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 PMCID: PMC11549936 DOI: 10.1124/pharmrev.124.000239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
Collins JM, Keane JM, Deady C, Khashan AS, McCarthy FP, O'Keeffe GW, Clarke G, Cryan JF, Caputi V, O'Mahony SM. Prenatal stress impacts foetal neurodevelopment: Temporal windows of gestational vulnerability. Neurosci Biobehav Rev 2024; 164:105793. [PMID: 38971516 DOI: 10.1016/j.neubiorev.2024.105793] [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: 04/09/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Prenatal maternal stressors ranging in severity from everyday occurrences/hassles to the experience of traumatic events negatively impact neurodevelopment, increasing the risk for the onset of psychopathology in the offspring. Notably, the timing of prenatal stress exposure plays a critical role in determining the nature and severity of subsequent neurodevelopmental outcomes. In this review, we evaluate the empirical evidence regarding temporal windows of heightened vulnerability to prenatal stress with respect to motor, cognitive, language, and behavioural development in both human and animal studies. We also explore potential temporal windows whereby several mechanisms may mediate prenatal stress-induced neurodevelopmental effects, namely, excessive hypothalamic-pituitary-adrenal axis activity, altered serotonin signalling and sympathetic-adrenal-medullary system, changes in placental function, immune system dysregulation, and alterations of the gut microbiota. While broadly defined developmental windows are apparent for specific psychopathological outcomes, inconsistencies arise when more complex cognitive and behavioural outcomes are considered. Novel approaches to track molecular markers reflective of the underlying aetiologies throughout gestation to identify tractable biomolecular signatures corresponding to critical vulnerability periods are urgently required.
Collapse
Affiliation(s)
- James M Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - James M Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Fergus P McCarthy
- The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Obstetrics and Gynaecology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Valentina Caputi
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | | |
Collapse
|
3
|
Alves LDF, Moore JB, Kell DB. The Biology and Biochemistry of Kynurenic Acid, a Potential Nutraceutical with Multiple Biological Effects. Int J Mol Sci 2024; 25:9082. [PMID: 39201768 PMCID: PMC11354673 DOI: 10.3390/ijms25169082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Kynurenic acid (KYNA) is an antioxidant degradation product of tryptophan that has been shown to have a variety of cytoprotective, neuroprotective and neuronal signalling properties. However, mammalian transporters and receptors display micromolar binding constants; these are consistent with its typically micromolar tissue concentrations but far above its serum/plasma concentration (normally tens of nanomolar), suggesting large gaps in our knowledge of its transport and mechanisms of action, in that the main influx transporters characterized to date are equilibrative, not concentrative. In addition, it is a substrate of a known anion efflux pump (ABCC4), whose in vivo activity is largely unknown. Exogeneous addition of L-tryptophan or L-kynurenine leads to the production of KYNA but also to that of many other co-metabolites (including some such as 3-hydroxy-L-kynurenine and quinolinic acid that may be toxic). With the exception of chestnut honey, KYNA exists at relatively low levels in natural foodstuffs. However, its bioavailability is reasonable, and as the terminal element of an irreversible reaction of most tryptophan degradation pathways, it might be added exogenously without disturbing upstream metabolism significantly. Many examples, which we review, show that it has valuable bioactivity. Given the above, we review its potential utility as a nutraceutical, finding it significantly worthy of further study and development.
Collapse
Affiliation(s)
- Luana de Fátima Alves
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - J. Bernadette Moore
- School of Food Science & Nutrition, University of Leeds, Leeds LS2 9JT, UK;
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| | - Douglas B. Kell
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| |
Collapse
|
4
|
Pedraz-Petrozzi B, Lamadé EK, Marszalek-Grabska M, Trzpil A, Lindner O, Meininger P, Fornal E, Turski WA, Witt SH, Gilles M, Deuschle M. Fetal Sex as Moderating Factor for the Relationship Between Maternal Childhood Trauma and Salivary Kynurenic Acid and Tryptophan in Pregnancy: A Pilot Study. Int J Tryptophan Res 2024; 17:11786469241244603. [PMID: 38660592 PMCID: PMC11041113 DOI: 10.1177/11786469241244603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/17/2024] [Indexed: 04/26/2024] Open
Abstract
Traumatic experiences and fetal development influence tryptophan (TRP) and its neuroactive byproduct, kynurenic acid (KYNA). Maternal TRP metabolite levels during pregnancy vary by fetal sex, with higher concentrations in mothers carrying male fetuses. This pilot study aimed to explore the relationship between offspring sex, maternal childhood trauma, and maternal salivary KYNA and TRP levels during pregnancy. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to determine KYNA and TRP levels in maternal saliva samples collected from 35 late-pregnancy participants. Maternal childhood trauma was assessed using the Childhood Trauma Questionnaire, including subscales for emotional abuse, physical abuse, sexual abuse, emotional neglect, and physical neglect. Among mothers pregnant with boys, salivary KYNA significantly correlated with physical and emotional neglect, and salivary TRP with emotional neglect. No significant correlations were found in mothers who delivered female offspring. Significant associations of childhood trauma and offspring sex were found for salivary KYNA but not TRP concentrations. Mothers with higher trauma levels who delivered boys exhibited higher levels of salivary KYNA compared to those with lower trauma levels. Moreover, mothers with higher trauma levels who delivered boys had higher salivary KYNA levels than those with higher trauma levels who delivered girls. This pilot study provides evidence of an association between maternal childhood trauma and TRP metabolism, measured in saliva, especially in mothers pregnant with boys. However, longitudinal studies with larger sample sizes are required to confirm these results.
Collapse
Affiliation(s)
- Bruno Pedraz-Petrozzi
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Eva Kathrin Lamadé
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| | | | - Alicja Trzpil
- Department of Bioanalytics, Medical University of Lublin, Poland
| | - Ole Lindner
- Center for Child and Adolescent Health, Pediatrics, University Hospital of Freiburg, Germany
| | - Pascal Meininger
- Department of Gynecology and Obstetrics, Westpfalz-Klinikum, Kaiserslautern, Germany
| | - Emilia Fornal
- Department of Bioanalytics, Medical University of Lublin, Poland
| | - Waldemar A Turski
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Poland
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Maria Gilles
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Michael Deuschle
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| |
Collapse
|
5
|
Pedraz-Petrozzi B, Marszalek-Grabska M, Kozub A, Szalaj K, Trzpil A, Stachniuk A, Lamadé EK, Gilles M, Deuschle M, Turski WA, Fornal E. LC-MS/MS-based quantification of tryptophan, kynurenine, and kynurenic acid in human placental, fetal membranes, and umbilical cord samples. Sci Rep 2023; 13:12554. [PMID: 37532780 PMCID: PMC10397233 DOI: 10.1038/s41598-023-39774-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023] Open
Abstract
Tryptophan breakdown metabolites formed along the kynurenine pathway play a significant role in pregnancy and fetal development. To understand their involvement, it is crucial to quantify the levels of tryptophan (TRP), kynurenine (KYN), and kynurenic acid (KYNA) in relevant biological samples such as the placenta, fetal membranes, and umbilical cord. This study used liquid chromatography-tandem mass spectrometry (LC-MS/MS) to determine TRP, KYN, and KYNA levels. The LC-MS/MS method was optimized for high sensitivity and specificity, demonstrating good reproducibility with a precision of < 10% CV and an accuracy of 85-115%. The lower limit of quantification for both TRP and KYN was 0.5 µg/ml, while for KYNA, it was 0.5 ng/mL. The method exhibited linearity within the examined range of concentrations in the homogenate, ranging from 0.5 to 30 µg/ml for TRP and KYN and from 0.5 to 25 ng/ml for KYNA. Using this method, we found significant differences in the concentrations of these substances in investigated maternal-fetal compartments. Placenta samples exhibited higher KYN and lower KYNA concentrations than the umbilical cord and fetal membrane, indicating a potentially important role for kynurenines in late pregnancy. Collectively, this finding may facilitate further research and provide inside into the involvement of the kynurenine pathway of TRP metabolism in fetal development.
Collapse
Affiliation(s)
- Bruno Pedraz-Petrozzi
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany.
| | - Marta Marszalek-Grabska
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Anna Kozub
- Department of Bioanalytics, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Klaudia Szalaj
- Department of Bioanalytics, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Alicja Trzpil
- Department of Bioanalytics, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Anna Stachniuk
- Department of Bioanalytics, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Eva Kathrin Lamadé
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Maria Gilles
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Michael Deuschle
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Waldemar A Turski
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Emilia Fornal
- Department of Bioanalytics, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| |
Collapse
|
6
|
Wright CJ, Milosavljevic S, Pocivavsek A. The stress of losing sleep: Sex-specific neurobiological outcomes. Neurobiol Stress 2023; 24:100543. [PMID: 37252645 PMCID: PMC10209346 DOI: 10.1016/j.ynstr.2023.100543] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Sleep is a vital and evolutionarily conserved process, critical to daily functioning and homeostatic balance. Losing sleep is inherently stressful and leads to numerous detrimental physiological outcomes. Despite sleep disturbances affecting everyone, women and female rodents are often excluded or underrepresented in clinical and pre-clinical studies. Advancing our understanding of the role of biological sex in the responses to sleep loss stands to greatly improve our ability to understand and treat health consequences of insufficient sleep. As such, this review discusses sex differences in response to sleep deprivation, with a focus on the sympathetic nervous system stress response and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We review sex differences in several stress-related consequences of sleep loss, including inflammation, learning and memory deficits, and mood related changes. Focusing on women's health, we discuss the effects of sleep deprivation during the peripartum period. In closing, we present neurobiological mechanisms, including the contribution of sex hormones, orexins, circadian timing systems, and astrocytic neuromodulation, that may underlie potential sex differences in sleep deprivation responses.
