1
|
Pocivavsek A, Schwarcz R, Erhardt S. Neuroactive Kynurenines as Pharmacological Targets: New Experimental Tools and Exciting Therapeutic Opportunities. Pharmacol Rev 2024; 76:978-1008. [PMID: 39304346 DOI: 10.1124/pharmrev.124.000239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Both preclinical and clinical studies implicate functional impairments of several neuroactive metabolites of the kynurenine pathway (KP), the major degradative cascade of the essential amino acid tryptophan in mammals, in the pathophysiology of neurologic and psychiatric diseases. A number of KP enzymes, such as tryptophan 2,3-dioxygenase (TDO2), indoleamine 2,3-dioxygenases (IDO1 and IDO2), kynurenine aminotransferases (KATs), kynurenine 3-monooxygenase (KMO), 3-hydroxyanthranilic acid oxygenase (3-HAO), and quinolinic acid phosphoribosyltransferase (QPRT), control brain KP metabolism in health and disease and are therefore increasingly considered to be promising targets for the treatment of disorders of the nervous system. Understanding the distribution, cellular expression, and regulation of KP enzymes and KP metabolites in the brain is therefore critical for the conceptualization and implementation of successful therapeutic strategies. SIGNIFICANCE STATEMENT: Studies have implicated the kynurenine pathway of tryptophan in the pathophysiology of neurologic and psychiatric diseases. Key enzymes of the kynurenine pathway regulate brain metabolism in both health and disease, making them promising targets for treating these disorders. Therefore, understanding the distribution, cellular expression, and regulation of these enzymes and metabolites in the brain is critical for developing effective therapeutic strategies. This review endeavors to describe these processes in detail.
Collapse
Affiliation(s)
- Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| | - Robert Schwarcz
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| | - Sophie Erhardt
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| |
Collapse
|
2
|
Kotańska M, Łanocha M, Bednarski M, Marcinkowska M. MM165 - A Small Hybrid Molecule Modulates the Kynurenine Pathway and Attenuates Lipopolysaccharide-Induced Memory Deficits and Inflammation. Neurochem Res 2024; 49:1200-1211. [PMID: 38381245 DOI: 10.1007/s11064-024-04105-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/22/2024]
Abstract
Cognitive dysfunctions are now recognized as core symptoms of various psychiatric disorders e.g., major depressive disorder. Sustained immune activation may leads to cognitive dysfunctions. Proinflammatory cytokines shunt the metabolism of tryptophan towards kynurenine and quinolinic acid may accumulate at toxic concentrations. This acid triggers an increase in neuronal nitric oxide synthase function and promotes oxidative stress. The searching for small molecules that can regulate tryptophan metabolites produced in the kynurenic pathway has become an important goal in developing treatments for various central nervous system diseases with an inflammatory component. Previously we have identified a small hybrid molecule - MM165 which significantly reduces depressive-like symptoms caused by inflammation induced by lipopolysaccharide administration. In the present study, we investigated whether this compound would mitigate cognitive deficits induced by lipopolysaccharide administration and whether treatment with it would affect the plasma or brain levels of quinolinic acid and kynurenic acid. Neuroinflammation was induced in rats by administering lipopolysaccharide at a dose of 0.5 mg/kg body weight for 10 days. We conducted two tests: novel object recognition and object location, to assess the effect on memory impairment in animals previously treated with lipopolysaccharide. In plasma collected from rats, the concentrations of C-reactive protein and tumor necrosis factor alfa were determined. The concentrations of kynurenic acid and quinolinic acid were determined in plasma and homogenates obtained from the cerebral cortex of rats. Interleukin 6 in the cerebral cortex of rats was determined. Additionally, the body and spleen mass and spontaneous activity were measured in rats. Our study shows that MM165 may mitigate cognitive deficits induced by inflammation after administration of lipopolysaccharide and alter the concentrations of tryptophan metabolites in the brain. Compounds exhibiting a mechanism of action analogous to that of MM165 may serve as foundational structures for the development of a new class of antidepressants.
Collapse
Affiliation(s)
- Magdalena Kotańska
- Department of Pharmacological Screening, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna, 30-688, Krakow, Poland.
| | - Michał Łanocha
- Department of Pharmacological Screening, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna, 30-688, Krakow, Poland
| | - Marek Bednarski
- Department of Pharmacological Screening, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna, 30-688, Krakow, Poland
| | - Monika Marcinkowska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna, 30-688, Krakow, Poland
| |
Collapse
|
3
|
Boyle CC, Bower JE, Eisenberger NI, Irwin MR. Stress to inflammation and anhedonia: Mechanistic insights from preclinical and clinical models. Neurosci Biobehav Rev 2023; 152:105307. [PMID: 37419230 DOI: 10.1016/j.neubiorev.2023.105307] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Anhedonia, as evidenced by impaired pleasurable response to reward, reduced reward motivation, and/or deficits in reward-related learning, is a common feature of depression. Such deficits in reward processing are also an important clinical target as a risk factor for depression onset. Unfortunately, reward-related deficits remain difficult to treat. To address this gap and inform the development of effective prevention and treatment strategies, it is critical to understand the mechanisms that drive impairments in reward function. Stress-induced inflammation is a plausible mechanism of reward deficits. The purpose of this paper is to review evidence for two components of this psychobiological pathway: 1) the effects of stress on reward function; and 2) the effects of inflammation on reward function. Within these two areas, we draw upon preclinical and clinical models, distinguish between acute and chronic effects of stress and inflammation, and address specific domains of reward dysregulation. By addressing these contextual factors, the review reveals a nuanced literature which might be targeted for additional scientific inquiry to inform the development of precise interventions.
