1
|
Adamczyk PM, Shaw A, Morella IM, More L. Neurobiology, molecular pathways, and environmental influences in antisocial traits and personality disorders. Neuropharmacology 2025; 269:110322. [PMID: 39864585 DOI: 10.1016/j.neuropharm.2025.110322] [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: 05/31/2024] [Revised: 12/17/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Personality disorders (PDs) are psychiatric conditions characterized by enduring patterns of cognition, emotion, and behaviour that deviate significantly from cultural norms, causing distress or impairment. The aetiology of PDs is complex, involving both genetic and environmental factors. Genetic studies estimate the heritability of PDs at 30%-60%, implicating genes involved in neurotransmitter regulation, such as those for serotonin transporters and dopamine receptors. Environmental factors, including childhood trauma and chronic stress, interact with genetic predispositions to induce epigenetic modifications like DNA methylation and histone modifications, contributing to PD development. Neurobiological research has identified structural and functional abnormalities in brain regions related to emotional regulation and social cognition, such as the amygdala, prefrontal cortex, and limbic system. These abnormalities are linked to impaired emotion processing and interpersonal functioning in PDs. This review focuses on how environmental factors shape maladaptive behaviours and endophenotypes central to many PDs. It explores the interaction between the Ras-ERK, p38, and mTOR molecular pathways in response to environmental stimuli, and examines the role of oxidative stress and mitochondrial metabolism in these processes. Also reviewed are various types of PDs and existing animal models that replicate key endophenotypes, highlighting changes in neurotransmitters and neurohormones. Identifying molecular biomarkers can lead to the development of "enviromimetic" drugs, which mimic environmental influences to activate molecular pathways, facilitating targeted, personalized treatments based on the molecular profiles of individuals with PDs. Ultimately, understanding the molecular mechanisms of PDs promises to enhance diagnostic accuracy, prognosis, and therapeutic outcomes for affected individuals.
Collapse
Affiliation(s)
- Patryk M Adamczyk
- School of Pharmacy and Biomedical Sciences, The University of Central Lancashire, Preston, UK
| | - Andrew Shaw
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK.
| | - Ilaria M Morella
- University of Pavia, Department of Biology and Biotechnology "Lazzaro Spallanzani", Pavia, Italy; Cardiff University, School of Medicine, Division of Psychological Medicine and Clinical Neurosciences, Cardiff, UK.
| | - Lorenzo More
- School of Pharmacy and Biomedical Sciences, The University of Central Lancashire, Preston, UK.
| |
Collapse
|
2
|
Fisher A, Boruah R, Mayne P, Monavari A, Crushell E, Knerr I. The metabolic landscape of tetrahydrobiopterin metabolism disorders in the Republic of Ireland. Mol Genet Metab Rep 2025; 42:101185. [PMID: 39844797 PMCID: PMC11751560 DOI: 10.1016/j.ymgmr.2024.101185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/24/2025] Open
Abstract
We present a case series of seven patients (5 males, 2 females, aged 7-38 yrs.) in Ireland with biopterin metabolism disorder. Five individuals had been diagnosed with dihydropteridine reductase (DHPR) deficiency and two with pyruvoyl tetrahydropterin synthase (PTPS) deficiency. While clinical symptoms were mainly neuro-developmental in nature, one of our patients with DHPR deficiency also had a mild pulmonary valve stenosis and patent arterial duct in infancy which subsequently resolved as a hitherto undescribed finding in this condition. Clinical outcomes in our patient cohort were overall satisfactory with the best outcomes in patients/siblings diagnosed on high-risk screening. In conclusion, early diagnosis, pathophysiology-driven treatments and frequent patient-specific treatment adjustments are crucial to sustain the best possible long-term outcomes. Ireland's cohort of tetrahydropterin metabolism disorders highlights that improved outcomes are achieved with an early diagnosis which may not be attainable through newborn screening alone.
Collapse
Affiliation(s)
- A. Fisher
- National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street, Dublin, Ireland
| | - R. Boruah
- National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street, Dublin, Ireland
| | - P.D. Mayne
- Department of Biochemistry, Children's Health Ireland at Temple Street, Dublin, Ireland
| | - A.A. Monavari
- National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street, Dublin, Ireland
| | - E. Crushell
- National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street, Dublin, Ireland
| | - I. Knerr
- National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street, Dublin, Ireland
| |
Collapse
|
3
|
Bekhbat M, Block AM, Dickinson SY, Tharp GK, Bosinger SE, Felger JC. Neurotransmitter and metabolic effects of interferon-alpha in association with decreased striatal dopamine in a non-human primate model of cytokine-Induced depression. Brain Behav Immun 2025; 125:308-318. [PMID: 39826580 PMCID: PMC11903159 DOI: 10.1016/j.bbi.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/13/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Inflammatory stimuli administered to humans and laboratory animals affect mesolimbic and nigrostriatal dopaminergic pathways in association with impaired motivation and motor activity. Alterations in dopaminergic corticostriatal reward and motor circuits have also been observed in depressed patients with increased peripheral inflammatory markers. The effects of peripheral inflammation on dopaminergic pathways and associated neurobiologic mechanisms and consequences have been difficult to measure in patients. Postmortem tissue (n = 11) from an established, translationally-relevant non-human primate model of cytokine-induced depressive behavior involving chronic interferon-alpha (IFN-a) administration was utilized herein to explore the molecular mechanisms of peripheral cytokine effects on striatal dopamine. Dopamine (but not serotonin or norepinephrine) was decreased in the nucleus accumbens (NAcc) and putamen of IFN-a-treated animals (p < 0.05). IFN-a had no effect on number of striatal neurons or dopamine terminal density, suggesting no overt neurodegenerative changes. RNA sequencing examined in the caudate, putamen, substantia nigra, and prefrontal cortical subregions revealed that while IFN-a nominally up-regulated limited numbers of genes enriching inflammatory signaling pathways in all regions, robust, whole genome-significant effects of IFN-a were observed specifically in putamen. Genes upregulated in the putamen primarily enriched synaptic signaling, glutamate receptor signaling, and inflammatory/metabolic pathways downstream of IFN-a, including MAPK and PI3K/AKT cascades. Conversely, gene transcripts reduced by IFN-a enriched oxidative phosphorylation (OXPHOS), protein translation, and pathways regulated by dopamine receptors. Unsupervised clustering identified a gene co-expression module in the putamen that was associated with both IFN-a treatment and low dopamine levels, which enriched similar inflammatory, metabolic, and synaptic signaling pathways. IFN-a-induced reductions in dopamine further correlated with genes related to excitotoxic glutamate, kynurenine, and altered dopamine receptor signaling (r = 0.78-97, p < 0.05). These findings provide insight into the immunologic mechanisms and neurobiological consequences of peripheral inflammation effects on dopamine, which may inform novel treatment strategies targeting inflammatory, metabolic or neurotransmitter systems in depressed patients with high inflammation.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Andrew M Block
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06030, USA
| | - Sarah Y Dickinson
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Gregory K Tharp
- Emory Nonhuman Primate Genomics Core, Division of Microbiology and Immunology, Emory National Primate Research Center (EPC), Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven E Bosinger
- Emory Nonhuman Primate Genomics Core, Division of Microbiology and Immunology, Emory National Primate Research Center (EPC), Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
4
|
Rashad S. Queuosine tRNA Modification: Connecting the Microbiome to the Translatome. Bioessays 2025; 47:e202400213. [PMID: 39600051 PMCID: PMC11755703 DOI: 10.1002/bies.202400213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
Transfer RNA (tRNA) modifications play an important role in regulating mRNA translation at the codon level. tRNA modifications can influence codon selection and optimality, thus shifting translation toward specific sets of mRNAs in a dynamic manner. Queuosine (Q) is a tRNA modification occurring at the wobble position. In eukaryotes, queuosine is synthesized by the tRNA-guanine trans-glycosylase (TGT) complex, which incorporates the nucleobase queuine (or Qbase) into guanine of the GUN anticodons. Queuine is sourced from gut bacteria and dietary intake. Q was recently shown to be critical for cellular responses to oxidative and mitochondrial stresses, as well as its potential role in neurodegenerative diseases and brain health. These unique features of Q provide an interesting insight into the regulation of mRNA translation by gut bacteria, and the potential health implications. In this review, Q biology is examined in the light of recent literature and nearly 4 decades of research. Q's role in neuropsychiatric diseases and cancer is highlighted and discussed. Given the recent interest in Q, and the new findings, more research is needed to fully comprehend its biological function and disease relevance, especially in neurobiology.
Collapse
Affiliation(s)
- Sherif Rashad
- Department of Neurosurgical Engineering and Translational NeuroscienceTohoku University Graduate School of MedicineSendaiJapan
- Department of Neurosurgical Engineering and Translational NeuroscienceGraduate School of Biomedical EngineeringTohoku UniversitySendaiJapan
| |
Collapse
|
5
|
Kajumba MM, Kakooza-Mwesige A, Nakasujja N, Koltai D, Canli T. Treatment-resistant depression: molecular mechanisms and management. MOLECULAR BIOMEDICINE 2024; 5:43. [PMID: 39414710 PMCID: PMC11485009 DOI: 10.1186/s43556-024-00205-y] [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: 03/20/2024] [Accepted: 09/03/2024] [Indexed: 10/18/2024] Open
Abstract
Due to the heterogeneous nature of depression, the underlying etiological mechanisms greatly differ among individuals, and there are no known subtype-specific biomarkers to serve as precise targets for therapeutic efficacy. The extensive research efforts over the past decades have not yielded much success, and the currently used first-line conventional antidepressants are still ineffective for close to 66% of patients. Most clinicians use trial-and-error treatment approaches, which seem beneficial to only a fraction of patients, with some eventually developing treatment resistance. Here, we review evidence from both preclinical and clinical studies on the pathogenesis of depression and antidepressant treatment response. We also discuss the efficacy of the currently used pharmacological and non-pharmacological approaches, as well as the novel emerging therapies. The review reveals that the underlying mechanisms in the pathogenesis of depression and antidepressant response, are not specific, but rather involve an interplay between various neurotransmitter systems, inflammatory mediators, stress, HPA axis dysregulation, genetics, and other psycho-neurophysiological factors. None of the current depression hypotheses sufficiently accounts for the interactional mechanisms involved in both its etiology and treatment response, which could partly explain the limited success in discovering efficacious antidepressant treatment. Effective management of treatment-resistant depression (TRD) requires targeting several interactional mechanisms, using subtype-specific and/or personalized therapeutic modalities, which could, for example, include multi-target pharmacotherapies in augmentation with psychotherapy and/or other non-pharmacological approaches. Future research guided by interaction mechanisms hypotheses could provide more insights into potential etiologies of TRD, precision biomarker targets, and efficacious therapeutic modalities.
