1
|
Barreira-Silva P, Lian Y, Kaufmann SHE, Moura-Alves P. The role of the AHR in host-pathogen interactions. Nat Rev Immunol 2025; 25:178-194. [PMID: 39415055 DOI: 10.1038/s41577-024-01088-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/18/2024]
Abstract
Host-microorganism encounters take place in many different ways and with different types of outcomes. Three major types of microorganisms need to be distinguished: (1) pathogens that cause harm to the host and must be controlled; (2) environmental microorganisms that can be ignored but must be controlled at higher abundance; and (3) symbiotic microbiota that require support by the host. Recent evidence indicates that the aryl hydrocarbon receptor (AHR) senses and initiates signalling and gene expression in response to a plethora of microorganisms and infectious conditions. It was originally identified as a receptor that binds xenobiotics. However, it was subsequently found to have a critical role in numerous biological processes, including immunity and inflammation and was recently classified as a pattern recognition receptor. Here we review the role of the AHR in host-pathogen interactions, focusing on AHR sensing of different microbial classes, the ligands involved, responses elicited and disease outcomes. Moreover, we explore the therapeutic potential of targeting the AHR in the context of infection.
Collapse
Affiliation(s)
- Palmira Barreira-Silva
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Yilong Lian
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, USA
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pedro Moura-Alves
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
2
|
Madison CA, Debler RA, Gallegos PL, Hillbrick L, Chapkin RS, Safe S, Eitan S. 1,4-dihydroxy-2-naphthoic acid prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced motor function deficits. Behav Pharmacol 2025; 36:40-46. [PMID: 39660867 PMCID: PMC11781791 DOI: 10.1097/fbp.0000000000000806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Parkinson's disease (PD), characterized by death of dopaminergic neurons in the substantia nigra, is the second most prevalent progressive neurodegenerative disease. However, the etiology of PD is largely elusive. This study employed the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) rodent model to examine the effectiveness of 1,4-dihydroxy-2-naphthoic acid (1,4-DHNA), an aryl hydrocarbon receptor (AhR) active gut bacteria-derived metabolite, in mitigating MPTP's motoric deficits, and the role of AhR in mediating these effects. Male C57BL/6 mice were fed daily with vehicle, 20 mg/kg 1,4-DHNA, or AhR-inactive isomer 3,7-DHNA, for 3 weeks before administration of 80 mg/kg MPTP or vehicle. Four weeks later, mice were assessed for motoric functions. Both 1,4-DHNA and 3,7-DHNA prevented MPTP-induced deficits in the motor pole test and in the adhesive strip removal test. Additionally, 1,4-DHNA improved balance beam performance and completely prevented MPTP-induced reduction in stride length. In contrast, 3,7-DHNA, an AhR-inactive compound, did not improve balance beam performance and had only a partial effect on stride length. This study suggests that natural metabolites of gut microbiota, such as 1,4-DHNA, could be beneficial to counteract the development of motor deficits observed in PD. Thus, this study further supports the hypothesis that pathological and mitigating processes in the gut could play an essential role in PD development. Moreover, this indicates that 1,4-DHNA's ability to combat various motor deficits is likely mediated via multiple underlying molecular mechanisms. Specifically, AhR is involved, at least partially, in control of gait and bradykinesia, but it likely does not mediate the effects on fine motor skills.
Collapse
Affiliation(s)
- Caitlin A. Madison
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Roanna A. Debler
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Paula L. Gallegos
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Lauren Hillbrick
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Robert S. Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466 USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| |
Collapse
|
3
|
Debler RA, Gallegos PL, Ojeda AC, Perttula AM, Lucio A, Chapkin RS, Safe S, Eitan S. TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) induces depression-like phenotype. Neurotoxicology 2024; 103:71-77. [PMID: 38838945 PMCID: PMC11288769 DOI: 10.1016/j.neuro.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
The etiology of major depressive disorder (MDD) remains poorly understood. Our previous studies suggest a role for the aryl hydrocarbon receptor (AhR) in depression. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a toxic environmental contaminant, with a high AhR binding affinity, and an established benchmark for assessing AhR activity. Therefore, this study examined the effect of TCDD on depression-like behaviors. Female mice were fed standard chow or a high-fat diet (HFD) for 11 weeks, and their weight was recorded. Subsequently, they were tested for baseline sucrose preference and splash test grooming. Then, TCDD (0.1 µg/kg/day) or vehicle was administered orally for 28 days, and mice were examined for their sucrose preference and performances in the splash test, forced swim test (FST), and Morris water maze (MWM) task. TCDD significantly decreased sucrose preference, increased FST immobility time, and decreased groom time in chow-fed mice. HFD itself significantly reduced sucrose preference. However, TCDD significantly increased FST immobility time and decreased groom time in HFD-fed mice. A small decrease in bodyweight was observed only at the fourth week of daily TCDD administration in chow-fed mice, and no significant effects of TCDD on bodyweights were observed in HFD-fed mice. TCDD did not have a significant effect on spatial learning in the MWM. Thus, this study demonstrated that TCDD induces a depression-like state, and the effects were not due to gross lethal toxicity. This study further suggests that more studies should examine a possible role for AhR and AhR-active environmental pollutants in precipitating or worsening MDD.
Collapse
Affiliation(s)
- Roanna A Debler
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Paula L Gallegos
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Alexandra C Ojeda
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Andrea M Perttula
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Ashley Lucio
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA.
| |
Collapse
|
4
|
Narita Y, Tamura A, Hatakeyama S, Uemura S, Miura A, Haga A, Tsuji N, Fujie N, Izumi Y, Sugawara T, Otaka M, Okamoto K, Lu P, Okuda S, Suzuki M, Nagata K, Shimizu H, Itoh H. The components of the AhR-molecular chaperone complex differ depending on whether the ligands are toxic or non-toxic. FEBS Lett 2024; 598:1478-1490. [PMID: 38605276 DOI: 10.1002/1873-3468.14871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/14/2024] [Accepted: 03/15/2024] [Indexed: 04/13/2024]
Abstract
The aryl hydrocarbon receptor (AhR) forms a complex with the HSP90-XAP2-p23 molecular chaperone when the cells are exposed to toxic compounds. Recently, 1,4-dihydroxy-2-naphthoic acid (DHNA) was reported to be an AhR ligand. Here, we investigated the components of the molecular chaperone complex when DHNA binds to AhR. Proteins eluted from the 3-Methylcolanthrene-affinity column were AhR-HSP90-XAP2-p23 complex. The AhR-molecular chaperone complex did not contain p23 in the eluents from the DHNA-affinity column. In 3-MC-treated cells, AhR formed a complex with HSP90-XAP2-p23 and nuclear translocation occurred within 30 min, while in DHNA-treated cells, AhR formed a complex with AhR-HSP90-XAP2, and translocation was slow from 60 min. Thus, the AhR activation mechanism may differ when DHNA is the ligand compared to toxic ligands.
Collapse
Affiliation(s)
- Yukihiko Narita
- Department of Neurosurgery, Akita University Graduate School of Medicine, Japan
| | - Arisa Tamura
- Department of Life Science, Graduate School of Engineering Science, Akita University, Japan
| | - Shiori Hatakeyama
- Department of Life Science, Graduate School of Engineering Science, Akita University, Japan
| | - Seiya Uemura
- Department of Life Science, Graduate School of Engineering Science, Akita University, Japan
| | - Atsuko Miura
- Department of Neurosurgery, Akita University Graduate School of Medicine, Japan
- Department of Life Science, Akita Cerebrospinal and Cardiovascular Center, Japan
| | - Asami Haga
- Department of Life Science, Graduate School of Engineering Science, Akita University, Japan
| | - Noriko Tsuji
- Department of Life Science, Graduate School of Engineering Science, Akita University, Japan
| | - Nozomi Fujie
- Department of Life Science, Graduate School of Engineering Science, Akita University, Japan
| | - Yukina Izumi
- Department of Life Science, Graduate School of Engineering Science, Akita University, Japan
| | - Taku Sugawara
- Department of Life Science, Akita Cerebrospinal and Cardiovascular Center, Japan
| | - Michiro Otaka
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Okamoto
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | - Peng Lu
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | - Suguru Okuda
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | - Michio Suzuki
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | - Koji Nagata
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | - Hiroaki Shimizu
- Department of Neurosurgery, Akita University Graduate School of Medicine, Japan
| | - Hideaki Itoh
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| |
Collapse
|
5
|
Bonati L, Motta S, Callea L. The AhR Signaling Mechanism: A Structural Point of View. J Mol Biol 2024; 436:168296. [PMID: 37797832 DOI: 10.1016/j.jmb.2023.168296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/23/2023] [Accepted: 09/28/2023] [Indexed: 10/07/2023]
Abstract
The Aryl hydrocarbon Receptor (AhR) is a well-known sensor of xenobiotics; moreover, it is considered a promising drug target as it is involved in the regulation of many patho-physiological processes. For these reasons the study of its ligand-activated transcription mechanism has stimulated several studies for over twenty years. In this review we highlight the key role of molecular structural information in understanding the different steps of the signaling mechanism. The architecture of the AhR cytosolic complex, encompassing the hsp90 chaperone protein and the XAP2 and p23 co-chaperones, has become available in the last year thanks to Cryo-EM experiments. The structure of the AhR ligand-binding (PAS-B) domain has remained elusive for a long time; it has been predicted by homology modelling, based on known PAS systems, and its ligand-bound forms were modelled through ligand molecular docking. Although very recently some structural information on this domain has become available, considerable efforts are still needed to determine the binding geometries of the AhR key ligands by experimental high-resolution studies. On the other hand, the dimeric structure of AhR with the ARNT protein, bound to the specific DNA responsive element, was partially determined by X-ray crystallography and it was completed by homology modelling. On the whole the current structural knowledge of the main protein complexes that form over the AhR mechanism opens the way to confirm and further investigate the main steps of the proposed ligand-activated transcription mechanism of the AhR.
Collapse
Affiliation(s)
- Laura Bonati
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy.
| | - Stefano Motta
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy.
| | - Lara Callea
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy.
| |
Collapse
|
6
|
Li S, De Groote Tavares C, Tolar JG, Ajo-Franklin CM. Selective bioelectronic sensing of pharmacologically relevant quinones using extracellular electron transfer in Lactiplantibacillus plantarum. Biosens Bioelectron 2024; 243:115762. [PMID: 37875059 DOI: 10.1016/j.bios.2023.115762] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 10/26/2023]
Abstract
Redox-active small molecules containing quinone functional groups play important roles as pharmaceuticals, but can be toxic if overdosed. Despite the need for a fast and quantitative method to detect quinone and its derivatives, current sensing strategies are often slow and struggle to differentiate between structural analogs. Leveraging the discovery that microorganisms use certain quinones to perform extracellular electron transfer (EET), we investigated the use of Lactiplantibacillus plantarum as a whole-cell bioelectronic sensor to selectively sense quinone analogs. By tailoring the native EET pathway in L. plantarum, we enabled quantitative quinone sensing of 1,4-dihydroxy-2-naphthoic acid (DHNA) - a gut bifidogenic growth stimulator. We found that L. plantarum could respond to environmental DHNA within seconds, producing concentration-dependent electrical signals. This sensing capacity was robust in different assay media and allowed for continuous monitoring of DHNA concentrations. In a simulated gut environment containing a mixed pool of quinone derivatives, this tailored EET pathway can selectively sense pharmacologically relevant quinone analogs, such as DHNA and menadione, amongst other structurally similar quinone derivatives. We also developed a multivariate model to describe the mechanism behind this selectivity and found a predictable correlation between quinone physiochemical properties and the corresponding electrical signals. Our work presents a new concept to selectively sense quinone using whole-cell bioelectronic sensors and opens the possibility of using probiotic L. plantarum for bioelectronic applications in human health.
