear aromatic hydrocarbons (PAH), benzo[alpha]pyrene (B[alpha]P) and 7,12-dimethylbenz[alpha]anthracene (DMBA) are toxic environmental agents distributed widely. The relative deleterious effects of these agents on growth and blood vasculature of fetus and placental tissues of the rat were studied. METHODS Pregnant rats (Day 1 sperm positive) with implantation sites confirmed by laparotomy were treated intraperitoneally (i.p.) on Pregnancy Days 10, 12, and 14 with these agents dissolved in corn oil at cumulated total doses 50, 100, and 200 mg/kg/rat, and control with corn oil only (3-20 dams/group). Fetal growth, tissue hemorrhage, and placental pathology were evaluated by different parameters on Pregnancy Day (PD) 20 in treated and control rats. RESULTS DMBA was relatively more deleterious compared to B[alpha]P indicated by increased lethality and progressive reduction of body weight of the mother with increasing doses. At 200 mg/kg/rat doses of these agents, maternal survival was 45% and 100% and body weight reduced 24% and 52% of controls, respectively. The fetal survival rates in live mothers were similar to that of controls. They induced marked fetal growth retardation and necrosis of placental tissues. B[alpha]P and DMBA produced significant toxicity to differentiating fetal blood vascular system as exhibited by rupture of blood vessels and hemorrhage, especially in the skin, cranial, and brain tissues. CONCLUSIONS Maternal PAH exposure induced placental toxicity and associated adverse fetal development and hemorrhage in different parts of the fetal body, in particular, marked intradermal and cranial hemorrhage, showing that developing fetal blood vasculature is a target of PAH toxicity.
Collapse
Affiliation(s)
- Mrinal K Sanyal
- Department of Obstetrics and Gynecology, Yale University Medical School, New Haven, CT, USA.
| | | |
Collapse
|