Collapse
Affiliation(s)
- Courtney J. Wright
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
7
|
Levin G, Ein-Dor T. A unified model of the biology of peripartum depression. Transl Psychiatry 2023; 13:138. [PMID: 37117197 PMCID: PMC10147643 DOI: 10.1038/s41398-023-02439-w] [Citation(s) in RCA: 12] [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: 10/14/2022] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023] Open
Abstract
Peripartum depression (PPD) is a prevalent and debilitating disorder that adversely affects the development of mothers and infants. Recently, there has been a plea for increased mental health screening during the peripartum period; however, currently, there is no accurate screening tool to identify women at risk of PPD. In addition, some women do not respond to current treatment schemes and develop treatment-resistant depression. The current perspective aims to propose a unified understanding of the biological underpinnings of PPD (UmPPD) that considers the heterogeneity in the onset, symptoms cluster, and severity of PPD. Such a model could promote basic and applied research on PPD and suggest new treatment avenues. The central hub of the model is the kynurenine pathway (KP) and the KP-serotonin ratio. The forces and specific processes at play that cause an imbalance within the KP and between KP and serotonin are inflammation, stress, reproductive hormones (especially estradiol and progesterone), and oxytocin. UmPPD predicts that the most severe PPD would comprise prolonged inflammation, ongoing or multiple stressors, excessive estrogen, progesterone resistance, and avoidance of breastfeeding, skin-to-skin contact, and social proximity. These factors would be associated with a higher likelihood of developing PPD, early onset, and more significant symptom severity. In addition, subtypes of PPD would consist of different compositions and expressions of these components, with one central common factor. UmPPD could aid in directing future research and possibly detecting critical processes that could help discover, develop, and utilize novel treatments for PPD.
Collapse
Affiliation(s)
- Gal Levin
- Reichman University. Please address all correspondence to: Tsachi Ein-Dor, Baruch Ivcher School of Psychology, Reichman University, University St. 8, Herzliya, 4610101, Israel
| | - Tsachi Ein-Dor
- Reichman University. Please address all correspondence to: Tsachi Ein-Dor, Baruch Ivcher School of Psychology, Reichman University, University St. 8, Herzliya, 4610101, Israel.
| |
Collapse
|
8
|
Murakami Y, Imamura Y, Kasahara Y, Yoshida C, Momono Y, Fang K, Sakai D, Konishi Y, Nishiyama T. Maternal Inflammation with Elevated Kynurenine Metabolites Is Related to the Risk of Abnormal Brain Development and Behavioral Changes in Autism Spectrum Disorder. Cells 2023; 12:1087. [PMID: 37048160 PMCID: PMC10093447 DOI: 10.3390/cells12071087] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
Several studies show that genetic and environmental factors contribute to the onset and progression of neurodevelopmental disorders. Maternal immune activation (MIA) during gestation is considered one of the major environmental factors driving this process. The kynurenine pathway (KP) is a major route of the essential amino acid L-tryptophan (Trp) catabolism in mammalian cells. Activation of the KP following neuro-inflammation can generate various endogenous neuroactive metabolites that may impact brain functions and behaviors. Additionally, neurotoxic metabolites and excitotoxicity cause long-term changes in the trophic support, glutamatergic system, and synaptic function following KP activation. Therefore, investigating the role of KP metabolites during neurodevelopment will likely promote further understanding of additional pathophysiology of neurodevelopmental disorders, including autism spectrum disorder (ASD). In this review, we describe the changes in KP metabolism in the brain during pregnancy and represent how maternal inflammation and genetic factors influence the KP during development. We overview the patients with ASD clinical data and animal models designed to verify the role of perinatal KP elevation in long-lasting biochemical, neuropathological, and behavioral deficits later in life. Our review will help shed light on new therapeutic strategies and interventions targeting the KP for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yuki Murakami
- Department of Hygiene and Public Health, Kansai Medical University, Hirakata 573-1010, Japan
| | - Yukio Imamura
- Department of Architecture and Architectual Systems Engineering, Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan
- Department of Traumatology and Acute Critical Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Suita 565-0871, Japan
| | - Yoshiyuki Kasahara
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Chihiro Yoshida
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yuta Momono
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Ke Fang
- Department of Hygiene and Public Health, Kansai Medical University, Hirakata 573-1010, Japan
| | - Daisuke Sakai
- Department of Biology, Kanazawa Medical University, Kanazawa 920-0293, Japan
| | - Yukuo Konishi
- Center for Baby Science, Doshisha University, Kyotanabe 619-0225, Japan
- Healthcare and Medical Data Multi-Level Integration Platform Group, RIKEN Medical Sciences Innovation Hub Program, Yokohama 230-0045, Japan
| | - Toshimasa Nishiyama
- Department of Hygiene and Public Health, Kansai Medical University, Hirakata 573-1010, Japan
| |
Collapse
|
9
|
Perrone S, Grassi F, Caporilli C, Boscarino G, Carbone G, Petrolini C, Gambini LM, Di Peri A, Moretti S, Buonocore G, Esposito SMR. Brain Damage in Preterm and Full-Term Neonates: Serum Biomarkers for the Early Diagnosis and Intervention. Antioxidants (Basel) 2023; 12:antiox12020309. [PMID: 36829868 PMCID: PMC9952571 DOI: 10.3390/antiox12020309] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/14/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
The Brain is vulnerable to numerous insults that can act in the pre-, peri-, and post-natal period. There is growing evidence that demonstrate how oxidative stress (OS) could represent the final common pathway of all these insults. Fetuses and newborns are particularly vulnerable to OS due to their inability to active the antioxidant defenses. Specific molecules involved in OS could be measured in biologic fluids as early biomarkers of neonatal brain injury with an essential role in neuroprotection. Although S-100B seems to be the most studied biomarker, its use in clinical practice is limited by the complexity of brain damage etiopathogenesis and the time of blood sampling in relation to the brain injury. Reliable early specific serum markers are currently lacking in clinical practice. It is essential to determine if there are specific biomarkers that can help caregivers to monitor the progression of the disease in order to active an early neuroprotective strategy. We aimed to describe, in an educational review, the actual evidence on serum biomarkers for the early identification of newborns at a high risk of neurological diseases. To move the biomarkers from the bench to the bedside, the assays must be not only be of a high sensitivity but suitable for the very rapid processing and return of the results for the clinical practice to act on. For the best prognosis, more studies should focus on the association of these biomarkers to the type and severity of perinatal brain damage.
Collapse
Affiliation(s)
- Serafina Perrone
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Correspondence:
| | - Federica Grassi
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Caporilli
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Giovanni Boscarino
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Giulia Carbone
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Petrolini
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Lucia Maria Gambini
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Antonio Di Peri
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Sabrina Moretti
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | | |
Collapse
|
10
|
Perspective: Gestational Tryptophan Fluctuation Altering Neuroembryogenesis and Psychosocial Development. Cells 2022; 11:cells11081270. [PMID: 35455949 PMCID: PMC9032700 DOI: 10.3390/cells11081270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/10/2022] Open
Abstract
Tryptophan, as the sole precursor of serotonin, mainly derived from diets, is essential for neurodevelopment and immunomodulation. Gestational tryptophan fluctuation may account for the maternal-fetal transmission in determining neuroembryogenesis with long-lasting effects on psychological development. Personality disorders and social exclusion are related to psychosocial problems, leading to impaired social functioning. However, it is not clear how the fluctuation in mother-child transmission regulates the neuroendocrine development and gut microbiota composition in progeny due to that tryptophan metabolism in pregnant women is affected by multiple factors, such as diets (tryptophan-enriched or -depleted diet), emotional mental states (anxiety, depression), health status (hypertension, diabetes), and social support as well as stresses and management skills. Recently, we have developed a non-mammal model to rationalize those discrepancies without maternal effects. This perspective article outlines the possibility and verified the hypothesis in bully-victim research with this novel model: (1). Summarizes the effects of the maternal tryptophan administration on the neuroendocrine and microbial development in their offspring; (2). Highlights the inconsistency and limitations in studying the relationship between gestational tryptophan exposure and psychosocial development in humans and viviparous animals; and (3). Evidences that embryonic exposure to tryptophan and its metabolite modify bullying interactions in the chicken model. With the current pioneer researches on the biomechanisms underlying the bully-victim interaction, the perspective article provides novel insights for developing appropriate intervention strategies to prevent psychological disorders among individuals, especially those who experienced prenatal stress, by controlling dietary tryptophan and medication therapy during pregnancy.