Collapse
Affiliation(s)
- Siliang Li
- Department of BioSciences, Rice University, Houston, TX, USA
| | | | - Joe G Tolar
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Caroline M Ajo-Franklin
- Department of BioSciences, Rice University, Houston, TX, USA; Department of Bioengineering, Rice University, Houston, TX, USA; Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, USA.
| |
Collapse
|
7
|
Chandrasekaran P, Han Y, Zerbe CS, Heller T, DeRavin SS, Kreuzberg SA, Marciano BE, Siu Y, Jones DR, Abraham RS, Stephens MC, Tsou AM, Snapper S, Conlan S, Subramanian P, Quinones M, Grou C, Calderon V, Deming C, Leiding JW, Arnold DE, Logan BR, Griffith LM, Petrovic A, Mousallem TI, Kapoor N, Heimall JR, Barnum JL, Kapadia M, Wright N, Rayes A, Chandra S, Broglie LA, Chellapandian D, Deal CL, Grunebaum E, Lim SS, Mallhi K, Marsh RA, Murguia-Favela L, Parikh S, Touzot F, Cowan MJ, Dvorak CC, Haddad E, Kohn DB, Notarangelo LD, Pai SY, Puck JM, Pulsipher MA, Torgerson TR, Kang EM, Malech HL, Segre JA, Bryant CE, Holland SM, Falcone EL. Intestinal microbiome and metabolome signatures in patients with chronic granulomatous disease. J Allergy Clin Immunol 2023; 152:1619-1633.e11. [PMID: 37659505 PMCID: PMC11279821 DOI: 10.1016/j.jaci.2023.07.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Chronic granulomatous disease (CGD) is caused by defects in any 1 of the 6 subunits forming the nicotinamide adenine dinucleotide phosphate oxidase complex 2 (NOX2), leading to severely reduced or absent phagocyte-derived reactive oxygen species production. Almost 50% of patients with CGD have inflammatory bowel disease (CGD-IBD). While conventional IBD therapies can treat CGD-IBD, their benefits must be weighed against the risk of infection. Understanding the impact of NOX2 defects on the intestinal microbiota may lead to the identification of novel CGD-IBD treatments. OBJECTIVE We sought to identify microbiome and metabolome signatures that can distinguish individuals with CGD and CGD-IBD. METHODS We conducted a cross-sectional observational study of 79 patients with CGD, 8 pathogenic variant carriers, and 19 healthy controls followed at the National Institutes of Health Clinical Center. We profiled the intestinal microbiome (amplicon sequencing) and stool metabolome, and validated our findings in a second cohort of 36 patients with CGD recruited through the Primary Immune Deficiency Treatment Consortium. RESULTS We identified distinct intestinal microbiome and metabolome profiles in patients with CGD compared to healthy individuals. We observed enrichment for Erysipelatoclostridium spp, Sellimonas spp, and Lachnoclostridium spp in CGD stool samples. Despite differences in bacterial alpha and beta diversity between the 2 cohorts, several taxa correlated significantly between both cohorts. We further demonstrated that patients with CGD-IBD have a distinct microbiome and metabolome profile compared to patients without CGD-IBD. CONCLUSION Intestinal microbiome and metabolome signatures distinguished patients with CGD and CGD-IBD, and identified potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | - Yu Han
- Division of Molecular Genetics and Pathology, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Md; Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Christa S Zerbe
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Theo Heller
- Translational Hepatology Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Md
| | - Suk See DeRavin
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Samantha A Kreuzberg
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Beatriz E Marciano
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Yik Siu
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY
| | - Roshini S Abraham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn; Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | | | - Amy M Tsou
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Mass; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, New York, NY
| | - Scott Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Sean Conlan
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Md
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, Md
| | - Mariam Quinones
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, Md
| | - Caroline Grou
- Bioinformatics Core, Montreal Clinical Research Institute (IRCM), Montreal, Quebec, Canada
| | - Virginie Calderon
- Bioinformatics Core, Montreal Clinical Research Institute (IRCM), Montreal, Quebec, Canada
| | - Clayton Deming
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Md
| | - Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University, Baltimore, Md
| | - Danielle E Arnold
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Md
| | - Brent R Logan
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wis
| | - Linda M Griffith
- Division of Allergy, Immunology, and Transplantation, NIAID, NIH, Bethesda, Md
| | - Aleksandra Petrovic
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital and Research Center, Seattle, Wash
| | - Talal I Mousallem
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Neena Kapoor
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, Calif
| | - Jennifer R Heimall
- Division of Allergy and Immunology, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Jessie L Barnum
- Division of Blood and Marrow Transplantation and Cellular Therapies, University of Pittsburgh Medical Center (UPMC) and Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Malika Kapadia
- Department of Pediatrics, Harvard University Medical School, Boston, Mass
| | - Nicola Wright
- Section of Hematology/Immunology, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Ahmad Rayes
- Intermountain Primary Children's Hospital, University of Utah, Salt Lake City, Utah
| | - Sharat Chandra
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Larisa A Broglie
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis
| | - Deepak Chellapandian
- Center for Cell and Gene Therapy for Non-Malignant Conditions, Johns Hopkins All Children's Hospital, St Petersburg, Fla
| | - Christin L Deal
- Division of Allergy and Immunology, UPMC, Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Eyal Grunebaum
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Stephanie Si Lim
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, Hawaii; University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, Hawaii
| | | | - Rebecca A Marsh
- Cincinnati Children's Hospital Medical Center, and University of Cincinnati, Cincinnati, Ohio
| | - Luis Murguia-Favela
- Section of Hematology/Immunology, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Suhag Parikh
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Ga
| | - Fabien Touzot
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Morton J Cowan
- University of California San Francisco Benioff Children's Hospital, San Francisco, Calif
| | - Christopher C Dvorak
- University of California San Francisco Benioff Children's Hospital, San Francisco, Calif
| | - Elie Haddad
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Donald B Kohn
- Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Calif
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Sung-Yun Pai
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Md
| | - Jennifer M Puck
- University of California San Francisco Benioff Children's Hospital, San Francisco, Calif
| | - Michael A Pulsipher
- Division of Pediatric Hematology and Oncology, Intermountain Primary Children's Hospital, Huntsman Cancer Institute at the University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, Utah
| | | | - Elizabeth M Kang
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Harry L Malech
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Julia A Segre
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Md
| | - Clare E Bryant
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Emilia Liana Falcone
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montreal, Quebec, Canada; Center for Immunity, Inflammation and Infectious Diseases, IRCM, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
8
|
Smith HB, Lee K, Freeman MJ, Stevenson DM, Amador-Noguez D, Sauer JD. Listeria monocytogenes requires DHNA-dependent intracellular redox homeostasis facilitated by Ndh2 for survival and virulence. Infect Immun 2023; 91:e0002223. [PMID: 37754681 PMCID: PMC10580952 DOI: 10.1128/iai.00022-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023] Open
Abstract
Listeria monocytogenes is a remarkably well-adapted facultative intracellular pathogen that can thrive in a wide range of ecological niches. L. monocytogenes maximizes its ability to generate energy from diverse carbon sources using a respiro-fermentative metabolism that can function under both aerobic and anaerobic conditions. Cellular respiration maintains redox homeostasis by regenerating NAD+ while also generating a proton motive force. The end products of the menaquinone (MK) biosynthesis pathway are essential to drive both aerobic and anaerobic cellular respirations. We previously demonstrated that intermediates in the MK biosynthesis pathway, notably 1,4-dihydroxy-2-naphthoate (DHNA), are required for the survival and virulence of L. monocytogenes independent of their role in respiration. Furthermore, we found that restoration of NAD+/NADH ratio through expression of water-forming NADH oxidase could rescue phenotypes associated with DHNA deficiency. Here, we extend these findings to demonstrate that endogenous production or direct supplementation of DHNA restored both the cellular redox homeostasis and metabolic output of fermentation in L. monocytogenes. Furthermore, exogenous supplementation of DHNA rescues the in vitro growth and ex vivo virulence of L. monocytogenes DHNA-deficient mutants. Finally, we demonstrate that exogenous DHNA restores redox balance in L. monocytogenes specifically through the recently annotated NADH dehydrogenase Ndh2, independent of its role in the extracellular electron transport pathway. These data suggest that the production of DHNA may represent an additional layer of metabolic adaptability by L. monocytogenes to drive energy metabolism in the absence of respiration-favorable conditions.
Collapse
Affiliation(s)
- Hans B. Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kijeong Lee
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Matthew J. Freeman
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David M. Stevenson
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Daniel Amador-Noguez
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
9
|
Vázquez-Gómez G, Petráš J, Dvořák Z, Vondráček J. Aryl hydrocarbon receptor (AhR) and pregnane X receptor (PXR) play both distinct and common roles in the regulation of colon homeostasis and intestinal carcinogenesis. Biochem Pharmacol 2023; 216:115797. [PMID: 37696457 DOI: 10.1016/j.bcp.2023.115797] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
Both aryl hydrocarbon receptor (AhR) and pregnane X receptor (PXR) belong among key regulators of xenobiotic metabolism in the intestinal tissue. AhR in particular is activated by a wide range of environmental and dietary carcinogens. The data accumulated over the last two decades suggest that both of these transcriptional regulators play a much wider role in the maintenance of gut homeostasis, and that both transcription factors may affect processes linked with intestinal tumorigenesis. Intestinal epithelium is continuously exposed to a wide range of AhR, PXR and dual AhR/PXR ligands formed by intestinal microbiota or originating from diet. Current evidence suggests that specific ligands of both AhR and PXR can protect intestinal epithelium against inflammation and assist in the maintenance of epithelial barrier integrity. AhR, and to a lesser extent also PXR, have been shown to play a protective role against inflammation-induced colon cancer, or, in mouse models employing overactivation of Wnt/β-catenin signaling. In contrast, other evidence suggests that both receptors may contribute to modulation of transformed colon cell behavior, with a potential to promote cancer progression and/or chemoresistance. The review focuses on both overlapping and separate roles of the two receptors in these processes, and on possible implications of their activity within the context of intestinal tissue.