Collapse
Affiliation(s)
- Mayanja M Kajumba
- Department of Mental Health and Community Psychology, Makerere University, P. O. Box 7062, Kampala, Uganda.
| | - Angelina Kakooza-Mwesige
- Department of Pediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
- Department of Pediatrics and Child Health, Mulago National Referral Hospital, Kampala, Uganda
| | - Noeline Nakasujja
- Department of Psychiatry, School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Deborah Koltai
- Duke Division of Global Neurosurgery and Neurology, Department of Neurosurgery, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, USA
| | - Turhan Canli
- Department of Psychology, Stony Brook University, New York, USA
- Department of Psychiatry, Stony Brook University, New York, USA
| |
Collapse
|
6
|
Koreki A, Sado M, Mitsukura Y, Tachimori H, Kubota A, Kanamori Y, Uchibori M, Usune S, Ninomiya A, Shirahama R, Fujimoto A, Inabe K, Miyata H, Mimura M. The association between salivary IL-6 and poor sleep quality assessed using Apple watches in stressed workers in Japan. Sci Rep 2024; 14:22620. [PMID: 39349506 PMCID: PMC11442746 DOI: 10.1038/s41598-024-70834-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/21/2024] [Indexed: 10/02/2024] Open
Abstract
Insomnia is a major concern among stressed workers worldwide. Although stress generally has a detrimental effect on sleep quality, the impact of biological stress, especially the immunological stress response, on sleep quality is not yet fully understood. Stressed workers were recruited through a screening process using a Brief Job Stress Questionnaire. The participants were asked to complete various occupation-related questionnaires. Additionally, saliva samples were collected to assess interleukin-6 (IL-6) levels as an immunological stress response. Subsequently, they were asked to wear an Apple Watch to record their sleep pattern for one week. Their sleep architecture was estimated using a previously published and validated method. Finally, data from 73 participants were analyzed. Our multivariable analysis revealed that shorter durations of slow-wave sleep (non-rapid eye movement sleep stage 3) were significantly associated with elevated IL-6 levels (p = 0.005) and greater daytime sleepiness (p = 0.002) after controlling for total sleep time. Our finding that a higher immunological stress response is associated with poor sleep quality contributes to a better understanding of insomnia in stressed workers and emphasizes the importance of stress management in this population.
Collapse
Affiliation(s)
- Akihiro Koreki
- Department of Neuropsychiatry, Keio University School of Medicine, Shinanomachi 35, Shinjuku-Ku, Tokyo, 160-8582, Japan
- Department of Psychiatry, NHO Shimofusa Psychiatric Medical Center, Chiba, Japan
- Mindfulness & Stress Research Center, Keio University, Tokyo, Japan
| | - Mitsuhiro Sado
- Department of Neuropsychiatry, Keio University School of Medicine, Shinanomachi 35, Shinjuku-Ku, Tokyo, 160-8582, Japan.
- Mindfulness & Stress Research Center, Keio University, Tokyo, Japan.
- Keio University Health Center, Tokyo, Japan.
| | - Yasue Mitsukura
- Department of Neuropsychiatry, Keio University School of Medicine, Shinanomachi 35, Shinjuku-Ku, Tokyo, 160-8582, Japan
- Department of System Design Engineering, Faculty of Science and Technology, Keio University, Tokyo, Japan
| | - Hisateru Tachimori
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
- Endowed Course for Health System Innovation, Keio University School of Medicine, Tokyo, Japan
| | - Anna Kubota
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiaki Kanamori
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Manae Uchibori
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Shiyori Usune
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Akira Ninomiya
- Department of Neuropsychiatry, Keio University School of Medicine, Shinanomachi 35, Shinjuku-Ku, Tokyo, 160-8582, Japan
- Mindfulness & Stress Research Center, Keio University, Tokyo, Japan
| | - Ryutaro Shirahama
- RESM Respiratory and Sleep Medical-Care Clinic, Tokyo/Yokohama, Japan
| | | | - Kanako Inabe
- Medical Headquarters, Eisai Co., Ltd, Tokyo, Japan
| | - Hiroaki Miyata
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Shinanomachi 35, Shinjuku-Ku, Tokyo, 160-8582, Japan.
- Mindfulness & Stress Research Center, Keio University, Tokyo, Japan.
- Center for Preventive Medicine, Keio University, Tokyo, Japan.
| |
Collapse
|
7
|
Allboani A, Kar S, Kavdia M. Computational modeling of neuronal nitric oxide synthase biochemical pathway: A mechanistic analysis of tetrahydrobiopterin and oxidative stress. Free Radic Biol Med 2024; 222:625-637. [PMID: 39004235 DOI: 10.1016/j.freeradbiomed.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/14/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Neuronal cell dysfunction plays an important role in neurodegenerative diseases. Oxidative stress can disrupt the redox balance within neuronal cells and may cause neuronal nitric oxide synthase (nNOS) to uncouple, contributing to the neurodegenerative processes. Experimental studies and clinical trials using nNOS cofactor tetrahydrobiopterin (BH4) and antioxidants in neuronal cell dysfunction have shown inconsistent results. A better mechanistic understanding of complex interactions of nNOS activity and oxidative stress in neuronal cell dysfunction is needed. In this study, we developed a computational model of neuronal cell using nNOS biochemical pathways to explore several key mechanisms that are known to influence neuronal cell redox homeostasis. We studied the effects of oxidative stress and BH4 synthesis on nNOS nitric oxide production and biopterin ratio (BH4/total biopterin). Results showed that nNOS remained coupled and maintained nitric oxide production for oxidative stress levels less than 230 nM/s. The results showed that neuronal oxidative stress above 230 nM/s increased the degree of nNOS uncoupling and introduced instability in the nitric oxide production. The nitric oxide production did not change irrespective of initial biopterin ratio of 0.05-0.99 for a given oxidative stress. Oxidative stress resulted in significant reduction in BH4 levels even when nitric oxide production was not affected. Enhancing BH4 synthesis or supplementation improved nNOS coupling, however the degree of improvement was determined by the levels of oxidative stress and BH4 synthesis. The results of our mechanistic analysis indicate that there is a potential for significant improvement in neuronal dysfunction by simultaneously increasing BH4 levels and reducing cellular oxidative stress.
Collapse
Affiliation(s)
- Amnah Allboani
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, 48202, USA
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Kuwait
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
8
|
Maldonado-García JL, García-Mena LH, Mendieta-Cabrera D, Pérez-Sánchez G, Becerril-Villanueva E, Alvarez-Herrera S, Homberg T, Vallejo-Castillo L, Pérez-Tapia SM, Moreno-Lafont MC, Ortuño-Sahagún D, Pavón L. Use of Extracellular Monomeric Ubiquitin as a Therapeutic Option for Major Depressive Disorder. Pharmaceuticals (Basel) 2024; 17:841. [PMID: 39065692 PMCID: PMC11279398 DOI: 10.3390/ph17070841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Major depressive disorder (MDD) is a mood disorder that has become a global health emergency according to the World Health Organization (WHO). It affects 280 million people worldwide and is a leading cause of disability and financial loss. Patients with MDD present immunoendocrine alterations like cortisol resistance and inflammation, which are associated with alterations in neurotransmitter metabolism. There are currently numerous therapeutic options for patients with MDD; however, some studies suggest a high rate of therapeutic failure. There are multiple hypotheses explaining the pathophysiological mechanisms of MDD, in which several systems are involved, including the neuroendocrine and immune systems. In recent years, inflammation has become an important target for the development of new therapeutic options. Extracellular monomeric ubiquitin (emUb) is a molecule that has been shown to have immunomodulatory properties through several mechanisms including cholinergic modulation and the generation of regulatory T cells. In this perspective article, we highlight the influence of the inflammatory response in MDD. In addition, we review and discuss the evidence for the use of emUb contained in Transferon as a concomitant treatment with selective serotonin reuptake inhibitors (SSRIs).
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (S.M.P.-T.)
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| | - Lissette Haydee García-Mena
- Departamento de Salud Digital, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico;
| | - Danelia Mendieta-Cabrera
- Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 14370, Mexico;
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| | - Samantha Alvarez-Herrera
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| | - Toni Homberg
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (T.H.); (L.V.-C.)
- Laboratorio Nacional Para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) Para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Mexico City 11340, Mexico
| | - Luis Vallejo-Castillo
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (T.H.); (L.V.-C.)
- Laboratorio Nacional Para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) Para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Mexico City 11340, Mexico
| | - Sonia Mayra Pérez-Tapia
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (S.M.P.-T.)
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (T.H.); (L.V.-C.)
- Laboratorio Nacional Para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) Para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Mexico City 11340, Mexico
| | - Martha C. Moreno-Lafont
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (S.M.P.-T.)
| | - Daniel Ortuño-Sahagún
- Instituto de Investigación en Ciencias Biomédicas (IICB), CUCS, Universidad de Guadalajara, Jalisco 44340, Mexico;
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| |
Collapse
|
9
|
Ferat-Osorio E, Maldonado-García JL, Pavón L. How inflammation influences psychiatric disease. World J Psychiatry 2024; 14:342-349. [PMID: 38617981 PMCID: PMC11008389 DOI: 10.5498/wjp.v14.i3.342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/16/2024] [Accepted: 02/18/2024] [Indexed: 03/19/2024] Open
Abstract
Recent studies highlight the strong correlation between infectious diseases and the development of neuropsychiatric disorders. In this editorial, we comment on the article "Anti-infective therapy durations predict psychological stress and laparoscopic surgery quality in pelvic abscess patients" by Zhang et al, published in the recent issue of the World Journal of Psychiatry 2023; 13 (11): 903-911. Our discussion highlighted the potential consequences of anxiety, depression, and psychosis, which are all linked to bacterial, fungal, and viral infections, which are relevant to the impact of inflammation on the sequelae in mental health as those we are observing after the coronavirus disease 2019 pandemic. We focus specifically on the immune mechanisms triggered by inflammation, the primary contributor to psychiatric complications. Importantly, pathophysiological mechanisms such as organ damage, post-injury inflammation, and infection-induced endocrine alterations, including hypocortisolism or autoantibody formation, significantly contribute to the development of chronic low-grade inflammation, promoting the emergence or development of psychiatric alterations in susceptible individuals. As inflammation can have long-term effects on patients, a multidisciplinary treatment plan can avoid complications and debilitating health issues, and it is crucial to recognize and address the mental health implications.
Collapse
Affiliation(s)
- Eduardo Ferat-Osorio
- División de Investigación Clínica de la Coordinación de Investigación en Salud, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - José Luis Maldonado-García
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán 04510, Ciudad de México, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| |
Collapse
|
10
|
Wilson SK, Thomas J. BH4 as a Therapeutic Target for ADHD: Relevance to Neurotransmitters and Stress-Driven Symptoms. J Atten Disord 2024; 28:161-167. [PMID: 37942650 DOI: 10.1177/10870547231204012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Tetrahydrobiopterin (BH4) is a critical cofactor in a variety of metabolic pathways that have been linked to ADHD. There have been no previous studies utilizing BH4 as a supplement for ADHD. BH4 has been approved as a treatment for phenylketonuria (PKU). Individuals with PKU and ADHD appear to have low DA levels in common, suggesting that the hypodopaminergic state seen in both illnesses could be a relationship between the two. Clinical research involving supplementation of BH4 has shown low occurrence of adverse. In experiments, BH4 has also been found to have good blood-brain barrier permeability. BH4 also has the ability in scavenging ROS activity, which is an implication of stress and is seen in ADHD. BH4's significance in ADHD is reviewed in this paper because of its involvement in numerous neurodevelopmental metabolic pathways, and we anticipate that exogenous BH4 can be used to treat ADHD.
