651
|
Kwiatkowska I, Hermanowicz JM, Mysliwiec M, Pawlak D. Oxidative Storm Induced by Tryptophan Metabolites: Missing Link between Atherosclerosis and Chronic Kidney Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6656033. [PMID: 33456671 PMCID: PMC7787774 DOI: 10.1155/2020/6656033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 02/08/2023]
Abstract
Chronic kidney disease (CKD) occurrence is rising all over the world. Its presence is associated with an increased risk of premature death from cardiovascular disease (CVD). Several explanations of this link have been put forward. It is known that in renal failure, an array of metabolites cannot be excreted, and they accumulate in the organism. Among them, some are metabolites of tryptophan (TRP), such as indoxyl sulfate and kynurenine. Scientists have become interested in them in the context of inducing vascular damage in the course of chronic kidney impairment. Experimental evidence suggests the involvement of TRP metabolites in the progression of chronic kidney disease and atherosclerosis separately and point to oxidative stress generation as one of the main mechanisms that is responsible for worsening those states. Since it is known that blood levels of those metabolites increase significantly in renal failure and that they generate reactive oxygen species (ROS), which lead to endothelial injury, it is reasonable to suspect that products of TRP metabolism are the missing link in frequently occurring atherosclerosis in CKD patients. This review focuses on reports that shed a light on TRP metabolites as contributing factors to vascular damage in the progression of impaired kidney function.
Collapse
Affiliation(s)
- Iwona Kwiatkowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
| | - Justyna M. Hermanowicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
- Department of Clinical Pharmacy, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
| | - Michal Mysliwiec
- Ist Department Nephrology and Transplantation, Medical University, Bialystok, Zurawia 14, 15-540 Bialystok, Poland
- Lomza State University of Applied Sciences, Akademicka 14, 18-400 Łomża, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
- Department of Pharmacology and Toxicology, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland
| |
Collapse
|
652
|
Miyajima M. Amino acids: key sources for immunometabolites and immunotransmitters. Int Immunol 2020; 32:435-446. [PMID: 32383454 DOI: 10.1093/intimm/dxaa019] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022] Open
Abstract
Immune-cell activation and functional plasticity are closely linked to metabolic reprogramming that is required to supply the energy and substrates for such dynamic transformations. During such processes, immune cells metabolize many kinds of molecules including nucleic acids, sugars and lipids, which is called immunometabolism. This review will mainly focus on amino acids and their derivatives among such metabolites and describe the functions of these molecules in the immune system. Although amino acids are essential for, and well known as, substrates for protein synthesis, they are also metabolized as energy sources and as substrates for functional catabolites. For example, glutamine is metabolized to produce energy through glutaminolysis and tryptophan is consumed to supply nicotinamide adenine dinucleotide, whereas arginine is metabolized to produce nitric acid and polyamine by nitric oxide synthase and arginase, respectively. In addition, serine is catabolized to produce nucleotides and to induce methylation reactions. Furthermore, in addition to their intracellular functions, amino acids and their derivatives are secreted and have extracellular functions as immunotransmitters. Many amino acids and their derivatives have been classified as neurotransmitters and their functions are clear as transmitters between nerve cells, or between nerve cells and immune cells, functioning as immunotransmitters. Thus, this review will describe the intracellular and external functions of amino acid from the perspective of immunometabolism and immunotransmission.
Collapse
Affiliation(s)
- Michio Miyajima
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Tsurumi-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
653
|
Sorgdrager F, van Der Ley CP, van Faassen M, Calus E, Nollen EA, Kema IP, van Dam D, De Deyn PP. The Effect of Tryptophan 2,3-Dioxygenase Inhibition on Kynurenine Metabolism and Cognitive Function in the APP23 Mouse Model of Alzheimer's Disease. Int J Tryptophan Res 2020; 13:1178646920972657. [PMID: 33447045 PMCID: PMC7780178 DOI: 10.1177/1178646920972657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 10/18/2020] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is associated with progressive endogenous neurotoxicity and hampered inflammatory regulation. The kynurenine (Kyn) pathway, which is controlled by tryptophan 2,3-dioxygenase (TDO), produces neuroactive and anti-inflammatory metabolites. Age-related Kyn pathway activation might contribute to AD pathology in humans, and inhibition of TDO was found to reduce AD-related cellular toxicity and behavioral deficits in animal models. To further explore the effect of aging on the Kyn pathway in the context of AD, we analyzed Kyn metabolite profiles in serum and brain tissue of the APP23 amyloidosis mouse model. We found that aging had genotype-independent effects on Kyn metabolite profiles in serum, cortex, hippocampus and cerebellum, whereas serum concentrations of many Kyn metabolites were reduced in APP23 mice. Next, to further establish the role of TDO in AD-related behavioral deficits, we investigated the effect of long-term pharmacological TDO inhibition on cognitive performance in APP23 mice. Our results indicated that TDO inhibition reversed recognition memory deficits without producing measurable changes in cerebral Kyn metabolites. TDO inhibition did not affect spatial learning and memory or anxiety-related behavior. These data indicate that age-related Kyn pathway activation is not specific for humans and could represent a cross-species phenotype of aging. These data warrant further investigation on the role of peripheral Kyn pathway disturbances and cerebral TDO activity in AD pathophysiology.
Collapse
Affiliation(s)
- Fjh Sorgdrager
- Laboratory of Neurochemistry and Behavior, Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands.,Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - C P van Der Ley
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M van Faassen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - E Calus
- Laboratory of Neurochemistry and Behavior, Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - E A Nollen
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - I P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - D van Dam
- Laboratory of Neurochemistry and Behavior, Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - P P De Deyn
- Laboratory of Neurochemistry and Behavior, Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands.,Department of Neurology, Memory Clinic of Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| |
Collapse
|
654
|
Mondanelli G, Volpi C. The double life of serotonin metabolites: in the mood for joining neuronal and immune systems. Curr Opin Immunol 2020; 70:1-6. [PMID: 33360496 DOI: 10.1016/j.coi.2020.11.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 12/22/2022]
Abstract
Neuroimmune system is nowadays considered as one complex, but unique example of coordination between cellular and molecular networks, only apparently segregated, but strictly collaborating for the maintenance of body integrity. Too often, serotonin and its metabolites have been considered merely as neurotransmitters, when they have multiple effects spreading from the modulation of mood and behavioral processes to the regulation of a wide range of physiologic and pathophysiologic processes in most human organs, not least the immune response. The purpose of this review is to highlight the importance of metabolites generated along the serotonin pathway in the constant dialogue between neuroendocrine and immune systems; moreover, we would like to point out that the molecules produced in the two main routes of tryptophan metabolism are involved in a loop of self-regulation aimed at maintaining the equilibrium between these two metabolic pathways in the neuroimmune system, in both physiologic and pathologic conditions.
Collapse
Affiliation(s)
- Giada Mondanelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudia Volpi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
655
|
Effect of a Single Bout of Aerobic Exercise on Kynurenine Pathway Metabolites and Inflammatory Markers in Prostate Cancer Patients-A Pilot Randomized Controlled Trial. Metabolites 2020; 11:metabo11010004. [PMID: 33374836 PMCID: PMC7823964 DOI: 10.3390/metabo11010004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 02/08/2023] Open
Abstract
The kynurenine (KYN) pathway gains growing research interest concerning the genesis, progression and therapy of solid tumors. Previous studies showed exercise-induced effects on metabolite levels along the KYN pathway. Modulations of the KYN pathway might be involved in the positive impact of exercise on prostate cancer progression and mortality. The objective of this trial was to investigate whether a single-physical exercise alters tryptophan (TRP) metabolism and related inflammatory markers in this population. We conducted a randomized controlled trial with 24 patients suffering from prostate cancer. While the control group remained inactive, the intervention group performed a 30-min aerobic exercise on a bicycle ergometer at 75% of individual VO2peak. Before (t0) and directly after the exercise intervention (t1) KYN, TRP, kynurenic acid, quinolinic acid as well as various inflammation markers (IL6, TNF-α, TGF-β) were measured in blood serum. At baseline, the present sample showed robust correlations between TRP, KYN, quinolinic acid and inflammatory markers. Regarding the exercise intervention, interaction effects for TRP, the KYN/TRP ratio and TGF-β were observed. The results show for the first time that acute physical exercise impacts TRP metabolism in prostate cancer patients. Moreover, baseline associations underline the relationship between inflammation and the KYN pathway in prostate cancer.
Collapse
|
656
|
Kashio S, Miura M. Kynurenine Metabolism in the Fat Body Non-autonomously Regulates Imaginal Disc Repair in Drosophila. iScience 2020; 23:101738. [PMID: 33376969 PMCID: PMC7756137 DOI: 10.1016/j.isci.2020.101738] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/07/2020] [Accepted: 10/23/2020] [Indexed: 12/28/2022] Open
Abstract
Tissue interactions are critical for maintaining homeostasis; however, little is known about how remote tissue regulates regeneration. Previously, we established a genetic dual system that induces cell ablation in Drosophila larval imaginal discs and simultaneously manipulates genes in non-damaged tissues. Using humoral metabolome analysis and a genetic damage system, we found that the Tryptophan (Trp)-Kynurenine (Kyn) pathway in the fat body is required for disc repair. Genetic manipulation of Trp-Kyn metabolism in the fat body impaired disc regeneration without affecting wing development. In particular, the fat body-derived humoral kynurenic acid (KynA) was required for disc repair. The impairment of S-adenosylmethionine (SAM) synthesis from methionine (Met) in the fat body hampers the maintenance of KynA levels in hemolymph at the early stage of disc repair, suggesting a connection between Met-SAM and Trp-Kyn metabolisms. Our data indicate KynA from the fat body acts as a permissive metabolite for tissue repair and regeneration. Trp-Kyn pathway in Drosophila larval fat body is remotely required for disc repair The fat body-derived humoral KynA is required for disc repair SAM synthesis in the fat body affects KynA levels in hemolymph during disc repair
Collapse
Affiliation(s)
- Soshiro Kashio
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
657
|
Karamitros CS, Murray K, Sugiyama Y, Kumada Y, Johnson KA, Georgiou G, D'Arcy S, Stone EM. Conformational Dynamics Contribute to Substrate Selectivity and Catalysis in Human Kynureninase. ACS Chem Biol 2020; 15:3159-3166. [PMID: 33275413 PMCID: PMC11104311 DOI: 10.1021/acschembio.0c00676] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Kynureninases (KYNases) are enzymes that play a key role in tryptophan catabolism through the degradation of intermediate kynurenine and 3'-hydroxy-kynurenine metabolites (KYN and OH-KYN, respectively). Bacterial KYNases exhibit high catalytic efficiency toward KYN and moderate activity toward OH-KYN, whereas animal KYNases are highly selective for OH-KYN, exhibiting only minimal activity toward the smaller KYN substrate. These differences reflect divergent pathways for KYN and OH-KYN utilization in the respective kingdoms. We examined the Homo sapiens and Pseudomonas fluorescens KYNases (HsKYNase and PfKYNase respectively) using pre-steady-state and hydrogen-deuterium exchange mass spectrometry (HDX-MS) methodologies. We discovered that the activity of HsKYNase critically depends on formation of hydrogen bonds with the hydroxyl group of OH-KYN to stabilize the entire active site and allow productive substrate turnover. With the preferred OH-KYN substrate, stabilization is observed at the substrate-binding site and the region surrounding the PLP cofactor. With the nonpreferred KYN substrate, less stabilization occurs, revealing a direct correlation with activity. This correlation holds true for PfKYNases; however there is only a modest stabilization at the substrate-binding site, suggesting that substrate discrimination is simply achieved by steric hindrance. We speculate that eukaryotic KYNases use dynamic mobility as a mechanism of substrate specificity to commit OH-KYN to nicotinamide synthesis and avoid futile hydrolysis of KYN. These findings have important ramifications for the engineering of HsKynase with high KYN activity as required for clinical applications in cancer immunotherapy. Our study shows how homologous enzymes with conserved active sites can use dynamics to discriminate between two highly similar substrates.