Collapse
Affiliation(s)
- Gerardo Vázquez-Gómez
- Department of Cytokinetics, Institute of Biophysics of the CAS, Královopolská 135, 61265 Brno, Czech Republic
| | - Jiří Petráš
- Department of Cytokinetics, Institute of Biophysics of the CAS, Královopolská 135, 61265 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the CAS, Královopolská 135, 61265 Brno, Czech Republic.
| |
Collapse
|
10
|
Debler RA, Madison CA, Hillbrick L, Gallegos P, Safe S, Chapkin RS, Eitan S. Selective aryl hydrocarbon receptor modulators can act as antidepressants in obese female mice. J Affect Disord 2023; 333:409-419. [PMID: 37084978 PMCID: PMC10561895 DOI: 10.1016/j.jad.2023.04.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/27/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND Obese females are more likely to suffer from depression and are also more likely to be resistant to current medications. This study examined the potential antidepressant-like effects of 1,4-dihydroxy-2-napthoic acid (DHNA), a selective aryl hydrocarbon receptor modulator (SAhRM), in obese female mice. METHODS Obesity was established by feeding C57BL/6N female mice a high fat diet (HFD) for 9-10 weeks. Subsequently, mice were subjected to unpredictable chronic mild stress (UCMS) or remained unstressed. Daily administration of vehicle or 20 mg/kg DHNA began three weeks prior or on the third week of UCMS. Mice were examined for depression-like behaviors (sucrose preference, forced swim test (FST), splash and tape groom tests), anxiety (open-field test, light/dark test, novelty-induced hypophagia), and cognition (object location recognition, novel object recognition, Morris water maze). RESULTS UCMS did not alter, and DHNA slightly increased, weight gain in HFD-fed females. HFD decreased sucrose preference, increased FST immobility time, but did not alter splash and tape tests' grooming time. UCMS did not have additional effects on sucrose preference. UCMS further increased FST immobility time and decreased splash and tape tests' grooming time; these effects were prevented and reversed by DHNA treatment. HFD did not affect behaviors in the cognitive tests. UCMS impaired spatial learning; this effect was not prevented nor reversed by DHNA. CONCLUSIONS DHNA protected against UCMS-induced depression-like behaviors in HFD-fed female mice. DHNA neither improved nor worsened UCMS-induced impairment of spatial learning. Our findings indicate that DHNA has high potential to act as an antidepressant in obese females.
Collapse
Affiliation(s)
- Roanna A Debler
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Caitlin A Madison
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Lauren Hillbrick
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Paula Gallegos
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA.
| |
Collapse
|
11
|
Pinto CJG, Ávila-Gálvez MÁ, Lian Y, Moura-Alves P, Nunes Dos Santos C. Targeting the aryl hydrocarbon receptor by gut phenolic metabolites: A strategy towards gut inflammation. Redox Biol 2023; 61:102622. [PMID: 36812782 PMCID: PMC9958510 DOI: 10.1016/j.redox.2023.102622] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The Aryl Hydrocarbon Receptor (AHR) is a ligand-dependent transcription factor able to control complex transcriptional processes in several cell types, which has been correlated with various diseases, including inflammatory bowel diseases (IBD). Numerous studies have described different compounds as ligands of this receptor, like xenobiotics, natural compounds, and several host-derived metabolites. Dietary (poly)phenols have been studied regarding their pleiotropic activities (e.g., neuroprotective and anti-inflammatory), but their AHR modulatory capabilities have also been considered. However, dietary (poly)phenols are submitted to extensive metabolism in the gut (e.g., gut microbiota). Thus, the resulting gut phenolic metabolites could be key players modulating AHR since they are the ones that reach the cells and may exert effects on the AHR throughout the gut and other organs. This review aims at a comprehensive search for the most abundant gut phenolic metabolites detected and quantified in humans to understand how many have been described as AHR modulators and what could be their impact on inflammatory gut processes. Even though several phenolic compounds have been studied regarding their anti-inflammatory capacities, only 1 gut phenolic metabolite, described as AHR modulator, has been evaluated on intestinal inflammatory models. Searching for AHR ligands could be a novel strategy against IBD.
Collapse
Affiliation(s)
- Catarina J G Pinto
- iNOVA4Health, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa, Lisboa, Portugal; IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - María Ángeles Ávila-Gálvez
- iNOVA4Health, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa, Lisboa, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal
| | - Yilong Lian
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7DQ, Oxford, United Kingdom
| | - Pedro Moura-Alves
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7DQ, Oxford, United Kingdom.
| | - Cláudia Nunes Dos Santos
- iNOVA4Health, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa, Lisboa, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal.
| |
Collapse
|
12
|
Madison CA, Hillbrick L, Kuempel J, Albrecht GL, Landrock KK, Safe S, Chapkin RS, Eitan S. Intestinal epithelium aryl hydrocarbon receptor is involved in stress sensitivity and maintaining depressive symptoms. Behav Brain Res 2023; 440:114256. [PMID: 36528169 PMCID: PMC9839636 DOI: 10.1016/j.bbr.2022.114256] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/03/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a key regulator in the microbiome-gut-brain axis, and AhR-active microbial metabolites modulate multiple neuronal responses. We recently demonstrated that 3,3'-diindolylmethane (DIM) and 1,4-dihydroxy-2-naphthoic acid (DHNA), two selective AhR modulators (SAhRMs), act as antidepressants in female mice. Thus, to examine the role of intestinal AhR in depression, anxiety, and spatial learning, this study employed transgenic mice in which the AhR was knockout only in the intestinal epithelium (AhRΔIEC). Additionally, this study examined whether the antidepressant effects of dietary DIM and DHNA is mediated by intestinal AhR. AhRΔIEC and WT female mice were fed daily with vehicle, 20 mg/kg DIM or DHNA for three weeks prior to four weeks of unpredictable chronic mild stress (UCMS). Mice were examined for weight gain, anhedonia-like behavior (sucrose preference test), anxiety levels (open field, light/dark, elevated plus maze, novelty-induced hypophagia, and marble burying tests), and spatial learning (Morris water maze). UCMS reduced weight gain in AhRΔIECs, but not WTs. Moreover, UCMS initially reduced sucrose preference in both AhRΔIECs and WTs, but over 4 weeks of UCMS, AhRΔIECs develop resilience to UCMS-induced anhedonia. Additionally, AhRΔIECs exhibit slightly reduced anxiety in certain tests and faster spatial learning. DIM and DHNA acted as antidepressants in both AhRΔIECs and WTs. Thus, this study suggests that intestinal AhR plays differential roles, mitigating stress effects on weight gain, and increasing stress effects on mood. However, the site of antidepressant action of SAhRMs, such as DIM and DHNA, is not dependent on the expression of intestinal AhR.
Collapse
Affiliation(s)
- Caitlin A Madison
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Lauren Hillbrick
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Jacob Kuempel
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Georgia Lee Albrecht
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Kerstin K Landrock
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA.
| |
Collapse
|
13
|
Madison CA, Debler RA, Vardeleon NI, Hillbrick L, Jayaraman A, Safe S, Chapkin RS, Eitan S. Sex-dependent differences in the stress mitigating and antidepressant effects of selective aryl hydrocarbon receptor modulators. J Affect Disord 2022; 319:213-220. [PMID: 36206882 PMCID: PMC10391660 DOI: 10.1016/j.jad.2022.09.155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/25/2022] [Accepted: 09/30/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Our recent study demonstrated that selective aryl hydrocarbon receptor modulators (SAhRMs), such as 1,4-dihydroxy-2-napthoic acid (DHNA) act as antidepressants in female mice. Given that some effects of certain SAhRMs are known to also be mediated via estrogen receptor signaling, this study examined whether the effects of SAhRMs on mood, emotional state, and cognition are sex-dependent. METHODS C57BL/6N mice were fed with vehicle or 20 mg/kg DHNA for three weeks prior to four weeks of unpredictable chronic mild stress (UCMS). Mice were examined for depression-like behaviors (sucrose preference, forced swim test (FST), splash test, tape groom test), emotional state (open-field test, light/dark test, marble burying, novelty-induced hypophagia, elevated-plus maze), and cognition (object location recognition, novel object recognition, Morris water maze). RESULTS In females, UCMS decreased sucrose preference and increased FST immobility time; both effects were prevented by DHNA. In males, UCMS increased FST immobility time, and increased the latency to groom in the splash test. These effects were not mitigated by DHNA. However, in males, UCMS induced an increase in novelty-induced locomotion, an increase in the time spent in the light compartment in the L/D test, and an increase in the time spent with an object in a novel location. These effects were prevented by DHNA. CONCLUSIONS Our findings indicate that DHNA has high potential to act as antidepressants in females. However, given classical interpretation, DHNA did not appear to act as an antidepressant in males. Nonetheless, our findings indicate that DHNA can mitigate stress effects and reactivity in males.
Collapse
Affiliation(s)
- Caitlin A Madison
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Roanna A Debler
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Nathan I Vardeleon
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Lauren Hillbrick
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA.
| |
Collapse
|
14
|
An overview of aryl hydrocarbon receptor ligands in the Last two decades (2002–2022): A medicinal chemistry perspective. Eur J Med Chem 2022; 244:114845. [DOI: 10.1016/j.ejmech.2022.114845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/28/2022] [Accepted: 10/08/2022] [Indexed: 11/21/2022]
|
15
|
Gatsios A, Kim CS, York AG, Flavell RA, Crawford JM. Cellular Stress-Induced Metabolites in Escherichia coli. JOURNAL OF NATURAL PRODUCTS 2022; 85:2626-2640. [PMID: 36346625 PMCID: PMC9949963 DOI: 10.1021/acs.jnatprod.2c00706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Escherichia coli isolates commonly inhabit the human microbiota, yet the majority of E. coli's small-molecule repertoire remains uncharacterized. We previously employed erythromycin-induced translational stress to facilitate the characterization of autoinducer-3 (AI-3) and structurally related pyrazinones derived from "abortive" tRNA synthetase reactions in pathogenic, commensal, and probiotic E. coli isolates. In this study, we explored the "missing" tryptophan-derived pyrazinone reaction and characterized two other families of metabolites that were similarly upregulated under erythromycin stress. Strikingly, the abortive tryptophanyl-tRNA synthetase reaction leads to a tetracyclic indole alkaloid metabolite (1) rather than a pyrazinone. Furthermore, erythromycin induced two naphthoquinone-functionalized metabolites (MK-hCys, 2; and MK-Cys, 3) and four lumazines (7-10). Using genetic and metabolite analyses coupled with biomimetic synthesis, we provide support that the naphthoquinones are derived from 4-dihydroxy-2-naphthoic acid (DHNA), an intermediate in the menaquinone biosynthetic pathway, and the amino acids homocysteine and cysteine. In contrast, the lumazines are dependent on a flavin intermediate and α-ketoacids from the aminotransferases AspC and TyrB. We show that one of the lumazine members (9), an indole-functionalized analogue, possesses antioxidant properties, modulates the anti-inflammatory fate of isolated TH17 cells, and serves as an aryl-hydrocarbon receptor (AhR) agonist. These three systems described here serve to illustrate that new metabolic branches could be more commonly derived from well-established primary metabolic pathways.