Collapse
Affiliation(s)
- Chloe C Boyle
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, UCLA, USA.
| | - Julienne E Bower
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, UCLA, USA; Department of Psychology, UCLA, Los Angeles, CA, USA
| | | | - Michael R Irwin
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, UCLA, USA
| |
Collapse
|
4
|
Phing AH, Makpol S, Nasaruddin ML, Wan Zaidi WA, Ahmad NS, Embong H. Altered Tryptophan-Kynurenine Pathway in Delirium: A Review of the Current Literature. Int J Mol Sci 2023; 24:5580. [PMID: 36982655 PMCID: PMC10056900 DOI: 10.3390/ijms24065580] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Delirium, a common form of acute brain dysfunction, is associated with increased morbidity and mortality, especially in older patients. The underlying pathophysiology of delirium is not clearly understood, but acute systemic inflammation is known to drive delirium in cases of acute illnesses, such as sepsis, trauma, and surgery. Based on psychomotor presentations, delirium has three main subtypes, such as hypoactive, hyperactive, and mixed subtype. There are similarities in the initial presentation of delirium with depression and dementia, especially in the hypoactive subtype. Hence, patients with hypoactive delirium are frequently misdiagnosed. The altered kynurenine pathway (KP) is a promising molecular pathway implicated in the pathogenesis of delirium. The KP is highly regulated in the immune system and influences neurological functions. The activation of indoleamine 2,3-dioxygenase, and specific KP neuroactive metabolites, such as quinolinic acid and kynurenic acid, could play a role in the event of delirium. Here, we collectively describe the roles of the KP and speculate on its relevance in delirium.
Collapse
Affiliation(s)
- Ang Hui Phing
- Department of Emergency Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.M.)
| | - Muhammad Luqman Nasaruddin
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.M.)
| | - Wan Asyraf Wan Zaidi
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Nurul Saadah Ahmad
- Department of Emergency Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Hashim Embong
- Department of Emergency Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
5
|
Bhatt S, Dhar AK, Samanta MK, Suttee A. Effects of Current Psychotropic Drugs on Inflammation and Immune System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:407-434. [PMID: 36949320 DOI: 10.1007/978-981-19-7376-5_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
The immune system and inflammation are involved in the pathological progression of various psychiatric disorders such as depression or major depressive disorder (MDD), generalized anxiety disorder (GAD) or anxiety, schizophrenia, Alzheimer's disease (AD), and Huntington's disease. It is observed that levels of inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and other markers are highly increased in the abovementioned disorders. The inflammation and immune component also lead to enhance the oxidative stress. The oxidative stress and increased production of reactive oxygen species (ROS) are considered as important factors that are involved in pathological progression of psychiatric disorders. Increase production of ROS is associated with excessive inflammation followed by cell necrosis and death. The psychotropic drugs are mainly work through modulations of neurotransmitter system. However, it is evident that inflammation and immune modulation are also having important role in the progression of psychiatric disorders. Rationale of the use of current psychotropic drugs is modulation of immune system by them. However, the effects of psychotropic drugs on the immune system and how these might contribute to their efficacy remain largely unclear. The drugs may act through modification of inflammation and related markers. The main purpose of this book chapter is to address the role of current psychotropic drugs on inflammation and immune system. Moreover, it will also address the role of inflammation in the progression of psychiatric disorders.
Collapse
Affiliation(s)
- Shvetank Bhatt
- School of Pharmacy, Dr. Vishwanath Karad MIT World Peace University, Pune, Maharashtra, India
- Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior, India
| | | | | | - Ashish Suttee
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
6
|
Chen G, Shi F, Yin W, Guo Y, Liu A, Shuai J, Sun J. Gut microbiota dysbiosis: The potential mechanisms by which alcohol disrupts gut and brain functions. Front Microbiol 2022; 13:916765. [PMID: 35966709 PMCID: PMC9372561 DOI: 10.3389/fmicb.2022.916765] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/29/2022] [Indexed: 11/24/2022] Open
Abstract
Alcohol use disorder (AUD) is a high-risk psychiatric disorder and a key cause of death and disability in individuals. In the development of AUD, there is a connection known as the microbiota-gut-brain axis, where alcohol use disrupts the gut barrier, resulting in changes in intestinal permeability as well as the gut microbiota composition, which in turn impairs brain function and worsens the patient’s mental status and gut activity. Potential mechanisms are explored by which alcohol alters gut and brain function through the effects of the gut microbiota and their metabolites on immune and inflammatory pathways. Alcohol and microbiota dysregulation regulating neurotransmitter release, including DA, 5-HT, and GABA, are also discussed. Thus, based on the above discussion, it is possible to speculate on the gut microbiota as an underlying target for the treatment of diseases associated with alcohol addiction. This review will focus more on how alcohol and gut microbiota affect the structure and function of the gut and brain, specific changes in the composition of the gut microbiota, and some measures to mitigate the changes caused by alcohol exposure. This leads to a potential intervention for alcohol addiction through fecal microbiota transplantation, which could normalize the disruption of gut microbiota after AUD.
Collapse
Affiliation(s)
- Ganggang Chen
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Fenglei Shi
- Department of Othopaedics, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Yin
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Yao Guo
- Shandong Provincial Mental Health Center, Jinan, China
| | - Anru Liu
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Jiacheng Shuai
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Jinhao Sun
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
- *Correspondence: Jinhao Sun,
| |
Collapse
|
7
|
Giménez-Gómez P, Ballestín R, Gil de Biedma-Elduayen L, Vidal R, Ferrer-Pérez C, Reguilón MD, O'Shea E, Miñarro J, Colado MI, Rodríguez-Arias M. Decreased kynurenine pathway potentiate resilience to social defeat effect on cocaine reward. Neuropharmacology 2021; 197:108753. [PMID: 34389399 DOI: 10.1016/j.neuropharm.2021.108753] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/26/2021] [Accepted: 08/08/2021] [Indexed: 02/08/2023]
Abstract
The kynurenine (KYN) pathway of tryptophan (TRP) degradation is activated by stress and inflammatory factors. It is now well established that social stress induces the activation of the immune system, with central inflammation and KYN metabolism being two of the main factors linking stress with depression. The aim of the present study was to evaluate the long-lasting changes in the KYN pathway induced by social defeat (SD) associated with the resilience or susceptibility to an increase in the conditioned rewarding effects of cocaine. Mice were exposed to repeated SD and 3 weeks later, a conditioned place preference (CPP) induced by a subthreshold dose of cocaine (1.5 mg/kg) was developed. KYN levels in plasma, cerebellum, hippocampus, striatum and limbic forebrain were studied at the end of the CPP procedure. Changes in the KYN pathway after exposure to pharmacological (oxytocin and indomethacin) and environmental interventions (environmental enrichment) were also evaluated. Our results showed that defeated susceptible (SD-S) mice had higher conditioning scores than resilient mice (SD-R). In addition, although KYN concentration was elevated in all defeated mice, SD-R mice showed smaller increases in KYN concentration in the cerebellum than SD-S mice. Oxytocin or Indomethacin treatment before SD normalized cocaine-induced CPP, although the increase in the KYN pathway was maintained. However, environmental enrichment before SD normalized cocaine-induced CPP and prevented the increase in the KYN pathway. The present study highlights the role of the KYN pathway and anti-inflammatory drugs acting on TRP metabolism as pharmacological targets to potentiate resilience to social stress effects.