Collapse
|
11
|
Kwon HK, Choi GB, Huh JR. Maternal inflammation and its ramifications on fetal neurodevelopment. Trends Immunol 2022; 43:230-244. [PMID: 35131181 PMCID: PMC9005201 DOI: 10.1016/j.it.2022.01.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
Abstract
Exposure to heightened inflammation in pregnancy caused by infections or other inflammatory insults has been associated with the onset of neurodevelopmental and psychiatric disorders in children. Rodent models have provided unique insights into how this maternal immune activation (MIA) disrupts brain development. Here, we discuss the key immune factors involved, highlight recent advances in determining the molecular and cellular pathways of MIA, and review how the maternal immune system affects fetal development. We also examine the roles of microbiomes in shaping maternal immune function and the development of autism-like phenotypes. A comprehensive understanding of the gut bacteria-immune-neuro interaction in MIA is essential for developing diagnostic and therapeutic measures for high-risk pregnant women and identifying targets for treating inflammation-induced neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ho-Keun Kwon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea; Pohang University of Science and Technology, Pohang, Korea.
| | - Gloria B. Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jun R. Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA.,Correspondence: Ho-Keun Kwon () and Jun R. Huh ()
| |
Collapse
|
12
|
FOXO1 Is a Critical Switch Molecule for Autophagy and Apoptosis of Sow Endometrial Epithelial Cells Caused by Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:1172273. [PMID: 34970413 PMCID: PMC8714345 DOI: 10.1155/2021/1172273] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/17/2021] [Accepted: 11/02/2021] [Indexed: 02/07/2023]
Abstract
Oxidative stress (OS) is involved in various reproductive diseases and can induce autophagy and apoptosis, which determine the different fates of cells. However, the sequence and the switch mechanism between autophagy and apoptosis are unclear. Here, we reported that chronic restraint stress (CRS) induced OS (decreased T-AOC, T-SOD, CAT and GSH-Px and increased MDA) and then disturbed the endocrine environment of sows during early pregnancy, including the hypothalamic-pituitary-ovarian (HPO) and the hypothalamic-pituitary-adrenal (HPA) axes. Meanwhile, after CRS, the KEAP1/NRF2 pathway was inhibited and attenuated the antioxidative ability to cause OS of the endometrium. The norepinephrine (NE) triggered β2-AR to activate the FOXO1/NF-κB pathway, which induced endometrial inflammation. CRS induced the caspase-dependent apoptosis pathway and caused MAP1LC3-II accumulation, SQSTM1/p62 degradation, and autophagosome formation to initiate autophagy. Furthermore, in vitro, a cellular OS model was established by adding hydrogen peroxide into cells. Low OS maintained the viability of endometrial epithelial cells by triggering autophagy, while high OS induced cell death by initiating caspase-dependent apoptosis. Autophagy preceded the occurrence of apoptosis, which depended on the subcellular localization of FOXO1. In the low OS group, FOXO1 was exported from the nucleus to be modified into Ac-FOXO1 and bound to ATG7 in the cytoplasm, which promoted autophagy to protect cells. In the high OS group, FOXO1 located in the nucleus to promote transcription of proapoptotic proteins and then induce apoptosis. Here, FOXO1, as a redox sensor switch, regulated the transformation of cell autophagy and apoptosis. In summary, the posttranslational modification of FOXO1 may become the target of OS treatment.
Collapse
|
13
|
Butler MI, Long-Smith C, Moloney GM, Morkl S, O'Mahony SM, Cryan JF, Clarke G, Dinan TG. The immune-kynurenine pathway in social anxiety disorder. Brain Behav Immun 2022; 99:317-326. [PMID: 34758380 DOI: 10.1016/j.bbi.2021.10.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/18/2021] [Accepted: 10/31/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The tryptophan-kynurenine pathway is of major interest in psychiatry and is altered in patients with depression, schizophrenia and panic disorder. Stress and immune alterations can impact this system, through cortisol- and cytokine-induced activation. In addition, there is emerging evidence that the kynurenine pathway is associated with suicidality. There have been no studies to date exploring the immune-kynurenine system in social anxiety disorder (SAD), and indeed very limited human studies on the kynurenine pathway in any clinical anxiety disorder. METHODS We investigated plasma levels of several kynurenine pathway markers, including kynurenine (KYN), tryptophan (TRYP) and kynurenic acid (KYNA), along with the KYN/TRYP and KYNA/KYN ratios, in a cohort of 32 patients with SAD and 36 healthy controls. We also investigated a broad array of both basal and lipopolysaccharide (LPS)-stimulated blood cytokine levels including IFN-γ, interleukin (IL)-10, IL-1β, IL-2, IL-4, IL-6, IL-8 and tumor necrosis factor (TNF)-α. RESULTS SAD patients had elevated plasma KYNA levels and an increased KYNA/KYN ratio compared to healthy controls. No differences in KYN, TRYP or the KYN/TRYP ratio were seen between the two groups. SAD patients with a history of past suicide attempt showed elevated plasma KYN levels and a higher KYN/TRYP ratio compared to patients without a history of suicide attempt. No differences were seen in basal or LPS-stimulated pro-inflammatory cytokine levels between the patients and controls. However, unstimulated IL-10, an anti-inflammatory cytokine, was significantly lower in the SAD group. A significant sex influence was evident with SAD males having lower levels of IL-10 compared to healthy males but no difference seen between SAD females and healthy females. CONCLUSIONS The peripheral kynurenine pathway is altered in SAD and preferentially directed towards KYNA synthesis. Additionally, kynurenine pathway activation, as evidenced by elevated KYN and KYN/TRYP ratio, is evident in SAD patients with a history of past suicide attempt. While no differences in pro-inflammatory cytokines is apparent in SAD patients, lower anti-inflammatory IL-10 levels are seen in SAD males. Further investigation of the role of the immune-kynurenine pathway in SAD and other clinical anxiety disorders is warranted.
Collapse
Affiliation(s)
- Mary I Butler
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| | | | - Gerard M Moloney
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Sabrina Morkl
- APC Microbiome Ireland, University College Cork, Ireland; Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Austria
| | - Siobhain M O'Mahony
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| |
Collapse
|
14
|
Murthy GG, Prideaux MA, Armstrong M, Kenney HM, Latchney SE, Susiarjo M, Murphy SP. Characterization of the temporal, cell-specific and interferon-inducible patterns of indoleamine 2,3 dioxygenase 1 (IDO1) expression in the human placenta across gestation. Placenta 2021; 115:129-138. [PMID: 34619429 DOI: 10.1016/j.placenta.2021.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/09/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION The human placenta performs multiple functions necessary for successful pregnancy, but the metabolic pathways and molecular mechanisms responsible for regulating placental development and functions remain incompletely understood. Catabolism of the essential amino acid tryptophan has numerous critical roles in normal physiology, including inflammation. The kynurenine pathway, which accounts for ∼90% of tryptophan breakdown, is mediated by indoleamine 2,3 dioxygenase 1 (IDO1) in the placenta. In pregnant mice, alterations of IDO1 activity or expression result in fetal resorption and a preeclampsia-like phenotype. Decreased IDO1 expression at the maternal-fetal interface has also been linked to preeclampsia, in utero growth restriction and recurrent miscarriage in humans. These collective observations suggest essential role(s) for IDO1 in maintaining healthy pregnancy. Despite these important roles, the precise temporal, cell-specific and inflammatory cytokine-mediated patterns of IDO1 expression in the human placenta have not been thoroughly characterized across gestation. METHODS Western blot and whole mount immunofluorescence (WMIF) were utilized to characterize and quantify basal and interferon (IFN)-inducible IDO1 expression in 1st trimester (7-13 weeks), 2nd trimester (14-22 weeks) and term (39-41 weeks) placental villi. RESULTS IDO1 expression is activated in the human placenta between the 13th and 14th weeks of pregnancy, increases through the 2nd trimester and remains elevated at term. Constitutive IDO1 expression is restricted to placental endothelial cells. Interestingly, different types of IFNs have distinct effects on IDO1 expression in the human placenta. DISCUSSION Our collective results are consistent with potential role(s) for IDO1 in the regulation of vascular functions in placental villi.