Collapse
Affiliation(s)
- Alexandra Gatsios
- Department of Chemistry, Yale University, New Haven, Connecticut 06520, United States
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, Connecticut 06516, United States
| | - Chung Sub Kim
- Department of Chemistry, Yale University, New Haven, Connecticut 06520, United States
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, Connecticut 06516, United States
- School of Pharmacy and Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Autumn G. York
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Jason M. Crawford
- Department of Chemistry, Yale University, New Haven, Connecticut 06520, United States
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, Connecticut 06516, United States
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
16
|
Park H, Jin UH, Martin G, Chapkin RS, Davidson LA, Lee K, Jayaraman A, Safe S. Structure-activity relationships among mono- and dihydroxy flavones as aryl hydrocarbon receptor (AhR) agonists or antagonists in CACO2 cells. Chem Biol Interact 2022; 365:110067. [PMID: 35917944 PMCID: PMC9667734 DOI: 10.1016/j.cbi.2022.110067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 12/30/2022]
Abstract
Unsubstituted flavone induced CYP1A1, CYP1B1 and UGT1A1 gene expression in Caco2 cells and was characterized as an aryl hydrocarbon receptor (AhR) agonist. The structure-activity relationships among 15 mono- and dihydroxyflavones showed that addition of one or two hydroxyl groups resulted in active (e.g.: 5- and 6- mono- and 5,6-dihydroxyflavones) and inactive (e.g.: 7-mono, 7,4' and 6,4'-dihydroxyflavones) AhR ligands. Ligand docking studies of flavone, mono- and dihydroxyflavones to the human AhR resulted in similar docking scores that varied from -3.48 to -4.58 kcal/mol and these values did not distinguish between AhR-active and AhR-inactive mono- and dihydroxyflavones. The AhR-inactive flavones were subsequently investigated as AhR antagonists by determining their activities as inhibitors of TCDD-induced expression of CYP1A1, CYP1AA2 and UGT 1A1 gene expression in Caco2 cells. Initial studies with 7,4'-dihydroxyflavone showed that this compound was an AhR antagonist in Caco2 cells and resembled the activity of the classical AhR antagonist CH223191. With few exceptions most of the remaining AhR-inactive compounds in terms of inducing AhR responsive genes were also AhR antagonists. Thus, based on modeling studies, mono- and dihydroxyflavones bind with similar affinities to the AhR and exhibit AhR agonist or antagonist activities, however, the structural requirements (substitution patterns) for predicting these opposing activities were not apparent and could only be determined using bioassays.
Collapse
Affiliation(s)
- Hyejin Park
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA, 77843
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA, 77843
| | - Gregory Martin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA, 77843
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX, USA, 77843
| | - Laurie A Davidson
- Department of Nutrition, Texas A&M University, College Station, TX, USA, 77843
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, USA, 02155
| | - Arul Jayaraman
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA, 77843
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA, 77843.
| |
Collapse
|
17
|
Zhang J, Cui S, Shen L, Gao Y, Liu W, Zhang C, Zhuang S. Promotion of Bladder Cancer Cell Metastasis by 2-Mercaptobenzothiazole via Its Activation of Aryl Hydrocarbon Receptor Transcription: Molecular Dynamics Simulations, Cell-Based Assays, and Machine Learning-Driven Prediction. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:13254-13263. [PMID: 36087060 DOI: 10.1021/acs.est.2c05178] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
2-Mercaptobenzothiazole (MBT) is an industrial chemical widely used for rubber products, corrosion inhibitors, and polymer materials with multiple environmental and exposure pathways. A growing body of evidence suggests its potential bladder cancer (BC) risk as a public health concern; however, the molecular mechanism remains poorly understood. Herein, we demonstrate the activation of the aryl hydrocarbon receptor (AhR) by MBT and reveal key events in carcinogenesis associated with BC. MBT alters conformational changes of AhR ligand binding domain (LBD) as revealed by 500 ns molecular dynamics simulations and activates AhR transcription with upregulation of AhR-target genes CYP1A1 and CYP1B1 to approximately 1.5-fold. MBT upregulates the expression of MMP1, the cancer cell metastasis biomarker, to 3.2-fold and promotes BC cell invasion through an AhR-mediated manner. MBT is further revealed to induce differentially expressed genes (DEGs) most enriched in cancer pathways by transcriptome profiling. The exposure of MBT at environmentally relevant concentrations induces BC risk via AhR signaling disruption, transcriptome aberration, and malignant cell metastasis. A machine learning-based model with an AUC value of 0.881 is constructed to successfully predict 31 MBT analogues. Overall, we provide molecular insight into the BC risk of MBT and develop an effective tool for rapid screening of AhR agonists.
Collapse
Affiliation(s)
- Jiachen Zhang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
- Women's Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Shixuan Cui
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lilai Shen
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuchen Gao
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weiping Liu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunlong Zhang
- Department of Environmental Sciences, University of Houston-Clear Lake, 2700 Bay Area Boulevard, Houston, Texas 77058, United States
| | - Shulin Zhuang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
- Women's Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| |
Collapse
|
18
|
Madison CA, Kuempel J, Albrecht GL, Hillbrick L, Jayaraman A, Safe S, Chapkin RS, Eitan S. 3,3'-Diindolylmethane and 1,4-dihydroxy-2-naphthoic acid prevent chronic mild stress induced depressive-like behaviors in female mice. J Affect Disord 2022; 309:201-210. [PMID: 35461819 PMCID: PMC9153281 DOI: 10.1016/j.jad.2022.04.106] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/06/2022] [Accepted: 04/14/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Current pharmaceutical treatments for depression are sometimes ineffective and may have unwanted side effects that interfere with patient compliance. This study examined the potential antidepressant-like effects of dietary- and microbial-derived aryl hydrocarbon receptor (AhR) ligands, 3,3'-diindolylmethane (DIM) and 1,4-dihydroxy-2-naphthoic acid (1,4-DHNA). METHODS Female C57BL/6 mice were subjected to unpredictable chronic mild stress (UCMS) or were unstressed. For three weeks prior to UCMS mice were fed daily with vehicle or 20 mg/kg DIM, 1,4-DHNA or AhR-inactive isomer 3,7-DHNA; another group was subjected to two weeks UCMS before ligand administration began. Mice were examined for anhedonia-like behavior as measured by the sucrose preference test. Additionally, anxiety levels of the mice were examined before UCMS and ligand administration began and at the end in the open field, light/dark, elevated plus maze, novelty-induced hypophagia, and marble burying tests. At the end of the experiment they were also examined in the Morris water maze (MWM) task. RESULTS Both DIM and 1,4-DHNA, but not 3,7-DHNA, successfully prevented and reversed UCMS-induced anhedonia-like behavior. Furthermore, both DIM and DHNA had little to no effect on anxiety levels and did not induce spatial learning deficits. LIMITATIONS Additional studies are required to determine to what degree the antidepressant-like effects of DIM and 1,4-DHNA can be attributed to their activities as AhR ligands. CONCLUSIONS Our findings indicate that dietary and microbial-derived AhR ligands may have clinical applications as potential antidepressants. Future studies are necessary to elucidate the role of AhR in depression-like states and the underlying mechanisms of action.
Collapse
Affiliation(s)
- Caitlin A Madison
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Jacob Kuempel
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Georgia Lee Albrecht
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Lauren Hillbrick
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA.
| |
Collapse
|
19
|
Current Therapeutic Landscape and Safety Roadmap for Targeting the Aryl Hydrocarbon Receptor in Inflammatory Gastrointestinal Indications. Cells 2022; 11:cells11101708. [PMID: 35626744 PMCID: PMC9139855 DOI: 10.3390/cells11101708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/30/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023] Open
Abstract
Target modulation of the AhR for inflammatory gastrointestinal (GI) conditions holds great promise but also the potential for safety liabilities both within and beyond the GI tract. The ubiquitous expression of the AhR across mammalian tissues coupled with its role in diverse signaling pathways makes development of a “clean” AhR therapeutically challenging. Ligand promiscuity and diversity in context-specific AhR activation further complicates targeting the AhR for drug development due to limitations surrounding clinical translatability. Despite these concerns, several approaches to target the AhR have been explored such as small molecules, microbials, PROTACs, and oligonucleotide-based approaches. These various chemical modalities are not without safety liabilities and require unique de-risking strategies to parse out toxicities. Collectively, these programs can benefit from in silico and in vitro methodologies that investigate specific AhR pathway activation and have the potential to implement thresholding parameters to categorize AhR ligands as “high” or “low” risk for sustained AhR activation. Exploration into transcriptomic signatures for AhR safety assessment, incorporation of physiologically-relevant in vitro model systems, and investigation into chronic activation of the AhR by structurally diverse ligands will help address gaps in our understanding regarding AhR-dependent toxicities. Here, we review the role of the AhR within the GI tract, novel therapeutic modality approaches to target the AhR, key AhR-dependent safety liabilities, and relevant strategies that can be implemented to address drug safety concerns. Together, this review discusses the emerging therapeutic landscape of modalities targeting the AhR for inflammatory GI indications and offers a safety roadmap for AhR drug development.
Collapse
|
20
|
Chapkin RS, Davidson LA, Park H, Jin UH, Fan YY, Cheng Y, Hensel ME, Landrock KK, Allred C, Menon R, Klemashevich C, Jayaraman A, Safe S. Role of the Aryl Hydrocarbon Receptor (AhR) in Mediating the Effects of Coffee in the Colon. Mol Nutr Food Res 2021; 65:e2100539. [PMID: 34406707 PMCID: PMC8530922 DOI: 10.1002/mnfr.202100539] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/26/2021] [Indexed: 12/14/2022]
Abstract
SCOPE This study investigates the mechanism of action and functional effects of coffee extracts in colonic cells, on intestinal stem cell growth, and inhibition of dextran sodium sulfate (DSS)-induced intestinal barrier damage in mice. METHODS AND RESULTS Aqueous coffee extracts induced Ah receptor (AhR) -responsive CYP1A1, CYP1B1, and UGT1A1 gene expression in colon-derived Caco2 and YAMC cells. Tissue-specific AhR knockout (AhRf/f x Lgr5-GFP-CreERT2 x Villin-Cre), wild-type (Lgr5-CreERT2 x Villin-Cre) mice are sources of stem cell enriched organoids and both coffee extracts and norharman, an AhR-active component of these extracts inhibited stem cell growth. Coffee extracts also inhibit DSS-induced damage to intestinal barrier function and DSS-induced mucosal inflammatory genes such as IL-6 and TGF-β1 in wild-type (AhR+/+ ) but not AhR-/- mice. In contrast, coffee does not exhibit protective effects in intestinal-specific AhR knockout mice. Coffee extracts also enhanced overall formation of AhR-active microbial metabolites. CONCLUSIONS In colon-derived cells and in the mouse intestine, coffee induced several AhR-dependent responses including gene expression, inhibition of intestinal stem cell-enriched organoid growth, and inhibition of DSS-induced intestinal barrier damage. We conclude that the anti-inflammatory effects of coffee in the intestine are due, in part, to activation of AhR signaling.