Collapse
Affiliation(s)
- Samson K Wilson
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala
| | - Jaya Thomas
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala
| |
Collapse
|
11
|
Kolks N, Neumaier F, Neumaier B, Zlatopolskiy BD. Preparation of NIn-Methyl-6-[ 18F]fluoro- and 5-Hydroxy-7-[ 18F]fluorotryptophans as Candidate PET-Tracers for Pathway-Specific Visualization of Tryptophan Metabolism. Int J Mol Sci 2023; 24:15251. [PMID: 37894930 PMCID: PMC10607147 DOI: 10.3390/ijms242015251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/09/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
Tryptophan (Trp) is an essential proteinogenic amino acid and metabolic precursor for several signaling molecules that has been implicated in many physiological and pathological processes. Since the two main branches of Trp metabolism-serotonin biosynthesis and kynurenine pathway-are differently affected by a variety of neurological and neoplastic diseases, selective visualization of these pathways is of high clinical relevance. However, while positron emission tomography (PET) with existing probes can be used for non-invasive assessment of total Trp metabolism, optimal imaging agents for pathway-specific PET imaging are still lacking. In this work, we describe the preparation of two 18F-labeled Trp derivatives, NIn-methyl-6-[18F]fluorotryptophan (NIn-Me-6-[18F]FTrp) and 5-hydroxy-7-[18F]fluorotryptophan (5-HO-7-[18F]FTrp). We also report feasible synthetic routes for the preparation of the hitherto unknown boronate radiolabeling precursors and non-radioactive reference compounds. Under optimized conditions, alcohol-enhanced Cu-mediated radiofluorination of the respective precursors afforded NIn-Me-6-[18F]FTrp and 5-HO-7-[18F]FTrp as application-ready solutions in radiochemical yields of 45 ± 7% and 29 ± 4%, respectively. As such, our work provides access to two promising candidate probes for pathway-specific visualization of Trp metabolism in amounts sufficient for their preclinical evaluation.
Collapse
Affiliation(s)
- Niklas Kolks
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Str., 52428 Jülich, Germany; (N.K.); (F.N.); (B.D.Z.)
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Felix Neumaier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Str., 52428 Jülich, Germany; (N.K.); (F.N.); (B.D.Z.)
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Str., 52428 Jülich, Germany; (N.K.); (F.N.); (B.D.Z.)
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Boris D. Zlatopolskiy
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Str., 52428 Jülich, Germany; (N.K.); (F.N.); (B.D.Z.)
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
12
|
Monchaux de Oliveira C, Morael J, Guille A, Amadieu C, Vancassel S, Gaudout D, Capuron L, Pourtau L, Castanon N. Saffron extract interferes with lipopolysaccharide-induced brain activation of the kynurenine pathway and impairment of monoamine neurotransmission in mice. Front Nutr 2023; 10:1267839. [PMID: 37867499 PMCID: PMC10585275 DOI: 10.3389/fnut.2023.1267839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/05/2023] [Indexed: 10/24/2023] Open
Abstract
Background Although activation of inflammatory processes is essential to fight infections, its prolonged impact on brain function is well known to contribute to the pathophysiology of many medical conditions, including neuropsychiatric disorders. Therefore, identifying novel strategies to selectively counter the harmful effects of neuroinflammation appears as a major health concern. In that context, this study aimed to test the relevance of a nutritional intervention with saffron, a spice known for centuries for its beneficial effect on health. Methods For this purpose, the impact of an acute oral administration of a standardized saffron extract, which was previously shown to display neuromodulatory properties and reduce depressive-like behavior, was measured in mice challenged with lipopolysaccharide (LPS, 830 μg/kg, ip). Results Pretreatment with saffron extract (6.5 mg/kg, per os) did not reduce LPS-induced sickness behavior, preserving therefore this adaptive behavioral response essential for host defense. However, it interfered with delayed changes of expression of cytokines, chemokines and markers of microglial activation measured 24 h post-LPS treatment in key brain areas for behavior and mood control (frontal cortex, hippocampus, striatum). Importantly, this pretreatment also counteracted by that time the impact of LPS on several neurobiological processes contributing to inflammation-induced emotional alterations, in particular the activation of the kynurenine pathway, assessed through the expression of its main enzymes, as well as concomitant impairment of serotonergic and dopaminergic neurotransmission. Conclusion Altogether, this study provides important clues on how saffron extract interferes with brain function in conditions of immune stimulation and supports the relevance of saffron-based nutritional interventions to improve the management of inflammation-related comorbidities.
Collapse
Affiliation(s)
- Camille Monchaux de Oliveira
- INRAE, NutriNeuro, UMR 1286, Bordeaux University, Bordeaux IPB, Bordeaux, France
- Activ’Inside, Beychac-et-Caillau, France
| | - Jennifer Morael
- INRAE, NutriNeuro, UMR 1286, Bordeaux University, Bordeaux IPB, Bordeaux, France
| | - Alexandrine Guille
- INRAE, NutriNeuro, UMR 1286, Bordeaux University, Bordeaux IPB, Bordeaux, France
| | - Camille Amadieu
- INRAE, NutriNeuro, UMR 1286, Bordeaux University, Bordeaux IPB, Bordeaux, France
| | - Sylvie Vancassel
- INRAE, NutriNeuro, UMR 1286, Bordeaux University, Bordeaux IPB, Bordeaux, France
| | | | - Lucile Capuron
- INRAE, NutriNeuro, UMR 1286, Bordeaux University, Bordeaux IPB, Bordeaux, France
| | | | - Nathalie Castanon
- INRAE, NutriNeuro, UMR 1286, Bordeaux University, Bordeaux IPB, Bordeaux, France
| |
Collapse
|
13
|
Hüfner K, Vedova S, Tymoszuk P, Nelles P, Bruckner T, Deisenhammer EA, Egeter J, Galffy M, Giesinger JM, Lehmann J, Oberhammer M, Rockenschaub J, Sacher M, Holzner B, Gostner JM, Sperner-Unterweger B. The effect of inflammation, SARS-CoV-2 infection, age and mental health on serotonin, and kynurenine and catecholamine pathway metabolites. Psychoneuroendocrinology 2023; 156:106334. [PMID: 37481962 DOI: 10.1016/j.psyneuen.2023.106334] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/12/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND A high prevalence of mental disorders following COVID-19 has been described. It is therefore essential to elucidate underlying biological mechanisms linking SARS-CoV-2 infection and mental health. The kynurenine and catecholamine metabolic pathways are modulated by inflammation and can affect systemic levels of serotonin and dopamine. Their activity may hence link physical disorders with mental health. We investigated factors that affect kynurenine and catecholamine pathway activity in SARS-CoV-2 infection and recovery. METHODS The cross-sectional SIMMUN (n = 165) and longitudinal INCOV cohort (n = 167, Su et al. 2022) were analyzed. Demographic and clinical characteristic, inflammatory markers, SARS-CoV-2 infection, symptoms of depression and anxiety (HADS), and mental stress (PSS-4) served as explanatory variables. Blood serotonin and markers of kynurenine (kynurenine/tryptophan ratio), and catecholamine pathway activity (dopamine 3-O-sulfate, phenylalanine/tyrosine ratio) were modeled by multi-parameter linear regression. RESULTS In the SIMMUN cohort, the inflammatory marker neopterin (β = 0.47 [95% CI: 0.34-0.61]), SARS-CoV-2-positivity (0.42 [0.16-0.68]), mental stress (0.18 [0.055-0.31]), and age (0.26 [0.12-0.39]) were positively associated with the kynurenine/tryptophan ratio. The phenylalanine/tyrosine ratio was lower in SARS-CoV-2-positive than uninfected participants (-0.38 [-0.68 to -0.08]). In the INCOV cohort, markers of inflammation were associated with lower serotonin (IL6: -0.22 [-0.38 to -0.053]) and dopamine 3-O-sulfate levels (interferon-gamma: -0.15 [-0.26 to -0.036]). Serotonin (0.76 [0.34-1.2]) and dopamine 3-O-sulfate levels (0.63 [0.28-0.99]) were higher during recovery than in acute SARS-CoV-2 infection. CONCLUSION SARS-CoV-2 infection, inflammation, age and mental stress are key independent predictors of kynurenine pathway activity, which may influence serotonin availability. The catecholamine pathway was also affected in SARS-CoV-2 infection. Altered activity of these pathways may contribute to impaired mental health following COVID-19.
Collapse
Affiliation(s)
- Katharina Hüfner
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria.
| | - Sophia Vedova
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Philipp Nelles
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Tobias Bruckner
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Eberhard A Deisenhammer
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria; University Hospital of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Jonas Egeter
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Matyas Galffy
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Johannes M Giesinger
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Jens Lehmann
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Maria Oberhammer
- University Hospital of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Joachim Rockenschaub
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Magdalena Sacher
- Department of Visceral, Transplant and Thoracic Surgery (VTT), Medical University Innsbruck, Innsbruck, Austria
| | - Bernhard Holzner
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Johanna M Gostner
- Institute of Medical Biochemistry, Medical University Innsbruck, Innsbruck, Austria
| | - Barbara Sperner-Unterweger
- University Hospital of Psychiatry II, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
14
|
Wu B, Gan A, Wang R, Lin F, Yan T, Jia Y. Alpinia oxyphylla Miq. volatile oil ameliorates depressive behaviors and inhibits neuroinflammation in CUMS-exposed mice by inhibiting the TLR4-medicated MyD88/NF-κB signaling pathway. J Chem Neuroanat 2023; 130:102270. [PMID: 37001682 DOI: 10.1016/j.jchemneu.2023.102270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/01/2023]
Abstract
This study aimed to explore the antidepressant effect and underlying mechanism of the Alpinia oxyphylla Miq. volatile oil (AOVO) in mice exposed to chronic unpredictable mild stress (CUMS). C57BL/6 mice were grouped and administered with different dosages of AOVO (0.25, 0.50, 1.00, or 2.00 mL/kg body weight, i.g.), TAK242 (a TLR4 inhibitor, 0.75 mg/kg body weight, i.p.), or TAK242 (0.75 mg/kg body weight, i.p.) + AOVO (0.50 mL/kg body weight, i.g.) for 21 days. Depression-like symptoms in the mice were then evaluated through their body weight gain (BW), the open field test (OFT), the sucrose preference test (SPT), the novelty-suppressed feeding test (NSFT), and forced swimming test (FST). The concentrations of interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), and 5-hydroxytyrptamine (5-HT) in the mice were determined using ELISA kits. Hematoxylin and eosin (HE) dying were performed for histopathological examination. The expression of inflammatory proteins was assessed through western blotting (WB) and immunofluorescence staining. AOVO was found to improve the behavioral indexes of CUMS-exposed mice behavioral and synergize TAK242 to mitigate both their depressive symptoms and neuroinflammation. Moreover, AOVO was found to inhibit the hippocampal damage, decrease inflammatory cytokines (Reduced IL-1β, IL-6, and TNF-α by 19.97 %, 22.87 %, and 24.13 %, respectively), and downregulate the expression of TLR4/MyD88/NF-κB signaling pathway-related proteins in the hippocampus of CUMS-exposed mice (Reduced TLR4, MyD88, and NF-κB by 46.14 %, 42.48 %, and 38.08 %, respectively). These findings demonstrate that AOVO can ameliorate depressive behaviors and mitigate neuroinflammation in the CUMS-exposed mice via suppressing the TLR4-medicated MyD88/NF-κB signaling pathway.