Collapse
Affiliation(s)
- Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Raúl Ballestín
- Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010, Valencia, Spain
| | - Leticia Gil de Biedma-Elduayen
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Rebeca Vidal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Carmen Ferrer-Pérez
- Departmento de Psicología and Sociología, Universidad de Zaragoza, C/ Ciudad Escolar s/n, 44003, Teruel, Spain
| | - Marina D Reguilón
- Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010, Valencia, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - José Miñarro
- Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010, Valencia, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - María Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Marta Rodríguez-Arias
- Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010, Valencia, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.
| |
Collapse
|
8
|
Notarangelo FM, Schwarcz R. A single prenatal lipopolysaccharide injection has acute, but not long-lasting, effects on cerebral kynurenine pathway metabolism in mice. Eur J Neurosci 2021; 54:5968-5981. [PMID: 34363411 DOI: 10.1111/ejn.15416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/26/2021] [Accepted: 08/03/2021] [Indexed: 11/28/2022]
Abstract
In rodents, a single injection of lipopolysaccharide (LPS) during gestation causes chemical and functional abnormalities in the offspring. These effects may involve changes in the kynurenine pathway (KP) of tryptophan degradation and may provide insights into the pathophysiology of psychiatric diseases. Using CD1 mice, we examined acute and long-term effects of prenatal LPS treatment on the levels of kynurenine and its neuroactive downstream products kynurenic acid (KYNA), 3-hydroxykynurenine (3-HK) and quinolinic acid. To this end, LPS (100 μg/kg, i.p.) was administered on gestational day 15, and KP metabolites were measured 4 and 24 h later or in adulthood. After 4 h, kynurenine, KYNA and 3-HK levels were elevated in the fetal brain, 3-HK and KYNA levels were increased in the maternal plasma, and kynurenine was increased in the maternal brain, whereas no changes were seen in the placenta. These effects were less prominent after 24 h, and prenatal LPS did not affect the basal levels of KP metabolites in the forebrain of adult animals. In addition, a second LPS injection (1 mg/kg) in adulthood in the offspring of prenatally saline- and LPS-treated mice caused a similar elevation in 3-HK levels in both groups after 24 h, but the effect was significantly more pronounced in male mice. Thus, acute immune activation during pregnancy has only short-lasting effects on KP metabolism and does not cause cerebral KP metabolites to be disproportionally affected by a second immune challenge in adulthood. However, prenatal KYNA elevations still contribute to functional abnormalities in the offspring.
Collapse
Affiliation(s)
- Francesca M Notarangelo
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Więdłocha M, Marcinowicz P, Janoska-Jaździk M, Szulc A. Gut microbiota, kynurenine pathway and mental disorders - Review. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110145. [PMID: 33203568 DOI: 10.1016/j.pnpbp.2020.110145] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023]
Abstract
The intestine and the gut-associated limphoid tissue constitute the largest immunity organ of the human body. Among several possible tryptophan metabolism routes, the kynurenine pathway can be influenced by the gut microbiota. Disturbances of gut biodiversity may cause increased gut permeability and cause systemic inflammation, also related to central nervous system. Proinflammatory cytokines induce kynurenine pathway enzymes resulting in formation of neuroactive metabolites, which are being associated with several psychiatric disorders. The kynurenine pathway may also be influenced by certain bacteria species directly. The aim of this review is to highlight the current knowledge on the interaction of gut microbiota and the central nervous system with the kynurenine pathway taken into special account. Up to date study results on specific psychiatric disorders such as schizophrenia, bipolar disorder, Alzheimer's disease, autism spectrum disorders, depression and alcoholism are presented. Available evidence suggests that toxicity of kynurenine metabolites may be reduced by adjunction of probiotics which can affect proinflammatory cytokines. Due to their potential for modulation of the kynurenine pathway, gut microbiota pose an interesting target for future therapies.
Collapse
Affiliation(s)
- Magdalena Więdłocha
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Poland.
| | - Piotr Marcinowicz
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Poland
| | | | - Agata Szulc
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Poland
| |
Collapse
|
10
|
Sellgren CM, Imbeault S, Larsson MK, Oliveros A, Nilsson IAK, Codeluppi S, Orhan F, Bhat M, Tufvesson-Alm M, Gracias J, Kegel ME, Zheng Y, Faka A, Svedberg M, Powell SB, Caldwell S, Kamenski ME, Vawter MP, Schulmann A, Goiny M, Svensson CI, Hökfelt T, Schalling M, Schwieler L, Cervenka S, Choi DS, Landén M, Engberg G, Erhardt S. GRK3 deficiency elicits brain immune activation and psychosis. Mol Psychiatry 2021; 26:6820-6832. [PMID: 33976392 PMCID: PMC8760053 DOI: 10.1038/s41380-021-01106-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 04/07/2021] [Indexed: 02/03/2023]
Abstract
The G protein-coupled receptor kinase (GRK) family member protein GRK3 has been linked to the pathophysiology of schizophrenia and bipolar disorder. Expression, as well as protein levels, of GRK3 are reduced in post-mortem prefrontal cortex of schizophrenia subjects. Here, we investigate functional behavior and neurotransmission related to immune activation and psychosis using mice lacking functional Grk3 and utilizing a variety of methods, including behavioral, biochemical, electrophysiological, molecular, and imaging methods. Compared to wildtype controls, the Grk3-/- mice show a number of aberrations linked to psychosis, including elevated brain levels of IL-1β, increased turnover of kynurenic acid (KYNA), hyper-responsiveness to D-amphetamine, elevated spontaneous firing of midbrain dopamine neurons, and disruption in prepulse inhibition. Analyzing human genetic data, we observe a link between psychotic features in bipolar disorder, decreased GRK expression, and increased concentration of CSF KYNA. Taken together, our data suggest that Grk3-/- mice show face and construct validity relating to the psychosis phenotype with glial activation and would be suitable for translational studies of novel immunomodulatory agents in psychotic disorders.