Collapse
Affiliation(s)
- Robert S. Chapkin
- Departments of Nutrition and Biochemistry & Biophysics Texas A&M University, College Station, TX, USA, 77843
| | - Laurie A. Davidson
- Departments of Nutrition and Biochemistry & Biophysics Texas A&M University, College Station, TX, USA, 77843
| | - Hyejin Park
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA, 77843
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA, 77843
| | - Yang-Yi Fan
- Departments of Nutrition and Biochemistry & Biophysics Texas A&M University, College Station, TX, USA, 77843
| | - Yating Cheng
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA, 77843
| | - Martha E. Hensel
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, USA, 77843
| | - Kerstin K. Landrock
- Departments of Nutrition and Biochemistry & Biophysics Texas A&M University, College Station, TX, USA, 77843
| | - Clinton Allred
- Departments of Nutrition and Biochemistry & Biophysics Texas A&M University, College Station, TX, USA, 77843
| | - Rani Menon
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA, 77843
| | - Cory Klemashevich
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA, 77843
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA, 77843
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA, 77843
| |
Collapse
|
21
|
Lujan H, Romer E, Salisbury R, Hussain S, Sayes C. Determining the Biological Mechanisms of Action for Environmental Exposures: Applying CRISPR/Cas9 to Toxicological Assessments. Toxicol Sci 2021; 175:5-18. [PMID: 32105327 DOI: 10.1093/toxsci/kfaa028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Toxicology is a constantly evolving field, especially in the area of developing alternatives to animal testing. Toxicological research must evolve and utilize adaptive technologies in an effort to improve public, environmental, and occupational health. The most commonly cited mechanisms of toxic action after exposure to a chemical or particle test substance is oxidative stress. However, because oxidative stress involves a plethora of genes and proteins, the exact mechanism(s) are not commonly defined. Exact mechanisms of toxicity can be revealed using an emerging laboratory technique referred to as CRISPR (clustered regularly interspaced short palindromic repeats). This article reviews the most common CRISPR techniques utilized today and how each may be applied in Toxicological Sciences. Specifically, the CRISPR/CRISPR-associated protein complex is used for single gene knock-outs, whereas CRISPR interference/activation is used for silencing or activating (respectively) ribonucleic acid. Finally, CRISPR libraries are used for knocking-out entire gene pathways. This review highlights the application of CRISPR in toxicology to elucidate the exact mechanism through which toxicants perturb normal cellular functions.
Collapse
Affiliation(s)
- Henry Lujan
- Department of Environmental Science, Baylor University, Waco, Texas 76706
| | - Eric Romer
- Molecular Bioeffects Branch, Bioeffects Division, 711th Human Performance Wing, Air Force Research Laboratory, Dayton, Ohio 45433
| | - Richard Salisbury
- Molecular Bioeffects Branch, Bioeffects Division, 711th Human Performance Wing, Air Force Research Laboratory, Dayton, Ohio 45433
| | - Saber Hussain
- Molecular Bioeffects Branch, Bioeffects Division, 711th Human Performance Wing, Air Force Research Laboratory, Dayton, Ohio 45433
| | - Christie Sayes
- Department of Environmental Science, Baylor University, Waco, Texas 76706
| |
Collapse
|
22
|
Listeria monocytogenes MenI Encodes a DHNA-CoA Thioesterase Necessary for Menaquinone Biosynthesis, Cytosolic Survival, and Virulence. Infect Immun 2021; 89:IAI.00792-20. [PMID: 33619030 DOI: 10.1128/iai.00792-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/13/2021] [Indexed: 12/14/2022] Open
Abstract
Listeria monocytogenes is a Gram-positive, intracellular pathogen that is highly adapted to invade and replicate in the cytosol of eukaryotic cells. Intermediate metabolites in the menaquinone biosynthesis pathway are essential for the cytosolic survival and virulence of L. monocytogenes, independent of the production of menaquinone (MK) and aerobic respiration. Determining which specific intermediate metabolite(s) are essential for cytosolic survival and virulence has been hindered by the lack of an identified 1,4-dihydroxy-2-naphthoyl-coenzyme A (DHNA-CoA) thioesterase essential for converting DHNA-CoA to DHNA in the MK synthesis pathway. Using the recently identified Escherichia coli DHNA-CoA thioesterase as a query, homology sequence analysis revealed a single homolog in L. monocytogenes, LMRG_02730 Genetic deletion of LMRG_02730 resulted in an ablated membrane potential, indicative of a nonfunctional electron transport chain (ETC) and an inability to aerobically respire. Biochemical kinetic analysis of LMRG_02730 revealed strong activity toward DHNA-CoA, similar to its E. coli homolog, further demonstrating that LMRG_02730 is a DHNA-CoA thioesterase. Functional analyses in vitro, ex vivo, and in vivo using mutants directly downstream and upstream of LMRG_02730 revealed that DHNA-CoA is sufficient to facilitate in vitro growth in minimal medium, intracellular replication, and plaque formation in fibroblasts. In contrast, protection against bacteriolysis in the cytosol of macrophages and tissue-specific virulence in vivo requires the production of 1,4-dihydroxy-2-naphthoate (DHNA). Taken together, these data implicate LMRG_02730 (renamed MenI) as a DHNA-CoA thioesterase and suggest that while DHNA, or an unknown downstream product of DHNA, protects the bacteria from killing in the macrophage cytosol, DHNA-CoA is necessary for intracellular bacterial replication.
Collapse
|
23
|
Park H, Jin UH, Karki K, Allred C, Davidson LA, Chapkin RS, Orr AA, Nowshad F, Jayaraman A, Tamamis P, Safe S. Hydroxylated Chalcones as Aryl Hydrocarbon Receptor Agonists: Structure-Activity Effects. Toxicol Sci 2021; 180:148-159. [PMID: 33263770 PMCID: PMC7916741 DOI: 10.1093/toxsci/kfaa179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Hydroxylated chalcones are phytochemicals which are biosynthetic precursors of flavonoids and their 1,3-diaryl-prop-2-en-1-one structure is used as a scaffold for drug development. In this study, the structure-dependent activation of aryl hydrocarbon receptor (AhR)-responsive CYP1A1, CYP1B1, and UGT1A1 genes was investigated in Caco2 colon cancer cells and in non-transformed young adult mouse colonocytes (YAMC) cells. The effects of a series of di- and trihydroxychalcones as AhR agonists was structure dependent with maximal induction of CYP1A1, CYP1B1, and UGT1A1 in Caco2 cells observed for compounds containing 2,2'-dihydroxy substituents and this included 2,2'-dihydroxy-, 2,2',4'-trihydroxy-, and 2,2',5'-trihydroxychalcones. In contrast, 2',4,5'-, 2'3',4'-, 2',4,4'-trihydroxy, and 2',3-, 2',4-, 2',4'-, and 2',5-dihydroxychalcones exhibited low to non-detectable AhR activity in Caco2 cells. In addition, all of the hydroxychalcones exhibited minimal to non-detectable activity in YAMC cells, whereas 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induced CYP1A1, CYP1B1, and UGT1A1 in Caco2 and YAMC cells. The activity of AhR-active chalcones was confirmed by determining their effects in AhR-deficient Caco2 cells. In addition, 2,2'-dihydroxychalcone induced CYP1A1 protein and formation of an AhR-DNA complex in an in vitro assay. Simulation and modeling studies of hydroxylated chalcones confirmed their interactions with the AhR ligand-binding domain and were consistent with their structure-dependent activity as AhR ligands. Thus, this study identifies hydroxylated chalcones as AhR agonists with potential for these phytochemicals to impact AhR-mediated colonic pathways.
Collapse
Affiliation(s)
- Hyejin Park
- Department of Veterinary Physiology and Pharmacology, College Station, Texas 77843, USA
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, College Station, Texas 77843, USA
| | - Keshav Karki
- Department of Veterinary Physiology and Pharmacology, College Station, Texas 77843, USA
| | - Clinton Allred
- Departments of Nutrition and Biochemistry and Biophysics, College Station, Texas 77843, USA
| | - Laurie A Davidson
- Departments of Nutrition and Biochemistry and Biophysics, College Station, Texas 77843, USA
| | - Robert S Chapkin
- Departments of Nutrition and Biochemistry and Biophysics, College Station, Texas 77843, USA
| | - Asuka A Orr
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Farrhin Nowshad
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Phanourios Tamamis
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, College Station, Texas 77843, USA
| |
Collapse
|
24
|
Han H, Davidson LA, Fan Y, Goldsby JS, Yoon G, Jin U, Wright GA, Landrock KK, Weeks BR, Wright RC, Allred CD, Jayaraman A, Ivanov I, Roper J, Safe SH, Chapkin RS. Loss of aryl hydrocarbon receptor potentiates FoxM1 signaling to enhance self-renewal of colonic stem and progenitor cells. EMBO J 2020; 39:e104319. [PMID: 32915464 PMCID: PMC7527924 DOI: 10.15252/embj.2019104319] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that senses xenobiotics, diet, and gut microbial-derived metabolites, is increasingly recognized as a key regulator of intestinal biology. However, its effects on the function of colonic stem and progenitor cells remain largely unexplored. Here, we observed that inducible deletion of AhR in Lgr5+ stem cells increases the percentage of colonic stem cells and enhances organoid initiating capacity and growth of sorted stem and progenitor cells, while AhR activation has the opposite effect. Moreover, intestinal-specific AhR knockout increases basal stem cell and crypt injury-induced cell proliferation and promotes colon tumorigenesis in a preclinical colitis-associated tumor model by upregulating FoxM1 signaling. Mechanistically, AhR transcriptionally suppresses FoxM1 expression. Activation of AhR in human organoids recapitulates phenotypes observed in mice, such as reduction in the percentage of colonic stem cells, promotion of stem cell differentiation, and attenuation of FoxM1 signaling. These findings indicate that the AhR-FoxM1 axis, at least in part, mediates colonic stem/progenitor cell behavior.