Collapse
|
15
|
Corrigan M, O'Rourke A, Moran B, Fletcher J, Harkin A. Inflammation in the pathogenesis of depression: a disorder of neuroimmune origin. Neuronal Signal 2023; 7:NS20220054. [PMID: 37457896 PMCID: PMC10345431 DOI: 10.1042/ns20220054] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
There are several hypotheses concerning the underlying pathophysiological mechanisms of major depression, which centre largely around adaptive changes in neuronal transmission and plasticity, neurogenesis, and circuit and regional connectivity. The immune and endocrine systems are commonly implicated in driving these changes. An intricate interaction of stress hormones, innate immune cells and the actions of soluble mediators of immunity within the nervous system is described as being associated with the symptoms of depression. Bridging endocrine and immune processes to neurotransmission and signalling within key cortical and limbic brain circuits are critical to understanding depression as a disorder of neuroimmune origins. Emergent areas of research include a growing recognition of the adaptive immune system, advances in neuroimaging techniques and mechanistic insights gained from transgenic animals. Elucidation of glial-neuronal interactions is providing additional avenues into promising areas of research, the development of clinically relevant disease models and the discovery of novel therapies. This narrative review focuses on molecular and cellular mechanisms that are influenced by inflammation and stress. The aim of this review is to provide an overview of our current understanding of depression as a disorder of neuroimmune origin, focusing on neuroendocrine and neuroimmune dysregulation in depression pathophysiology. Advances in current understanding lie in pursuit of relevant biomarkers, as the potential of biomarker signatures to improve clinical outcomes is yet to be fully realised. Further investigations to expand biomarker panels including integration with neuroimaging, utilising individual symptoms to stratify patients into more homogenous subpopulations and targeting the immune system for new treatment approaches will help to address current unmet clinical need.
Collapse
Affiliation(s)
- Myles Corrigan
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Transpharmation Ireland, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Aoife M. O'Rourke
- School of Biochemistry and Immunology, Trinity Biosciences Institute, Trinity College, Dublin, Ireland
| | - Barry Moran
- School of Biochemistry and Immunology, Trinity Biosciences Institute, Trinity College, Dublin, Ireland
| | - Jean M. Fletcher
- School of Biochemistry and Immunology, Trinity Biosciences Institute, Trinity College, Dublin, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| |
Collapse
|
16
|
Piber D, Olmstead R, Cho JH, Guzman M, Irwin MR. Interferon- γ moderation of poor sleep maintenance and depressed mood in community-dwelling older adults. Psychol Med 2023; 53:3548-3556. [PMID: 35144705 DOI: 10.1017/s0033291722000113] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Depressive symptoms, such as depressed mood, are common in older adults and associated with an increased risk for morbidity and mortality. Given the evidence that sleep disturbance and alterations in interferon (IFN)-γ biology are associated with depression risk, this study examines the separate and joint contributions of poor sleep maintenance and IFN-γ to depressed mood in older adults. METHODS Community-dwelling, non-depressed older adults (n = 36, 72.1 ± 6.8 years) underwent a night of polysomnography to assess sleep maintenance [i.e. wake time after sleep onset (WASO)]. The morning after polysomnography, plasma levels of IFN-γ were evaluated along with self-reported depressed mood throughout the day. Multivariate linear regression tested associations of WASO and IFN-γ with the severity of depressed mood. In addition, moderation and mediation models examined the role of IFN-γ for the relationship between WASO and depressed mood. RESULTS A greater amount of WASO (p < 0.05) and higher levels of IFN-γ (p < 0.01) were both associated with the severity of depressed mood. Moreover, IFN-γ moderated the relationship between WASO and depressed mood (p < 0.01), such that WASO was more strongly related to the depressed mood among those with higher IFN-γ, than among those with lower IFN-γ. However, IFN-γ did not mediate the relationship between WASO and depressed mood. CONCLUSION In this study of older adults, poor sleep maintenance and higher levels of IFN-γ were both related to depressed mood. Moreover, IFN-γ moderated the relationship between poor sleep maintenance and depressed mood. Together, these findings suggest that older adults with higher IFN-γ are at heightened risk for depressive symptoms following sleep disturbance.
Collapse
Affiliation(s)
- Dominique Piber
- Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Psychiatry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Richard Olmstead
- Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Joshua H Cho
- Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Miguel Guzman
- Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Michael R Irwin
- Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Psychology, College of Arts and Sciences, University of California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Lambregts BIHM, Vassena E, Jansen A, Stremmelaar DE, Pickkers P, Kox M, Aarts E, van der Schaaf ME. Fatigue during acute systemic inflammation is associated with reduced mental effort expenditure while task accuracy is preserved. Brain Behav Immun 2023:S0889-1591(23)00131-9. [PMID: 37257522 DOI: 10.1016/j.bbi.2023.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/12/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Earlier work within the physical domain showed that acute inflammation changes motivational prioritization and effort allocation rather than physical abilities. It is currently unclear whether a similar motivational framework accounts for the mental fatigue and cognitive symptoms of acute sickness. Accordingly, this study aimed to assess the relationship between fatigue, cytokines and mental effort-based decision making during acute systemic inflammation. METHODS Eighty-five participants (41 males; 18-30 years (M=23.0, SD=2.4)) performed a mental effort-based decision-making task before, 2 hours after, and 5 hours after intravenous administration of 1 ng/kg bacterial lipopolysaccharide (LPS) to induce systemic inflammation. Plasma concentrations of cytokines (interleukin (IL)-6, IL-8 and tumor necrosis factor (TNF)) and fatigue levels were assessed at similar timepoints. In the task, participants decided whether they wanted to perform (i.e., 'accepted') arithmetic calculations of varying difficulty (3 levels: easy, medium, hard) in order to obtain rewards (3 levels: 5, 6 or 7 points). Acceptance rates were analyzed using a binomial generalized estimated equation (GEE) approach with effort, reward and time as independent variables. Arithmetic performance was measured per effort level prior to the decisions and included as a covariate. Associations between acceptance rates, fatigue (self-reported) and cytokine concentrations levels were analyzed using partial correlation analyses. RESULTS Plasma cytokine concentrations and fatigue were increased at 2 hours post-LPS compared to baseline and 5 hours post-LPS administration. Acceptance rates decreased for medium, but not for easy or hard effort levels at 2 hours post-LPS versus baseline and 5 hours post-LPS administration, irrespective of reward level. This reduction in acceptance rates occurred despite improved accuracy on the arithmetic calculations itself. Reduced acceptance rates for medium effort were associated with increased fatigue, but not with increased cytokines. CONCLUSION Fatigue during acute systemic inflammation is associated with alterations in mental effort allocation, similarly as observed previously for physical effort-based choice. Specifically, willingness to exert mental effort depended on effort and not reward information, while task accuracy was preserved. These results extend the motivational account of inflammation to the mental domain and suggest that inflammation may not necessarily affect domain-specific mental abilities, but rather affects domain-general effort-allocation processes.
Collapse
Affiliation(s)
- B I H M Lambregts
- Department of Psychiatry, Radboud University Medical Center Postbus 9101, 6500 HB Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Postbus 9104, HE Nijmegen, The Netherlands.
| | - E Vassena
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Postbus 9104, HE Nijmegen, The Netherlands; Experimental Psychopathology and Treatment, Behavioural Science Institute Radboud University Nijmegen Postbus 9104, 6500 HE Nijmegen, The Netherlands.
| | - A Jansen
- Department of Intensive Care Medicine, Radboud University Medical Center Postbus 9101, 6500 HB Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center Postbus 9101, 6500 HB Nijmegen, The Netherlands.
| | - D E Stremmelaar
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Postbus 9104, HE Nijmegen, The Netherlands.
| | - P Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center Postbus 9101, 6500 HB Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center Postbus 9101, 6500 HB Nijmegen, The Netherlands.
| | - M Kox
- Department of Intensive Care Medicine, Radboud University Medical Center Postbus 9101, 6500 HB Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center Postbus 9101, 6500 HB Nijmegen, The Netherlands.
| | - E Aarts
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Postbus 9104, HE Nijmegen, The Netherlands.
| | - M E van der Schaaf
- Department of Psychiatry, Radboud University Medical Center Postbus 9101, 6500 HB Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Postbus 9104, HE Nijmegen, The Netherlands; Department of Cognitive Neuropsychology, Tilburg University Postbus 90153, 5000 LE Tilburg, The Netherlands.
| |
Collapse
|
18
|
Mir HD, Giorgini G, Di Marzo V. The emerging role of the endocannabinoidome-gut microbiome axis in eating disorders. Psychoneuroendocrinology 2023; 154:106295. [PMID: 37229916 DOI: 10.1016/j.psyneuen.2023.106295] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Among the sources of chemical signals regulating food intake, energy metabolism and body weight, few have attracted recently as much attention as the expanded endocannabinoid system, or endocannabinoidome (eCBome), and the gut microbiome, the two systems on which this review article is focussed. Therefore, it is legitimate to expect that these two systems also play a major role in the etiopathology of eating disorders (EDs), in particular of anorexia nervosa, bulimia nervosa and binge-eating disorder. The major mechanisms through which, also via interactions with other endogenous signaling systems, the eCBome, with its several lipid mediators and receptors, and the gut microbiome, via its variety of microbial kingdoms, phyla and species, and armamentarium of metabolites, intervene in these disorders, are described here, based on several published studies in either experimental models or patients. Additionally, in view of the emerging multi-faceted cross-talk mechanisms between these two complex systems, we discuss the possibility that the eCBome-gut microbiome axis is also involved in EDs.
Collapse
Affiliation(s)
- Hayatte-Dounia Mir
- Centre de Recherche de l'Institut Universitaire de Pneumologie et Cardiologie (CRIUCPQ), Université Laval, Québec, Canada; Department of Medicine, Faculty of Medicine (FMED), Université Laval, Québec, Canada; Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, Canada
| | - Giada Giorgini
- Centre de Recherche de l'Institut Universitaire de Pneumologie et Cardiologie (CRIUCPQ), Université Laval, Québec, Canada; Department of Medicine, Faculty of Medicine (FMED), Université Laval, Québec, Canada; Unité Mixte Internationale en Recherche Chimique et Biomoléculaire sur le Microbiome et son Impact sur la Santé Métabolique et la Nutrition (UMI-MicroMeNu) entre l'Université Laval, Québec, Canada, et le Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry (ICB-CNR), Pozzuoli, Italy; Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, Canada
| | - Vincenzo Di Marzo
- Centre de Recherche de l'Institut Universitaire de Pneumologie et Cardiologie (CRIUCPQ), Université Laval, Québec, Canada; Department of Medicine, Faculty of Medicine (FMED), Université Laval, Québec, Canada; Unité Mixte Internationale en Recherche Chimique et Biomoléculaire sur le Microbiome et son Impact sur la Santé Métabolique et la Nutrition (UMI-MicroMeNu) entre l'Université Laval, Québec, Canada, et le Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry (ICB-CNR), Pozzuoli, Italy; Centre Nutrition, Santé et Société (NUTRISS), Université Laval, Québec, Canada; Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec, Canada; École de nutrition, Faculté des Sciences de l'agriculture et de l'alimentation (FSAA), Université Laval, Québec, Canada; Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, Canada.