Collapse
Affiliation(s)
- Carl M. Sellgren
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden ,grid.4714.60000 0004 1937 0626Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm & Stockholm Health Care Services, Region Stockholm, Sweden
| | - Sophie Imbeault
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Markus K. Larsson
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Alfredo Oliveros
- grid.66875.3a0000 0004 0459 167XDepartment of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN USA
| | - Ida A. K. Nilsson
- grid.4714.60000 0004 1937 0626Translational Psychiatry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Simone Codeluppi
- grid.4714.60000 0004 1937 0626Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Funda Orhan
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Bhat
- grid.418151.80000 0001 1519 6403Research and Development, Innovative Medicines, Personalised Healthcare and Biomarkers, Translational Science Centre, Science for Life Laboratory, AstraZeneca, Solna, Sweden ,grid.4714.60000 0004 1937 0626Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Maximilian Tufvesson-Alm
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jessica Gracias
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Magdalena E. Kegel
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Yiran Zheng
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anthi Faka
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Svedberg
- grid.4714.60000 0004 1937 0626Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Susan B. Powell
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Sorana Caldwell
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Mary E. Kamenski
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Marquis P. Vawter
- grid.266093.80000 0001 0668 7243Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine School of Medicine, Irvine, CA USA
| | - Anton Schulmann
- grid.416868.50000 0004 0464 0574Human Genetics Branch, National Institute of Mental Health, Bethesda, MD USA
| | - Michel Goiny
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Camilla I. Svensson
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Hökfelt
- grid.4714.60000 0004 1937 0626Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Schalling
- grid.4714.60000 0004 1937 0626Translational Psychiatry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lilly Schwieler
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Simon Cervenka
- grid.4714.60000 0004 1937 0626Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm & Stockholm Health Care Services, Region Stockholm, Sweden
| | - Doo-Sup Choi
- grid.66875.3a0000 0004 0459 167XDepartment of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN USA ,grid.66875.3a0000 0004 0459 167XDepartment of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN USA
| | - Mikael Landén
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden ,grid.4714.60000 0004 1937 0626Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Göran Engberg
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Sophie Erhardt
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Repeated administration of LPS exaggerates amphetamine-induced locomotor response and causes learning deficits in mice. J Neuroimmunol 2020; 349:577401. [PMID: 33002724 DOI: 10.1016/j.jneuroim.2020.577401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 09/03/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
Immune activation contributes to the pathophysiology of psychiatric disorders. Administration of a single dose of lipopolysaccharides (LPS) has been shown to induce depressive- and anxiety-like behaviors in rodents through activation of the kynurenine pathway, increasing levels of the N-methyl-d-aspartate (NMDA) receptor agonist quinolinic acid. Conversely, repeated administration of LPS produces increased levels of the NMDA receptor antagonist kynurenic acid. Here we show that repeated LPS administration increases sensitivity to D-amphetamine and produces cognitive deficits and anxiety-like behavior. Together, our behavioral data suggests that repeated LPS administration may be useful to study the contribution of inflammation to psychiatric disorders such as schizophrenia.
Collapse
|
12
|
You MJ, Bang M, Park HS, Yang B, Jang KB, Yoo J, Hwang DY, Kim M, Kim B, Lee SH, Kwon MS. Human umbilical cord-derived mesenchymal stem cells alleviate schizophrenia-relevant behaviors in amphetamine-sensitized mice by inhibiting neuroinflammation. Transl Psychiatry 2020; 10:123. [PMID: 32341334 PMCID: PMC7186225 DOI: 10.1038/s41398-020-0802-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/22/2020] [Accepted: 03/03/2020] [Indexed: 12/11/2022] Open
Abstract
At present, therapeutic options available for treating schizophrenia are limited to monoamine-based antipsychotic drugs. Recent genome wide association study (GWAS) indicated a close relationship between immune system and schizophrenia. To leverage the GWAS finding for therapeutic strategy, we conducted a mechanism and effect study on application of human umbilical cord-derived mesenchymal stem cells (hUC-MSC) with potent immune-modulatory effect in an animal model useful for the study of schizophrenia. Schizophrenia-relevant behaviors were induced by amphetamine administration (amphetamine-sensitized mice) and the effect of a single intravenous administration of hUC-MSC was examined in the amphetamine-sensitized mice. Schizophrenia-relevant behaviors were assessed by open field test, light/dark box, social interaction test, latent inhibition, prepulse inhibition, tail suspension test, and forced swimming test. Our results indicated that neuroinflammation along with peripheral TNF-α elevation is associated with schizophrenia-relevant behaviors in amphetamine-sensitized mice. In addition, hUC-MSC inhibited schizophrenia-relevant and the neuroinflammatory changes. The main mechanism of hUC-MSC was associated with the induction of Treg and production of the anti-inflammatory cytokine, IL-10 in periphery. In vitro study revealed that amphetamine did not directly induce a neuroinflammatory reaction, while recombinant TNF-α (rTNF-α) increased mRNA expression of TNF-α, KMO, and IL-1β in several microglial cell lines. Moreover, recombinant IL-10 (rIL-10) and MSC conditioned media inhibited the inflammatory response in rTNF-α-treated microglial cells. Assuming that hUC-MSCs rarely reach the CNS and do not remain in the body for an extended time, these findings suggest that a single hUC-MSC infusion have long-term beneficial effect via regulatory T cell induction and secretion of IL-10 in amphetamine-sensitized mice.