Collapse
Affiliation(s)
- Christos S Karamitros
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Kyle Murray
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Yusuke Sugiyama
- Department of Molecular Chemistry and Engineering, Graduate School of Science and Technology, Kyoto Institute of Technology, Kyoto, Japan
| | - Yoichi Kumada
- Department of Molecular Chemistry and Engineering, Graduate School of Science and Technology, Kyoto Institute of Technology, Kyoto, Japan
| | - Kenneth A Johnson
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, United States
| | - George Georgiou
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, United States
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Oncology, University of Texas Dell Medical School, LiveSTRONG Cancer Institutes, Austin, Texas 78712, United States
| | - Sheena D'Arcy
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Everett M Stone
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Oncology, University of Texas Dell Medical School, LiveSTRONG Cancer Institutes, Austin, Texas 78712, United States
| |
Collapse
|
658
|
Ory P, Hamani V, Bodet PE, Murillo L, Graber M. The variegated scallop, Mimachlamys varia, undergoes alterations in several of its metabolic pathways under short-term zinc exposure. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2020; 37:100779. [PMID: 33360397 DOI: 10.1016/j.cbd.2020.100779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/19/2020] [Accepted: 11/20/2020] [Indexed: 01/25/2023]
Abstract
The variegated scallop (Mimachlamys varia) is a filter feeder bivalve encountered in marine regions of the Atlantic coast. In particular, it is present in the La Rochelle marina (France), where it is used for the biomonitoring of marine pollution, due to its ability to strongly bioaccumulate pollutants. In this semi-closed environment, contamination generated by port activities leads to an accumulation of both organic and metal pollutants. Zinc is one of these pollutants, present at a dose of up to 150 μg.L-1. This study investigated the effects of 48 h zinc exposure upon the metabolic profiles of Mimachlamys varia using UHPLC/QToF (ultra-high performance liquid chromatography-quadrupole time-of-flight) tandem mass spectrometry metabolomics. After acclimation in mesocosms recreating in situ conditions, both controls and exposed with Zn2+ (150 μg.L-1) bivalves were dissected to recover the gills after 48 h and stored at -80 °C before metabolites extraction. UHPLC/QToF tandem mass spectrometry was performed to study metabolite composition of samples. Statistical analysis of results using multivariate techniques showed a good classification between control and exposed groups. Eleven identified metabolites were found to be down-modulated in exposed scallops. These variations could reflect potential zinc effects on several of the biological processes, such as energy metabolism, osmoregulation and defense against oxidative stress. Among the eleven metabolites highlighted, four were reported for the first time in an aquatic organism exposed to Zn. This study demonstrates once again the diversity of interactions between bivalves and metals and the complexity of the physiological response of marine bivalves to pollutants.
Collapse
Affiliation(s)
- P Ory
- Littoral Environnement et Sociétés (LIENSs), UMR 7266, CNRS-Université de La Rochelle, 2 rue Olympe de Gouges, F-17042 La Rochelle Cedex 01, France
| | - V Hamani
- Littoral Environnement et Sociétés (LIENSs), UMR 7266, CNRS-Université de La Rochelle, 2 rue Olympe de Gouges, F-17042 La Rochelle Cedex 01, France
| | - P-E Bodet
- Littoral Environnement et Sociétés (LIENSs), UMR 7266, CNRS-Université de La Rochelle, 2 rue Olympe de Gouges, F-17042 La Rochelle Cedex 01, France
| | - L Murillo
- Littoral Environnement et Sociétés (LIENSs), UMR 7266, CNRS-Université de La Rochelle, 2 rue Olympe de Gouges, F-17042 La Rochelle Cedex 01, France
| | - M Graber
- Littoral Environnement et Sociétés (LIENSs), UMR 7266, CNRS-Université de La Rochelle, 2 rue Olympe de Gouges, F-17042 La Rochelle Cedex 01, France.
| |
Collapse
|
659
|
Török N, Tanaka M, Vécsei L. Searching for Peripheral Biomarkers in Neurodegenerative Diseases: The Tryptophan-Kynurenine Metabolic Pathway. Int J Mol Sci 2020; 21:E9338. [PMID: 33302404 PMCID: PMC7762583 DOI: 10.3390/ijms21249338] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/28/2020] [Accepted: 11/29/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases are multifactorial, initiated by a series of the causative complex which develops into a certain clinical picture. The pathogenesis and disease course vary from patient to patient. Thus, it should be likewise to the treatment. Peripheral biomarkers are to play a central role for tailoring a personalized therapeutic plan for patients who suffered from neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis, among others. Nevertheless, the use of biomarkers in clinical practice is still underappreciated and data presented in biomarker research for clinical use is still uncompelling, compared to the abundant data available for drug research and development. So is the case with kynurenines (KYNs) and the kynurenine pathway (KP) enzymes, which have been associated with a wide range of diseases including cancer, autoimmune diseases, inflammatory diseases, neurologic diseases, and psychiatric disorders. This review article discusses current knowledge of KP alterations observed in the central nervous system as well as the periphery, its involvement in pathogenesis and disease progression, and emerging evidence of roles of microbiota in the gut-brain axis, searching for practical peripheral biomarkers which ensure personalized treatment plans for neurodegenerative diseases.
Collapse
Affiliation(s)
- Nóra Török
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (N.T.); (M.T.)
| | - Masaru Tanaka
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (N.T.); (M.T.)
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - László Vécsei
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (N.T.); (M.T.)
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
660
|
Mo Y, Lu Z, Wang L, Ji C, Zou C, Liu X. The Aryl Hydrocarbon Receptor in Chronic Kidney Disease: Friend or Foe? Front Cell Dev Biol 2020; 8:589752. [PMID: 33415104 PMCID: PMC7784643 DOI: 10.3389/fcell.2020.589752] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that promotes cell responses to small molecules derived from the diet, microorganisms, metabolism and pollutants. The AhR signal regulates many basic cellular processes, including cell cycle progression, adhesion, migration, apoptosis and cell proliferation. Many studies have shown that AhR is associated with chronic kidney disease (CKD) and its complications. This article reviews the current knowledge about the role of AhR in CKD, showing that AhR mediates CKD complications, including cardiovascular disease, anemia, bone disorders, cognitive dysfunction and malnutrition, and that it influences drug metabolism in individuals with CKD. AhR enhances the intestinal barrier function to reduce the harmful effects of uremic toxins. Therefore, understanding the complex roles of AhR during CKD is important to be able to target this transcription factor safely and effectively for CKD prevention and treatment.
Collapse
Affiliation(s)
- Yenan Mo
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhaoyu Lu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lixin Wang
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunlan Ji
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuan Zou
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xusheng Liu
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
661
|
Huang Q, Lei Y, Li X, Guo F, Liu M. A Highlight of the Mechanisms of Immune Checkpoint Blocker Resistance. Front Cell Dev Biol 2020; 8:580140. [PMID: 33344447 PMCID: PMC7746821 DOI: 10.3389/fcell.2020.580140] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 10/28/2020] [Indexed: 02/05/2023] Open
Abstract
In recent years, as our understanding of tumor immunology is continuously improved, immunotherapy has come to the center stage of cancer therapy and is deemed as the most promising approach for cancer control. Although immunotherapy, particularly immune checkpoint blockade (ICB), has achieved a milestone in several types of tumors, the majority of cancer patients do not benefit from immunotherapy. The dismal outcome of cancer immunotherapy is mainly due to primary or acquired resistance arising from tumor immune evasion. Exploring the mechanisms of tumor immune evasion in the course of immunotherapy may identify biological targets to conquer tumor resistance to immunotherapy. In this review, we highlight tumor cell-intrinsic and -extrinsic factors that may underlie tumor resistance to immune checkpoint blockers. Targeting these factors in combination with immune checkpoint blockers points to the future direction of cancer immunotherapy.
Collapse
Affiliation(s)
- Qian Huang
- Laboratory of Signal Transduction and Molecular Targeted Therapy, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yanna Lei
- Laboratory of Signal Transduction and Molecular Targeted Therapy, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoying Li
- Laboratory of Signal Transduction and Molecular Targeted Therapy, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Ming Liu
- Laboratory of Signal Transduction and Molecular Targeted Therapy, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
662
|
Hartstra AV, Schüppel V, Imangaliyev S, Schrantee A, Prodan A, Collard D, Levin E, Dallinga-Thie G, Ackermans MT, Winkelmeijer M, Havik SR, Metwaly A, Lagkouvardos I, Nier A, Bergheim I, Heikenwalder M, Dunkel A, Nederveen AJ, Liebisch G, Mancano G, Claus SP, Benítez-Páez A, la Fleur SE, Bergman JJ, Gerdes V, Sanz Y, Booij J, Kemper E, Groen AK, Serlie MJ, Haller D, Nieuwdorp M. Infusion of donor feces affects the gut-brain axis in humans with metabolic syndrome. Mol Metab 2020; 42:101076. [PMID: 32916306 PMCID: PMC7536740 DOI: 10.1016/j.molmet.2020.101076] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Increasing evidence indicates that intestinal microbiota play a role in diverse metabolic processes via intestinal butyrate production. Human bariatric surgery data suggest that the gut-brain axis is also involved in this process, but the underlying mechanisms remain unknown. METHODS We compared the effect of fecal microbiota transfer (FMT) from post-Roux-en-Y gastric bypass (RYGB) donors vs oral butyrate supplementation on (123I-FP-CIT-determined) brain dopamine transporter (DAT) and serotonin transporter (SERT) binding as well as stable isotope-determined insulin sensitivity at baseline and after 4 weeks in 24 male and female treatment-naïve metabolic syndrome subjects. Plasma metabolites and fecal microbiota were also determined at these time points. RESULTS We observed an increase in brain DAT after donor FMT compared to oral butyrate that reduced this binding. However, no effect on body weight and insulin sensitivity was demonstrated after post-RYGB donor feces transfer in humans with metabolic syndrome. Increases in fecal levels of Bacteroides uniformis were significantly associated with an increase in DAT, whereas increases in Prevotella spp. showed an inverse association. Changes in the plasma metabolites glycine, betaine, methionine, and lysine (associated with the S-adenosylmethionine cycle) were also associated with altered striatal DAT expression. CONCLUSIONS Although more and larger studies are needed, our data suggest a potential gut microbiota-driven modulation of brain dopamine and serotonin transporters in human subjects with obese metabolic syndrome. These data also suggest the presence of a gut-brain axis in humans that can be modulated. NTR REGISTRATION 4488.