Collapse
Affiliation(s)
- Gayathri Guru Murthy
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Mallory A Prideaux
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Madison Armstrong
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - H Mark Kenney
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Sarah E Latchney
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Martha Susiarjo
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Shawn P Murphy
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
15
|
Rentschler KM, Baratta AM, Ditty AL, Wagner NTJ, Wright CJ, Milosavljevic S, Mong JA, Pocivavsek A. Prenatal Kynurenine Elevation Elicits Sex-Dependent Changes in Sleep and Arousal During Adulthood: Implications for Psychotic Disorders. Schizophr Bull 2021; 47:1320-1330. [PMID: 33823027 PMCID: PMC8379538 DOI: 10.1093/schbul/sbab029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Dysregulation of the kynurenine pathway (KP) of tryptophan catabolism has been implicated in psychotic disorders, including schizophrenia and bipolar disorder. Kynurenic acid (KYNA) is a KP metabolite synthesized by kynurenine aminotransferases (KATs) from its biological precursor kynurenine and acts as an endogenous antagonist of N-methyl-D-aspartate and α7-nicotinic acetylcholine receptors. Elevated KYNA levels found in postmortem brain tissue and cerebrospinal fluid of patients are hypothesized to play a key role in the etiology of cognitive symptoms observed in psychotic disorders. Sleep plays an important role in memory consolidation, and sleep disturbances are common among patients. Yet, little is known about the effect of altered KP metabolism on sleep-wake behavior. We presently utilized a well-established experimental paradigm of embryonic kynurenine (EKyn) exposure wherein pregnant dams are fed a diet laced with kynurenine the last week of gestation and hypothesized disrupted sleep-wake behavior in adult offspring. We examined sleep behavior in adult male and female offspring using electroencephalogram and electromyogram telemetry and determined sex differences in sleep and arousal in EKyn offspring. EKyn males displayed reduced rapid eye movement sleep, while female EKyn offspring were hyperaroused compared to controls. We determined that EKyn males maintain elevated brain KYNA levels, while KYNA levels were unchanged in EKyn females, yet the activity levels of KAT I and KAT II were reduced. Our findings indicate that elevated prenatal kynurenine exposure elicits sex-specific changes in sleep-wake behavior, arousal, and KP metabolism.
Collapse
Affiliation(s)
- Katherine M Rentschler
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | | | - Audrey L Ditty
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Nathan T J Wagner
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Courtney J Wright
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Jessica A Mong
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
16
|
Kelly JR, Minuto C, Cryan JF, Clarke G, Dinan TG. The role of the gut microbiome in the development of schizophrenia. Schizophr Res 2021; 234:4-23. [PMID: 32336581 DOI: 10.1016/j.schres.2020.02.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a heterogeneous neurodevelopmental disorder involving the convergence of a complex and dynamic bidirectional interaction of genetic expression and the accumulation of prenatal and postnatal environmental risk factors. The development of the neural circuitry underlying social, cognitive and emotional domains requires precise regulation from molecular signalling pathways, especially during critical periods or "windows", when the brain is particularly sensitive to the influence of environmental input signalling. Many of the brain regions involved, and the molecular substrates sub-serving these domains are responsive to life-long microbiota-gut-brain (MGB) axis signalling. This intricate microbial signalling system communicates with the brain via the vagus nerve, immune system, enteric nervous system, enteroendocrine signalling and production of microbial metabolites, such as short-chain fatty acids. Preclinical data has demonstrated that MGB axis signalling influences neurotransmission, neurogenesis, myelination, dendrite formation and blood brain barrier development, and modulates cognitive function and behaviour patterns, such as, social interaction, stress management and locomotor activity. Furthermore, preliminary clinical studies suggest altered gut microbiota profiles in schizophrenia. Unravelling MGB axis signalling in the context of an evolving dimensional framework in schizophrenia may provide a more complete understanding of the neurobiological architecture of this complex condition and offers the possibility of translational interventions.
Collapse
Affiliation(s)
- John R Kelly
- Department of Psychiatry, Trinity College Dublin, Ireland
| | - Chiara Minuto
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
17
|
Prenatal stress-induced disruptions in microbial and host tryptophan metabolism and transport. Behav Brain Res 2021; 414:113471. [PMID: 34280459 DOI: 10.1016/j.bbr.2021.113471] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/22/2021] [Accepted: 07/14/2021] [Indexed: 12/19/2022]
Abstract
The aromatic amino acid tryptophan (Trp) is a precursor for multiple metabolites that can steer proper immune and neurodevelopment as well as social behavior in later life. Dysregulation in the Trp metabolic pathways and abundance of Trp or its derivatives, including indoles, kynurenine (Kyn), and particularly serotonin, has been associated with behavioral deficits and neuropsychiatric disorders including autism spectrum disorders (ASD) and schizophrenia. Previously, we have shown that prenatal stress (PNS) alters placental Trp and serotonin, and reduces Trp-metabolizing members of the maternal colonic microbiota. Given that PNS also results in alterations in offspring neurodevelopment, behavior and immune function, we hypothesized that PNS affects Trp metabolism and transport in both the maternal and fetal compartments, and that these alterations continue into adolescence. We surmised that this is due to reductions in Trp-metabolizing microbes that would otherwise reduce the Trp pool under normal metabolic conditions. To test this, pregnant mice were exposed to a restraint stressor and gene expression of enzymes involved in Trp and serotonin metabolism were measured. Specifically, tryptophan 2,3-dioxygenase, aryl hydrocarbon receptor, and solute carrier proteins, were altered due to PNS both prenatally and postnatally. Additionally, Parasutterella and Bifidobacterium, which metabolize Trp in the gut, were reduced in both the dam and the offspring. Together, the reductions of Trp-associated microbes and concomitant dysregulation in Trp metabolic machinery in dam and offspring suggest that PNS-induced Trp metabolic dysfunction may mediate aberrant fetal neurodevelopment.
Collapse
|
18
|
Karahoda R, Robles M, Marushka J, Stranik J, Abad C, Horackova H, Tebbens JD, Vaillancourt C, Kacerovsky M, Staud F. Prenatal inflammation as a link between placental expression signature of tryptophan metabolism and preterm birth. Hum Mol Genet 2021; 30:2053-2067. [PMID: 34169316 PMCID: PMC8561419 DOI: 10.1093/hmg/ddab169] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/06/2023] Open
Abstract
Spontaneous preterm birth is a serious medical condition responsible for substantial perinatal morbidity and mortality. Its phenotypic characteristics, preterm labor with intact membranes (PTL) and preterm premature rupture of the membranes (PPROM), are associated with significantly increased risks of neurological and behavioral alterations in childhood and later life. Recognizing the inflammatory milieu associated with PTL and PPROM, here, we examined expression signatures of placental tryptophan metabolism, an important pathway in prenatal brain development and immunotolerance. The study was performed in a well-characterized clinical cohort of healthy term pregnancies (n = 39) and 167 preterm deliveries (PTL, n = 38 and PPROM, n = 129). Within the preterm group, we then investigated potential mechanistic links between differential placental tryptophan pathway expression, preterm birth and both intra-amniotic markers (such as amniotic fluid interleukin-6) and maternal inflammatory markers (such as maternal serum C-reactive protein and white blood cell count). We show that preterm birth is associated with significant changes in placental tryptophan metabolism. Multifactorial analysis revealed similarities in expression patterns associated with multiple phenotypes of preterm delivery. Subsequent correlation computations and mediation analyses identified links between intra-amniotic and maternal inflammatory markers and placental serotonin and kynurenine pathways of tryptophan catabolism. Collectively, the findings suggest that a hostile inflammatory environment associated with preterm delivery underlies the mechanisms affecting placental endocrine/transport functions and may contribute to disruption of developmental programming of the fetal brain.
Collapse
Affiliation(s)
- Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Morgane Robles
- INRS-Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QC, Canada
| | - Julia Marushka
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Jaroslav Stranik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Hana Horackova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Jurjen Duintjer Tebbens
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Cathy Vaillancourt
- INRS-Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QC, Canada
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| |
Collapse
|
19
|
Koola MM. Alpha7 nicotinic-N-methyl-D-aspartate hypothesis in the treatment of schizophrenia and beyond. Hum Psychopharmacol 2021; 36:1-16. [PMID: 32965756 DOI: 10.1002/hup.2758] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Development of novel treatments for positive, cognitive, and negative symptoms continue to be a high-priority area of schizophrenia research and a major unmet clinical need. Given that all randomized controlled trials (RCTs) conducted to date failed with one add-on medication/mechanism of action, future RCTs with the same approach are not warranted. Even if the field develops a medication for cognition, others are still needed to treat negative and positive symptoms. Therefore, fixing one domain does not completely solve the problem. Also, targeting the cholinergic system, glutamatergic system, and cholinergic plus alpha7 nicotinic and N-methyl-D-aspartate (NMDA) receptors failed independently. Hence, targeting other less important pathophysiological mechanisms/targets is unlikely to be successful. Meta-analyses of RCTs targeting major pathophysiological mechanisms have found some efficacy signal in schizophrenia; thus, combination treatments with different mechanisms of action may enhance the efficacy signal. The objective of this article is to highlight the importance of conducting RCTs with novel combination treatments in schizophrenia to develop antischizophrenia treatments. Positive RCTs with novel combination treatments that target the alpha7 nicotinic and NMDA receptors simultaneously may lead to a disease-modifying therapeutic armamentarium in schizophrenia. Novel combination treatments that concurrently improve the three domains of psychopathology and several prognostic and theranostic biomarkers may facilitate therapeutic discovery in schizophrenia.