Collapse
Affiliation(s)
- Min-Jung You
- grid.410886.30000 0004 0647 3511Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do 13488 Republic of Korea
| | - Minji Bang
- grid.410886.30000 0004 0647 3511Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do 13496 Republic of Korea
| | - Hyun-Sun Park
- grid.410886.30000 0004 0647 3511Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do 13488 Republic of Korea
| | - Bohyun Yang
- grid.410886.30000 0004 0647 3511Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do 13488 Republic of Korea
| | - Kyu Beom Jang
- grid.410886.30000 0004 0647 3511Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do 13488 Republic of Korea
| | - Jongman Yoo
- grid.410886.30000 0004 0647 3511Department of Microbiology, School of Medicine, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do 13488 Republic of Korea
| | - Dong-Youn Hwang
- grid.410886.30000 0004 0647 3511Department of Microbiology, School of Medicine, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do 13488 Republic of Korea
| | - MinYoung Kim
- grid.410886.30000 0004 0647 3511Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, 13497 Republic of Korea
| | - Borah Kim
- grid.410886.30000 0004 0647 3511Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do 13496 Republic of Korea
| | - Sang-Hyuk Lee
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| | - Min-Soo Kwon
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
13
|
Hong J, Bang M. Anti-inflammatory Strategies for Schizophrenia: A Review of Evidence for Therapeutic Applications and Drug Repurposing. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2020; 18:10-24. [PMID: 31958901 PMCID: PMC7006977 DOI: 10.9758/cpn.2020.18.1.10] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/12/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a debilitating psychiatric disorder with a substantial socioeconomic and humanistic burden. Currently available treatment strategies mostly rely on antipsychotic drugs, which block dopaminergic effects in the mesolimbic pathway of the brain. Although antipsychotic drugs help relieve psychotic symptoms, a definitive cure for schizophrenia has yet to be achieved. Recent advances in neuroinflammation research suggest that proinflammatory processes in the brain could cause alterations in neurobehavioral development and increase vulnerability to schizophrenia. With a growing need for novel strategies in the treatment of schizophrenia, it would be meaningful to review the current evidence supporting the therapeutic potential of anti-inflammatory strategies. This review details the key findings of clinical trials that investigate the efficacy of anti-inflammatory agents as adjuvants to antipsychotic treatment. We further discuss the possibilities of repurposing anti-inflammatory agents and developing novel strategies for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Jonghee Hong
- CHA University School of Medicine, Seongnam, Korea
| | - Minji Bang
- Department of Psychiatry, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| |
Collapse
|
14
|
Effects of IDO1 and TDO2 inhibition on cognitive deficits and anxiety following LPS-induced neuroinflammation. Acta Neuropsychiatr 2020; 32:43-53. [PMID: 31753057 DOI: 10.1017/neu.2019.44] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Sustained immune activation leads to cognitive dysfunctions, depression-, and anxiety-like behaviours in humans and rodents. It is modelled by administration of lipopolysaccharides (LPS) to induce expression of pro-inflammatory cytokines that then activate indoleamine 2,3 dioxygenase (IDO1), the rate-limiting enzyme in the kynurenine pathway of tryptophan metabolism. Here, we ask whether chronic IDO1 inhibition by 1-methyl-tryptophan (1-MT, added at 2 g/l in the drinking water) or chronic inhibition of tryptophan 2,3 dioxygenase (TDO2), another enzyme capable of converting tryptophan to kynurenine, by 680C91 (15 mg/kg per os), can rescue LPS-induced (0.83-mg/kg intraperitoneally) anxiety and cognitive deficits. We also investigate the acute effects of 680C91 on serotonergic, dopaminergic, and kynurenine pathway metabolites. METHODS We examined LPS-induced deficits in trace fear conditioning and anxiety in the light-dark box and elevated plus maze (EPM) in group-housed C57Bl6/N mice. Kynurenine pathway metabolites and monoamine levels were measured via high-performance liquid chromatography. RESULTS Chronic blockade of IDO1 with 1-MT did not rescue cognitive deficits or abrogate the anxiogenic behaviour caused by LPS despite a decrease in the brain kynurenine:tryptophan ratio. However, 1-MT by itself demonstrated anxiolytic properties in the EPM. Acute and chronic inhibition of TDO2 elevated brain levels of tryptophan, while chronic inhibition of TDO2 was unsuccessful in rescuing cognitive deficits and abrogating the anxiety caused by LPS. CONCLUSIONS In line with previous studies, we show that LPS administration induces anxiety and cognitive dysfunctions in mice that however were not reversed by chronic blockade of IDO1 or TDO2 at the doses used.
Collapse
|
15
|
Peyton L, Oliveros A, Tufvesson-Alm M, Schwieler L, Starski P, Engberg G, Erhardt S, Choi DS. Lipopolysaccharide Increases Cortical Kynurenic Acid and Deficits in Reference Memory in Mice. Int J Tryptophan Res 2019; 12:1178646919891169. [PMID: 31896932 PMCID: PMC6920585 DOI: 10.1177/1178646919891169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/06/2019] [Indexed: 01/24/2023] Open
Abstract
Kynurenic acid (KYNA), a glial-derived metabolite of tryptophan metabolism, is an
antagonist of the alpha 7 nicotinic acetylcholine receptor and the
glycine-binding site of N-methyl-d-aspartate (NMDA)
receptors. Kynurenic acid levels are increased in both the brain and
cerebrospinal fluid of several psychiatric disorders including bipolar disorder,
schizophrenia, and Alzheimer disease. In addition, pro-inflammatory cytokines
have been found to be elevated in the blood of schizophrenic patients suggesting
inflammation may play a role in psychiatric illness. As both pro-inflammatory
cytokines and KYNA can be elevated in the brain by peripheral lipopolysaccharide
(LPS) injection, we therefore sought to characterize the role of
neuroinflammation on learning and memory using a well-described dual-LPS
injection model. Mice were injected with an initial injection (0.25 mg/kg LPS,
0.50 mg/kg, or saline) of LPS and then administrated a second injection 16 hours
later. Our results indicate both 0.25 and 0.50 mg/kg dual-LPS treatment
increased l-kynurenine and KYNA levels in the medial pre-frontal cortex
(mPFC). Mice exhibited impaired acquisition of CS+ (conditioned stimulus)
Pavlovian conditioning. Notably, mice showed impairment in reference memory
while working memory was normal in an 8-arm maze. Taken together, our findings
suggest that neuroinflammation induced by peripheral LPS administration
contributes to cognitive dysfunction.