Collapse
Affiliation(s)
- Annick V Hartstra
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Valentina Schüppel
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Sultan Imangaliyev
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Anouk Schrantee
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Andrei Prodan
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Didier Collard
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Geesje Dallinga-Thie
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Mariette T Ackermans
- Laboratory of Endocrinology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Maaike Winkelmeijer
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Stefan R Havik
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Amira Metwaly
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Ilias Lagkouvardos
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Anika Nier
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Austria
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Austria
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | - Andreas Dunkel
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, Freising, Germany
| | - Aart J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Gerhard Liebisch
- Department of Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Giulia Mancano
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Sandrine P Claus
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Alfonso Benítez-Páez
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Susanne E la Fleur
- Laboratory of Endocrinology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Jacques J Bergman
- Department of Gastroenterology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Victor Gerdes
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Yolanda Sanz
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Elles Kemper
- Department of Clinical Pharmacy, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Albert K Groen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany; ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands.
| |
Collapse
|
663
|
Tan X, van Egmond LT, Cedernaes J, Benedict C. The role of exercise-induced peripheral factors in sleep regulation. Mol Metab 2020; 42:101096. [PMID: 33045432 PMCID: PMC7585947 DOI: 10.1016/j.molmet.2020.101096] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/25/2020] [Accepted: 10/06/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Recurrently disrupted sleep is a widespread phenomenon in our society. This is worrisome as chronically impaired sleep increases the risk of numerous diseases that place a heavy burden on health services worldwide, including type 2 diabetes, obesity, depression, cardiovascular disease, and dementia. Therefore, strategies mitigating the current societal sleep crisis are needed. SCOPE OF REVIEW Observational and interventional studies have found that regular moderate to intensive exercise is associated with better subjective and objective sleep in humans, with and without pre-existing sleep disturbances. Here, we summarize recent findings from clinical studies in humans and animal experiments suggesting that molecules that are expressed, produced, and released by the skeletal muscle in response to exercise may contribute to the sleep-improving effects of exercise. MAJOR CONCLUSIONS Exercise-induced skeletal muscle recruitment increases blood concentrations of signaling molecules, such as the myokine brain-derived neurotrophic factor (BDNF), which has been shown to increase the depth of sleep in animals. As reviewed herein, BDNF and other muscle-induced factors are likely to contribute to the sleep-promoting effects of exercise. Despite progress in the field, however, several fundamental questions remain. For example, one central question concerns the optimal time window for exercise to promote sleep. It is also unknown whether the production of muscle-induced peripheral factors promoting sleep is altered by acute and chronic sleep disturbances, which has become increasingly common in the modern 24/7 lifestyle.
Collapse
Affiliation(s)
- Xiao Tan
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | | | | | | |
Collapse
|
664
|
Venkatesan D, Iyer M, Narayanasamy A, Siva K, Vellingiri B. Kynurenine pathway in Parkinson's disease-An update. eNeurologicalSci 2020; 21:100270. [PMID: 33134567 PMCID: PMC7585940 DOI: 10.1016/j.ensci.2020.100270] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/05/2020] [Accepted: 08/26/2020] [Indexed: 12/24/2022] Open
Abstract
Parkinson's disease (PD) is a complex multi-factorial neurodegenerative disorder where various altered metabolic pathways contribute to the progression of the disease. Tryptophan (TRP) is a major precursor in kynurenine pathway (KP) and it has been discussed in various in vitro studies that the metabolites quinolinic acid (QUIN) causes neurotoxicity and kynurenic acid (KYNA) acts as neuroprotectant respectively. More studies are also focused on the effects of other KP metabolites and its enzymes as it has an association with ageing and PD pathogenesis. Until now, very few studies have targeted the role of genetic mutations in abnormal KP metabolism in adverse conditions of PD. Therefore, the present review gives an updated research studies on KP in connection with PD. Moreover, the review emphasizes on the urge for the development of biomarkers and also this would be an initiative in generating an alternative therapeutic approach for PD.
Collapse
Key Words
- 3-HAA, 3-hydroxyanthranilic acid
- 3-HK, 3-hydroxykynurenine
- 6-OHDA, 6-hydroxydopamine
- AA, anthranilic acid
- ACMSD, amino-carboxymuconatesemialdehyde decarboxylase
- AD, Alzheimer's disease
- ATP, adenosine triphosphate
- Ageing
- AhR, aryl hydrocarbon receptor
- Biomarkers
- CNS, central nervous system
- CSF, cerebrospinal fluid
- DA, dopaminergic
- FAM, formamidase
- IDO-1, indoleamine-2,3-dioxygenases
- IFN-γ, interferon-γ
- KATs, kynurenine aminotransferases
- KMO, kynurenine −3-monooxygenase
- KP, Kynurenine pathway
- KYN, kynurenine
- KYNA, kynurenic acid
- Kynurenine pathway (KP)
- L-DOPA, L-dopamine
- LID, L-DOPA-induced dyskinesia
- MPTP, 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine
- NAD+, nicotinamide adenine dinucleotide
- NADPH, nicotinamide adenine dinucleotide phosphate
- NFK, N′-formylkynurenine
- NMDA, N-methyl-d-aspartate
- PA, picolinic acid
- PD, Parkinson's disease
- Parkinson's disease (PD)
- QUIN, quinolinic acid
- RBCs, red blood cells
- SNpc, substantianigra pars compacta
- TDO, tryptophan 2,3-dioxygenase
- TRP, tryptophan
- Therapeutics
- XA, xanthurenic acid
- ZNS, zonisamide
- α-synuclein, αSyn
Collapse
Affiliation(s)
- Dhivya Venkatesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641 043, Tamil Nadu, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Kamalakannan Siva
- National Centre for Disease Control, Ministry of Health and Family Welfare, Government of India, New Delhi 110054, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| |
Collapse
|
665
|
Pfeffer TJ, Herrmann J, Berliner D, König T, Winter L, Ricke‐Hoch M, Ponimaskin E, Schuchardt S, Thum T, Hilfiker‐Kleiner D, Bauersachs J, Kahl KG. Assessment of major mental disorders in a German peripartum cardiomyopathy cohort. ESC Heart Fail 2020; 7:4394-4398. [PMID: 32909398 PMCID: PMC7754901 DOI: 10.1002/ehf2.12967] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/07/2020] [Accepted: 08/06/2020] [Indexed: 12/20/2022] Open
Abstract
AIMS Peripartum cardiomyopathy (PPCM) is a heart disease affecting women during the last month of pregnancy or in the first months after delivery. The impact of the disease on mental health is largely unknown. METHODS AND RESULTS Major mental disorders were assessed by a structured clinical interview in 40 patients with a confirmed PPCM diagnosis, and the data were compared with published prevalence in postpartum women. Circulating biomarkers associated with mental health, such as kynurenine, serotonin, and microRNA (miR)-30e, were evaluated in PPCM and compared with matched healthy pregnancy-matched postpartum controls (PP-Ctrl). Major mental disorders were diagnosed in 65% (26/40) of the PPCM cohort. The prevalence for major depressive disorders was 4-fold, for post-traumatic stress disorder 14-fold, and for panic disorder 6-fold higher in PPCM patients compared with postpartum women without a PPCM diagnosis. Compared with PP-Ctrl, PPCM patients displayed elevated levels of serum kynurenine (P < 0.01), reduced levels of serum serotonin (P < 0.05), and elevated levels of plasma miR-30e (P < 0.05). CONCLUSIONS The majority of PPCM patients in the present cohort displayed mental disorders with a higher prevalence of major depressive disorders, post-traumatic stress disorder (PTBS), and panic disorder, compared with postpartum women without a PPCM diagnosis. This higher prevalence was associated with an impaired tryptophan metabolism and elevated levels of the depression-associated miR-30e, suggesting a potential predisposition for mental disorders at the time of PPCM diagnosis. Consequently, physicians should be aware of the increased risk for mental disorders in PPCM patients, and psychiatric assessment should be included in the diagnosis and management of PPCM patients.
Collapse
Affiliation(s)
- Tobias J. Pfeffer
- Department of Cardiology and AngiologyHannover Medical SchoolCarl‐Neuberg‐Str. 1Hannover30625Germany
| | - Julian Herrmann
- Department of Psychiatry, Social Psychiatry and PsychotherapyHannover Medical SchoolHannoverGermany
| | - Dominik Berliner
- Department of Cardiology and AngiologyHannover Medical SchoolCarl‐Neuberg‐Str. 1Hannover30625Germany
| | - Tobias König
- Department of Cardiology and AngiologyHannover Medical SchoolCarl‐Neuberg‐Str. 1Hannover30625Germany
| | - Lotta Winter
- Department of Psychiatry, Social Psychiatry and PsychotherapyHannover Medical SchoolHannoverGermany
| | - Melanie Ricke‐Hoch
- Department of Cardiology and AngiologyHannover Medical SchoolCarl‐Neuberg‐Str. 1Hannover30625Germany
| | | | - Sven Schuchardt
- Department of Bio and Environmental AnalyticsFraunhofer Institute for Toxicology and Experimental MedicineHannoverGermany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic StrategiesHannover Medical SchoolHannoverGermany
| | - Denise Hilfiker‐Kleiner
- Department of Cardiology and AngiologyHannover Medical SchoolCarl‐Neuberg‐Str. 1Hannover30625Germany
| | - Johann Bauersachs
- Department of Cardiology and AngiologyHannover Medical SchoolCarl‐Neuberg‐Str. 1Hannover30625Germany
| | - Kai G. Kahl
- Department of Psychiatry, Social Psychiatry and PsychotherapyHannover Medical SchoolHannoverGermany
| |
Collapse
|
666
|
Jung TW, Park J, Sun JL, Ahn SH, Abd El-Aty AM, Hacimuftuoglu A, Kim HC, Shim JH, Shin S, Jeong JH. Administration of kynurenic acid reduces hyperlipidemia-induced inflammation and insulin resistance in skeletal muscle and adipocytes. Mol Cell Endocrinol 2020; 518:110928. [PMID: 32702471 DOI: 10.1016/j.mce.2020.110928] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/17/2022]
Abstract
Kynurenic acid (KA), an endogenous product of L-tryptophan metabolism in the kynurenine pathway, regulates adipose tissue energy homeostasis and inflammation. However, its role in palmitate-induced insulin resistance and detailed underlying mechanisms in skeletal muscles and adipose tissues are unclear. Herein, we report that KA ameliorated palmitate-induced inflammation and insulin resistance in differentiated C2C12 and 3T3-L1 cell lines as well as soleus skeletal muscle and subcutaneous adipose tissues in mice. Palmitate-induced inflammatory markers, such as nuclear factor κB translocation, inhibitory κBα phosphorylation, pro-inflammatory cytokine expression, and impaired insulin signaling, were markedly attenuated by KA both in vitro and in vivo. KA significantly increased AMP-activated protein kinase (AMPK) phosphorylation and sirtuin 6 (SIRT6) expressions in C2C12 myocytes and 3T3-L1 adipocytes and skeletal muscle and adipose tissues of mice. siRNA-mediated AMPK or SIRT6 inhibition significantly mitigated the suppressive effects of KA on palmitate-induced inflammation and insulin resistance. KA significantly stimulated expression of genes involved in fatty acid oxidation in C2C12 myocytes and skeletal muscle of mice. Moreover, KA inhibits lipogenesis in 3T3-L1 adipocytes. AMPK or SIRT6 siRNA markedly reversed these changes. The siRNA targeting Gpr35 abrogated the effects of KA on AMPK phosphorylation in C2C12 myocytes and 3T3-L1 adipocytes, except SIRT6 expression. It has therefore been shown that KA could potentially alleviate inflammation and insulin resistance in skeletal muscle and adipose tissues through Gpr35/AMPK and SIRT6-mediated pathways.
Collapse
Affiliation(s)
- Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, 221, Heuksuk-dong, Dongjak-gu, Seoul, 156-756, Republic of Korea.
| | - Jinwoo Park
- Department of Pharmacology, College of Medicine, Chung-Ang University, 221, Heuksuk-dong, Dongjak-gu, Seoul, 156-756, Republic of Korea.
| | - Jaw Long Sun
- Department of Pharmacology, College of Medicine, Chung-Ang University, 221, Heuksuk-dong, Dongjak-gu, Seoul, 156-756, Republic of Korea.
| | - Sung Ho Ahn
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| | - A M Abd El-Aty
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, Jinan, 250353, China; Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211, Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey.
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey.
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea.
| | - Jae-Han Shim
- Natural Products Chemistry Laboratory, College of Agriculture and Life Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju, 500-757, Republic of Korea.
| | - SungShik Shin
- Laboratory of Parasitology, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju, 500-757, Republic of Korea.