Collapse
Affiliation(s)
- Huajun Han
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of Biochemistry & BiophysicsTexas A&M UniversityCollege StationTXUSA
| | - Laurie A Davidson
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Yang‐Yi Fan
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Jennifer S Goldsby
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Grace Yoon
- Department of StatisticsTexas A&M UniversityCollege StationTXUSA
| | - Un‐Ho Jin
- Veterinary Physiology and PharmacologyTexas A&M UniversityCollege StationTXUSA
| | - Gus A Wright
- Department of Veterinary PathobiologyTexas A&M UniversityCollege StationTXUSA
| | - Kerstin K Landrock
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Bradley R Weeks
- Department of Veterinary PathobiologyTexas A&M UniversityCollege StationTXUSA
| | - Rachel C Wright
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | | | - Arul Jayaraman
- Department of Chemical EngineeringTexas A&M UniversityCollege StationTXUSA
| | - Ivan Ivanov
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Veterinary Physiology and PharmacologyTexas A&M UniversityCollege StationTXUSA
| | - Jatin Roper
- Department of MedicineDivision of GastroenterologyDuke University School of MedicineDurhamNCUSA
| | - Stephen H Safe
- Veterinary Physiology and PharmacologyTexas A&M UniversityCollege StationTXUSA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of Biochemistry & BiophysicsTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| |
Collapse
|
25
|
Faber SC, Giani Tagliabue S, Bonati L, Denison MS. The Cellular and Molecular Determinants of Naphthoquinone-Dependent Activation of the Aryl Hydrocarbon Receptor. Int J Mol Sci 2020; 21:ijms21114111. [PMID: 32526934 PMCID: PMC7312509 DOI: 10.3390/ijms21114111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/28/2020] [Accepted: 06/05/2020] [Indexed: 12/30/2022] Open
Abstract
1,2-naphthoquinone (1,2-NQ) and 1,4-naphthoquinone (1,4-NQ) are clinically promising biologically active chemicals that have been shown to stimulate the aryl hydrocarbon receptor (AhR) signaling pathway, but whether they are direct or indirect ligands or activate the AhR in a ligand-independent manner is unknown. Given the structural diversity of AhR ligands, multiple mechanisms of AhR activation of gene expression, and species differences in AhR ligand binding and response, we examined the ability of 1,2-NQ and 1,4-NQ to bind to and activate the mouse and human AhRs using a series of in vitro AhR-specific bioassays and in silico modeling techniques. Both NQs induced AhR-dependent gene expression in mouse and human hepatoma cells, but were more potent and efficacious in human cells. 1,2-NQ and 1,4-NQ stimulated AhR transformation and DNA binding in vitro and was inhibited by AhR antagonists. Ligand binding analysis confirmed the ability of 1,2-NQ and 1,4-NQ to competitively bind to the AhR ligand binding cavity and the molecular determinants for interactions were predicted by molecular modeling methods. NQs were shown to bind distinctly differently from that of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and differences were also observed between species. Mutation of amino acid residues (F289, M334, and M342) involved in critical NQ:AhR binding interactions, decreased NQ- and AhR-dependent gene expression, consistent with a role for these residues in binding and activation of the AhR by NQs. These studies provide insights into the molecular mechanism of action of NQs and contribute to the development of emerging NQ-based therapeutics.
Collapse
Affiliation(s)
- Samantha C. Faber
- Department of Environmental Toxicology, University of California, Davis, CA 95616, USA;
| | - Sara Giani Tagliabue
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, 20126 Milan, Italy; (S.G.T.); (L.B.)
| | - Laura Bonati
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, 20126 Milan, Italy; (S.G.T.); (L.B.)
| | - Michael S. Denison
- Department of Environmental Toxicology, University of California, Davis, CA 95616, USA;
- Correspondence: ; Tel.: +1-(530)-752-3879
| |
Collapse
|
26
|
Liu Y, Wei Y, Zhang S, Yan X, Zhu H, Xu L, Zhao B, Xie HQ, Yan B. Regulation of Aryl Hydrocarbon Receptor Signaling Pathway and Dioxin Toxicity by Novel Agonists and Antagonists. Chem Res Toxicol 2020; 33:614-624. [PMID: 31878777 DOI: 10.1021/acs.chemrestox.9b00431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dioxins, mostly through activation of aryl hydrocarbon receptor (AhR), are potent toxic substances widely distributed in the environment, while moderated suppression of AhR also exhibits anti-tumor effects. Therefore, the proper modulation of AhR activity may counteract AhR-mediated toxicities and certain diseases. In this investigation, we identified several novel AhR moderate agonists and antagonists using chemical biology approaches. The mechanisms and mode of interactions with AhR by these hits were also revealed using both experimental and computational studies. The newly identified AhR moderate agonists and antagonists were predicted to bind to AhR and modulate AhR signaling. The structure-activity relationships of moderate agonists and antagonists and their unique binding features with AhR have created a solid framework for further optimization of the next generation of AhR modulators.
Collapse
Affiliation(s)
- Yin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yongyi Wei
- Institute of Environmental Research at Greater Bay, Guangzhou University, Key Laboratory for Water Quality and Conservation of the Pearl River Delta , Ministry of Education , Guangzhou 510006 , China.,School of Environmental Science and Engineering , Shandong University , Qingdao 266237 , China
| | - Songyan Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Xiliang Yan
- School of Environmental Science and Engineering , Shandong University , Qingdao 266237 , China.,The Rutgers Center for Computational and Integrative Biology , Camden , New Jersey 08102 , United States
| | - Hao Zhu
- The Rutgers Center for Computational and Integrative Biology , Camden , New Jersey 08102 , United States
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Bing Yan
- Institute of Environmental Research at Greater Bay, Guangzhou University, Key Laboratory for Water Quality and Conservation of the Pearl River Delta , Ministry of Education , Guangzhou 510006 , China.,School of Environmental Science and Engineering , Shandong University , Qingdao 266237 , China
| |
Collapse
|
27
|
Chen GY, Kao CY, Smith HB, Rust DP, Powers ZM, Li AY, Sauer JD. Mutation of the Transcriptional Regulator YtoI Rescues Listeria monocytogenes Mutants Deficient in the Essential Shared Metabolite 1,4-Dihydroxy-2-Naphthoate (DHNA). Infect Immun 2019; 88:e00366-19. [PMID: 31685546 PMCID: PMC6921671 DOI: 10.1128/iai.00366-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/17/2019] [Indexed: 12/30/2022] Open
Abstract
Listeria monocytogenes, a Gram-positive, facultative intracellular pathogen, survives and replicates in the cytosol of host cells. Synthesis of 1,4-dihydroxy-2-naphthoate (DHNA), an intermediate of menaquinone biosynthesis, is essential for cytosolic survival of L. monocytogenes independent from its role in respiration. Here, we demonstrate that DHNA is essential for virulence in a murine model of listeriosis due to both respiration-dependent and -independent functions. In addition, DHNA can be both secreted and utilized as an extracellular shared metabolite to promote cytosolic survival inside host macrophages. To understand the role(s) of DHNA in L. monocytogenes intracellular survival and virulence, we isolated DHNA-deficient (ΔmenD strain) suppressor mutants that formed plaques in monolayers of fibroblasts. Five ΔmenD suppressor (mds) mutants additionally rescued at least 50% of the cytosolic survival defect of the parent ΔmenD mutant. Whole-genome sequencing revealed that four of the five suppressor mutants had independent missense mutations in a putative transcriptional regulator, ytoI (lmo1576). Clean deletion and complementation in trans confirmed that loss of ytoI could restore plaquing and cytosolic survival of DHNA-deficient L. monocytogenes RNA-seq transcriptome analysis revealed five genes (lmo0944, lmo1575, lmo1577, lmo2005, and lmo2006) expressed at a higher level in the ΔytoI strain than in the wild-type strain, whereas two genes (lmo1917 and lmo2103) demonstrated lower expression in the ΔytoI mutant. Intriguingly, the majority of these genes are involved in controlling pyruvate flux. Metabolic analysis confirmed that acetoin, acetate, and lactate flux were altered in a ΔytoI mutant, suggesting a critical role for regulating these metabolic programs. In conclusion, we have demonstrated that, similar to findings in select other bacteria, DHNA can act as a shared resource, and it is essential for cytosolic survival and virulence of L. monocytogenes Furthermore, we have identified a novel transcriptional regulator in L. monocytogenes and determined that its metabolic regulation is implicated in cytosolic survival of L. monocytogenes.
Collapse
Affiliation(s)
- Grischa Y Chen
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison Wisconsin, USA
| | - Cheng-Yen Kao
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison Wisconsin, USA
| | - Hans B Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison Wisconsin, USA
| | - Drew P Rust
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison Wisconsin, USA
| | - Zachary M Powers
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison Wisconsin, USA
| | - Alexandria Y Li
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison Wisconsin, USA
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison Wisconsin, USA
| |
Collapse
|
28
|
Park H, Jin UH, Orr AA, Echegaray SP, Davidson LA, Allred CD, Chapkin RS, Jayaraman A, Lee K, Tamamis P, Safe S. Isoflavones as Ah Receptor Agonists in Colon-Derived Cell Lines: Structure-Activity Relationships. Chem Res Toxicol 2019; 32:2353-2364. [PMID: 31621310 PMCID: PMC6938648 DOI: 10.1021/acs.chemrestox.9b00352] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many of the protective responses observed for flavonoids in the gastrointestinal track resemble aryl hydrocarbon receptor (AhR)-mediated effects. Therefore, we examined the structure-activity relationships of isoflavones and isomeric flavone and flavanones as AhR ligands on the basis of their induction of CYP1A1, CYP1B1, and UGT1A1 gene expression in colon cancer Caco2 cells and young adult mouse colonocyte (YAMC) cells. Caco2 cells were significantly more Ah-responsive than YAMC cells, and this was due, in part, to flavonoid-induced cytotoxicity in the latter cell lines. The structure-activity relationships for the flavonoids were complex and both response and cell context specific; however, there was significant variability in the AhR activities of the isomeric substituted isoflavones and flavones. For example, 4',5,7-trihydroxyisoflavone (genistein) was AhR-inactive whereas 4',5,7-trihydroxyflavone (apigenin) induced CYP1A1, CYP1B1, and UGT1A1 in Caco2 cells. In contrast, both 5,7-dihydroxy-4-methoxy substituted isoflavone (biochanin A) and flavone (acacetin) induced all three AhR-responsive genes; 4',5,7-trimethoxyisoflavone was a potent AhR agonist, and the isomeric flavone was AhR-inactive. These results coupled with simulation studies modeling flavonoid interaction within the AhR binding pocket demonstrate that the orientation of the substituted phenyl ring at C-2 (flavones) or C-3 (isoflavones) on the common 4-H-chromen-4-one ring strongly influences the activities of isoflavones and flavones as AhR agonists.