Collapse
Affiliation(s)
- Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Alfredo Oliveros
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | | | - Lilly Schwieler
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Phillip Starski
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Göran Engberg
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Sopie Erhardt
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
16
|
Giménez-Gómez P, Pérez-Hernández M, O'Shea E, Caso JR, Martín-Hernandez D, Cervera LA, Centelles MLGL, Gutiérrez-Lopez MD, Colado MI. Changes in brain kynurenine levels via gut microbiota and gut-barrier disruption induced by chronic ethanol exposure in mice. FASEB J 2019; 33:12900-12914. [PMID: 31509716 DOI: 10.1096/fj.201900491rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammatory processes have been shown to modify tryptophan (Trp) metabolism. Gut microbiota appears to play a significant role in the induction of peripheral and central inflammation. Ethanol (EtOH) exposure alters gut permeability, but its effects on Trp metabolism and the involvement of gut microbiota have not been studied. We analyzed several parameters of gut-barrier and of peripheral and central Trp metabolism following 2 different EtOH consumption patterns in mice, the binge model, drinking in the dark (DID), and the chronic intermittent (CI) consumption paradigm. Antibiotic treatment was used to evaluate gut microbiota involvement in the CI model. Mice exposed to CI EtOH intake, but not DID, show bacterial translocation and increased plasma LPS immediately after EtOH removal. Gut-barrier permeability to FITC-dextran is increased by CI, and, furthermore, intestinal epithelial tight-junction (TJ) disruption is observed (decreased expression of zonula occludens 1 and occludin) associated with increased matrix metalloproteinase (MMP)-9 activity and iNOS expression. CI EtOH, but not DID, increases kynurenine (Kyn) levels in plasma and limbic forebrain. Intestinal bacterial decontamination prevents the LPS increase but not the permeability to FITC-dextran, TJ disruption, or the increase in MMP-9 activity and iNOS expression. Although plasma Kyn levels are not affected by antibiotic treatment, the elevation of Kyn in brain is prevented, pointing to an involvement of microbiota in CI EtOH-induced changes in brain Trp metabolism. Additionally, CI EtOH produces depressive-like symptoms of anhedonia, which are prevented by the antibiotic treatment thus pointing to an association between anhedonia and the increase in brain Kyn and to the involvement of gut microbiota.-Giménez-Gómez, P., Pérez-Hernández, M., O'Shea, E., Caso, J. R., Martín-Hernández, D., Cervera, L. A., Centelles. M. L. G.-L., Gutiérrez-Lopez, M. D., Colado, M. I. Changes in brain kynurenine levels via gut microbiota and gut-barrier disruption induced by chronic ethanol exposure in mice.
Collapse
Affiliation(s)
- Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Mercedes Pérez-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Javier R Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Salud Mental (CIBERSAM), Madrid, Spain
| | - David Martín-Hernandez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Salud Mental (CIBERSAM), Madrid, Spain
| | - Luis Alou Cervera
- Área de Microbiología, Departamento de Medicina, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | | | - María Dolores Gutiérrez-Lopez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Maria Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| |
Collapse
|
17
|
Clark SM, Notarangelo FM, Li X, Chen S, Schwarcz R, Tonelli LH. Maternal immune activation in rats blunts brain cytokine and kynurenine pathway responses to a second immune challenge in early adulthood. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:286-294. [PMID: 30267854 PMCID: PMC6249106 DOI: 10.1016/j.pnpbp.2018.09.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/21/2018] [Accepted: 09/22/2018] [Indexed: 01/07/2023]
Abstract
Maternal immune activation (MIA) with the viral mimic poly I:C provides an established rodent model for studying schizophrenia (SZ) and other human neurodevelopmental disorders. Postnatal infections are additional risk factors in SZ and may cumulatively contribute to the emergence of pathophysiology. Underlying mechanisms may involve metabolites of the kynurenine pathway (KP) of tryptophan degradation, which is readily induced by inflammatory stimuli. Here we compared the expression of selected cytokines and KP enzymes, and the levels of selected KP metabolites, in the brain of MIA offspring following a second, acute immune challenge with lipopolysaccharides (LPS) on postnatal day (PND) 35 (adolescence) or PND 60 (early adulthood). Assessed in adolescence, MIA did not alter the expression of pro-inflammatory cytokines (except TNF-α) or KP metabolite levels compared to controls, but substantially reduced the expression of the anti-inflammatory cytokines IL-4 and IL-10 and influenced the expression of two of the four KP enzymes examined (IDO1 and TDO2). LPS treatment caused distinct changes in the expression of pro- and anti-inflammatory cytokines, as well as KP enzymes in MIA offspring, but had no effect on KP metabolites compared to control rats. Several of these effects were blunted in MIA offspring receiving LPS on PND 60. Notably, LPS caused a significant reduction in brain kynurenine levels in these animals. Of relevance for SZ-related hypotheses, these results indicate that MIA leads to an increasingly defective, rather than an overactive, immune regulation of cerebral KP metabolism during the postnatal period.
Collapse
Affiliation(s)
- Sarah M Clark
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Francesca M Notarangelo
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xin Li
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Leonardo H Tonelli
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
Time and frequency dependent changes in resting state EEG functional connectivity following lipopolysaccharide challenge in rats. PLoS One 2018; 13:e0206985. [PMID: 30418990 PMCID: PMC6231634 DOI: 10.1371/journal.pone.0206985] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
Research has shown that inflammatory processes affect brain function and behavior through several neuroimmune pathways. However, high order brain functions affected by inflammation largely remain to be defined. Resting state functional connectivity of synchronized oscillatory activity is a valid approach to understand network processing and high order brain function under different experimental conditions. In the present study multi-electrode EEG recording in awake, freely moving rats was used to study resting state connectivity after administration of lipopolysaccharides (LPS). Male Wistar rats were implanted with 10 cortical surface electrodes and administered with LPS (2 mg/kg) and monitored for symptoms of sickness at 3, 6 and 24 h. Resting state connectivity and power were computed at baseline, 6 and 24 h. Three prominent connectivity bands were identified using a method resistant to spurious correlation: alpha (5–15 Hz), beta-gamma (20–80 Hz), and high frequency oscillation (150–200 Hz). The most prominent connectivity band, alpha, was strongly reduced 6 h after LPS administration, and returned to baseline at 24 h. Beta-gamma connectivity was also reduced at 6 h and remained reduced at 24 h. Interestingly, high frequency oscillation connectivity remained unchanged at 6 h and was impaired 24 h after LPS challenge. Expected elevations in delta and theta power were observed at 6 h after LPS administration, when behavioral symptoms of sickness were maximal. Notably, gamma and high frequency power were reduced 6 h after LPS and returned to baseline by 24 h, when the effects on connectivity were more evident. Finally, increases in cross-frequency coupling elicited by LPS were detected at 6 h for theta-gamma and at 24 h for theta-high frequency oscillations. These studies show that LPS challenge profoundly affects EEG connectivity across all identified bands in a time-dependent manner indicating that inflammatory processes disrupt both bottom-up and top-down communication across the cortex during the peak and resolution of inflammation.