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, 221, Heuksuk-dong, Dongjak-gu, Seoul, 156-756, Republic of Korea.
| |
Collapse
|
667
|
Muneer A. Kynurenine Pathway of Tryptophan Metabolism in Neuropsychiatric Disorders: Pathophysiologic and Therapeutic Considerations. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2020; 18:507-526. [PMID: 33124585 PMCID: PMC7609208 DOI: 10.9758/cpn.2020.18.4.507] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/04/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022]
Abstract
Under physiological conditions 95% of the ingested essential amino acid tryptophan is metabolized by the kynurenine pathway (KP) to yield the ubiquitous co-enzyme nicotinamide adenine dinucleotide, fulfilling cellular energy require-ments. Importantly, the intermediaries of KP exert crucial effects throughout the body, including the central nervous system. Besides, KP metabolites are implicated in diverse disease processes such as inflammation/immune disorders, endocrine/metabolic conditions, cancers and neuropsychiatric diseases. A burgeoning body of research indicates that the KP plays a pathogenic role in major psychiatric diseases like mood disorders and schizophrenia. Triggered by inflammatory processes, the balance between neurotoxic and neuroprotective branches of the KP is disturbed. In preclinical models these discrepancies result in behaviors reminiscent of depression and psychosis. In clinical samples, recent studies are discovering key kynurenine pathway abnormalities which incriminate it in the pathogenesis of the main psychiatric disorders. Harnessing this knowledge has the potential to find disease biomarkers helpful in identifying and prognosticating neuropsychiatric disorders. Concurrently, earnest research efforts directed towards manipulating the KP hold the promise of discovering novel pharmacological agents that have therapeutic value. In this manuscript, an in-depth appraisal of the extant literature is done to understand the working of KP as this applies to neuropsychiatric disorders. It is concluded that this pathway plays an overarching role in the development of major psychiatric disorders, the KP metabolites have the potential to serve as disease markers and new medications based on KP modulation can bring lasting cures for patients suffering from these intractable conditions.
Collapse
Affiliation(s)
- Ather Muneer
- Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan
| |
Collapse
|
668
|
İriz A, Şemsi R, Eser B, Arslan B, Dinçel AS. The evaluation of serum tryptophan and kynurenine levels in patients with obstructive sleep apnea syndrome. Sleep Breath 2020; 25:1389-1398. [PMID: 33222028 DOI: 10.1007/s11325-020-02250-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE The kynurenine (Kyn) pathway may play a role in certain physiological functions such as behavior, sleep, thermoregulation, and pregnancy. Tryptophan (Trp) is oxidized with tryptophan 2,3-dioxygenase and indolamine 2,3-dioxygenase (IDO). Under normal conditions, hepatic kynurenine is a transcription factor and IDO expression in healthy tissues is very low. The ratio of Kyn to Trp can be used as an indicator to assess IDO activity. This study aimed to determine the relationship between Kyn/Trp ratio and obstructive sleep apnea syndrome (OSAS) disease activity. METHODS Study participants were categorized in 3 groups: Group 1 included patients with mild OSAS, Group 2, patients with moderate to severe OSAS, and Group 3, individuals considered normal to serve as controls. The demographic characteristics of the patients were recorded. Apnea-hypopnea index (AHI) and oxygen desaturation index (ODI) measurements were performed by diagnostic polysomnography (PSG). Trp and Kyn levels were determined by HPLC-UV method. RESULTS Group 1 included 30 patients (18 men) with mild OSAS; Group 2 included 42 patients (31 men) with moderate to severe OSAS; and Group 3 included 25 controls (13 men). While there was no statistically significant difference between the levels of tryptophan and kynurenine in the groups, a significant difference was found between the Kyn/Trp ratios. A significant correlation was observed in individuals with a body mass index less than 25 with the Kyn/Trp ratio. In individuals with mild OSAS, a significant correlation was observed between ODI and BMI. In individuals with moderate to severe OSAS, there was a significant correlation between ODI, AHI, and BMI. CONCLUSION In this study, there was no relationship between OSAS disease severity and IDO activity as assessed by immunoreactivity via the Kyn/Trp pathway.
Collapse
Affiliation(s)
- Ayşe İriz
- Department of Otorhinolaryngology (Ear-Nose-Throat), Gazi University Faculty of Medicine, Ankara, Turkey
| | - Rabia Şemsi
- Department of Medical Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey.
| | - Burcu Eser
- Department of Medical Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
- Gülhane Institute of Health Sciences, R&D Centre, Chromatography Laboratory, The University of Health Sciences, Ankara, Turkey
| | - Burak Arslan
- Department of Medical Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
- Department of Medical Biochemistry, Ercis State Hospital, Van, Turkey
| | - Aylin Sepici Dinçel
- Department of Medical Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
669
|
Garcia-Lino AM, Gomez-Gomez A, Garcia-Mateos D, de la Fuente A, Alvarez AI, Pozo OJ, Merino G. Analysis of the interaction between tryptophan-related compounds and ATP-binding cassette transporter G2 (ABCG2) using targeted metabolomics. Food Chem 2020; 344:128665. [PMID: 33250293 DOI: 10.1016/j.foodchem.2020.128665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 11/04/2020] [Accepted: 11/14/2020] [Indexed: 12/19/2022]
Abstract
ATP-binding cassette transporter G2 (ABCG2) is involved in the secretion of several compounds in milk. The in vitro and in vivo interactions between tryptophan-related compounds and ABCG2 were investigated. The tryptophan metabolome was determined by liquid chromatography-tandem mass spectrometry in milk and plasma from wild-type and Abcg2-/- mice as well as dairy cows carrying the ABCG2 Y581S polymorphism (Y/S) and noncarrier animals (Y/Y). The milk-to-plasma ratios of tryptophan, kynurenic acid, kynurenine, anthranilic acid, and xanthurenic acid were higher in wild-type mice than in Abcg2-/- mice. The ratio was 2-fold higher in Y/S than in Y/Y cows for kynurenine. In vitro transport assays confirmed that some of these compounds were in vitro substrates of the transporter and validated the differences observed between the two variants of the bovine protein. These findings show that the secretion of metabolites belonging to the kynurenine pathway into milk is mediated by ABCG2.
Collapse
Affiliation(s)
- Alba M Garcia-Lino
- Department of Biomedical Sciences-Physiology, Faculty of Veterinary Medicine, Animal Health Institute (INDEGSAL), University of Leon, Campus de Vegazana, 24071 Leon, Spain
| | - Alex Gomez-Gomez
- Integrative Pharmacology and Systems Neuroscience Group, IMIM-Hospital del Mar Medical Research Institute, 88 Doctor Aiguader, 08003 Barcelona, Spain
| | - Dafne Garcia-Mateos
- Department of Biomedical Sciences-Physiology, Faculty of Veterinary Medicine, Animal Health Institute (INDEGSAL), University of Leon, Campus de Vegazana, 24071 Leon, Spain
| | - Alvaro de la Fuente
- Department of Biomedical Sciences-Physiology, Faculty of Veterinary Medicine, Animal Health Institute (INDEGSAL), University of Leon, Campus de Vegazana, 24071 Leon, Spain
| | - Ana I Alvarez
- Department of Biomedical Sciences-Physiology, Faculty of Veterinary Medicine, Animal Health Institute (INDEGSAL), University of Leon, Campus de Vegazana, 24071 Leon, Spain
| | - Oscar J Pozo
- Integrative Pharmacology and Systems Neuroscience Group, IMIM-Hospital del Mar Medical Research Institute, 88 Doctor Aiguader, 08003 Barcelona, Spain
| | - Gracia Merino
- Department of Biomedical Sciences-Physiology, Faculty of Veterinary Medicine, Animal Health Institute (INDEGSAL), University of Leon, Campus de Vegazana, 24071 Leon, Spain.
| |
Collapse
|
670
|
Zhang HY, Tian JX, Lian FM, Li M, Liu WK, Zhen Z, Liao JQ, Tong XL. Therapeutic mechanisms of traditional Chinese medicine to improve metabolic diseases via the gut microbiota. Biomed Pharmacother 2020; 133:110857. [PMID: 33197760 DOI: 10.1016/j.biopha.2020.110857] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/01/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022] Open
Abstract
Metabolic diseases such as obesity, type 2 diabetes mellitus, and hyperlipidemia are associated with the dysfunction of gut microbiota. Traditional Chinese medicines (TCMs) have shown considerable effects in the treatment of metabolic disorders by regulating the gut microbiota. However, the underlying mechanisms are unclear. Studies have shown that TCMs significantly affect glucose and lipid metabolism by modulating the gut microbiota, particularly mucin-degrading bacteria, bacteria with anti-inflammatory properties, lipopolysaccharide- and short-chain fatty acid (SCFA)-producing bacteria, and bacteria with bile-salt hydrolase activity. In this review, we explored potential mechanisms by which TCM improved metabolic disorders via regulating gut microbiota composition and functional structure. In particular, we focused on the protection of the intestinal barrier function, modulation of metabolic endotoxemia and inflammatory responses, regulation of the effects of SCFAs, modulation of the gut-brain axis, and regulation of bile acid metabolism and tryptophan metabolism as therapeutic mechanisms of TCMs in metabolic diseases.
Collapse
Affiliation(s)
- Hai-Yu Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate College, Beijing University of Traditional Chinese Medicine, Beijing, 100029, China
| | - Jia-Xing Tian
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Feng-Mei Lian
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Min Li
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Wen-Ke Liu
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Zhong Zhen
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jiang-Quan Liao
- Department of National Integrated Traditional and Western Medicine Center for Cardiovascular Disease, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiao-Lin Tong
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
671
|
Jang IY, Park JH, Kim JH, Lee S, Lee E, Lee JY, Park SJ, Kim DA, Hamrick MW, Kim BJ. The association of circulating kynurenine, a tryptophan metabolite, with frailty in older adults. Aging (Albany NY) 2020; 12:22253-22265. [PMID: 33188590 PMCID: PMC11623974 DOI: 10.18632/aging.104179] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/05/2020] [Indexed: 06/11/2023]
Abstract
Despite the accumulating evidence from in vitro and animal experiments supporting the role of kynurenine (a tryptophan metabolite) in a number of degenerative age-related changes, the relationship between kynurenine and frailty in older adults is not well understood. We collected blood samples from 73 participants who underwent a comprehensive geriatric assessment, measuring kynurenine levels using liquid chromatography-tandem mass spectrometry. We assessed the phenotypic frailty and the deficit accumulation frailty index using widely validated approaches proposed by Fried et al. and Rockwood et al., respectively. After adjusting for sex, age, and body mass index, the frail participants presented 52.9% and 34.3% higher serum kynurenine levels than those with robustness and prefrailty, respectively (P = 0.005 and 0.014, respectively). Serum kynurenine levels were positively associated with the frailty index, time to complete 5 chair stands, and patient health questionnaire-2 score and inversely associated with grip strength and gait speed (P = 0.042 to <0.001). Furthermore, the odds ratio per increase in serum kynurenine level for phenotypic frailty was approximately 2.62 (95% confidence interval = 1.22-5.65, P = 0.014). These data provide clinical evidence that circulating kynurenine might be a potential biomarker for assessing the risk of frailty in humans.
Collapse
Affiliation(s)
- Il-Young Jang
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jin Hoon Park
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jeoung Hee Kim
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seungjoo Lee
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Eunju Lee
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jin Young Lee
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - So Jeong Park
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Da Ae Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Mark W. Hamrick
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Beom-Jun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
672
|
Wang W, Zou W. Amino Acids and Their Transporters in T Cell Immunity and Cancer Therapy. Mol Cell 2020; 80:384-395. [PMID: 32997964 PMCID: PMC7655528 DOI: 10.1016/j.molcel.2020.09.006] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/01/2020] [Accepted: 09/07/2020] [Indexed: 12/25/2022]
Abstract
Metabolism reprogramming is critical for both cancer progression and effective immune responses in the tumor microenvironment. Amino acid metabolism in different cells and their cross-talk shape tumor immunity and therapy efficacy in patients with cancer. In this review, we focus on multiple amino acids and their transporters, solute carrier (SLC) members. We discuss their involvement in regulation of immune responses in the tumor microenvironment and assess their associations with cancer immunotherapy, chemotherapy, and radiation therapy, and we review their potential as targets for cancer therapy. We stress the necessity to understand individual amino acids and their transporters in different cell subsets, the molecular intersection between amino acid metabolism, and effective T cell immunity and its relevance in cancer therapies.