| |
Collapse
|
19
|
Cavaleri D, Bartoli F, Capogrosso CA, Guzzi P, Moretti F, Riboldi I, Misiak B, Kishi T, Rubin RT, Fuchs D, Crocamo C, Carrà G. Blood concentrations of neopterin and biopterin in subjects with depression: A systematic review and meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110633. [PMID: 36089162 DOI: 10.1016/j.pnpbp.2022.110633] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 10/14/2022]
Abstract
INTRODUCTION Pteridines, such as neopterin, biopterin, and tetrahydrobiopterin (BH4), may be involved in depression pathophysiology owing to their links to immune-inflammatory response, oxidative and nitrosative stress, and monoaminergic transmission. Nonetheless, studies assessing pteridines in depression are inconsistent. We conducted a systematic review and meta-analysis of observational studies comparing blood pteridine concentrations between subjects with depression and healthy controls (HCs). METHODS We searched Embase, MEDLINE, and PsycInfo for articles indexed through November 2021. Study quality was appraised, evaluating age and gender comparability between groups, sample representativeness, and methods to assess depression. Random-effects meta-analyses were carried out, generating pooled standardized mean differences (SMDs). Heterogeneity across studies was estimated using the I2 statistic. RESULTS Twenty-four studies, involving 3075 subjects, were included. Individuals with depression showed blood neopterin concentrations higher than HCs (k = 19; SMD = 0.36; p < 0.001) with moderate heterogeneity across studies (I2 = 58.2%). No moderating role of age, gender, or type of blood sample was found. Sensitivity analyses showed no impact of inconsistency and quality of studies on findings. Neopterin concentrations were higher among individuals with major depressive disorder compared to HCs (SMD = 0.44; p < 0.001). This held true also when considering only drug-free subjects (SMD = 0.68; p = 0.003). No differences in biopterin concentrations were found between subjects with depression and HCs (k = 5; SMD = -0.35; p = 0.086), though this result was limited by inconsistency of findings (I2 = 77.9%) and quality of studies. Finally, no sufficient data were available for a meta-analysis on BH4. CONCLUSIONS As a whole, our work partly supports the hypothesis of an imbalance of pteridine metabolism in depression.
Collapse
Affiliation(s)
- Daniele Cavaleri
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Francesco Bartoli
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy.
| | - Chiara A Capogrosso
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Pierluca Guzzi
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Federico Moretti
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Ilaria Riboldi
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Błażej Misiak
- Department of Psychiatry, Division of Consultation Psychiatry and Neuroscience, Wroclaw Medical University, Pasteura 10 Street, 50-367 Wroclaw, Poland
| | - Taro Kishi
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
| | - Robert T Rubin
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Community Memorial Health System, Ventura County Medical Center, 147 N Brent St, Ventura, CA 93003, United States
| | - Dietmar Fuchs
- Institute of Biological Chemistry, Biocentre, Medical University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Cristina Crocamo
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Giuseppe Carrà
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy; Division of Psychiatry, University College London, Maple House 149, London W1T 7BN, United Kingdom
| |
Collapse
|
20
|
Amin NS, El Tayebi HM. More gain, less pain: How resistance training affects immune system functioning in multiple sclerosis patients: A review. Mult Scler Relat Disord 2023; 69:104401. [PMID: 36403379 DOI: 10.1016/j.msard.2022.104401] [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: 09/08/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
Multiple sclerosis (MS) is characterized by a complex etiology that is mirrored by the perplexing and inconsistent treatment responses observed across different patients. Although epigenetic research has garnered rightful interest in its efforts towards demystifying and understanding aberrant responses to treatment, the interim undoubtedly requires alternative non-pharmacological approaches towards attaining more effective management strategies. Of particular interest in this review is resistance training (RT) as a non-pharmacological exercise-based interventional strategy and its potential role as a disease-modifying tool. RT has been reported across literature to positively influence numerous aspects in the quality of life (QoL) and functional capacity of MS patients, and one of the attributes of these benefits may be a shift in the immune system of these individuals. RT has also been proven to affect different immune system key players associated with MS pathology. Ultimately, this brief review aims to provide a potential yet crucial link between RT, alterations in the expression profile of the immune system, and finally an imminent improvement in the overall well-being and QoL of MS patients, suggesting that utilizing RT as an interventional exercise modality may be an effective strategy that would aid in managing such a complex and debilitating disease.
Collapse
Affiliation(s)
- Nada Sherif Amin
- Department of Pharmacology and Toxicology, Molecular Pharmacology Research Group, Faculty of Pharmacy and Biotechnology, Head of Molecular Genetics and Pharmacology Research Group, German University in Cairo, Cairo 11835, Egypt
| | - Hend M El Tayebi
- Department of Pharmacology and Toxicology, Molecular Pharmacology Research Group, Faculty of Pharmacy and Biotechnology, Head of Molecular Genetics and Pharmacology Research Group, German University in Cairo, Cairo 11835, Egypt.
| |
Collapse
|
21
|
Nani JV, Almeida PGC, Noto C, Bressan RA, Brietzke E, Hayashi MAF. Unraveiling the correlation among neurodevelopmental and inflammatory biomarkers in patients with chronic schizophrenia. Nord J Psychiatry 2022; 76:559-564. [PMID: 36189960 DOI: 10.1080/08039488.2021.2023217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Nuclear distribution element like-1 (Ndel1) is a cytosolic oligopeptidase, which was suggested as a potential biomarker of aberrant neurodevelopment and early stage of schizophrenia (SCZ). The involvement of Ndel1 in neurite outgrowth, neuronal migration and neurodevelopment was demonstrated. Moreover, Ndel1 cleaves neuropeptides, including the endogenous antipsychotic peptide neurotensin, and lower Ndel1 activity was reported in SCZ patients compared with healthy controls (HCs). Changes in brain-derived neurotrophic factor (BDNF) and inflammatory cytokines levels were also implicated in SCZ. OBJECTIVE This preliminary study aimed to investigate the interactions between these immune and neurodevelopmental/neurotrophic biomarkers, namely BDNF and the recently identified SCZ biomarker Ndel1. RESULTS We observed lower Ndel1 activity and IL-4 levels, and higher BDNF levels, in plasma of SCZ (N = 23) compared with HCs (N = 29). Interestingly, significant correlation between Ndel1 activity and IL-4 levels was observed in SCZ, while no correlation with any other evaluated interleukins (namely IL-2, IL-8, IL-10 and IL-17A) or BDNF levels was noticed. CONCLUSION Although this hypothesis needs to be further explored for a better understanding of the mechanisms by which these altered pathways are associated to each other in SCZ, we suggest that Ndel1 and the inflammatory marker IL-4 are directly correlated.
Collapse
Affiliation(s)
- João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq), Sao Paulo, Brazil
| | - Priscila G C Almeida
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Cristiano Noto
- Department of Psychiatry, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Rodrigo A Bressan
- Department of Psychiatry, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Elisa Brietzke
- Department of Psychiatry, Queen's University School of Medicine, Kingston, Canada
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq), Sao Paulo, Brazil
| |
Collapse
|
22
|
Vancassel S, Fanet H, Castanon N, Monchaux De Oliveira C, Cussotto S, Capuron L. Tetrahydrobiopterin modulates the behavioral neuroinflammatory response to an LPS challenge in mice. Brain Behav Immun 2022; 105:139-148. [PMID: 35781010 DOI: 10.1016/j.bbi.2022.06.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022] Open
Abstract
Tetrahydrobiopterin (BH4) is a necessary cofactor for the synthesis of monoamines from essential amino-acids, phenylalanine, tyrosine and tryptophan. The BH4 synthesis pathway is induced by inflammatory factors but highly regulated processes maintain levels in a physiological range. However, BH4 activity can be durably altered in inflammation-related pathologies, such as certain types of depression, potentially involving impairment of dopaminergic neurotransmission. The purpose of this study was to investigate the response of the brain BH4 pathway to the inflammatory stimulus induced by lipopolysaccharide (LPS) in mice. Brain expression of genes related to BH4 synthesis, levels of BH4, changes in L-aromatic amino acid precursors of monoamines and dopamine levels were determined. As secondary aim, the effect of acute BH4 supply under the inflammatory challenge was tested on these parameters and on the expression of inflammatory cytokines. Mice were also submitted to the sucrose preference test and to the open-field in order to asses hedonic and locomotor responses to LPS, in addition to their modulation by BH4 supply. The LPS challenge resulted in decreased striatal DA levels and increased Phenylalanine/Tyrosine ratio, suggesting reduced BH4 activity. BH4 supply was effective to increase striatal BH4 levels, to restore the LPS-induced decreased in DA levels in striatum and to dampen the LPS-induced expression of inflammatory cytokines. At the behavioral level, BH4 supply was able to restore the loss of locomotor response to amphetamine in the LPS treated mice, suggesting a modulation of the dopaminergic neurotransmission. These data suggest that BH4 can be considered as a potential add-on molecule, helping to maintain or restore dopaminergic neurotransmission in neuroinflammatory conditions..
Collapse
Affiliation(s)
- S Vancassel
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France.
| | - H Fanet
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France; OptiNutriBrain, International Associated Laboratory (NutriNeuro France-INAF Canada), Quebec City, Canada
| | - N Castanon
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | | | - S Cussotto
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - L Capuron
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| |
Collapse
|
23
|
Emerging roles of brain metabolism in cognitive impairment and neuropsychiatric disorders. Neurosci Biobehav Rev 2022; 142:104892. [PMID: 36181925 DOI: 10.1016/j.neubiorev.2022.104892] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 08/18/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022]
Abstract
Here we discuss the role of diverse environmental manipulations affecting cognition with special regard to psychiatric conditions. We present evidence supporting a direct causal correlation between the valence of the environmental stimulation and some psychopathological traits and how the environment influences brain structure and function with special regard to oxidative stress and mitochondrial activity. Increasing experimental evidence supports a role for mitochondrial dysfunctions in neuropsychiatric disorders. Brain mitochondria are considered crucial mediators of allostasis, that is the capability to adapt to stress via a complex interaction between the autonomic, metabolic, and immune systems to maintain cellular homeostasis. In this process, mitochondria act as highly dynamic integrators by sensing and transducing stressors into adaptation mechanisms via metabolic stress mediators, such as glucocorticoids and catecholamines. Alterations in cellular homeostasis induced by chronic stress are thought to predispose to disease by triggering the so-called "mitochondrial allostatic load". This process is characterized by functional and structural changes of the mitochondria, ultimately leading to oxidative stress, inflammation, mitochondrial DNA damage and apoptosis. In this review we discuss the role of diverse environmental manipulations to affect cognition with special regard to psychiatric conditions. How the environment influences brain structure and function, and the interactions between rearing conditions, oxidative stress and mitochondrial activity are fundamental questions that are still poorly understood. As will be discussed, increasing experimental evidence supports a role for mitochondrial dysfunctions in neuropsychiatric disorders. Brain mitochondria are considered crucial mediators of allostasis, that is the capability to adapt to stress via a complex interaction between the autonomic, metabolic, and immune systems to maintain cellular homeostasis. In this process, mitochondria act as highly dynamic integrators by sensing and transducing stressors into adaptation mechanisms via metabolic stress mediators, such as glucocorticoids and catecholamines. Alterations in cellular homeostasis induced by chronic stress are thought to predispose to disease by triggering the so-called "mitochondrial allostatic load". This process is characterized by functional and structural changes of the mitochondria, ultimately leading to oxidative stress, inflammation, mitochondrial DNA damage and apoptosis. The brain requires considerable mitochondrial reserve not only to sustain basal neuronal needs but a also to provide increasing energy demands during stress. Consistently with these high energetic requirements, it is reasonable to hypothesise that the brain is particularly vulnerable to mitochondrial defects. Thus, even subtle metabolic alterations might have a substantial impact on cognitive functions. Over the last decade, several experimental evidence supported the hypothesis that a suboptimal mitochondrial function, which could be of genetic origin or acquired following adverse life events, is a key vulnerability factor for stress-related psychopathologies. Chronic psychological stress is a major promoter of anxiety as well as of oxidative damage, as shown in several studies. Recent evidence from mouse models harbouring mutations in mitochondrial genes demonstrated the role of mitochondria in modulating the response to acute psychological stress. However, it has yet to be determined whether mitochondrial dysfunctions are the cause or the consequence of anxiety. In this review, we discuss how adverse psychosocial environments can impact mitochondrial bioenergetics at the molecular level and we gather evidence from several studies linking energy metabolism and stress resilience/vulnerability. Moreover, we review recent findings supporting that metabolic dysfunction can underlie deficits in complex social behaviours. As will be discussed, aberrations in mitochondrial functionality have been found in the nucleus accumbens of highly anxious mice and mediate low social competitiveness. In addition, alterations in sociability can be reversed by enhancing mitochondrial functions. Recent evidence also demonstrated that a specific mutation in mitochondrial DNA, previously linked to autism spectrum disorder, produces autistic endophenotypes in mice by altering respiration chain and reactive oxygen species (ROS) production. Finally, we discuss a "Negative Enrichment" model that can explain some of the psychopathological conditions relevant to humans. Evidence of a direct causal correlation of valence of environmental stimulation and psychopathological traits will be presented, and possible molecular mechanisms that focus on oxidative stress. Collectively, the findings described here have been achieved with a wide set of behavioural and cognitive tasks with translational validity. Thus, they will be useful for future work aimed to elucidate the fine metabolic alterations in psychopathologies and devise novel approaches targeting mitochondria to alleviate these conditions.