Collapse
|
19
|
Beggiato S, Notarangelo FM, Sathyasaikumar KV, Giorgini F, Schwarcz R. Maternal genotype determines kynurenic acid levels in the fetal brain: Implications for the pathophysiology of schizophrenia. J Psychopharmacol 2018; 32:1223-1232. [PMID: 30354938 DOI: 10.1177/0269881118805492] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Several studies suggest a pathophysiologically relevant association between increased brain levels of the neuroinhibitory tryptophan metabolite kynurenic acid and cognitive dysfunctions in people with schizophrenia. Elevated kynurenic acid in schizophrenia may be secondary to a genetic alteration of kynurenine 3-monooxygenase, a pivotal enzyme in the kynurenine pathway of tryptophan degradation. In rats, prenatal exposure to kynurenine, the direct bioprecursor of kynurenic acid, induces cognitive impairments reminiscent of schizophrenia in adulthood, suggesting a developmental dimension to the link between kynurenic acid and schizophrenia. AIM The purpose of this study was to explore the possible impact of the maternal genotype on kynurenine pathway metabolism. METHODS We exposed pregnant wild-type ( Kmo+/+ ) and heterozygous ( Kmo+/-) mice to kynurenine (10 mg/day) during the last week of gestation and determined the levels of kynurenic acid and two other neuroactive kynurenine pathway metabolites, 3-hydroxykynurenine and quinolinic acid, in fetal brain and placenta on embryonic day 17/18. RESULTS Maternal kynurenine treatment raised kynurenic acid levels significantly more in the brain of heterozygous offspring of Kmo+/- than in the brain of Kmo+/+ offspring. Conversely, 3-hydroxykynurenine and quinolinic acid levels in the fetal brain tended to be lower in heterozygous animals derived from kynurenine-treated Kmo+/- mice than in corresponding Kmo+/+ offspring. Genotype-related effects on the placenta were qualitatively similar but less pronounced. Kynurenine treatment also caused a preferential elevation in cerebral kynurenic acid levels in Kmo+/- compared to Kmo+/+ dams. CONCLUSIONS The disproportionate kynurenic acid increase in the brain of Kmo+/- animals indicates that the maternal Kmo genotype may play a key role in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Sarah Beggiato
- 1 Department of Life Sciences and Biotechnologies, University of Ferrara, Ferrara, Italy.,2 Laboratory for the Technology of Advanced Therapies (LTTA Centre), University of Ferrara, Ferrara, Italy
| | - Francesca M Notarangelo
- 3 Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Flaviano Giorgini
- 4 Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Robert Schwarcz
- 3 Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Clemens V, Regen F, Le Bret N, Heuser I, Hellmann-Regen J. Anti-inflammatory effects of minocycline are mediated by retinoid signaling. BMC Neurosci 2018; 19:58. [PMID: 30241502 PMCID: PMC6151010 DOI: 10.1186/s12868-018-0460-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 09/17/2018] [Indexed: 01/23/2023] Open
Abstract
Background Minocycline is a lipophilic tetracycline of increasing appeal in neuroscience as it inhibits microglial activation, a mechanism involved in numerous neuropsychiatric disorders. Own data point towards retinoid-mediated effects of minocycline in murine brain and skin, and towards a vicious cycle of neuroinflammation which is driven by microglial activation-induced breakdown of local retinoids such as retinoic acid (RA). We therefore sought to study minocycline’s anti-inflammatory effects on human microglial-like monocyte-derived cells in the context of retinoid signaling. Results As hypothesized, minocycline exposure resulted in a substantial increase of RA levels in the human monocytic cell line THP-1. While pro-inflammatory stimulation with lipopolysaccharides resulted in increased tryptophane-degrading indoleamine-2,3-dioxygenase IDO-expression and TNF-α levels in primary human monocyte-derived microglial-like cells, this effect was attenuated by minocycline only in the presence of retinoids. The anti-inflammatory effects of minocycline on TNF-α expression were completely abolished by a pharmacological blockage of retinoic acid receptors (RARs) using BMS-493 and unaffected by selectively blocking retinoid-X-receptors using UVI-3003. Conclusions Our data indicate for the first time a RA-dependent, anti-inflammatory effect for minocycline in human microglial-like cells via inhibition of local RA turnover. The RA-dependent mode of action for minocycline appears to be predominantly mediated through RAR-signaling.
Collapse
Affiliation(s)
- Vera Clemens
- Section Clinical Neurobiology, Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - University Medicine Berlin, Hindenburgdamm 30, 12203, Berlin, Germany.
| | - Francesca Regen
- Section Clinical Neurobiology, Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - University Medicine Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Nathalie Le Bret
- Section Clinical Neurobiology, Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - University Medicine Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Isabella Heuser
- Section Clinical Neurobiology, Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - University Medicine Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Julian Hellmann-Regen
- Section Clinical Neurobiology, Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - University Medicine Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| |
Collapse
|
21
|
2, 3, 5, 4'-Tetrahydroxystilbene-2-O-β-D-glucoside prevention of lipopolysaccharide-induced depressive-like behaviors in mice involves neuroinflammation and oxido-nitrosative stress inhibition. Behav Pharmacol 2018; 28:365-374. [PMID: 28410266 DOI: 10.1097/fbp.0000000000000307] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although numerous hypotheses have been raised in recent years, the exact mechanisms that promote the development of major depression are largely unknown. Recently, strategies targeting the process of neuroinflammation and oxidative stress in depression have been attracting greater attention. 2, 3, 5, 4'-Tetrahydroxystilbene-2-O-β-D-glucoside (TSG), a compound purified from a traditional Chinese herbal medicine polygonummultiflorum, has been widely reported to inhibit neuroinflammation and oxidative stress. In this context, we investigated whether TSG affects lipopolysaccharide (LPS)-induced depressive-like behaviors in a manner associated with neuroinflammation and oxido-nitrosative stress. Results showed that administration of ICR mice with 0.83 mg/kg of LPS-induced typical depressive-like behaviors in the experiments of the tail-suspension test, the forced-swimming test, and sucrose preference, and these behaviors were prevented by TSG treatment (30 and 60 mg/kg). Further analysis showed that TSG pretreatment at the doses of 30 and 60 mg/kg not only inhibited the production of proinflammatory cytokines induced by LPS, such as interleukin-1β, interleukin-6, and tumor necrosis factor-α, but also prevented the LPS-induced enhancement of oxido-nitrosative stress in mouse hippocampus and prefrontal cortex. The LPS-induced decreases in brain-derived neurotrophic factor levels in the hippocampus and prefrontal cortex were also prevented by TSG treatment. Generally, our data provide evidence to show that TSG could be used to cope with depressive-like symptoms by inhibition of neuroinflammation and oxido-nitrosative stress.