Collapse
Affiliation(s)
- Hyejin Park
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, United States
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, United States
| | - Asuka A. Orr
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Stephanie P. Echegaray
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Laurie A. Davidson
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843, United States
| | - Clinton D. Allred
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843, United States
| | - Robert S. Chapkin
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843, United States
| | - Arul Jayaraman
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Phanourios Tamamis
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
29
|
Giani Tagliabue S, Faber SC, Motta S, Denison MS, Bonati L. Modeling the binding of diverse ligands within the Ah receptor ligand binding domain. Sci Rep 2019; 9:10693. [PMID: 31337850 PMCID: PMC6650409 DOI: 10.1038/s41598-019-47138-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 07/02/2019] [Indexed: 01/08/2023] Open
Abstract
The Ah receptor (AhR) is a ligand-dependent transcription factor belonging to the basic helix-loop-helix Per-Arnt-Sim (bHLH-PAS) superfamily. Binding to and activation of the AhR by a variety of chemicals results in the induction of expression of diverse genes and production of a broad spectrum of biological and toxic effects. The AhR also plays important roles in several physiological responses, which has led it to become a novel target for the development of therapeutic drugs. Differences in the interactions of various ligands within the AhR ligand binding domain (LBD) may contribute to differential modulation of AhR functionality. We combined computational and experimental analyses to investigate the binding modes of a group of chemicals representative of major classes of AhR ligands. On the basis of a novel computational approach for molecular docking to the homology model of the AhR LBD that includes the receptor flexibility, we predicted specific residues within the AhR binding cavity that play a critical role in binding of three distinct groups of chemicals. The prediction was validated by site-directed mutagenesis and evaluation of the relative ligand binding affinities for the mutant AhRs. These results provide an avenue for understanding ligand modulation of the AhR functionality and for rational drug design.
Collapse
Affiliation(s)
- Sara Giani Tagliabue
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Samantha C Faber
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stefano Motta
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Michael S Denison
- Department of Environmental Toxicology, University of California, Davis, CA, USA
| | - Laura Bonati
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy.
| |
Collapse
|
30
|
Yoon K, Chen CC, Orr AA, Barreto PN, Tamamis P, Safe S. Activation of COUP-TFI by a Novel Diindolylmethane Derivative. Cells 2019; 8:220. [PMID: 30866413 PMCID: PMC6468570 DOI: 10.3390/cells8030220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
Chicken ovalbumin upstream promoter-transcription factor I (COUP-TFI) is an orphan receptor and member of the nuclear receptor superfamily. Among a series of methylene substituted diindolylmethanes (C-DIMs) containing substituted phenyl and heteroaromatic groups, we identified 1,1-bis(3'-indolyl)-1-(4-pyridyl)-methane (DIM-C-Pyr-4) as an activator of COUP-TFI. Structure activity studies with structurally diverse heteroaromatic C-DIMs showed that the pyridyl substituted compound was active and the 4-pyridyl substituent was more potent than the 2- or 3-pyridyl analogs in transactivation assays in breast cancer cells. The DIM-C-Pyr-4 activated chimeric GAL4-COUP-TFI constructs containing full length, C- or N-terminal deletions, and transactivation was inhibited by phosphatidylinositol-3-kinase and protein kinase A inhibitors. However, DIM-C-Pyr-4 also induced transactivation and interactions of COUP-TFI and steroid receptor coactivators-1 and -2 in mammalian two-hybrid assays, and ligand-induced interactions of the C-terminal region of COUP-TFI were not affected by kinase inhibitors. We also showed that DIM-C-Pyr-4 activated COUP-TFI-dependent early growth response 1 (Egr-1) expression and this response primarily involved COUP-TFI interactions with Sp3 and to a lesser extent Sp1 bound to the proximal region of the Egr-1 promoter. Modeling studies showed interactions of DIM-C-Pyr-4 within the ligand binding domain of COUP-TFI. This report is the first to identify a COUP-TFI agonist and demonstrate activation of COUP-TFI-dependent Egr-1 expression.
Collapse
Affiliation(s)
- Kyungsil Yoon
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA.
- Division of Translational Science, National Cancer Center, Goyang-si, Gyeonggi-do 10408, Korea.
| | - Chien-Cheng Chen
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA.
| | - Asuka A Orr
- Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA.
| | - Patricia N Barreto
- Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA.
| | - Phanourios Tamamis
- Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA.
| | - Stephen Safe
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA.
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
31
|
Karlgren M, Simoff I, Keiser M, Oswald S, Artursson P. CRISPR-Cas9: A New Addition to the Drug Metabolism and Disposition Tool Box. Drug Metab Dispos 2018; 46:1776-1786. [PMID: 30126863 DOI: 10.1124/dmd.118.082842] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/03/2018] [Indexed: 02/06/2023] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated protein 9 (Cas9), i.e., CRISPR-Cas9, has been extensively used as a gene-editing technology during recent years. Unlike earlier technologies for gene editing or gene knockdown, such as zinc finger nucleases and RNA interference, CRISPR-Cas9 is comparably easy to use, affordable, and versatile. Recently, CRISPR-Cas9 has been applied in studies of drug absorption, distribution, metabolism, and excretion (ADME) and for ADME model generation. To date, about 50 papers have been published describing in vitro or in vivo CRISPR-Cas9 gene editing of ADME and ADME-related genes. Twenty of these papers describe gene editing of clinically relevant genes, such as ATP-binding cassette drug transporters and cytochrome P450 drug-metabolizing enzymes. With CRISPR-Cas9, the ADME tool box has been substantially expanded. This new technology allows us to develop better and more predictive in vitro and in vivo ADME models and map previously underexplored ADME genes and gene families. In this mini-review, we give an overview of the CRISPR-Cas9 technology and summarize recent applications of CRISPR-Cas9 within the ADME field. We also speculate about future applications of CRISPR-Cas9 in ADME research.
Collapse
Affiliation(s)
- M Karlgren
- Department of Pharmacy (M.Ka., P.A.), Uppsala University Drug Optimization and Pharmaceutical Profiling Platform, Department of Pharmacy (I.S.), and Science for Life Laboratory (P.A.), Uppsala University, Uppsala, Sweden; and Department of Clinical Pharmacology, Center of Drug Absorption and Transport, University Medicine of Greifswald, Germany (M.Ke., S.O.)
| | - I Simoff
- Department of Pharmacy (M.Ka., P.A.), Uppsala University Drug Optimization and Pharmaceutical Profiling Platform, Department of Pharmacy (I.S.), and Science for Life Laboratory (P.A.), Uppsala University, Uppsala, Sweden; and Department of Clinical Pharmacology, Center of Drug Absorption and Transport, University Medicine of Greifswald, Germany (M.Ke., S.O.)
| | - M Keiser
- Department of Pharmacy (M.Ka., P.A.), Uppsala University Drug Optimization and Pharmaceutical Profiling Platform, Department of Pharmacy (I.S.), and Science for Life Laboratory (P.A.), Uppsala University, Uppsala, Sweden; and Department of Clinical Pharmacology, Center of Drug Absorption and Transport, University Medicine of Greifswald, Germany (M.Ke., S.O.)
| | - S Oswald
- Department of Pharmacy (M.Ka., P.A.), Uppsala University Drug Optimization and Pharmaceutical Profiling Platform, Department of Pharmacy (I.S.), and Science for Life Laboratory (P.A.), Uppsala University, Uppsala, Sweden; and Department of Clinical Pharmacology, Center of Drug Absorption and Transport, University Medicine of Greifswald, Germany (M.Ke., S.O.)
| | - P Artursson
- Department of Pharmacy (M.Ka., P.A.), Uppsala University Drug Optimization and Pharmaceutical Profiling Platform, Department of Pharmacy (I.S.), and Science for Life Laboratory (P.A.), Uppsala University, Uppsala, Sweden; and Department of Clinical Pharmacology, Center of Drug Absorption and Transport, University Medicine of Greifswald, Germany (M.Ke., S.O.)
| |
Collapse
|
32
|
Orr AA, Shaykhalishahi H, Mirecka EA, Jonnalagadda SVR, Hoyer W, Tamamis P. Elucidating the multi-targeted anti-amyloid activity and enhanced islet amyloid polypeptide binding of β-wrapins. Comput Chem Eng 2018; 116:322-332. [PMID: 30405276 PMCID: PMC6217933 DOI: 10.1016/j.compchemeng.2018.02.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
β-wrapins are engineered binding proteins stabilizing the β-hairpin conformations of amyloidogenic proteins islet amyloid polypeptide (IAPP), amyloid-β, and α-synuclein, thus inhibiting their amyloid propensity. Here, we use computational and experimental methods to investigate the molecular recognition of IAPP by β-wrapins. We show that the multi-targeted, IAPP, amyloid-β, and α-synuclein, binding properties of β-wrapins originate mainly from optimized interactions between β-wrapin residues and sets of residues in the three amyloidogenic proteins with similar physicochemical properties. Our results suggest that IAPP is a comparatively promiscuous β-wrapin target, probably due to the low number of charged residues in the IAPP β-hairpin motif. The sub-micromolar affinity of β-wrapin HI18, specifically selected against IAPP, is achieved in part by salt-bridge formation between HI18 residue Glu10 and the IAPP N-terminal residue Lys1, both located in the flexible N-termini of the interacting proteins. Our findings provide insights towards developing novel protein-based single- or multi-targeted therapeutics.
Collapse
Affiliation(s)
- Asuka A. Orr
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Hamed Shaykhalishahi
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf 40204, Germany
| | - Ewa A. Mirecka
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf 40204, Germany
| | - Sai Vamshi R. Jonnalagadda
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Wolfgang Hoyer
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf 40204, Germany
- Institute of Structural Biochemistry (ICS-6), Research Centre Jülich, Jülich 52425, Germany
| | - Phanourios Tamamis
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
| |
Collapse
|
33
|
Orr AA, Gonzalez-Rivera JC, Wilson M, Bhikha PR, Wang D, Contreras LM, Tamamis P. A high-throughput and rapid computational method for screening of RNA post-transcriptional modifications that can be recognized by target proteins. Methods 2018; 143:34-47. [DOI: 10.1016/j.ymeth.2018.01.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/14/2018] [Accepted: 01/26/2018] [Indexed: 12/25/2022] Open
|
34
|
Jin UH, Park H, Li X, Davidson LA, Allred C, Patil B, Jayaprakasha G, Orr AA, Mao L, Chapkin RS, Jayaraman A, Tamamis P, Safe S. Structure-Dependent Modulation of Aryl Hydrocarbon Receptor-Mediated Activities by Flavonoids. Toxicol Sci 2018; 164:205-217. [PMID: 29584932 PMCID: PMC6016704 DOI: 10.1093/toxsci/kfy075] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Dietary flavonoids are used in treatment of multiple diseases, and their antiinflammatory effects in the intestine are due, in part, to interactions with gut microflora and possibly due to modulation of aryl hydrocarbon receptor (AhR) signaling. In this study, we investigated the structure-dependent AhR activity of 14 flavonoids in Caco2 colon cancer cells using induction of CYP1A1 and UGT1A1 gene expression as endpoints. A major structural determinant for AhR activation was the number of hydroxyl groups where pentahydroxyflavonoids (with the exception of morin) > hexahydroxyflavonoids > tetra-/trihydroxyflavonoids, and some of the latter compounds such as apigenin exhibited AhR antagonist activity for induction of CYP1A1. Simulations suggest that while quercetin and apigenin interact primarily with the same residues, the strength of interactions between specific AhR residues with CYP1A1 agonist, quercetin, in comparison with CYP1A1 antagonist, apigenin, is different; thus, such interactions are presumably indicative of potential switches for modulating CYP1A1 activity. The structure-dependent effects of the hydroxyl flavonoids on induction of UGT1A1 were similar to that observed for induction of CYP1A1 except that luteolin and apigenin induced UGT1A1 levels similar to that observed for TCDD, whereas both compounds were AhR antagonists for CYP1A1. Thus, the effects of the flavonoids in Caco2 cells on Ah-responsiveness and interactions with butyrate were both ligand structure- and response-dependent and these activities are consistent with hydroxyflavonoids being selective AhR modulators.