Collapse
Affiliation(s)
- Weimin Wang
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Graduate Program in Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Graduate Program in Cancer Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
| |
Collapse
|
673
|
Transplantation of microbiota from drug-free patients with schizophrenia causes schizophrenia-like abnormal behaviors and dysregulated kynurenine metabolism in mice. Mol Psychiatry 2020; 25:2905-2918. [PMID: 31391545 DOI: 10.1038/s41380-019-0475-4] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/28/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that gut microbiota plays a role in the pathogenesis of schizophrenia via the microbiota-gut-brain axis. This study sought to investigate whether transplantation of fecal microbiota from drug-free patients with schizophrenia into specific pathogen-free mice could cause schizophrenia-like behavioral abnormalities. The results revealed that transplantation of fecal microbiota from schizophrenic patients into antibiotic-treated mice caused behavioral abnormalities such as psychomotor hyperactivity, impaired learning and memory in the recipient animals. These mice also showed elevation of the kynurenine-kynurenic acid pathway of tryptophan degradation in both periphery and brain, as well as increased basal extracellular dopamine in prefrontal cortex and 5-hydroxytryptamine in hippocampus, compared with their counterparts receiving feces from healthy controls. Furthermore, colonic luminal filtrates from the mice transplanted with patients' fecal microbiota increased both kynurenic acid synthesis and kynurenine aminotransferase II activity in cultured hepatocytes and forebrain cortical slices. Sixty species of donor-derived bacteria showed significant difference between the mice colonized with the patients' and the controls' fecal microbiota, highlighting 78 differentially enriched functional modules including tryptophan biosynthesis function. In conclusion, our study suggests that the abnormalities in the composition of gut microbiota contribute to the pathogenesis of schizophrenia partially through the manipulation of tryptophan-kynurenine metabolism.
Collapse
|
674
|
Liu Y, Hou Y, Wang G, Zheng X, Hao H. Gut Microbial Metabolites of Aromatic Amino Acids as Signals in Host-Microbe Interplay. Trends Endocrinol Metab 2020; 31:818-834. [PMID: 32284282 DOI: 10.1016/j.tem.2020.02.012] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023]
Abstract
Gut microbial metabolism is intimately coupled with host health and disease. Aromatic amino acid (AAA) catabolism by the gut microbiome yields numerous metabolites that may regulate immune, metabolic, and neuronal responses at local and distant sites. Such a chemical dialog between host cells and the gut microbiome is shaped by environmental cues, and may become dysregulated in gastrointestinal and systems diseases. Increasing knowledge of the bacterial pathway and signaling basis may shed additional light on metabolic host-microbiome crosstalk that remains untapped for drug discovery. Here, we update our understanding of microbial AAA metabolism and its impacts on host physiology and disease. We also consider open questions related to therapeutically mining these signaling metabolites and how recent concepts and tools may drive this area forward.
Collapse
Affiliation(s)
- Yali Liu
- Laboratory of Metabolism and Drug Target Discovery, State Key Laboratory of Natural Medicines, College of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuanlong Hou
- Laboratory of Metabolism and Drug Target Discovery, State Key Laboratory of Natural Medicines, College of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Laboratory of Metabolism and Drug Target Discovery, State Key Laboratory of Natural Medicines, College of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiao Zheng
- Laboratory of Metabolism and Drug Target Discovery, State Key Laboratory of Natural Medicines, College of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Haiping Hao
- Laboratory of Metabolism and Drug Target Discovery, State Key Laboratory of Natural Medicines, College of Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
675
|
Negatu DA, Gengenbacher M, Dartois V, Dick T. Indole Propionic Acid, an Unusual Antibiotic Produced by the Gut Microbiota, With Anti-inflammatory and Antioxidant Properties. Front Microbiol 2020; 11:575586. [PMID: 33193190 PMCID: PMC7652848 DOI: 10.3389/fmicb.2020.575586] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
Most antibiotics are produced by soil microbes and typically interfere with macromolecular synthesis processes as their antibacterial mechanism of action. These natural products are often large and suffer from poor chemical tractability. Here, we discuss discovery, mechanism of action, and the therapeutic potentials of an unusual antibiotic, indole propionic acid (IPA). IPA is produced by the human gut microbiota. The molecule is small, chemically tractable, and targets amino acid biosynthesis. IPA is active against a broad spectrum of mycobacteria, including drug resistant Mycobacterium tuberculosis and non-tuberculous mycobacteria (NTM). Interestingly, the microbiota-produced metabolite is detectable in the serum of healthy individuals, tuberculosis (TB) patients, and several animal models. Thus, the microbiota in our gut may influence susceptibility to mycobacterial diseases. If a gut-lung microbiome axis can be demonstrated, IPA may have potential as a biomarker of disease progression, and development of microbiota-based therapies could be explored. In addition to its antimycobacterial activity, the molecule displays anti-inflammatory and antioxidant properties. This raises the possibility that IPA has therapeutic potential as both antibiotic and add-on host-directed drug for the treatment of TB in patient populations where disease morbidity and mortality is driven by excessive inflammation and tissue damage, such as TB-associated immune reconstitution inflammatory syndrome, TB-meningitis, and TB-diabetes.
Collapse
Affiliation(s)
- Dereje Abate Negatu
- Center for Innovative Drug Development and Therapeutic Trials for Africa, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia.,Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Martin Gengenbacher
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States.,Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States.,Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States.,Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, United States.,Department of Microbiology and Immunology, Georgetown University, Washington, DC, United States
| |
Collapse
|
676
|
Effect of Tryptophan-Derived AhR Ligands, Kynurenine, Kynurenic Acid and FICZ, on Proliferation, Cell Cycle Regulation and Cell Death of Melanoma Cells-In Vitro Studies. Int J Mol Sci 2020; 21:ijms21217946. [PMID: 33114713 PMCID: PMC7663343 DOI: 10.3390/ijms21217946] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 12/18/2022] Open
Abstract
Tryptophan metabolites: kynurenine (KYN), kynurenic acid (KYNA) and 6-formylindolo[3,2-b]carbazole (FICZ) are considered aryl hydrocarbon receptor (AhR) ligands. AhR is mainly expressed in barrier tissues, including skin, and is involved in various physiological and pathological processes in skin. We studied the effect of KYN, KYNA and FICZ on melanocyte and melanoma A375 and RPMI7951 cell toxicity, proliferation and cell death. KYN and FICZ inhibited DNA synthesis in both melanoma cell lines, but RPMI7951 cells were more resistant to pharmacological treatment. Tested compounds were toxic to melanoma cells but not to normal human adult melanocytes. Changes in the protein level of cyclin D1, CDK4 and retinoblastoma tumor suppressor protein (Rb) phosphorylation revealed different mechanisms of action of individual AhR ligands. Importantly, all tryptophan metabolites induced necrosis, but only KYNA and FICZ promoted apoptosis in melanoma A375 cells. This effect was not observed in RPMI7951 cells. KYN, KYNA and FICZ in higher concentrations inhibited the protein level of AhR but did not affect the gene expression. To conclude, despite belonging to the group of AhR ligands, KYN, KYNA and FICZ exerted different effects on proliferation, toxicity and induction of cell death in melanoma cells in vitro.
Collapse
|
677
|
Bekki S, Hashimoto S, Yamasaki K, Komori A, Abiru S, Nagaoka S, Saeki A, Suehiro T, Kugiyama Y, Beppu A, Kuroki T, Nakamura M, Ito M, Yatsuhashi H. Serum kynurenine levels are a novel biomarker to predict the prognosis of patients with hepatocellular carcinoma. PLoS One 2020; 15:e0241002. [PMID: 33085694 PMCID: PMC7577466 DOI: 10.1371/journal.pone.0241002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND We examined serum kynurenine levels in patients with chronic hepatitis C virus infection, and the relationship between serum kynurenine and prognosis in patients with hepatocellular carcinoma (HCC) and chronic hepatitis C. METHODS We retrospectively analyzed 604 patients with HCC diagnosed between January 1999 and December 2015, and 288 patients without HCC who were seen at the National Hospital Organization Nagasaki Medical Center between October 2014 and November 2017. The association between serum kynurenine and prognosis was evaluated using the Cox's proportional hazards regression analysis. RESULTS Patients with HCC had significantly higher values of serum kynurenine than patients without HCC (median: 557.1 vs. 464.2 ng/mL, p<0.001). Five-year survival rates of HCC patients with serum kynurenine ≥900 (n = 65), 600-899 (n = 194), and <600 ng/mL (n = 345) were 30.6%, 47.4%, and 61.4%, respectively (p = 0.001, log-rank test). Multivariate analysis identified serum kynurenine as an independent predictor for prognosis of HCC patients. The hazard ratio of serum kynurenine ≥900, and 600-899 compared with serum kynurenine <600 ng/mL were 1.91 (p<0.001) and 1.37 (p = 0.015), respectively. CONCLUSIONS A high level of serum kynurenine correlated with poor prognosis of HCC. Serum kynurenine levels may be a novel biomarker to predict the prognosis of patients with HCC. The development of drugs that inhibit kynurenine production is expected to help improve the prognosis of patients with HCC.
Collapse
Affiliation(s)
- Shigemune Bekki
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
- Department of Viral Hepatitis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Satoru Hashimoto
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Kazumi Yamasaki
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Atsumasa Komori
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Seigo Abiru
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Shinya Nagaoka
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Akira Saeki
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Tomoyuki Suehiro
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Yuki Kugiyama
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Asami Beppu
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Tamotsu Kuroki
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
- Department of Viral Hepatitis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Minoru Nakamura
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
- Department of Viral Hepatitis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Masahiro Ito
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
- Department of Viral Hepatitis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Hiroshi Yatsuhashi
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Nagasaki, Japan
- Department of Viral Hepatitis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
678
|
Pang L, Cui M, Dai W, Kong J, Chen H, Wu S. Can Intraoperative Low-Dose R, S-Ketamine Prevent Depressive Symptoms After Surgery? The First Meta-Analysis of Clinical Trials. Front Pharmacol 2020; 11:586104. [PMID: 33192527 PMCID: PMC7604489 DOI: 10.3389/fphar.2020.586104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Postoperative depression is a common complication after surgery that profoundly affects recovery and prognosis. New research indicates that (R,S)-ketamine is a potent antidepressant that exerts a rapid and sustained antidepressive effect. However, there is no consensus on whether intraoperative low-dose (R,S)-ketamine prevents postoperative depression. Objectives: This study aimed to investigate the safety, feasibility, and short-term complications of intraoperative low-dose (R,S)-ketamine in preventing postoperative depressive symptoms. Methods: The Web of Science, Cochrane, PubMed, and CNKI databases were systematically searched (last search February 28, 2020) to identify studies involving ketamine. Sensitivity and metaregression analyses were performed to identify potential confounders. The meta-analysis was performed using Review Manager 5.3. Results: A total of 13 studies (seven in Chinese and six in English) representing 1,148 cases of patients who were treated with (R,S)-ketamine and 874 cases of patients who received other treatments were included in the meta-analysis. Anesthesia duration and blood loss did not significantly differ between the two groups, demonstrating that (R,S)-ketamine was safe (odds ratio,OR: 0.27; 95% CI: -1.14 to 1.68; P = 0.71) for prophylactic treatment of postoperative depression. Blood loss (OR: -1.83; 95% CI: -8.34 to 4.68; P = 0.58), the number of postoperative depressive patients (95% CI: 0.8-1.07; P = 0.08; (R,S)-ketamine: control = 12.9%:15.8%), and postoperative complications (OR: 0.83, 95% CI: 0.44-1.58; P = 0.57; (R,S)-ketamine: control = 19.3%:19.3%) were all similar across groups. Intra-operative low-dose (R,S)-ketamine reduced extubation time (OR: -2.84; 95% CI: -5.48 to -0.21; P = 0.03). Conclusions: The prophylactic anti-depressant effect of (R,S)-ketamine did not significantly differ between the (R,S)-ketamine and control groups in patients undergoing general or spinal anesthesia. However, (R,S)-ketamine use led to a higher incidence of adverse reactions in patients under 40 years of age who underwent a Cesarean section under spinal anesthesia.