Collapse
|
22
|
Quantitative Analysis of Kynurenine Aminotransferase II in the Adult Rat Brain Reveals High Expression in Proliferative Zones and Corpus Callosum. Neuroscience 2017; 369:1-14. [PMID: 29126954 DOI: 10.1016/j.neuroscience.2017.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/06/2017] [Accepted: 11/01/2017] [Indexed: 12/27/2022]
Abstract
Kynurenic acid, a metabolite of the kynurenine pathway of tryptophan degradation, acts as an endogenous antagonist of alpha7 nicotinic and NMDA receptors and is implicated in a number of neurophysiological and neuropathological processes including cognition and neurodegenerative events. Therefore, kynurenine aminotransferase II (KAT II/AADAT), the enzyme responsible for the formation of the majority of neuroactive kynurenic acid in the brain, has prompted significant interest. Using immunohistochemistry, this enzyme was localized primarily in astrocytes throughout the adult rat brain, but detailed neuroanatomical studies are lacking. Here, we employed quantitative in situ hybridization to analyze the relative expression of KAT II mRNA in the brain of rats under normal conditions and 6 h after the administration of lipopolysaccharides (LPSs). Specific hybridization signals for KAT II were detected, with the highest expression in the subventricular zone (SVZ), the rostral migratory stream and the floor of the third ventricle followed by the corpus callosum and the hippocampus. This pattern of mRNA expression was paralleled by differential protein expression, determined by serial dilutions of antibodies (up to 1:1 million), and was confirmed to be primarily astrocytic in nature. The mRNA signal in the SVZ and the hippocampus was substantially increased by the LPS treatment without detectable changes elsewhere. These results demonstrate that KAT II is expressed in the rat brain in a region-specific manner and that gene expression is sensitive to inflammatory processes. This suggests an unrecognized role for kynurenic acid in the brain's germinal zones.
Collapse
|
23
|
Brown AG, Tulina NM, Barila GO, Hester MS, Elovitz MA. Exposure to intrauterine inflammation alters metabolomic profiles in the amniotic fluid, fetal and neonatal brain in the mouse. PLoS One 2017; 12:e0186656. [PMID: 29049352 PMCID: PMC5648237 DOI: 10.1371/journal.pone.0186656] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/04/2017] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Exposure to prenatal inflammation is associated with diverse adverse neurobehavioral outcomes in exposed offspring. The mechanism by which inflammation negatively impacts the developing brain is poorly understood. Metabolomic profiling provides an opportunity to identify specific metabolites, and novel pathways, which may reveal mechanisms by which exposure to intrauterine inflammation promotes fetal and neonatal brain injury. Therefore, we investigated whether exposure to intrauterine inflammation altered the metabolome of the amniotic fluid, fetal and neonatal brain. Additionally, we explored whether changes in the metabolomic profile from exposure to prenatal inflammation occurs in a sex-specific manner in the neonatal brain. METHODS CD-1, timed pregnant mice received an intrauterine injection of lipopolysaccharide (50 μg/dam) or saline on embryonic day 15. Six and 48 hours later mice were sacrificed and amniotic fluid, and fetal brains were collected (n = 8/group). Postnatal brains were collected on day of life 1 (n = 6/group/sex). Global biochemical profiles were determined using ultra performance liquid chromatography/tandem mass spectrometry (Metabolon Inc.). Statistical analyses were performed by comparing samples from lipopolysaccharide and saline treated animals at each time point. For the P1 brains, analyses were stratified by sex. RESULTS/CONCLUSIONS Exposure to intrauterine inflammation induced unique, temporally regulated changes in the metabolic profiles of amniotic fluid, fetal brain and postnatal brain. Six hours after exposure to intrauterine inflammation, the amniotic fluid and the fetal brain metabolomes were dramatically altered with significant enhancements of amino acid and purine metabolites. The amniotic fluid had enhanced levels of several members of the (hypo) xanthine pathway and this compound was validated as a potential biomarker. By 48 hours, the number of altered biochemicals in both the fetal brain and the amniotic fluid had declined, yet unique profiles existed. Neonatal pups exposed to intrauterine inflammation have significant alterations in their lipid metabolites, in particular, fatty acids. These sex-specific metabolic changes within the newborn brain offer an explanation regarding the sexual dimorphism of certain psychiatric and neurobehavioral disorders associated with exposure to prenatal inflammation.
Collapse
Affiliation(s)
- Amy G. Brown
- Maternal Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - Natalia M. Tulina
- Maternal Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Guillermo O. Barila
- Maternal Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael S. Hester
- Maternal Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michal A. Elovitz
- Maternal Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
24
|
Oliveros A, Wininger K, Sens J, Larsson MK, Liu XC, Choi S, Faka A, Schwieler L, Engberg G, Erhardt S, Choi DS. LPS-induced cortical kynurenic acid and neurogranin-NFAT signaling is associated with deficits in stimulus processing during Pavlovian conditioning. J Neuroimmunol 2017; 313:1-9. [PMID: 29153599 DOI: 10.1016/j.jneuroim.2017.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 05/04/2017] [Accepted: 09/27/2017] [Indexed: 12/13/2022]
Abstract
The N-Methyl-d-Aspartate receptor (NMDAR) antagonist kynurenic acid (KYNA) and the post-synaptic calmodulin binding protein neurogranin (Nrgn) have been implicated in neurological and neuropsychiatric conditions including Alzheimer's disease and schizophrenia. This study indicates that systemic dual-lipopolysaccharide (LPS) injections increases KYNA in the medial prefrontal cortex (mPFC), which is accompanied with increased phosphorylation of nuclear factor kappa chain of activated B cells (NFκB) and activation of the nuclear factor of activated T- cells (NFAT). Our results also indicate that dual-LPS increases Nrgn phosphorylation and concomitantly reduces phosphorylation of calmodulin kinase-II (CaMKII). We confirmed that systemic blockade of kynurenine-3 monooxygenase in conjunction with kynurenine administration results in significant increases in Nrgn phosphorylation and a significant reduction of CaMKII phosphorylation in the mPFC. Consequently, dual-LPS administration induced significant impairments in stimulus processing during Pavlovian conditioning. Taken together, our study indicates that elevations in KYNA in the mPFC can directly regulate NMDA-Nrgn-CaMKII signaling, suggesting that neuroinflammatory conditions affecting this pathway may be associated with cognitive dysfunction.
Collapse
Affiliation(s)
- A Oliveros
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - K Wininger
- Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - J Sens
- Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - M K Larsson
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - X C Liu
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - S Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - A Faka
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - L Schwieler
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - G Engberg
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - S Erhardt
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - D S Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|