Collapse
Affiliation(s)
- Maju Mathew Koola
- Department of Psychiatry and Behavioral Health, Stony Brook University Renaissance School of Medicine, Stony Brook, New York, USA
| |
Collapse
|
20
|
Bhat A, Pires AS, Tan V, Babu Chidambaram S, Guillemin GJ. Effects of Sleep Deprivation on the Tryptophan Metabolism. Int J Tryptophan Res 2020; 13:1178646920970902. [PMID: 33281456 PMCID: PMC7686593 DOI: 10.1177/1178646920970902] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Sleep has a regulatory role in maintaining metabolic homeostasis and cellular functions. Inadequate sleep time and sleep disorders have become more prevalent in the modern lifestyle. Fragmentation of sleep pattern alters critical intracellular second messengers and neurotransmitters which have key functions in brain development and behavioral functions. Tryptophan metabolism has also been found to get altered in SD and it is linked to various neurodegenerative diseases. The kynurenine pathway is a major regulator of the immune response. Adequate sleep alleviates neuroinflammation and facilitates the cellular clearance of metabolic toxins produced within the brain, while sleep deprivation activates the enzymatic degradation of tryptophan via the kynurenine pathway, which results in an increased accumulation of neurotoxic metabolites. SD causes increased production and accumulation of kynurenic acid in various regions of the brain. Higher levels of kynurenic acid have been found to trigger apoptosis, leads to cognitive decline, and inhibit neurogenesis. This review aims to link the impact of sleep deprivation on tryptophan metabolism and associated complication in the brain.
Collapse
Affiliation(s)
- Abid Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Ananda Staats Pires
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Vanessa Tan
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| | - Gilles J Guillemin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
21
|
Fitzgerald E, Hor K, Drake AJ. Maternal influences on fetal brain development: The role of nutrition, infection and stress, and the potential for intergenerational consequences. Early Hum Dev 2020; 150:105190. [PMID: 32948364 PMCID: PMC7481314 DOI: 10.1016/j.earlhumdev.2020.105190] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
An optimal early life environment is crucial for ensuring ideal neurodevelopmental outcomes. Brain development consists of a finely tuned series of spatially and temporally constrained events, which may be affected by exposure to a sub-optimal intra-uterine environment. Evidence suggests brain development may be particularly vulnerable to factors such as maternal nutrition, infection and stress during pregnancy. In this review, we discuss how maternal factors such as these can affect brain development and outcome in offspring, and we also identify evidence which suggests that the outcome can, in many cases, be stratified by socio-economic status (SES), with individuals in lower brackets typically having a worse outcome. We consider the relevant epidemiological evidence and draw parallels to mechanisms suggested by preclinical work where appropriate. We also discuss possible transgenerational effects of these maternal factors and the potential mechanisms involved. We conclude that modifiable factors such as maternal nutrition, infection and stress are important contributors to atypical brain development and that SES also likely has a key role.
Collapse
Affiliation(s)
- Eamon Fitzgerald
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Kahyee Hor
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Amanda J Drake
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
22
|
Abad C, Karahoda R, Kastner P, Portillo R, Horackova H, Kucera R, Nachtigal P, Staud F. Profiling of Tryptophan Metabolic Pathways in the Rat Fetoplacental Unit During Gestation. Int J Mol Sci 2020; 21:ijms21207578. [PMID: 33066440 PMCID: PMC7589826 DOI: 10.3390/ijms21207578] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/11/2020] [Accepted: 10/11/2020] [Indexed: 01/11/2023] Open
Abstract
Placental homeostasis of tryptophan is essential for fetal development and programming. The two main metabolic pathways (serotonin and kynurenine) produce bioactive metabolites with immunosuppressive, neurotoxic, or neuroprotective properties and their concentrations in the fetoplacental unit must be tightly regulated throughout gestation. Here, we investigated the expression/function of key enzymes/transporters involved in tryptophan pathways during mid-to-late gestation in rat placenta and fetal organs. Quantitative PCR and heatmap analysis revealed the differential expression of several genes involved in serotonin and kynurenine pathways. To identify the flux of substrates through these pathways, Droplet Digital PCR, western blot, and functional analyses were carried out for the rate-limiting enzymes and transporters. Our findings show that placental tryptophan metabolism to serotonin is crucial in mid-gestation, with a subsequent switch to fetal serotonin synthesis. Concurrently, at term, the close interplay between transporters and metabolizing enzymes of both placenta and fetal organs orchestrates serotonin homeostasis and prevents hyper/hypo-serotonemia. On the other hand, the placental production of kynurenine increases during pregnancy, with a low contribution of fetal organs throughout gestation. Any external insult to this tightly regulated harmony of transporters and enzymes within the fetoplacental unit may affect optimal in utero conditions and have a negative impact on fetal programming.
Collapse
Affiliation(s)
- Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (C.A.); (R.K.); (R.P.); (H.H.)
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (C.A.); (R.K.); (R.P.); (H.H.)
| | - Petr Kastner
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (P.K.); (R.K.)
| | - Ramon Portillo
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (C.A.); (R.K.); (R.P.); (H.H.)
| | - Hana Horackova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (C.A.); (R.K.); (R.P.); (H.H.)
| | - Radim Kucera
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (P.K.); (R.K.)
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic;
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (C.A.); (R.K.); (R.P.); (H.H.)
- Correspondence: ; Tel.: +420-495-067-407
| |
Collapse
|
23
|
Li YJ, Yang LP, Hou JL, Li XM, Chen L, Zhu JH, Wang QY, Li G, Zhao PY, Liu XH, Shi ZJ. Prenatal Stress Impairs Postnatal Learning and Memory Development via Disturbance of the cGMP-PKG Pathway and Oxidative Phosphorylation in the Hippocampus of Rats. Front Mol Neurosci 2020; 13:158. [PMID: 33013315 PMCID: PMC7509422 DOI: 10.3389/fnmol.2020.00158] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/03/2020] [Indexed: 12/30/2022] Open
Abstract
Clinical and animal studies have found that prenatal stress can lead to pathological changes in embryos and fetuses. However, the mechanisms through which this occurs have not been made clear. In the present study, pregnant rats were subjected to chronic psychological stress during gestational days using an improved communication box system, and the changes in behavioral performance and proteins in the hippocampus of offspring were analyzed. It was found that prenatal stress caused postnatal growth retardation and impairment in spatial learning and memory. Furthermore, in isobaric tags for relative and absolute quantitation-based proteomics analyses, 158 significantly differentially expressed proteins (DEPs) were found between the two groups. Further analyses showed that these DEPs are involved in different molecular function categories and participate in several biological processes, such as energy metabolism, learning or memory, and synaptic plasticity. Moreover, the enrichment of pathways showed that the learning and memory impairment was primarily connected with the cyclic guanosine monophosphate–protein kinase G (cGMP–PKG) pathway and oxidative phosphorylation. At the same time, the cGMP level and the expression of PKG protein were significantly decreased, and the neuronal mitochondria appeared to have a swollen and irregular shape in the hippocampus of offspring of stressed rats. These results suggest that the chronic psychological stress that pregnant rats were subjected to during gestational days may have impaired the spatial learning and memory of offspring. This affected the hippocampal oxidative phosphorylation and inhibited the cGMP–PKG pathway.