Collapse
Affiliation(s)
- Liwei Pang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Meiying Cui
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wanling Dai
- Innovation Institute of China Medical University, Shenyang, China
| | - Jing Kong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongzhi Chen
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Shuodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
679
|
Chen X, Xu J, Wang H, Luo J, Wang Z, Chen G, Jiang D, Cao R, Huang H, Luo D, Xiao X, Hu J. Profiling the differences of gut microbial structure between schizophrenia patients with and without violent behaviors based on 16S rRNA gene sequencing. Int J Legal Med 2020; 135:131-141. [PMID: 33067643 DOI: 10.1007/s00414-020-02439-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
Understanding the violence behaviors in schizophrenia patients has always been the focus of forensic psychiatry. Although many studies show gut microbiota could regulate behavior, to our knowledge, no studies have profiled the gut microbiota structure in schizophrenia patients with violence. We profiled the characteristics of gut microbiota structure in 26 schizophrenia patients with violence (V.SCZ) by comparing with that of 16 schizophrenia patients without violence (NV.SCZ) under the control of confounders, and found the differences of gut microbiota structure between the two groups. Violence was assessed by the MacArthur Community Violence Instrument. Psychiatric symptoms were assessed by the Positive and Negative Syndrome Scale. The 16S rRNA gene sequencing was used to identify and relatively quantify gut microbial composition. Bioinformatics analysis was used to find differential gut microbial composition between the V.SCZ and NV.SCZ groups. Fifty-nine differential microbial taxonomic compositions were found between the two groups. Fifteen gut microbial compositions were the key microbial taxonomic compositions responsible for the differences between the V.SCZ and NV.SCZ groups, including five enriched microbial taxonomic compositions (p_Bacteroidetes, c_Bacteroidia, o_Bacteroidales, f_Prevotellaceae, s_Bacteroides_uniformis), and ten impoverished microbial taxonomic compositions (p_Actinobacteria, c_unidentified_Actinobacteria, o_Bifidobacteriales, f_ Enterococcaceae, f_Veillonellaceae, f_Bifidobacteriaceae, g_Enterococcus, g_Candidatus_Saccharimonas, g_Bifidobacterium, and s_Bifidobacterium_pseudocatenulatum). This study profiled the differences of gut microbiota between schizophrenia patients with violence and without violence. These results could enrich the etiological understanding of violence in schizophrenia and might be helpful to violence management in the future.
Collapse
Affiliation(s)
- Xiacan Chen
- Institute of Forensic Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jiajun Xu
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, China.
| | - Hongren Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jiaguo Luo
- Jinxin Mental Health Center, Chengdu, China
| | - Zheng Wang
- Institute of Forensic Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Gang Chen
- Jinxin Mental Health Center, Chengdu, China
| | - Dan Jiang
- Jinxin Mental Health Center, Chengdu, China
| | - Ruochen Cao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Haolan Huang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Dan Luo
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, China
| | - Xiao Xiao
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University and the Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Junmei Hu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
680
|
Proietti E, Rossini S, Grohmann U, Mondanelli G. Polyamines and Kynurenines at the Intersection of Immune Modulation. Trends Immunol 2020; 41:1037-1050. [PMID: 33055013 DOI: 10.1016/j.it.2020.09.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Polyamines (i.e., putrescine, spermidine, and spermine) are bioactive polycations capable of binding nucleic acids and proteins and modulating signaling pathways. Polyamine functions have been studied most extensively in tumors, where they can promote cell transformation and proliferation. Recently, spermidine was found to exert protective effects in an experimental model of multiple sclerosis (MS) and to confer immunoregulatory properties on dendritic cells (DCs), via the indoleamine 2,3-dioxygenase 1 (IDO1) enzyme. IDO1 converts l-tryptophan into metabolites, collectively known as kynurenines, endowed with several immunoregulatory effects via activation of the arylhydrocarbon receptor (AhR). Because AhR activation increases polyamine production, the emerging scenario has identified polyamines and kynurenines as actors of an immunoregulatory circuitry with potential implications for immunotherapy in autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Elisa Proietti
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Sofia Rossini
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| |
Collapse
|
681
|
The specific metabolome profiling of patients infected by SARS-COV-2 supports the key role of tryptophan-nicotinamide pathway and cytosine metabolism. Sci Rep 2020; 10:16824. [PMID: 33033346 PMCID: PMC7544910 DOI: 10.1038/s41598-020-73966-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/21/2020] [Indexed: 01/10/2023] Open
Abstract
The biological mechanisms involved in SARS-CoV-2 infection are only partially understood. Thus we explored the plasma metabolome of patients infected with SARS-CoV-2 to search for diagnostic and/or prognostic biomarkers and to improve the knowledge of metabolic disturbance in this infection. We analyzed the plasma metabolome of 55 patients infected with SARS-CoV-2 and 45 controls by LC-HRMS at the time of viral diagnosis (D0). We first evaluated the ability to predict the diagnosis from the metabotype at D0 in an independent population. Next, we assessed the feasibility of predicting the disease evolution at the 7th and 15th day. Plasma metabolome allowed us to generate a discriminant multivariate model to predict the diagnosis of SARS-CoV-2 in an independent population (accuracy > 74%, sensitivity, specificity > 75%). We identified the role of the cytosine and tryptophan-nicotinamide pathways in this discrimination. However, metabolomic exploration modestly explained the disease evolution. Here, we present the first metabolomic study in SARS-CoV-2 patients which showed a high reliable prediction of early diagnosis. We have highlighted the role of the tryptophan-nicotinamide pathway clearly linked to inflammatory signals and microbiota, and the involvement of cytosine, previously described as a coordinator of cell metabolism in SARS-CoV-2. These findings could open new therapeutic perspectives as indirect targets.
Collapse
|
682
|
Gut microbial metabolites as multi-kingdom intermediates. Nat Rev Microbiol 2020; 19:77-94. [PMID: 32968241 DOI: 10.1038/s41579-020-0438-4] [Citation(s) in RCA: 754] [Impact Index Per Article: 150.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
The gut microbiota contributes to host physiology through the production of a myriad of metabolites. These metabolites exert their effects within the host as signalling molecules and substrates for metabolic reactions. Although the study of host-microbiota interactions remains challenging due to the high degree of crosstalk both within and between kingdoms, metabolite-focused research has identified multiple actionable microbial targets that are relevant for host health. Metabolites, as the functional output of combined host and microorganism interactions, provide a snapshot in time of an extraordinarily complex multi-organism system. Although substantial work remains towards understanding host-microbiota interactions and the underlying mechanisms, we review the current state of knowledge for each of the major classes of microbial metabolites with emphasis on clinical and translational research implications. We provide an overview of methodologies available for measurement of microbial metabolites, and in addition to discussion of key challenges, we provide a potential framework for integration of discovery-based metabolite studies with mechanistic work. Finally, we highlight examples in the literature where this approach has led to substantial progress in understanding host-microbiota interactions.
Collapse
|
683
|
Runtsch MC, Ferrara G, Angiari S. Metabolic determinants of leukocyte pathogenicity in neurological diseases. J Neurochem 2020; 158:36-58. [PMID: 32880969 DOI: 10.1111/jnc.15169] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/31/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
Neuroinflammatory and neurodegenerative diseases are characterized by the recruitment of circulating blood-borne innate and adaptive immune cells into the central nervous system (CNS). These leukocytes sustain the detrimental response in the CNS by releasing pro-inflammatory mediators that induce activation of local glial cells, blood-brain barrier (BBB) dysfunction, and neural cell death. However, infiltrating peripheral immune cells could also dampen CNS inflammation and support tissue repair. Recent advances in the field of immunometabolism demonstrate the importance of metabolic reprogramming for the activation and functionality of such innate and adaptive immune cell populations. In particular, an increasing body of evidence suggests that the activity of metabolites and metabolic enzymes could influence the pathogenic potential of immune cells during neuroinflammatory and neurodegenerative disorders. In this review, we discuss the role of intracellular metabolic cues in regulating leukocyte-mediated CNS damage in Alzheimer's and Parkinson's disease, multiple sclerosis and stroke, highlighting the therapeutic potential of drugs targeting metabolic pathways for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Marah C Runtsch
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | | | - Stefano Angiari
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
684
|
Impact of Exercise on Immunometabolism in Multiple Sclerosis. J Clin Med 2020; 9:jcm9093038. [PMID: 32967206 PMCID: PMC7564219 DOI: 10.3390/jcm9093038] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple Sclerosis (MS) is a chronic, autoimmune condition characterized by demyelinating lesions and axonal degradation. Even though the cause of MS is heterogeneous, it is known that peripheral immune invasion in the central nervous system (CNS) drives pathology at least in the most common form of MS, relapse-remitting MS (RRMS). The more progressive forms’ mechanisms of action remain more elusive yet an innate immune dysfunction combined with neurodegeneration are likely drivers. Recently, increasing studies have focused on the influence of metabolism in regulating immune cell function. In this regard, exercise has long been known to regulate metabolism, and has emerged as a promising therapy for management of autoimmune disorders. Hence, in this review, we inspect the role of key immunometabolic pathways specifically dysregulated in MS and highlight potential therapeutic benefits of exercise in modulating those pathways to harness an anti-inflammatory state. Finally, we touch upon current challenges and future directions for the field of exercise and immunometabolism in MS.
Collapse
|
685
|
Nutritional Therapy to Modulate Tryptophan Metabolism and Aryl Hydrocarbon-Receptor Signaling Activation in Human Diseases. Nutrients 2020; 12:nu12092846. [PMID: 32957545 PMCID: PMC7551725 DOI: 10.3390/nu12092846] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a nuclear protein which, upon association with certain endogenous and exogenous ligands, translocates into the nucleus, binds DNA and regulates gene expression. Tryptophan (Trp) metabolites are one of the most important endogenous AhR ligands. The intestinal microbiota is a critical player in human intestinal homeostasis. Many of its effects are mediated by an assembly of metabolites, including Trp metabolites. In the intestine, Trp is metabolized by three main routes, leading to kynurenine, serotonin, and indole derivative synthesis under the direct or indirect involvement of the microbiota. Disturbance in Trp metabolism and/or AhR activation is strongly associated with multiple gastrointestinal, neurological and metabolic disorders, suggesting Trp metabolites/AhR signaling modulation as an interesting therapeutic perspective. In this review, we describe the most recent advances concerning Trp metabolism and AhR signaling in human health and disease, with a focus on nutrition as a potential therapy to modulate Trp metabolites acting on AhR. A better understanding of the complex balance between these pathways in human health and disease will yield therapeutic opportunities.