Collapse
|
24
|
Severe psychiatric disorders and general medical comorbidities: inflammation-related mechanisms and therapeutic opportunities. Clin Sci (Lond) 2022; 136:1257-1280. [PMID: 36062418 DOI: 10.1042/cs20211106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022]
Abstract
Individuals with severe psychiatric disorders, such as mood disorders and schizophrenia, are at increased risk of developing other medical conditions, especially cardiovascular and metabolic diseases. These medical conditions are underdiagnosed and undertreated in these patients contributing to their increased morbidity and mortality. The basis for this increased comorbidity is not well understood, possibly reflecting shared risks factors (e.g. lifestyle risk factors), shared biological mechanisms and/or reciprocal interactions. Among overlapping pathophysiological mechanisms, inflammation and related factors, such as dysbiosis and insulin resistance, stand out. Besides underlying the association between psychiatric disorders and cardiometabolic diseases, these mechanisms provide several potential therapeutic targets.
Collapse
|
25
|
Zakaria FH, Samhani I, Mustafa MZ, Shafin N. Pathophysiology of Depression: Stingless Bee Honey Promising as an Antidepressant. Molecules 2022; 27:molecules27165091. [PMID: 36014336 PMCID: PMC9416360 DOI: 10.3390/molecules27165091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/30/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Depression is a debilitating psychiatric disorder impacting an individual’s quality of life. It is the most prevalent mental illness across all age categories, incurring huge socio-economic impacts. Most depression treatments currently focus on the elevation of neurotransmitters according to the monoamine hypothesis. Conventional treatments include tricyclic antidepressants (TCAs), norepinephrine–dopamine reuptake inhibitors (NDRIs), monoamine oxidase inhibitors (MAOIs), and serotonin reuptake inhibitors (SSRIs). Despite numerous pharmacological strategies utilising conventional drugs, the discovery of alternative medicines from natural products is a must for safer and beneficial brain supplement. About 30% of patients have been reported to show resistance to drug treatments coupled with functional impairment, poor quality of life, and suicidal ideation with a high relapse rate. Hence, there is an urgency for novel discoveries of safer and highly effective depression treatments. Stingless bee honey (SBH) has been proven to contain a high level of antioxidants compared to other types of honey. This is a comprehensive review of the potential use of SBH as a new candidate for antidepressants from the perspective of the monoamine, inflammatory and neurotrophin hypotheses.
Collapse
Affiliation(s)
- Fatin Haniza Zakaria
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
| | - Ismail Samhani
- Faculty of Medicine, Universiti Sultan Zainal Abidin (UniSZA), Medical Campus, Jalan Sultan Mahmud, Kuala Terengganu 20400, Malaysia
| | - Mohd Zulkifli Mustafa
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
- Correspondence: (M.Z.M.); (N.S.); Tel.: +609-7673000 (M.Z.M. & N.S.)
| | - Nazlahshaniza Shafin
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
- Correspondence: (M.Z.M.); (N.S.); Tel.: +609-7673000 (M.Z.M. & N.S.)
| |
Collapse
|
26
|
Sun Y, Wang Z, Hou J, Shi J, Tang Z, Wang C, Zhao H. Shuangxinfang Prevents S100A9-Induced Macrophage/Microglial Inflammation to Improve Cardiac Function and Depression-Like Behavior in Rats After Acute Myocardial Infarction. Front Pharmacol 2022; 13:832590. [PMID: 35814253 PMCID: PMC9263923 DOI: 10.3389/fphar.2022.832590] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/06/2022] [Indexed: 01/07/2023] Open
Abstract
Background: Depression is a common complication of cardiovascular disease, which deteriorates cardiac function. Shuangxinfang (psycho-cardiology formula, PCF) was reported to alleviate myocardial ischemia injury and improve depression-like behavior. Interestingly, our previous proteomics study predicted that the protein S100A9 appeared as an important target, and macrophage/microglial inflammation might be involved in the process of PCF improving depression induced by acute myocardial infarction (AMI). This study aims to validate the proteomics results. Methods: AMI rat models were established in vivo, followed by the administration of PCF or ABR-215757 (also named paquinimod, inhibiting S100A9 binding to TLR4) for 5 days. Forced swimming test (FST) and open field test (OFT) were applied to record depression-like behavior, and echocardiography was employed to evaluate cardiac function. Morphological changes of cardiomyocytes were assessed by HE staining and TUNEL staining on day 7 after cardiac surgery, as well as Masson trichrome staining on day 21. Hippocampal neurogenesis was determined by Nissl staining, while 5-hydroxytryptamine (5-HT), tryptophan/kynurenine ratio, and brain-derived neurotrophic factor (BDNF) in the hippocampus were analyzed as biochemical indicators of depression. We employed RT-qPCR, western blotting, and immunofluorescence to detect the expression of pathway-related genes and proteins. Myocardial and hippocampal expression of inflammatory factors were performed by ELISA. The activation of macrophage and microglia was assessed via immunoreaction using CD68 and Iba1, respectively. For in vitro confirmation, BV2 cells were primed with recombinant protein S100A9 and then treated with PCF serum or ferulic acid to determine alterations in microglial inflammation. Results: Rats in the AMI group showed heart function deterioration and depression-like behavior. Coronary ligation not only brought about myocardial inflammation, cell apoptosis, and fibrosis but also reduced the neurogenesis, elevated the tryptophan/kynurenine ratio, and decreased the content of 5-HT. PCF could ameliorate the pathological and phenotypic changes in the heart and brain and inhibit the expression of the S100A9 protein, the activation of the microglial cell, and the secretion of IL-1β and TNF-α raised by AMI. ABR-215757 showed therapeutic effect and molecular biological mechanisms similar to PCF. Treatment with PCF serum or ferulic acid in vitro was proved to efficiently block the hyperactivation of BV2 cells and increment of cytokine contents induced by recombinant protein S100A9. Conclusion: We identify S100A9 as a novel and potent regulator of inflammation in both the heart and brain. Macrophage/microglia inflammation mediated by S100A9 is considered a pivotal pathogenic in depression after AMI and a major pathway for the treatment of PCF, suggesting that PCF is a promising therapeutic candidate for psycho-cardiology disease.
Collapse
Affiliation(s)
- Yize Sun
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zheyi Wang
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiqiu Hou
- Oriental Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinyu Shi
- Oriental Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhuoran Tang
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wang
- Oriental Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Chao Wang, ; Haibin Zhao,
| | - Haibin Zhao
- Oriental Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Chao Wang, ; Haibin Zhao,
| |
Collapse
|
27
|
Catale C, Lo Iacono L, Martini A, Heil C, Guatteo E, Mercuri NB, Viscomi MT, Palacios D, Carola V. Early Life Social Stress Causes Sex- and Region-Dependent Dopaminergic Changes that Are Prevented by Minocycline. Mol Neurobiol 2022; 59:3913-3932. [PMID: 35435618 PMCID: PMC9148283 DOI: 10.1007/s12035-022-02830-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/02/2022] [Indexed: 02/03/2023]
Abstract
Early life stress (ELS) is known to modify trajectories of brain dopaminergic development, but the mechanisms underlying have not been determined. ELS perturbs immune system and microglia reactivity, and inflammation and microglia influence dopaminergic transmission and development. Whether microglia mediate the effects of ELS on dopamine (DA) system development is still unknown. We explored the effects of repeated early social stress on development of the dopaminergic system in male and female mice through histological, electrophysiological, and transcriptomic analyses. Furthermore, we tested whether these effects could be mediated by ELS-induced altered microglia/immune activity through a pharmacological approach. We found that social stress in early life altered DA neurons morphology, reduced dopamine transporter (DAT) and tyrosine hydroxylase expression, and lowered DAT-mediated currents in the ventral tegmental area but not substantia nigra of male mice only. Notably, stress-induced DA alterations were prevented by minocycline, an inhibitor of microglia activation. Transcriptome analysis in the developing male ventral tegmental area revealed that ELS caused downregulation of dopaminergic transmission and alteration in hormonal and peptide signaling pathways. Results from this study offer new insight into the mechanisms of stress response and altered brain dopaminergic maturation after ELS, providing evidence of neuroimmune interaction, sex differences, and regional specificity.