Collapse
Affiliation(s)
- Yu-Jie Li
- Pharmacology Laboratory, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Li-Ping Yang
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jun-Lin Hou
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xin-Min Li
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lei Chen
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiang-Hui Zhu
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qi-Yang Wang
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Gai Li
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Pei-Yuan Zhao
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xi-Hong Liu
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhan-Jiang Shi
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
24
|
Buck SA, Baratta AM, Pocivavsek A. Exposure to elevated embryonic kynurenine in rats: Sex-dependent learning and memory impairments in adult offspring. Neurobiol Learn Mem 2020; 174:107282. [PMID: 32738461 PMCID: PMC7506508 DOI: 10.1016/j.nlm.2020.107282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 12/14/2022]
Abstract
Distinct abnormalities in kynurenine pathway (KP) metabolism have been reported in various psychiatric disorders, including schizophrenia (SZ). Kynurenic acid (KYNA), a neuroactive metabolite of the KP, is elevated in individuals diagnosed with SZ and has been linked to cognitive impairments seen in the disorder. To further understand the role of KYNA in SZ etiology, we developed a prenatal insult model where kynurenine (100 mg/day) is fed to pregnant Wistar rats from embryonic day (ED) 15 to ED 22. As sex differences in the prevalence and severity of SZ have been observed, we presently investigated the impact of prenatal kynurenine exposure on KP metabolism and spatial learning and memory in male and female offspring. Specifically, brain tissue and plasma from offspring (control: ECon; kynurenine-treated: EKyn) in prepuberty (postnatal day (PD) 21), adolescence (PD 32-35), and adulthood (PD 56-85) were collected. Separate cohorts of adult offspring were tested in the Barnes maze to assess hippocampus- and prefrontal cortex-mediated learning and memory. Plasma tryptophan, kynurenine, and KYNA were unchanged between ECon and EKyn offspring across all three ages. Hippocampal and frontal cortex KYNA were elevated in male EKyn offspring only in adulthood, compared to ECon, while brain KYNA levels were unchanged in adult females. Male EKyn offspring were significantly impaired during acquisition of the Barnes maze and during reversal learning in the task. In female EKyn offspring, learning and memory remained relatively intact. Taken together, our data demonstrate that exposure to elevated kynurenine during the last week of gestation results in intriguing sex differences and further support the EKyn model as an attractive tool to study the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Silas A Buck
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Annalisa M Baratta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ana Pocivavsek
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| |
Collapse
|
25
|
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
|
26
|
Klausing AD, Fukuwatari T, Bucci DJ, Schwarcz R. Stress-induced impairment in fear discrimination is causally related to increased kynurenic acid formation in the prefrontal cortex. Psychopharmacology (Berl) 2020; 237:1931-1941. [PMID: 32215686 PMCID: PMC7308198 DOI: 10.1007/s00213-020-05507-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/10/2020] [Indexed: 12/11/2022]
Abstract
RATIONALE Stress is related to cognitive impairments which are observed in most major brain diseases. Prior studies showed that the brain concentration of the tryptophan metabolite kynurenic acid (KYNA) is modulated by stress, and that changes in cerebral KYNA levels impact cognition. However, the link between these phenomena has not been tested directly so far. OBJECTIVES To investigate a possible causal relationship between acute stress, KYNA, and fear discrimination. METHODS Adult rats were exposed to one of three acute stressors-predator odor, restraint, or inescapable foot shocks (ISS)-and KYNA in the prefrontal cortex was measured using microdialysis. Corticosterone was analyzed in a subset of rats. Another cohort underwent a fear discrimination procedure immediately after experiencing stress. Different auditory conditioned stimuli (CSs) were either paired with foot shock (CS+) or were non-reinforced (CS-). One week later, fear was assessed by re-exposing rats to each CS. Finally, to test whether stress-induced changes in KYNA causally impacted fear discrimination, a group of rats that received ISS were pre-treated with the selective KYNA synthesis inhibitor PF-04859989. RESULTS ISS caused the greatest increase in circulating corticosterone levels and raised extracellular KYNA levels by ~ 85%. The two other stressors affected KYNA much less (< 25% increase). Moreover, only rats that received ISS were unable to discriminate between CS+ and CS-. PF-04859989 abolished the stress-induced KYNA increase and also prevented the impairment in fear discrimination in animals that experienced ISS. CONCLUSIONS These findings demonstrate a causal connection between stress-induced KYNA increases and cognitive deficits. Pharmacological manipulation of KYNA synthesis therefore offers a novel approach to modulate cognitive processes in stress-related disorders.
Collapse
Affiliation(s)
- Alex D Klausing
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, 55 Wade Avenue, Baltimore, MD, 21228, USA
| | - Tsutomu Fukuwatari
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, 55 Wade Avenue, Baltimore, MD, 21228, USA
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga, 522-8533, Japan
| | - David J Bucci
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, 55 Wade Avenue, Baltimore, MD, 21228, USA.
| |
Collapse
|
27
|
Rock KD, St Armour G, Horman B, Phillips A, Ruis M, Stewart AK, Jima D, Muddiman DC, Stapleton HM, Patisaul HB. Effects of Prenatal Exposure to a Mixture of Organophosphate Flame Retardants on Placental Gene Expression and Serotonergic Innervation in the Fetal Rat Brain. Toxicol Sci 2020; 176:203-223. [PMID: 32243540 PMCID: PMC7357193 DOI: 10.1093/toxsci/kfaa046] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is a growing need to understand the potential neurotoxicity of organophosphate flame retardants (OPFRs) and plasticizers because use and, consequently, human exposure, is rapidly expanding. We have previously shown in rats that developmental exposure to the commercial flame retardant mixture Firemaster 550 (FM 550), which contains OPFRs, results in sex-specific behavioral effects, and identified the placenta as a potential target of toxicity. The placenta is a critical coordinator of fetal growth and neurodevelopment, and a source of neurotransmitters for the developing brain. We have shown in rats and humans that flame retardants accumulate in placental tissue, and induce functional changes, including altered neurotransmitter production. Here, we sought to establish if OPFRs (triphenyl phosphate and a mixture of isopropylated triarylphosphate isomers) alter placental function and fetal forebrain development, with disruption of tryptophan metabolism as a primary pathway of interest. Wistar rat dams were orally exposed to OPFRs (0, 500, 1000, or 2000 μg/day) or a serotonin (5-HT) agonist 5-methoxytryptamine for 14 days during gestation and placenta and fetal forebrain tissues collected for analysis by transcriptomics and metabolomics. Relative abundance of genes responsible for the transport and synthesis of placental 5-HT were disrupted, and multiple neuroactive metabolites in the 5-HT and kynurenine metabolic pathways were upregulated. In addition, 5-HTergic projections were significantly longer in the fetal forebrains of exposed males. These findings suggest that OPFRs have the potential to impact the 5-HTergic system in the fetal forebrain by disrupting placental tryptophan metabolism.
Collapse
Affiliation(s)
- Kylie D Rock
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27619
| | - Genevieve St Armour
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27619
| | - Brian Horman
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27619
| | - Allison Phillips
- Nicholas School of the Environment, Duke University, Durham, North Carolina 27708
| | - Matthew Ruis
- Nicholas School of the Environment, Duke University, Durham, North Carolina 27708
| | - Allison K Stewart
- Molecular Education, Technology, and Research Innovation Center, North Carolina State University, Raleigh, North Carolina 27695
| | - Dereje Jima
- Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27695
| | - David C Muddiman
- Molecular Education, Technology, and Research Innovation Center, North Carolina State University, Raleigh, North Carolina 27695
| | - Heather M Stapleton
- Nicholas School of the Environment, Duke University, Durham, North Carolina 27708
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27619
- Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695
| |
Collapse
|
28
|
Possibility of Amino Acid Treatment to Prevent the Psychiatric Disorders via Modulation of the Production of Tryptophan Metabolite Kynurenic Acid. Nutrients 2020; 12:nu12051403. [PMID: 32414200 PMCID: PMC7284450 DOI: 10.3390/nu12051403] [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: 03/14/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 01/06/2023] Open
Abstract
Kynurenic acid, a metabolite of the kynurenine pathway of tryptophan catabolism, acts as an antagonist for both the α7 nicotinic acetylcholine receptor and glycine coagonist sites of the N-methyl-d-aspartic acid receptor at endogenous brain concentrations. Elevation of brain kynurenic acid levels reduces the release of neurotransmitters such as dopamine and glutamate, and kynurenic acid is considered to be involved in psychiatric disorders such as schizophrenia and depression. Thus, the control of kynurenine pathway, especially kynurenic acid production, in the brain is an important target for the improvement of brain function or the effective treatment of brain disorders. Astrocytes uptake kynurenine, the immediate precursor of kynurenic acid, via large neutral amino acid transporters, and metabolize kynurenine to kynurenic acid by kynurenine aminotransferases. The former transport both branched-chain and aromatic amino acids, and the latter have substrate specificity for amino acids and their metabolites. Recent studies have suggested the possibility that amino acids may suppress kynurenic acid production via the blockade of kynurenine transport or via kynurenic acid synthesis reactions. This approach may be useful in the treatment and prevention of neurological and psychiatric diseases associated with elevated kynurenic acid levels.
Collapse
|
29
|
Baratta AM, Kanyuch NR, Cole CA, Valafar H, Deslauriers J, Pocivavsek A. Acute sleep deprivation during pregnancy in rats: Rapid elevation of placental and fetal inflammation and kynurenic acid. Neurobiol Stress 2019; 12:100204. [PMID: 32258253 PMCID: PMC7109515 DOI: 10.1016/j.ynstr.2019.100204] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 11/27/2019] [Accepted: 12/11/2019] [Indexed: 01/19/2023] Open
Abstract
The kynurenine pathway (KP) is the dominant pathway for tryptophan degradation in the mammalian body and emerging evidence suggests that acute episodes of sleep deprivation (SD) disrupt tryptophan metabolism via the KP. Increases in the neuroactive KP metabolite kynurenic acid (KYNA) during pregnancy may lead to a higher risk for disrupted neurodevelopment in the offspring. As pregnancy is a critical period during which several factors, including sleep disruptions, could disrupt the fetal environment, we presently explored the relationship between maternal SD and KP metabolism and immune pathways in maternal, placenta, and fetal tissues. Pregnant Wistar rat dams were sleep deprived by gentle handling for 5 h from zeitgeber time (ZT) 0 to ZT 5. Experimental cohorts included: i) controls, ii) one session of SD on embryonic day (ED) 18 or iii) three sessions of SD occurring daily on ED 16, ED 17 and ED 18. Maternal (plasma, brain), placental and fetal (plasma, brain) tissues were collected immediately after the last session of SD or after 24 h of recovery from SD. Respective controls were euthanized at ZT 5 on ED 18 or ED 19. Maternal plasma corticosterone and fetal brain KYNA were significantly elevated only after one session of SD on ED 18. Importantly, maternal plasma corticosterone levels correlated significantly with fetal brain KYNA levels. In addition, placental levels of the proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-6 (IL-6) were increased following maternal SD, suggesting a relationship between placental immune response to SD and fetal brain KYNA accumulation. Collectively, our results demonstrate that sleep loss during the last week of gestation can adversely impact maternal stress, placental immune function, and fetal brain KYNA levels. We introduce KYNA as a novel molecular target influenced by sleep loss during pregnancy. Prenatal sleep deprivation influences kynurenine pathway metabolism in utero. Fetal brain kynurenic acid (KYNA) is elevated after maternal sleep deprivation. Maternal plasma corticosterone is increased after sleep deprivation. Prenatal sleep deprivation induces placental and fetal brain cytokines. These data support an interplay with stress, in utero inflammation, and KYNA.