Collapse
|
686
|
Cabbia A, Hilbers PA, van Riel NA. A Distance-Based Framework for the Characterization of Metabolic Heterogeneity in Large Sets of Genome-Scale Metabolic Models. PATTERNS (NEW YORK, N.Y.) 2020; 1:100080. [PMID: 33205127 PMCID: PMC7660451 DOI: 10.1016/j.patter.2020.100080] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/29/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022]
Abstract
Gene expression and protein abundance data of cells or tissues belonging to healthy and diseased individuals can be integrated and mapped onto genome-scale metabolic networks to produce patient-derived models. As the number of available and newly developed genome-scale metabolic models increases, new methods are needed to objectively analyze large sets of models and to identify the determinants of metabolic heterogeneity. We developed a distance-based workflow that combines consensus machine learning and metabolic modeling techniques and used it to apply pattern recognition algorithms to collections of genome-scale metabolic models, both microbial and human. Model composition, network topology and flux distribution provide complementary aspects of metabolic heterogeneity in patient-specific genome-scale models of skeletal muscle. Using consensus clustering analysis we identified the metabolic processes involved in the individual responses to endurance training in older adults.
Collapse
Affiliation(s)
- Andrea Cabbia
- Computational Biology, Eindhoven University of Technology, Groene Loper 5, 5612 AE Eindhoven, the Netherlands
| | - Peter A.J. Hilbers
- Computational Biology, Eindhoven University of Technology, Groene Loper 5, 5612 AE Eindhoven, the Netherlands
| | - Natal A.W. van Riel
- Computational Biology, Eindhoven University of Technology, Groene Loper 5, 5612 AE Eindhoven, the Netherlands
- Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| |
Collapse
|
687
|
Ma N, He T, Johnston LJ, Ma X. Host-microbiome interactions: the aryl hydrocarbon receptor as a critical node in tryptophan metabolites to brain signaling. Gut Microbes 2020; 11:1203-1219. [PMID: 32401136 PMCID: PMC7524279 DOI: 10.1080/19490976.2020.1758008] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tryptophan (Trp) is not only a nutrient enhancer but also has systemic effects. Trp metabolites signaling through the well-known aryl hydrocarbon receptor (AhR) constitute the interface of microbiome-gut-brain axis. However, the pathway through which Trp metabolites affect central nervous system (CNS) function have not been fully elucidated. AhR participates in a broad variety of physiological and pathological processes that also highly relevant to intestinal homeostasis and CNS diseases. Via the AhR-dependent mechanism, Trp metabolites connect bidirectional signaling between the gut microbiome and the brain, mediated via immune, metabolic, and neural (vagal) signaling mechanisms, with downstream effects on behavior and CNS function. These findings shed light on the complex Trp regulation of microbiome-gut-brain axis and add another facet to our understanding that dietary Trp is expected to be a promising noninvasive approach for alleviating systemic diseases.
Collapse
Affiliation(s)
- Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lee J. Johnston
- West Central Research & Outreach Center, University of Minnesota, Morris, MN, USA
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China,CONTACT Xi Ma State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing100193, China
| |
Collapse
|
688
|
Targeting Metabolic Pathways in Kidney Cancer: Rationale and Therapeutic Opportunities. ACTA ACUST UNITED AC 2020; 26:407-418. [PMID: 32947309 DOI: 10.1097/ppo.0000000000000472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alterations in cellular sugar, amino acid and nucleic acid, and lipid metabolism, as well as in mitochondrial function, are a hallmark of renal cell carcinoma (RCC). The activation of oncogenes such as hypoxia-inducible factor and loss of the von Hippel-Lindau function and other tumor suppressors frequently occur early on during tumorigenesis and are the drivers for these changes, collectively known as "metabolic reprogramming," which promotes cellular growth, proliferation, and stress resilience. However, tumor cells can become addicted to reprogrammed metabolism. Here, we review the current knowledge of metabolic addictions in clear cell RCC, the most common form of RCC, and to what extent this has created therapeutic opportunities to interfere with such altered metabolic pathways to selectively target tumor cells. We highlight preclinical and emerging clinical data on novel therapeutics targeting metabolic traits in clear cell RCC to provide a comprehensive overview on current strategies to exploit metabolic reprogramming clinically.
Collapse
|
689
|
Zhao J, Jiang W, Wang X, Cai Z, Liu Z, Liu G. Exercise, brain plasticity, and depression. CNS Neurosci Ther 2020; 26:885-895. [PMID: 32491278 PMCID: PMC7415205 DOI: 10.1111/cns.13385] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/25/2022] Open
Abstract
Depression is a common mental disorder characterized by high incidence, high disability, and high fatality, causing great burden to the society, families, and individuals. The changes in brain plasticity may be a main reason for depression. Recent studies have shown that exercise plays a positive role in depression, but systematic and comprehensive studies are lacking on brain plasticity changes in depression. To further understand the antidepressive effect of exercise and the changes in brain plasticity, we retrieved related literatures using key words "depression," "depressive disorder," "exercise," "brain plasticity," "brain structure," and "brain function" from the database of Web of Science, PubMed, EBSCO host, and CNKI, hoping to provide evidence for exercise in preventing and treating depression. Increase in exercise has been found negatively correlated with the risk of depression. Randomized controlled experiments have shown that aerobic exercise, resistance exercise, and mind-body exercise can improve depressive symptoms and levels. The intensity and long-term effect of exercise are now topical research issues. Exercise has been proven to reshape the brain structure of depression patients, activate the function of related brain areas, promote behavioral adaptation changes, and maintain the integrity of hippocampal and white matter volume, thus improving the brain neuroprocessing and delaying cognitive degradation in depression patients. Future studies are urgently needed to establish accurate exercise prescriptions for improving depressive symptoms, and studies on different depressive populations and studies using multimodal brain imaging combined with multiple analytical methods are also needed.
Collapse
Affiliation(s)
- Jin‐Lei Zhao
- School of Physical Education and HealthShanghai Lixin University of Accounting and FinanceShanghaiChina
| | - Wan‐Ting Jiang
- Department of Physical Education and Sport TrainingShanghai University of SportShanghaiChina
| | - Xing Wang
- Department of Physical Education and Sport TrainingShanghai University of SportShanghaiChina
| | - Zhi‐Dong Cai
- Department of Physical Education and Sport TrainingShanghai University of SportShanghaiChina
| | - Zu‐Hong Liu
- Department of Physical Education and Sport TrainingShanghai University of SportShanghaiChina
| | - Guo‐Rong Liu
- School of Physical Education and HealthShanghai Lixin University of Accounting and FinanceShanghaiChina
| |
Collapse
|
690
|
Valenzuela PL, Castillo-García A, Morales JS, de la Villa P, Hampel H, Emanuele E, Lista S, Lucia A. Exercise benefits on Alzheimer's disease: State-of-the-science. Ageing Res Rev 2020; 62:101108. [PMID: 32561386 DOI: 10.1016/j.arr.2020.101108] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 05/29/2020] [Accepted: 06/06/2020] [Indexed: 01/15/2023]
Abstract
Although there is no unanimity, growing evidence supports the value of regular physical exercise to prevent Alzheimer's disease as well as cognitive decline in affected patients. Together with an introductory summary on epidemiological evidence, the aim of this review is to summarize the current knowledge on the potential biological mechanisms underlying exercise benefits in this condition. Regular physical exercise has proven to be beneficial for traditional cardiovascular risk factors (e.g., reduced vascular flow, diabetes) involved in the pathogenesis of Alzheimer's disease. Exercise also promotes neurogenesis via increases in exercise-induced metabolic factors (e.g., ketone bodies, lactate) and muscle-derived myokines (cathepsin-B, irisin), which in turn stimulate the production of neurotrophins such as brain-derived neurotrophic factor. Finally, regular exercise exerts anti-inflammatory effects and improves the brain redox status, thereby ameliorating the pathophysiological hallmarks of Alzheimer's disease (e.g., amyloid-β deposition). In summary, physical exercise might provide numerous benefits through different pathways that might, in turn, help prevent risk and progression of Alzheimer's disease. More evidence is needed, however, based on human studies.
Collapse
|
691
|
Therapeutic Use of the Ketogenic Diet in Refractory Epilepsy: What We Know and What Still Needs to Be Learned. Nutrients 2020; 12:nu12092616. [PMID: 32867258 PMCID: PMC7551948 DOI: 10.3390/nu12092616] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 01/22/2023] Open
Abstract
Ketogenic diet (KD) has been used to treat epilepsy for 100 years. It is a high-fat, low-carbohydrate, and sufficient-protein-for-growth diet that mimics the metabolic changes occurring during starvation. Except for classic KD, its modified counterparts, including modified Atkins diet and low-glycemic-index treatment, have gained grounds to increase palatability and adherence. Strong evidence exists that the KD offers protection against seizures in difficult-to-treat epilepsy and possesses long-lasting anti-epileptic activity, improving long-term disease outcome. The KD can also provide symptomatic and disease-modifying activity in a wide range of neurodegenerative diseases. In an era of highly available new anti-seizure medications (ASMs), the challenge of refractory epilepsy has still not been solved. This metabolic therapy is increasingly considered due to unique mechanisms and turns out to be a powerful tool in the hands of a skillful team. Despite decades of extensive research to explain the mechanism of its efficacy, the precise mechanism of action is to date still largely unknown. The key feature of this successful diet is the fact that energy is derived largely from fat but not from carbohydrates. Consequently, fundamental change occurs regarding the method of energy production that causes alterations in numerous biochemical pathways, thus restoring energetic and metabolic homeostasis of the brain. There are barriers during the use of this special and individualized therapy in many clinical settings worldwide. The aim of this review is to revisit the current state of the art of therapeutic application of KD in refractory epilepsy.
Collapse
|
692
|
Westbrook R, Chung T, Lovett J, Ward C, Joca H, Yang H, Khadeer M, Tian J, Xue QL, Le A, Ferrucci L, Moaddel R, de Cabo R, Hoke A, Walston J, Abadir PM. Kynurenines link chronic inflammation to functional decline and physical frailty. JCI Insight 2020; 5:136091. [PMID: 32814718 PMCID: PMC7455140 DOI: 10.1172/jci.insight.136091] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 07/15/2020] [Indexed: 12/27/2022] Open
Abstract
Chronic inflammation is associated with physical frailty and functional decline in older adults; however, the molecular mechanisms of this linkage are not understood. A mouse model of chronic inflammation showed reduced motor function and partial denervation at the neuromuscular junction. Metabolomic profiling of these mice and further validation in frail human subjects showed significant dysregulation in the tryptophan degradation pathway, including decreased tryptophan and serotonin, and increased levels of some neurotoxic kynurenines. In humans, kynurenine strongly correlated with age, frailty status, TNF-αR1 and IL-6, weaker grip strength, and slower walking speed. To study the effects of elevated neurotoxic kynurenines on motor neuronal cell viability and axonal degeneration, we used motor neuronal cells treated with 3-hydroxykynurenine and quinolinic acid and observed neurite degeneration in a dose-dependent manner and potentiation of toxicity between 3-hydroxykynurenine and quinolinic acid. These results suggest that kynurenines mediate neuromuscular dysfunction associated with chronic inflammation and aging. Tryptophan-related toxic metabolites known as kynurenines are altered with chronic inflammation, which damages nerves in aged and frail mice and humans.