Collapse
Affiliation(s)
- Clarissa Catale
- Division of Experimental Neuroscience, Neurobiology of Behavior Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Luisa Lo Iacono
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, Rome, Italy
| | - Alessandro Martini
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Constantin Heil
- Division of Experimental Neuroscience, Epigenetics and Signal Transduction Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Ezia Guatteo
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Motor Science and Wellness, University of Naples Parthenope, Naples, Italy
| | - Nicola Biagio Mercuri
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Systems Medicine, Università Degli Studi Di Roma Tor Vergata, Rome, Italy
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health, Section of Histology and Embryology, Università Cattolica Del S. Cuore, Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Daniela Palacios
- Division of Experimental Neuroscience, Epigenetics and Signal Transduction Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
- Department of Life Science and Public Health, Section of Biology, Università Cattolica Del S. Cuore, Rome, Italy
| | - Valeria Carola
- Division of Experimental Neuroscience, Neurobiology of Behavior Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, Rome, Italy.
| |
Collapse
|
28
|
Wang W, Zheng Q, Yu C, Pan C, Luo P, Chen J. WSV056 Inhibits Shrimp Nitric Oxide Synthase Activity by Downregulating Litopenaeus vannamei Sepiapterin Reductase to Promote White Spot Syndrome Virus Replication. Front Microbiol 2022; 12:796049. [PMID: 35003027 PMCID: PMC8733705 DOI: 10.3389/fmicb.2021.796049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/19/2021] [Indexed: 02/05/2023] Open
Abstract
Sepiapterin reductase (Spr) plays an essential role in the biosynthesis of tetrahydrobiopterin (BH4), a key cofactor of multiple enzymes involved in various physiological and immune processes. Suppression of Spr could result in BH4 deficiency-caused diseases in human and murine models. However, information on the biological function of Spr in invertebrates is limited. In this study, two Sprs (CG12116 and Sptr) from Drosophila melanogaster were found to be downregulated in transgenic flies overexpressing white spot syndrome virus (WSSV) immediate-early protein WSV056. CG12116 and Sptr exerted an inhibitory effect on the replication of the Drosophila C virus. A Litopenaeus vannamei Spr (LvSpr) exhibiting similarity of 64.1–67.5% and 57.3–62.2% to that of invertebrate and vertebrate Sprs, respectively, were cloned. L. vannamei challenged with WSSV revealed a significant decrease in LvSpr transcription and Spr activity in hemocytes. In addition, the BH4 co-factored nitric oxide synthase (Nos) activity in shrimp hemocytes was reduced in WSSV-infected and LvSpr knockdown shrimp, suggesting WSSV probably inhibits the LvNos activity through LvSpr downregulation to limit the production of nitric oxide (NO). Knockdown of LvSpr and LvNos caused the reduction in NO level in hemocytes and the increase of viral copy numbers in WSSV-infected shrimp. Supplementation of NO donor DETA/NO or double gene knockdown of WSV056 + LvSpr and WSV056 + LvNos recovered the NO production, whereas the WSSV copy numbers were decreased. Altogether, the findings demonstrated that LvSpr and LvNos could potentially inhibit WSSV. In turn, the virus has evolved to attenuate NO production via LvSpr suppression by WSV056, allowing evasion of host antiviral response to ensure efficient replication.
Collapse
Affiliation(s)
- Wei Wang
- Institute of Oceanography, Minjiang University, Fuzhou, China
| | - Qin Zheng
- Institute of Oceanography, Minjiang University, Fuzhou, China
| | - Chen Yu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Changkun Pan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Luo
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Jianming Chen
- Institute of Oceanography, Minjiang University, Fuzhou, China
| |
Collapse
|
29
|
Macaluso M. L-Methylfolate in Antidepressant Non-responders: The Impact of Body Weight and Inflammation. Front Psychiatry 2022; 13:840116. [PMID: 35370812 PMCID: PMC8968318 DOI: 10.3389/fpsyt.2022.840116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/18/2022] [Indexed: 12/02/2022] Open
Abstract
This summary provides context for the role of L-methylfolate (LMF) in treating antidepressant non-responders. Bidirectional relationships have been observed between obesity and/or inflammation and depression. Studies have shown an increased prevalence of depression among patients with elevated body mass index and/or chronic inflammation and an increased risk of becoming obese and experiencing chronic inflammation in those with depression. These relationships can negatively affect the pathophysiology of depression. Elevated cytokine levels have been found to be among the factors that correlate with poor antidepressant treatment responsiveness. Low baseline neurotransmitter levels (e.g., serotonin) can also be associated with reduced effectiveness of commonly used antidepressants [e.g., selective serotonin reuptake inhibitors (SSRIs)]. LMF is an approved nutritional adjunctive antidepressant therapy that increases central neurotransmitter levels and thereby improves the effectiveness of antidepressant therapy. LMF can increase clinical response when used adjunctively in patients with major depressive disorder (MDD) and who are SSRI-resistant. In 2 randomized controlled trials, the pooled results showed increased response rates (32.3 vs. 14.6%; P = 0.04) as measured by a ≥50% reduction or final score ≤ 7 on the Hamilton Depression Rating Scale (HAM-D) and greater mean HAM-D reductions (-5.6 vs. -3.0; P = 0.05) when LMF was added to an SSRI compared with an SSRI plus placebo. Additionally, LMF has demonstrated effectiveness in real-world studies, with 67.9% of patients responding to therapy, using the 9-item Patient Health Questionnaire (P < 0.001). Post-hoc analyses found that patients with inflammation and/or obesity responded better to adjunctive LMF therapy compared with the overall sample (mean HAM-D reduction: -2.74 vs. +0.99).
Collapse
Affiliation(s)
- Matthew Macaluso
- Department of Psychiatry and Behavioral Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
30
|
Prevention of Stress-Induced Depressive-like Behavior by Saffron Extract Is Associated with Modulation of Kynurenine Pathway and Monoamine Neurotransmission. Pharmaceutics 2021; 13:pharmaceutics13122155. [PMID: 34959434 PMCID: PMC8709346 DOI: 10.3390/pharmaceutics13122155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 12/28/2022] Open
Abstract
Depressive disorders are a major public health concern. Despite currently available treatment options, their prevalence steadily increases, and a high rate of therapeutic failure is often reported, together with important antidepressant-related side effects. This highlights the need to improve existing therapeutic strategies, including by using nutritional interventions. In that context, saffron recently received particular attention for its beneficial effects on mood, although the underlying mechanisms are poorly understood. This study investigated in mice the impact of a saffron extract (Safr’Inside™; 6.25 mg/kg, per os) on acute restraint stress (ARS)-induced depressive-like behavior and related neurobiological alterations, by focusing on hypothalamic–pituitary–adrenal axis, inflammation-related metabolic pathways, and monoaminergic systems, all known to be altered by stress and involved in depressive disorder pathophysiology. When given before stress onset, Safr’Inside administration attenuated ARS-induced depressive-like behavior in the forced swim test. Importantly, it concomitantly reversed several stress-induced monoamine dysregulations and modulated the expression of key enzymes of the kynurenine pathway, likely reducing kynurenine-related neurotoxicity. These results show that saffron pretreatment prevents the development of stress-induced depressive symptoms and improves our understanding about the underlying mechanisms, which is a central issue to validate the therapeutic relevance of nutritional interventions with saffron in depressed patients.
Collapse
|
31
|
A new experimental design to study inflammation-related versus non-inflammation-related depression in mice. J Neuroinflammation 2021; 18:290. [PMID: 34895261 PMCID: PMC8666053 DOI: 10.1186/s12974-021-02330-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/24/2021] [Indexed: 12/28/2022] Open
Abstract
Background Major depressive disorder (MDD) represents a major public health concern, particularly due to its steadily rising prevalence and the poor responsiveness to standard antidepressants notably in patients afflicted with chronic inflammatory conditions, such as obesity. This highlights the need to improve current therapeutic strategies, including by targeting inflammation based on its role in the pathophysiology and treatment responsiveness of MDD. Nevertheless, dissecting the relative contribution of inflammation in the development and treatment of MDD remains a major issue, further complicated by the lack of preclinical depression models suitable to experimentally dissociate inflammation-related vs. inflammation-unrelated depression. Methods While current models usually focus on one particular MDD risk factor, we compared in male C57BL/6J mice the behavioral, inflammatory and neurobiological impact of chronic exposure to high-fat diet (HFD), a procedure known to induce inflammation-related depressive-like behaviors, and unpredictable chronic mild stress (UCMS), a stress-induced depression model notably renowned for its responsivity to antidepressants. Results While both paradigms induced neurovegetative, depressive-like and anxiety-like behaviors, inflammation and downstream neurobiological pathways contributing to inflammation-driven depression were specifically activated in HFD mice, as revealed by increased circulating levels of inflammatory factors, as well as brain expression of microglial activation markers and enzymes from the kynurenine and tetrahydrobiopterin (BH4) pathways. In addition, serotoninergic and dopaminergic systems were differentially impacted, depending on the experimental condition. Conclusions These data validate an experimental design suitable to deeply study the mechanisms underlying inflammation-driven depression comparatively to non-inflammatory depression. This design could help to better understand the pathophysiology of treatment resistant depression. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02330-9.
Collapse
|
32
|
Chen Y, Zhou YD, Laborda P, Wang HL, Wang R, Chen X, Liu FQ, Yang DJ, Wang SY, Shi XC, Laborda P. Mode of action and efficacy of quinolinic acid for the control of Ceratocystis fimbriata on sweet potato. PEST MANAGEMENT SCIENCE 2021; 77:4564-4571. [PMID: 34086397 DOI: 10.1002/ps.6495] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/27/2021] [Accepted: 06/04/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Ceratocystis fimbriata is a hazardous fungal pathogen able to cause black rot disease on sweet potato. The management of C. fimbriata strongly relies on the use of toxic fungicides, and there is a lack of efficient alternative strategies. RESULTS The antifungal properties of quinolinic acid (QA) were studied for the first time, indicating that QA shows selective antifungal activity against C. fimbriata. QA inhibited completely the mycelial growth of C. fimbriata at less than 0.8 mg mL-1 concentration (pH 4), and was able to produce alterations in the fungal cell wall, and to impede spore agglutination and mycelium formation. QA significantly reduced the concentration of ergosterol, and was able to associate to iron (II), suggesting that QA may be a lanosterol 14-α demethylase inhibitor. In preventive applications, QA reduced the disease incidence of C. fimbriata on sweet potato by 75%, achieving higher control efficacy in comparison with commercial fungicides prochloraz and carbendazim. CONCLUSIONS The first selective antifungal agent against C. fimbriata was discovered in this work, and showed suitable antifungal properties for the management of black rot disease. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ying Chen
- School of Life Sciences, Nantong University, Nantong, China
| | - Yi-Dong Zhou
- School of Life Sciences, Nantong University, Nantong, China
| | - Pablo Laborda
- Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Hai-Lin Wang
- School of Life Sciences, Nantong University, Nantong, China
| | - Rui Wang
- School of Life Sciences, Nantong University, Nantong, China
| | - Xian Chen
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Feng-Quan Liu
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Dong-Jing Yang
- Xuzhou Institute of Agricultural Sciences in Xuhuai District, Sweet Potato Research Institute, Xuzhou, China
| | - Su-Yan Wang
- School of Life Sciences, Nantong University, Nantong, China
| | - Xin-Chi Shi
- School of Life Sciences, Nantong University, Nantong, China
| | - Pedro Laborda
- School of Life Sciences, Nantong University, Nantong, China
| |
Collapse
|
33
|
Qiu W, Cai X, Zheng C, Qiu S, Ke H, Huang Y. Update on the Relationship Between Depression and Neuroendocrine Metabolism. Front Neurosci 2021; 15:728810. [PMID: 34531719 PMCID: PMC8438205 DOI: 10.3389/fnins.2021.728810] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/11/2021] [Indexed: 12/27/2022] Open
Abstract
Through the past decade of research, the correlation between depression and metabolic diseases has been noticed. More and more studies have confirmed that depression is comorbid with a variety of metabolic diseases, such as obesity, diabetes, metabolic syndrome and so on. Studies showed that the underlying mechanisms of both depression and metabolic diseases include chronic inflammatory state, which is significantly related to the severity. In addition, they also involve endocrine, immune systems. At present, the effects of clinical treatments of depression is limited. Therefore, exploring the co-disease mechanism of depression and metabolic diseases is helpful to find a new clinical therapeutic intervention strategy. Herein, focusing on the relationship between depression and metabolic diseases, this manuscript aims to provide an overview of the comorbidity of depression and metabolic.