Collapse
Affiliation(s)
- Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology
| | - Hyejin Park
- Department of Veterinary Physiology and Pharmacology
| | - Xi Li
- Department of Veterinary Physiology and Pharmacology
| | | | | | | | | | - Asuka A Orr
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843
| | - Leevin Mao
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843
| | | | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843
| | - Phanourios Tamamis
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology
| |
Collapse
|
35
|
Mohan R, Wilson M, Gorham RD, Harrison RES, Morikis VA, Kieslich CA, Orr AA, Coley AV, Tamamis P, Morikis D. Virtual Screening of Chemical Compounds for Discovery of Complement C3 Ligands. ACS OMEGA 2018; 3:6427-6438. [PMID: 30221234 PMCID: PMC6130793 DOI: 10.1021/acsomega.8b00606] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/04/2018] [Indexed: 06/08/2023]
Abstract
The complement system is our first line of defense against foreign pathogens, but when it is not properly regulated, complement is implicated in the pathology of several autoimmune and inflammatory disorders. Compstatin is a peptidic complement inhibitor that acts by blocking the cleavage of complement protein C3 to the proinflammatory fragment C3a and opsonin fragment C3b. In this study, we aim to identify druglike small-molecule complement inhibitors with physicochemical, geometric, and binding properties similar to those of compstatin. We employed two approaches using various high-throughput virtual screening methods, which incorporate molecular dynamics (MD) simulations, pharmacophore model design, energy calculations, and molecular docking and scoring. We have generated a library of 274 chemical compounds with computationally predicted binding affinities for the compstatin binding site of C3. We have tested subsets of these chemical compounds experimentally for complement inhibitory activity, using hemolytic assays, and for binding affinity, using microscale thermophoresis. As a result, although none of the compounds showed inhibitory activity, compound 29 was identified to exhibit weak competitive binding against a potent compstatin analogue, therefore validating our computational approaches. Additional docking and MD simulation studies suggest that compound 29 interacts with C3 residues, which have been shown to be important in binding of compstatin to the C3c fragment of C3. Compound 29 is amenable to physicochemical optimization to acquire inhibitory properties. Additionally, it is possible that some of the untested compounds will demonstrate binding and inhibition in future experimental studies.
Collapse
Affiliation(s)
- Rohith
R. Mohan
- Department
of Bioengineering, University of California,
Riverside, 900 University
Avenue, Riverside, California 92521, United States
| | - Mark Wilson
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, 3122 TAMU, College Station, Texas 77843, United
States
| | - Ronald D. Gorham
- Department
of Bioengineering, University of California,
Riverside, 900 University
Avenue, Riverside, California 92521, United States
| | - Reed E. S. Harrison
- Department
of Bioengineering, University of California,
Riverside, 900 University
Avenue, Riverside, California 92521, United States
| | - Vasilios A. Morikis
- Department
of Bioengineering, University of California,
Riverside, 900 University
Avenue, Riverside, California 92521, United States
| | - Chris A. Kieslich
- Department
of Bioengineering, University of California,
Riverside, 900 University
Avenue, Riverside, California 92521, United States
| | - Asuka A. Orr
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, 3122 TAMU, College Station, Texas 77843, United
States
| | - Alexis V. Coley
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, 3122 TAMU, College Station, Texas 77843, United
States
| | - Phanourios Tamamis
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, 3122 TAMU, College Station, Texas 77843, United
States
| | - Dimitrios Morikis
- Department
of Bioengineering, University of California,
Riverside, 900 University
Avenue, Riverside, California 92521, United States
| |
Collapse
|
36
|
Dolciami D, Gargaro M, Cerra B, Scalisi G, Bagnoli L, Servillo G, Fazia MAD, Puccetti P, Quintana FJ, Fallarino F, Macchiarulo A. Binding Mode and Structure-Activity Relationships of ITE as an Aryl Hydrocarbon Receptor (AhR) Agonist. ChemMedChem 2018; 13:270-279. [PMID: 29266750 DOI: 10.1002/cmdc.201700669] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/15/2017] [Indexed: 12/13/2022]
Abstract
Discovered as a modulator of the toxic response to environmental pollutants, aryl hydrocarbon receptor (AhR) has recently gained attention for its involvement in various physiological and pathological pathways. AhR is a ligand-dependent transcription factor activated by a large array of chemical compounds, which include metabolites of l-tryptophan (l-Trp) catabolism as endogenous ligands of the receptor. Among these, 2-(1'H-indole-3'-carbonyl)thiazole-4-carboxylic acid methyl ester (ITE) has attracted interest in the scientific community, being endowed with nontoxic, immunomodulatory, and anticancer AhR-mediated functions. So far, no information about the binding mode and interactions of ITE with AhR is available. In this study, we used docking and molecular dynamics to propose a putative binding mode of ITE into the ligand binding pocket of AhR. Mutagenesis studies were then instrumental in validating the proposed binding mode, identifying His 285 and Tyr 316 as important key residues for ligand-dependent receptor activation. Finally, a set of ITE analogues was synthesized and tested to further probe molecular interactions of ITE to AhR and characterize the relevance of specific functional groups in the chemical structure for receptor activity.
Collapse
Affiliation(s)
- Daniela Dolciami
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Marco Gargaro
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Bruno Cerra
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Giulia Scalisi
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Luana Bagnoli
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Giuseppe Servillo
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Maria Agnese Della Fazia
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Francesca Fallarino
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| |
Collapse
|
37
|
Abstract
Docking algorithms have been widely used to elucidate ligand:receptor interactions that are important in biological function. Here, we introduce an in-house developed docking-refinement protocol that combines the following innovative features. (1) The use of multiple short molecular dynamics (MD) docking simulations, with residues within the binding pocket of the receptor unconstrained, so that the binding modes of the ligand in the binding pocket may be exhaustively examined. (2) The initial positioning of the ligand within the binding pocket based on complementary shape, and the use of both harmonic and quartic spherical potentials to constrain the ligand in the binding pocket during multiple short docking simulations. (3) The selection of the most probable binding modes generated by the short docking simulations using interaction energy calculations, as well as the subsequent application of all-atom MD simulations and physical-chemistry based free energy calculations to elucidate the most favorable binding mode of the ligand in complex with the receptor. In this chapter, we provide step-by-step instructions on how to computationally investigate the binding of small-molecule ligands to protein receptors by examining as control and test cases, respectively, the binding of L-serine and R-3,4-dihydroxymandelic acid (R-DHMA) to the Escherichia coli chemoreceptor Tsr. Similar computational strategies can be used for the molecular modeling of a series of ligand:protein receptor interactions.
Collapse
|
38
|
Safe S. Carbidopa: a selective Ah receptor modulator (SAhRM). Biochem J 2017; 474:3763-3765. [PMID: 29109131 DOI: 10.1042/bcj20170728] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 09/25/2017] [Accepted: 09/27/2017] [Indexed: 01/23/2023]
Abstract
The aryl hydrocarbon receptor (AhR) was discovered as the intracellular receptor that bound with high affinity to the environmental toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), and the AhR is required for mediating the toxicity induced by TCDD. Subsequent studies show that the AhR binds structurally diverse chemicals including plant-derived compounds that promote health and several AhR-active pharmaceuticals that exhibit anticancer activity. In this issue, there is a report that carbidopa, a drug used for treating Parkinson's disease, is also an AhR ligand, and this compound inhibits pancreatic cancer cell and tumor growth. These results are consistent with activities of other AhR-active compounds that inhibit carcinogenesis. Like carbidopa, these chemicals are selective AhR modulators with potential clinical applications that are AhR-dependent.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, U.S.A.
| |
Collapse
|
39
|
Jin UH, Cheng Y, Park H, Davidson LA, Callaway ES, Chapkin RS, Jayaraman A, Asante A, Allred C, Weaver EA, Safe S. Short Chain Fatty Acids Enhance Aryl Hydrocarbon (Ah) Responsiveness in Mouse Colonocytes and Caco-2 Human Colon Cancer Cells. Sci Rep 2017; 7:10163. [PMID: 28860561 PMCID: PMC5579248 DOI: 10.1038/s41598-017-10824-x] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/15/2017] [Indexed: 02/07/2023] Open
Abstract
Aryl hydrocarbon receptor (AhR) ligands are important for gastrointestinal health and play a role in gut inflammation and the induction of T regulatory cells, and the short chain fatty acids (SCFAs) butyrate, propionate and acetate also induce similar protective responses. Initial studies with butyrate demonstrated that this compound significantly increased expression of Ah-responsive genes such as Cyp1a1/CYP1A1 in YAMC mouse colonocytes and Caco-2 human colon cancer cell lines. Butyrate synergistically enhanced AhR ligand-induced Cyp1a1/CYP1A1 in these cells with comparable enhancement being observed for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and also microbiota-derived AhR ligands tryptamine, indole and 1,4-dihydroxy-2-naphthoic acid (DHNA). The effects of butyrate on enhancing induction of Cyp1b1/CYP1B1, AhR repressor (Ahrr/AhRR) and TCDD-inducible poly(ADP-ribose)polymerase (Tiparp/TiPARP) by AhR ligands were gene- and cell context-dependent with the Caco-2 cells being the most responsive cell line. Like butyrate and propionate, the prototypical hydroxyamic acid-derived histone deacetylase (HDAC) inhibitors Panobinostat and Vorinostat also enhanced AhR ligand-mediated induction and this was accompanied by enhanced histone acetylation. Acetate also enhanced basal and ligand-inducible Ah responsiveness and histone acetylation, demonstrating that acetate was an HDAC inhibitor. These results demonstrate SCFA-AhR ligand interactions in YAMC and Caco-2 cells where SCFAs synergistically enhance basal and ligand-induced expression of AhR-responsive genes.
Collapse
Affiliation(s)
- Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Yating Cheng
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Hyejin Park
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Laurie A Davidson
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Evelyn S Callaway
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Robert S Chapkin
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Arul Jayaraman
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Andrew Asante
- Department of Biology, Alabama State University, Montgomery, AL, 36101, USA
| | - Clinton Allred
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Evelyn A Weaver
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|