Collapse
Affiliation(s)
| | - Tae Chung
- Department of Physical Medicine and Rehabilitation, and.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Chris Ward
- Department of Orthopedics and Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Humberto Joca
- Department of Orthopedics and Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Huanle Yang
- Division of Geriatric Medicine and Gerontology
| | | | - Jing Tian
- Division of Geriatric Medicine and Gerontology
| | - Qian-Li Xue
- Division of Geriatric Medicine and Gerontology
| | - Anne Le
- Department of Oncology and.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Luigi Ferrucci
- National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Ruin Moaddel
- National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Rafa de Cabo
- National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Ahmet Hoke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeremy Walston
- Division of Geriatric Medicine and Gerontology.,Department of Medicine, Kyung Hee University, Seoul, South Korea
| | | |
Collapse
|
693
|
IL4I1 Is a Metabolic Immune Checkpoint that Activates the AHR and Promotes Tumor Progression. Cell 2020; 182:1252-1270.e34. [PMID: 32818467 DOI: 10.1016/j.cell.2020.07.038] [Citation(s) in RCA: 325] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/25/2020] [Accepted: 07/28/2020] [Indexed: 01/01/2023]
Abstract
Aryl hydrocarbon receptor (AHR) activation by tryptophan (Trp) catabolites enhances tumor malignancy and suppresses anti-tumor immunity. The context specificity of AHR target genes has so far impeded systematic investigation of AHR activity and its upstream enzymes across human cancers. A pan-tissue AHR signature, derived by natural language processing, revealed that across 32 tumor entities, interleukin-4-induced-1 (IL4I1) associates more frequently with AHR activity than IDO1 or TDO2, hitherto recognized as the main Trp-catabolic enzymes. IL4I1 activates the AHR through the generation of indole metabolites and kynurenic acid. It associates with reduced survival in glioma patients, promotes cancer cell motility, and suppresses adaptive immunity, thereby enhancing the progression of chronic lymphocytic leukemia (CLL) in mice. Immune checkpoint blockade (ICB) induces IDO1 and IL4I1. As IDO1 inhibitors do not block IL4I1, IL4I1 may explain the failure of clinical studies combining ICB with IDO1 inhibition. Taken together, IL4I1 blockade opens new avenues for cancer therapy.
Collapse
|
694
|
Wang Y, Liu KF, Yang Y, Davis I, Liu A. Observing 3-hydroxyanthranilate-3,4-dioxygenase in action through a crystalline lens. Proc Natl Acad Sci U S A 2020; 117:19720-19730. [PMID: 32732435 PMCID: PMC7443976 DOI: 10.1073/pnas.2005327117] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The synthesis of quinolinic acid from tryptophan is a critical step in the de novo biosynthesis of nicotinamide adenine dinucleotide (NAD+) in mammals. Herein, the nonheme iron-based 3-hydroxyanthranilate-3,4-dioxygenase responsible for quinolinic acid production was studied by performing time-resolved in crystallo reactions monitored by UV-vis microspectroscopy, electron paramagnetic resonance (EPR) spectroscopy, and X-ray crystallography. Seven catalytic intermediates were kinetically and structurally resolved in the crystalline state, and each accompanies protein conformational changes at the active site. Among them, a monooxygenated, seven-membered lactone intermediate as a monodentate ligand of the iron center at 1.59-Å resolution was captured, which presumably corresponds to a substrate-based radical species observed by EPR using a slurry of small-sized single crystals. Other structural snapshots determined at around 2.0-Å resolution include monodentate and subsequently bidentate coordinated substrate, superoxo, alkylperoxo, and two metal-bound enol tautomers of the unstable dioxygenase product. These results reveal a detailed stepwise O-atom transfer dioxygenase mechanism along with potential isomerization activity that fine-tunes product profiling and affects the production of quinolinic acid at a junction of the metabolic pathway.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, TX 78249
| | - Kathy Fange Liu
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Yu Yang
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, TX 78249
| | - Ian Davis
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, TX 78249
| | - Aimin Liu
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, TX 78249;
| |
Collapse
|
695
|
Hanswijk SI, Spoelder M, Shan L, Verheij MMM, Muilwijk OG, Li W, Liu C, Kolk SM, Homberg JR. Gestational Factors throughout Fetal Neurodevelopment: The Serotonin Link. Int J Mol Sci 2020; 21:E5850. [PMID: 32824000 PMCID: PMC7461571 DOI: 10.3390/ijms21165850] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/24/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
Serotonin (5-HT) is a critical player in brain development and neuropsychiatric disorders. Fetal 5-HT levels can be influenced by several gestational factors, such as maternal genotype, diet, stress, medication, and immune activation. In this review, addressing both human and animal studies, we discuss how these gestational factors affect placental and fetal brain 5-HT levels, leading to changes in brain structure and function and behavior. We conclude that gestational factors are able to interact and thereby amplify or counteract each other's impact on the fetal 5-HT-ergic system. We, therefore, argue that beyond the understanding of how single gestational factors affect 5-HT-ergic brain development and behavior in offspring, it is critical to elucidate the consequences of interacting factors. Moreover, we describe how each gestational factor is able to alter the 5-HT-ergic influence on the thalamocortical- and prefrontal-limbic circuitry and the hypothalamo-pituitary-adrenocortical-axis. These alterations have been associated with risks to develop attention deficit hyperactivity disorder, autism spectrum disorders, depression, and/or anxiety. Consequently, the manipulation of gestational factors may be used to combat pregnancy-related risks for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sabrina I. Hanswijk
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Marcia Spoelder
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Ling Shan
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands;
| | - Michel M. M. Verheij
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Otto G. Muilwijk
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Weizhuo Li
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China; (W.L.); (C.L.)
| | - Chunqing Liu
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China; (W.L.); (C.L.)
| | - Sharon M. Kolk
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 AJ Nijmegen, The Netherlands;
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| |
Collapse
|
696
|
Tryptophan and kynurenine stimulate human decidualization via activating Aryl hydrocarbon receptor: Short title: Kynurenine action on human decidualization. Reprod Toxicol 2020; 96:282-292. [PMID: 32781018 DOI: 10.1016/j.reprotox.2020.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/27/2020] [Indexed: 12/30/2022]
Abstract
Decidualization is essential for successful pregnancy in rodents and primates. Although L-Tryptophan and its metabolites are essential for mammalian pregnancy, the underlying mechanism is poorly defined. We explored effects of tryptophan and kynurenine on human in vitro decidualization in human endometrial stromal cell line and primary endometrial stromal cells. Tryptophan significantly stimulates the expression of prolactin and insulin growth factor binding protein 1, reliable markers for human decidualization. When stromal cells are treated with tryptophan, tryptophan hydroxylase-1 remains unchanged, but indoleamine 2,3-dioxygenase 1 is significantly increased, suggesting tryptophan is mainly metabolized through kynurenine pathway. Kynurenine significantly stimulates insulin growth factor binding protein 1 expression. Aryl hydrocarbon receptor and its target genes (P450 1A1 and P450 1B1) are significantly increased by tryptophan and kynurenine. The induction of tryptophan and kynurenine on insulin growth factor binding protein 1 is abrogated by CH223191, an aryl hydrocarbon receptor inhibitor. Cytochrome P450 1A1 and P450 1B1 catalyze the oxidative metabolism of estradiol to catechol estrogens (2-hydroxy estradiol and 4-hydroxy estradiol), respectively. Insulin growth factor binding protein 1 is up-regulated by 2-hydroxy estradiol and 4-hydroxy estradiol. Interferon-γ significantly induces the expression of indoleamine 2,3-dioxygenase 1, aryl hydrocarbon receptor and insulin growth factor binding protein 1. All the data are also verified in primary human stromal cells. Our data indicate that Interferon-γ-induced kynurenine pathway promotes human decidualization via aryl hydrocarbon receptor signaling.
Collapse
|
697
|
Abstract
Antiretroviral therapies efficiently block HIV-1 replication but need to be maintained for life. Moreover, chronic inflammation is a hallmark of HIV-1 infection that persists despite treatment. There is, therefore, an urgent need to better understand the mechanisms driving HIV-1 pathogenesis and to identify new targets for therapeutic intervention. In the past few years, the decisive role of cellular metabolism in the fate and activity of immune cells has been uncovered, as well as its impact on the outcome of infectious diseases. Emerging evidence suggests that immunometabolism has a key role in HIV-1 pathogenesis. The metabolic pathways of CD4+ T cells and macrophages determine their susceptibility to infection, the persistence of infected cells and the establishment of latency. Immunometabolism also shapes immune responses against HIV-1, and cell metabolic products are key drivers of inflammation during infection. In this Review, we summarize current knowledge of the links between HIV-1 infection and immunometabolism, and we discuss the potential opportunities and challenges for therapeutic interventions.
Collapse
|
698
|
Kubacka J, Stefańska A, Sypniewska G. Kynurenine pathway: the link between depressive disorders and inflammation. POSTEP HIG MED DOSW 2020. [DOI: 10.5604/01.3001.0014.3454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Depression is highly prevalent worldwide and the leading cause of disability. It is believed that currently more than 300 million people of all ages suffer from depression. However, the unambiguous cause of the depression remains unknown. It is suggested that the occurrence of this disease is primarily affected by genetic factors, psychological factors and atypical brain structure or function. Recently, an increasingly important role is attributed to the inflammatory response, which is considered to be the main cause of depression. Activation of the kynurenine pathway (KP) is one of the described mechanisms by which inflammation can induce depression. Kynurenine pathway activation is associated with several neuropsychiatric diseases, including major depression disorder (MDD). The imbalance between the neuroprotective and neurotoxic metabolites in the kynurenine pathway and the associated serotonin and melatonin deficiency, may contribute to the manifestation of depressive symptoms. In this review we discuss the role of the major enzymes of the tryptophan KP: tryptophan 2,3-dioxygenase (TDO) and indoleamine 2,3-dioxygenase (IDO) and the role of selected kynurenic metabolites in the depressive disorders. Particular attention was also paid to the genetic basis of depressive disorders and to the summary of current knowledge on the effectiveness of treatment and supplementation with tryptophan and 5-hydroxytryptophan in depression.
Collapse
Affiliation(s)
- Justyna Kubacka
- Department of Laboratory Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Anna Stefańska
- Department of Laboratory Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Grażyna Sypniewska
- Department of Laboratory Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| |
Collapse
|
699
|
Jena A, Montoya CA, Mullaney JA, Dilger RN, Young W, McNabb WC, Roy NC. Gut-Brain Axis in the Early Postnatal Years of Life: A Developmental Perspective. Front Integr Neurosci 2020; 14:44. [PMID: 32848651 PMCID: PMC7419604 DOI: 10.3389/fnint.2020.00044] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence suggests that alterations in the development of the gastrointestinal (GI) tract during the early postnatal period can influence brain development and vice-versa. It is increasingly recognized that communication between the GI tract and brain is mainly driven by neural, endocrine, immune, and metabolic mediators, collectively called the gut-brain axis (GBA). Changes in the GBA mediators occur in response to the developmental changes in the body during this period. This review provides an overview of major developmental events in the GI tract and brain in the early postnatal period and their parallel developmental trajectories under physiological conditions. Current knowledge of GBA mediators in context to brain function and behavioral outcomes and their synthesis and metabolism (site, timing, etc.) is discussed. This review also presents hypotheses on the role of the GBA mediators in response to the parallel development of the GI tract and brain in infants.
Collapse
Affiliation(s)
- Ankita Jena
- School of Food & Advanced Technology, College of Sciences, Massey University, Palmerston North, New Zealand.,The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
| | - Carlos A Montoya
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
| | - Jane A Mullaney
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Wayne Young
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Warren C McNabb
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C Roy
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Liggins Institute, The University of Auckland, Auckland, New Zealand.,Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| |
Collapse
|
700
|
Brown J, Robusto B, Morel L. Intestinal Dysbiosis and Tryptophan Metabolism in Autoimmunity. Front Immunol 2020; 11:1741. [PMID: 32849620 PMCID: PMC7417361 DOI: 10.3389/fimmu.2020.01741] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022] Open
Abstract
The development of autoimmunity involves complex interactions between genetics and environmental triggers. The gut microbiota is an important environmental constituent that can heavily influence both local and systemic immune reactivity through distinct mechanisms. It is therefore a relevant environmental trigger or amplifier to consider in autoimmunity. This review will examine recent evidence for an association between intestinal dysbiosis and autoimmune diseases, and the mechanisms by which the gut microbiota may contribute to autoimmune activation. We will specifically focus on recent studies connecting tryptophan metabolism to autoimmune disease pathogenesis and discuss evidence for a microbial origin. This will be discussed in the context of our current understanding of how tryptophan metabolites regulate immune responses, and how it may, or may not, be applicable to autoimmunity.
Collapse
Affiliation(s)
- Josephine Brown
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Brian Robusto
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|