Collapse
Affiliation(s)
- Annalisa M Baratta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nickole R Kanyuch
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Casey A Cole
- College of Engineering and Computing, University of South Carolina, Columba, South Carolina, USA
| | - Homayoun Valafar
- College of Engineering and Computing, University of South Carolina, Columba, South Carolina, USA
| | - Jessica Deslauriers
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, USA
| | - Ana Pocivavsek
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
30
|
Reduced adolescent risk-assessment and lower nicotinic beta-2 expression in rats exposed to nicotine through lactation by forcedly drinking dams. Neuroscience 2019; 413:64-76. [DOI: 10.1016/j.neuroscience.2019.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 01/06/2023]
|
31
|
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzes the first and rate-limiting reaction of l-tryptophan (Trp) conversion into l-kynurenine (Kyn). The depletion of Trp, and the accumulation of Kyn have been proposed as mechanisms that contribute to the suppression of the immune response-primarily evidenced by in vitro study. IDO1 is therefore considered to be an immunosuppressive modulator and quantification of IDO1 metabolism may be critical to understanding its role in select immunopathologies, including autoimmune- and oncological-conditions, as well as for determining the potency of IDO1 enzyme inhibitors. Because tryptophan 2,3-dioxygenase (TDO), and to a significantly lesser extent, IDO2, also catabolize Trp into Kyn, it's important to differentiate the contribution of each enzyme to Trp catabolism and Kyn generation. Moreover, a great variety of detection methods have been developed for the quantification of Trp metabolites, but choosing the suitable protocol remains challenging. Here, we review the differential expression of IDO1/TDO/IDO2 in normal and malignant tissues, followed by a comprehensive analysis of methodologies for quantifying Trp and Kyn in vitro and in vivo, with an emphasis on the advantages/disadvantages for each application.
Collapse
|
32
|
Notarangelo FM, Beggiato S, Schwarcz R. Assessment of Prenatal Kynurenine Metabolism Using Tissue Slices: Focus on the Neosynthesis of Kynurenic Acid in Mice. Dev Neurosci 2019; 41:102-111. [PMID: 31117076 DOI: 10.1159/000499736] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/19/2019] [Indexed: 01/17/2023] Open
Abstract
Several lines of evidence support the hypothesis that abnormally elevated brain levels of kynurenic acid (KYNA), a metabolite of the kynurenine pathway (KP) of tryptophan degradation, play a pathophysiologically significant role in schizophrenia and other major neurodevelopmental disorders. Studies in experimental animal models suggest that KP impairments in these diseases may originate already in utero since prenatal administration of KYNA's bioprecursor, kynurenine, leads to biochemical and structural abnormalities as well as distinct cognitive impairments in adulthood. As KP metabolism during pregnancy is still insufficiently understood, we designed this study to examine the de novo synthesis of KYNA and 3-hydroxykynurenine (3-HK), an alternative biologically active product of kynurenine degradation, in tissue slices obtained from pregnant mice on gestational day (GD) 18. Fetal brain and liver, placenta, and maternal brain and liver were collected, and the tissues were incubated in vitroin the absence or presence of micromolar concentrations of kynurenine. KYNA and 3-HK were measured in the extracellular milieu. Basal and newly produced KYNA was detected in all cases. As KYNA formation exceeded 3-HK production by 2-3 orders of magnitude in the placenta and maternal brain, and as very little 3-HK neosynthesis was detectable in fetal brain tissue, detailed follow-up experiments focused on KYNA only. The fetal brain produced 3-4 times more KYNA than the maternal brain and placenta, though less than the maternal and fetal liver. No significant differences were observed when using tissues obtained on GD 14 and GD 18. Pharmacological inhibition of KYNA's main biosynthetic enzymes, kynurenine aminotransferase (KAT) I and KAT II, revealed qualitative and quantitative differences between the tissues, with a preferential role of KAT I in the fetal and maternal brain and of KAT II in the fetal and maternal liver. Findings using tissue slices from KAT II knockout mice confirmed these conclusions. Together, these results clarify the dynamics of KP metabolism during pregnancy and provide the basis for the conceptualization of interventions aimed at manipulating cerebral KP function in the prenatal period.
Collapse
Affiliation(s)
- Francesca M Notarangelo
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA,
| | - Sarah Beggiato
- 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
|
33
|
Kalinina DS, Vasilev DS, Volnova AB, Nalivaeva NN, Zhuravin IA. Age-Dependent Electrocorticogram Dynamics and Epileptogenic Responsiveness in Rats Subjected to Prenatal Hypoxia. Dev Neurosci 2019; 41:56-66. [PMID: 30904914 DOI: 10.1159/000497224] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 01/24/2019] [Indexed: 11/19/2022] Open
Abstract
Using electrocorticogram (ECoG) analysis, we compared age-related dynamics of general neuronal activity and convulsive epileptiform responsiveness induced by intracortical microinjections of 4-aminopyridine (4-AP) in control Wistar rats and those subjected to prenatal hypoxia (Hx; E14; 7% O2, 3 h). The studies were carried out in three age periods roughly corresponding to childhood (P20-27), adolescence (P30-45), and adulthood (P90-120). It was found that in the process of postnatal development of the control rats, the peak of the ECoG power spectrum density (PSD) of the theta rhythm during wakefulness shifted from the low to the higher frequency, while in the Hx rats this shift had the opposite direction. Moreover, the Hx rats had different frequency characteristics of the ECoG PSD and longer episodes of spike-and-wave discharges caused by 4-AP injections compared to the controls. The total ECoG PSD of slow-wave sleep (1-5 Hz) was also dramatically decreased in the process of development of the Hx rats. Such alterations in PSD could be explained by the changes in balance of the excitation and inhibition processes in the cortical networks. Analyzing protein levels of neurotransmitter transporters in the brain structures of the Hx rats, we found that the content of the glutamate transporter EAAT1 was higher in the parietal cortex in all age groups of Hx rats while in the hippocampus it decreased during postnatal development compared to controls. Furthermore, the content of the vesicular acetylcholine transporter in the parietal cortex, and of the inhibitory GABA transporter 1 in the hippocampus, was also affected by prenatal Hx. These data suggest that prenatal Hx results in a shift in the excitatory and inhibitory balance in the rat cortex towards excitation, making the rat's brain more vulnerable to the effects of proconvulsant drugs and predisposing animals to epileptogenesis during postnatal life.
Collapse
Affiliation(s)
- Daria S Kalinina
- Saint Petersburg State University, Saint Petersburg, Russian Federation.,I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, Russian Federation
| | - Dmitrii S Vasilev
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, Russian Federation.,Saint Petersburg State Pediatric Medical University, Saint Petersburg, Russian Federation
| | - Anna B Volnova
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Natalia N Nalivaeva
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, Russian Federation
| | - Igor A Zhuravin
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, Russian Federation, .,Saint Petersburg State Pediatric Medical University, Saint Petersburg, Russian Federation,
| |
Collapse
|
34
|
Effect of substituents on 3(S)-amino-1-hydroxy-3,4-dihydroquinolin-2(1H)-one: a DFT study. Theor Chem Acc 2019. [DOI: 10.1007/s00214-018-2403-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
35
|
Shah J, Deas SB, Ren C, Jilling T, Brawner KM, Martin CA. The Effects of Gestational Psychological Stress on Neonatal Mouse Intestinal Development. J Surg Res 2018; 235:621-628. [PMID: 30691851 DOI: 10.1016/j.jss.2018.10.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 10/03/2018] [Accepted: 10/30/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Psychological stress during pregnancy has been shown to cause subsequent harm to the fetus and newborn. Many studies focus on neurodevelopmental outcomes, but little is known about the effect of gestational stress on intestinal immunity and development. The purpose of this study was to determine the effect of psychological stress during pregnancy on intestinal architecture and growth in newborns. METHODS Eight-week-old C57BL6 littermates underwent timed breeding. Pregnant dams were subjected to 1 h of daily psychological stress by using a well-established restraint model during days E7-E14. The distal ileum of 2-wk-old offspring of stressed mothers and nonstressed controls was harvested for histologic analysis. Slides were blinded to measure villus height and crypt depth and surface area. Serum was obtained to measure serum corticosterone levels. An explant model was used to measure corticosterone on the intestinal stem cell marker Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) and growth factors epidermal growth factor receptor and insulin-like growth factor-1. RESULTS The villus height, crypt depth, and surface area were significantly decreased in newborn exposed to stress during gestation. In addition, corticosterone levels were elevated in 2-wk-old mice exposed to stress. Real-time polymerase chain reaction revealed that explants exposed to corticosterone had a decrease in LGR5 compared with controls and an increase in epidermal growth factor receptor. CONCLUSIONS Here, we establish that neonatal mice from mothers that were subjected to psychological stress during pregnancy have significantly shorter villi and crypts compared with controls. In addition, pups from stressed mothers have decreased expression levels of the intestinal stem cell marker LGR5. These findings will aid in determining the effect of gestational psychological stress on intestinal development and stem cell plasticity.
Collapse
Affiliation(s)
- Juhi Shah
- Division of Pediatric Surgery, Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sara Beth Deas
- Division of Pediatric Surgery, Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Changchun Ren
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tamas Jilling
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kyle M Brawner
- Division of Pediatric Surgery, Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Colin A Martin
- Division of Pediatric Surgery, Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
36
|
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: 1.4] [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
|
37
|
Dostal CR, Gamsby NS, Lawson MA, McCusker RH. Glia- and tissue-specific changes in the Kynurenine Pathway after treatment of mice with lipopolysaccharide and dexamethasone. Brain Behav Immun 2018; 69:321-335. [PMID: 29241670 PMCID: PMC5857427 DOI: 10.1016/j.bbi.2017.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 12/09/2017] [Accepted: 12/09/2017] [Indexed: 12/31/2022] Open
Abstract
Behavioral symptoms associated with mood disorders have been intimately linked with immunological and psychological stress. Induction of immune and stress pathways is accompanied by increased tryptophan entry into the Kynurenine (Kyn) Pathway as governed by the rate-limiting enzymes indoleamine/tryptophan 2,3-dioxygenases (DO's: Ido1, Ido2, Tdo2). Indeed, elevated DO expression is associated with inflammation- and stress-related depression symptoms. Here we examined central (brain, astrocyte and microglia) and peripheral (lung, liver and spleen) DO expression in mice treated intraperitoneally with lipopolysaccharide (LPS) and dexamethasone (DEX) to model the response of the Kyn Pathway to inflammation and glucocorticoids. LPS-induced expression of cytokines in peripheral tissues was attenuated by DEX, confirming inflammatory and anti-inflammatory responses, respectively. Increased Kyn levels following LPS and DEX administration verified Kyn Pathway activation. Expression of multiple mRNA isoforms for each DO, which we have shown to be differentially utilized and regulated, were quantified including reference/full-length (FL) and variant (v) transcripts. LPS increased Ido1-FL in brain (∼1000-fold), a response paralleled by increased expression in both astrocytes and microglia. Central Ido1-FL was not changed by DEX; however, LPS-induced Ido1-FL was decreased by DEX in peripheral tissues. In contrast, DEX increased Ido1-v1 expression by astrocytes and microglia, but not peripheral tissues. In comparison, brain Ido2 was minimally induced by LPS or DEX. Uniquely, Ido2-v6 was LPS- and DEX-inducible in astrocytes, suggesting a unique role for astrocytes in response to inflammation and glucocorticoids. Only DEX increased central Tdo2 expression; however, peripheral Tdo2 was upregulated by either LPS or DEX. In summary, specific DO isoforms are increased by LPS and DEX, but LPS-dependent Ido1 and Ido2 induction are attenuated by DEX only in the periphery indicating that elevated DO expression and Kyn production within the brain can occur independent of the periphery. These findings demonstrate a plausible interaction between immune activation and glucocorticoids associated with depression.
Collapse
Affiliation(s)
- Carlos R. Dostal
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States,Medical Scholars Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States,Integrative Immunology and Behavior Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Nicolaus S. Gamsby
- School of Earth, Society and Environment, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Marcus A. Lawson
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States,Integrative Immunology and Behavior Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Robert H. McCusker
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States,Integrative Immunology and Behavior Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States,Department of Pathology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| |
Collapse
|
38
|
Goeden N, Notarangelo FM, Pocivavsek A, Beggiato S, Bonnin A, Schwarcz R. Prenatal Dynamics of Kynurenine Pathway Metabolism in Mice: Focus on Kynurenic Acid. Dev Neurosci 2017; 39:519-528. [PMID: 29080891 DOI: 10.1159/000481168] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/31/2017] [Indexed: 01/09/2023] Open
Abstract
The kynurenine pathway (KP), the major catabolic route of tryptophan in mammals, contains several neuroactive metabolites, including kynurenic acid (KYNA) and 3-hydroxykynurenine (3-HK). KP metabolism, and especially the fate of KYNA, during pregnancy is poorly understood, yet it may play a significant role in the development of psychiatric disorders later in life. The present study was designed to investigate the prenatal features of KP metabolism in vivo, with special focus on KYNA. To this end, pregnant CD-1 mice were treated systemically with kynurenine (100 mg/kg), KYNA (10 mg/kg), or saline on embryonic day 18. As expected, administration of either kynurenine or KYNA increased KYNA levels in the maternal plasma and placenta. Maternal kynurenine treatment also raised kynurenine levels in the fetal plasma and brain, demonstrating the ability of this pivotal KP metabolite to cross the placenta and increase the levels of both KYNA and 3-HK in the fetal brain. In contrast, maternal administration of KYNA caused only a small, nonsignificant elevation in KYNA levels in fetal plasma and brain. Complementary experiments using an ex vivo placental perfusion procedure confirmed the significant transplacental transfer of kynurenine and demonstrated that only a very small fraction of maternal kynurenine is converted to KYNA in the placenta and released into the fetal compartment under physiological conditions. Jointly, these results help to clarify the contributions of the maternal circulation and the placenta to fetal KYNA in the late prenatal period.
Collapse
Affiliation(s)
- Nick Goeden
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
39
|
Kelly JR, Minuto C, Cryan JF, Clarke G, Dinan TG. Cross Talk: The Microbiota and Neurodevelopmental Disorders. Front Neurosci 2017; 11:490. [PMID: 28966571 PMCID: PMC5605633 DOI: 10.3389/fnins.2017.00490] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/17/2017] [Indexed: 12/11/2022] Open
Abstract
Humans evolved within a microbial ecosystem resulting in an interlinked physiology. The gut microbiota can signal to the brain via the immune system, the vagus nerve or other host-microbe interactions facilitated by gut hormones, regulation of tryptophan metabolism and microbial metabolites such as short chain fatty acids (SCFA), to influence brain development, function and behavior. Emerging evidence suggests that the gut microbiota may play a role in shaping cognitive networks encompassing emotional and social domains in neurodevelopmental disorders. Drawing upon pre-clinical and clinical evidence, we review the potential role of the gut microbiota in the origins and development of social and emotional domains related to Autism spectrum disorders (ASD) and schizophrenia. Small preliminary clinical studies have demonstrated gut microbiota alterations in both ASD and schizophrenia compared to healthy controls. However, we await the further development of mechanistic insights, together with large scale longitudinal clinical trials, that encompass a systems level dimensional approach, to investigate whether promising pre-clinical and initial clinical findings lead to clinical relevance.
Collapse
Affiliation(s)
- John R Kelly
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Chiara Minuto
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College CorkCork, Ireland.,Department of Anatomy and Neuroscience, University College CorkCork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| |
Collapse
|
40
|
Xu K, Liu H, Bai M, Gao J, Wu X, Yin Y. Redox Properties of Tryptophan Metabolism and the Concept of Tryptophan Use in Pregnancy. Int J Mol Sci 2017; 18:E1595. [PMID: 28737706 PMCID: PMC5536082 DOI: 10.3390/ijms18071595] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/11/2017] [Accepted: 07/19/2017] [Indexed: 12/30/2022] Open
Abstract
During pregnancy, tryptophan (Trp) is required for several purposes, and Trp metabolism varies over time in the mother and fetus. Increased oxidative stress (OS) with high metabolic, energy and oxygen demands during normal pregnancy or in pregnancy-associated disorders has been reported. Taking the antioxidant properties of Trp and its metabolites into consideration, we made four hypotheses. First, the use of Trp and its metabolites is optional based on their antioxidant properties during pregnancy. Second, dynamic Trp metabolism is an accommodation mechanism in response to OS. Third, regulation of Trp metabolism could be used to control/attenuate OS according to variations in Trp metabolism during pregnancy. Fourth, OS-mediated injury could be alleviated by regulation of Trp metabolism in pregnancy-associated disorders. Future studies in normal/abnormal pregnancies and in associated disorders should include measurements of free Trp, total Trp, Trp metabolites, and activities of Trp-degrading enzymes in plasma. Abnormal pregnancies and some associated disorders may be associated with disordered Trp metabolism related to OS. Mounting evidence suggests that the investigation of the use of Trp and its metabolites in pregnancy will be meanful.
Collapse
Affiliation(s)
- Kang Xu
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Hongnan Liu
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Miaomiao Bai
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Jing Gao
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Xin Wu
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Yulong Yin
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| |
Collapse
|