Collapse
Affiliation(s)
- Wenxin Qiu
- Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaodan Cai
- Fujian Medical University, Fuzhou, Fujian, China
| | | | - Shumin Qiu
- Fujian Medical University, Fuzhou, Fujian, China
| | - Hanyang Ke
- Fujian Medical University, Fuzhou, Fujian, China
| | - Yinqiong Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
34
|
Maldonado-García JL, Pérez-Sánchez G, Becerril Villanueva E, Alvarez-Herrera S, Pavón L, Gutiérrez-Ospina G, López-Santiago R, Maldonado-Tapia JO, Pérez-Tapia SM, Moreno-Lafont MC. Behavioral and Neurochemical Shifts at the Hippocampus and Frontal Cortex Are Associated to Peripheral Inflammation in Balb/c Mice Infected with Brucella abortus 2308. Microorganisms 2021; 9:1937. [PMID: 34576830 PMCID: PMC8470318 DOI: 10.3390/microorganisms9091937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/15/2021] [Accepted: 09/01/2021] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is a zoonosis affecting 50,000,000 people annually. Most patients progress to a chronic phase of the disease in which neuropsychiatric symptoms upsurge. The biological processes underlying the progression of these symptoms are yet unclear. Peripheral inflammation mounted against Brucella may condition neurochemical shifts and hence unchained neuropsychiatric disorders. Our work aimed at establishing whether neurological, behavioral, and neurochemical disarrays are circumstantially linked to peripheral inflammation uprise secondary to Brucella abortus 2308 infections. We then evaluated, in control and Brucella-infected mice, skeletal muscle strength, movement coordination, and balance and motivation, as well as dopamine, epinephrine, norepinephrine, and serotonin availability in the cerebellum, frontal cortex, and hippocampus. Serum levels of proinflammatory cytokines and corticosterone in vehicle-injected and -infected mice were also estimated. All estimates were gathered at the infection acute and chronic phases. Our results showed that infected mice displayed motor disabilities, muscular weakness, and reduced motivation correlated with neurochemical and peripheral immunological disturbances that tended to decrease after 21 days of infection. The present observations support that disturbed peripheral inflammation and the related neurochemical disruption might lead to mood disorders in infected mice. Future experiments must be aimed at establishing causal links and to explore whether similar concepts might explain neurological and mood disorders in humans affected by brucellosis.
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 14370, Mexico; (J.L.M.-G.); (G.P.-S.); (E.B.V.); (S.A.-H.)
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (R.L.-S.); (J.O.M.-T.)
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 14370, Mexico; (J.L.M.-G.); (G.P.-S.); (E.B.V.); (S.A.-H.)
| | - Enrique Becerril Villanueva
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 14370, Mexico; (J.L.M.-G.); (G.P.-S.); (E.B.V.); (S.A.-H.)
| | - Samantha Alvarez-Herrera
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 14370, Mexico; (J.L.M.-G.); (G.P.-S.); (E.B.V.); (S.A.-H.)
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 14370, Mexico; (J.L.M.-G.); (G.P.-S.); (E.B.V.); (S.A.-H.)
| | - Gabriel Gutiérrez-Ospina
- Laboratorio de Biología de Sistemas, Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas y Coordinación de Psicobiología y Neurociencias, Facultad de Psicología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Rubén López-Santiago
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (R.L.-S.); (J.O.M.-T.)
| | - Jesús Octavio Maldonado-Tapia
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (R.L.-S.); (J.O.M.-T.)
| | - Sonia Mayra Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico;
| | - Martha C. Moreno-Lafont
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (R.L.-S.); (J.O.M.-T.)
| |
Collapse
|
35
|
Hüfner K, Giesinger JM, Gostner JM, Egeter J, Koudouovoh-Tripp P, Vill T, Fuchs D, Sperner-Unterweger B. Neurotransmitter Precursor Amino Acid Ratios Show Differential, Inverse Correlations with Depression Severity in the Low and High Depression Score Range. Int J Tryptophan Res 2021; 14:11786469211039220. [PMID: 34483668 PMCID: PMC8414612 DOI: 10.1177/11786469211039220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/20/2021] [Indexed: 12/28/2022] Open
Abstract
The immunomodulatory capacity of mental stress is one of the basic concepts of
psychoneuroimmunology. The current prospective longitudinal study was designed
to evaluate the effect of acute mental stress on neurotransmitter precursor
amino acid levels in individuals with depression at 2 time points. Ten
physically healthy patients with a diagnosis of major depressive episode and
Montgomery–Åsberg Depression Rating Scale scores (MADRAS) ⩾20 points at
inclusion were assessed on 2 study days (once with higher MADRAS scores, once
with lower MADRAS scores; median 34.5 days apart) and subjected to a
standardized acute mental stress test on each study day. Blood was collected at
4 time points: once prior to and at 3 time points (0, 30 minutes, 60 minutes)
following mental stress. Neurotransmitter precursor amino acid levels, that is
kynurenine/tryptophan (KYN/TRP) and phenylalanine/tyrosine (PHE/TYR), as well as
neopterin and nitrite were analyzed in a total of 80 individual blood samples.
Regression and correlation analyses were performed. Regression analyses of
PHE/TYR (R2 = .547) and KYN/TRP
(R2 = .440) in relation to MADRAS depression
severity showed a quadratic curve fit. This was reflected by a negative linear
correlation between MADRAS scores and PHE/TYR as well as KYN/TRP in the lower
score range (r = −.805, P < .001 and
r = −.586, P < .001 respectively) and a
positive correlation in the higher MADRAS score range
(r = .713, P < .001 and
r = .379, P = .016 respectively). No effect of
acute mental stress was found. This analysis exemplifies the implications of
sampling as well as data distributions on results. The crosstalk of biological
mechanisms that orchestrate metabolic and immunological signaling may vary
depending on depression severity resulting in non-linear associations that may
explain the heterogeneity of results found in the literature.
Collapse
Affiliation(s)
- Katharina Hüfner
- University Clinic for Psychiatry II,
Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University of
Innsbruck, Austria
- Katharina Hüfner, University Clinic for
Psychiatry II, Department of Psychiatry, Psychotherapy and Psychosomatics,
Medical University of Innsbruck, Anichstr. 35, Innsbruck 6020, Austria.
| | - Johannes M Giesinger
- University Clinic for Psychiatry II,
Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University of
Innsbruck, Austria
| | - Johanna M Gostner
- Institute of Medical Biochemistry,
Biocenter, Medical University of Innsbruck, Austria
| | - Jonas Egeter
- University Clinic for Psychiatry II,
Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University of
Innsbruck, Austria
| | - Pia Koudouovoh-Tripp
- University Clinic for Psychiatry II,
Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University of
Innsbruck, Austria
- Division of Psychiatry and
Psychotherapeutic Medicine, Hospital St. Vinzenz Zams, Austria
| | - Theresa Vill
- University Clinic for Psychiatry II,
Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University of
Innsbruck, Austria
| | - Dietmar Fuchs
- Institute of Biological Chemistry,
Biocenter, Medical University of Innsbruck, Austria
| | - Barbara Sperner-Unterweger
- University Clinic for Psychiatry II,
Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University of
Innsbruck, Austria
| |
Collapse
|
36
|
Mampay M, Flint MS, Sheridan GK. Tumour brain: Pretreatment cognitive and affective disorders caused by peripheral cancers. Br J Pharmacol 2021; 178:3977-3996. [PMID: 34029379 DOI: 10.1111/bph.15571] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/12/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
People that develop extracranial cancers often display co-morbid neurological disorders, such as anxiety, depression and cognitive impairment, even before commencement of chemotherapy. This suggests bidirectional crosstalk between non-CNS tumours and the brain, which can regulate peripheral tumour growth. However, the reciprocal neurological effects of tumour progression on brain homeostasis are not well understood. Here, we review brain regions involved in regulating peripheral tumour development and how they, in turn, are adversely affected by advancing tumour burden. Tumour-induced activation of the immune system, blood-brain barrier breakdown and chronic neuroinflammation can lead to circadian rhythm dysfunction, sleep disturbances, aberrant glucocorticoid production, decreased hippocampal neurogenesis and dysregulation of neural network activity, resulting in depression and memory impairments. Given that cancer-related cognitive impairment diminishes patient quality of life, reduces adherence to chemotherapy and worsens cancer prognosis, it is essential that more research is focused at understanding how peripheral tumours affect brain homeostasis.
Collapse
Affiliation(s)
- Myrthe Mampay
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Melanie S Flint
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Graham K Sheridan
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
37
|
Branchi I, Poggini S, Capuron L, Benedetti F, Poletti S, Tamouza R, Drexhage HA, Penninx BWJH, Pariante CM. Brain-immune crosstalk in the treatment of major depressive disorder. Eur Neuropsychopharmacol 2021; 45:89-107. [PMID: 33386229 DOI: 10.1016/j.euroneuro.2020.11.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/04/2020] [Accepted: 11/30/2020] [Indexed: 02/08/2023]
Abstract
A growing number of studies are pointing out the need for a conceptual shift from a brain-centered to a body-inclusive approach in mental health research. In this perspective, the link between the immune and the nervous system, which are deeply interconnected and continuously interacting, is one of the most important novel theoretical framework to investigate the biological bases of major depressive disorder and, more in general, mental illness. Indeed, depressed patients show high levels of inflammatory markers, administration of pro-inflammatory drugs triggers a depressive symptomatology and antidepressant efficacy is reduced by excessive immune system activation. A number of molecular and cellular mechanisms have been hypothesized to act as a link between the immune and brain function, thus representing potential pharmacologically targetable processes for the development of novel and effective therapeutic strategies. These include the modulation of the kynurenine pathway, the crosstalk between metabolic and inflammatory processes, the imbalance in acquired immune responses, in particular T cell responses, and the interplay between neural plasticity and immune system activation. In the personalized medicine approach, the assessment and regulation of these processes have the potential to lead, respectively, to novel diagnostic approaches for the prediction of treatment outcome according to the patient's immunological profile, and to improved efficacy of antidepressant compounds through immune modulation.
Collapse
Affiliation(s)
- Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Silvia Poggini
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Lucile Capuron
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Francesco Benedetti
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Sara Poletti
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Ryad Tamouza
- Département Medico-Universitaire de Psychiatrie et d'Addictologie (DMU ADAPT), Laboratoire Neuro-psychiatrie translationnelle, AP-HP, Université Paris Est Créteil, INSERM U955, IMRB, Hôpital Henri Mondor, Fondation FondaMental, F-94010 Créteil, France
| | - Hemmo A Drexhage
- Department of Immunology, ErasmusMC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC, Department of Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Carmine M Pariante
- Department of Psychological Medicine, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | -
- Département Medico-Universitaire de Psychiatrie et d'Addictologie (DMU ADAPT), Laboratoire Neuro-psychiatrie translationnelle, AP-HP, Université Paris Est Créteil, INSERM U955, IMRB, Hôpital Henri Mondor, Fondation FondaMental, F-94010 Créteil, France
| |
Collapse
|
38
|
Sen ZD, Danyeli LV, Woelfer M, Lamers F, Wagner G, Sobanski T, Walter M. Linking atypical depression and insulin resistance-related disorders via low-grade chronic inflammation: Integrating the phenotypic, molecular and neuroanatomical dimensions. Brain Behav Immun 2021; 93:335-352. [PMID: 33359233 DOI: 10.1016/j.bbi.2020.12.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Insulin resistance (IR) and related disorders, such as T2DM, increase the risk of major depressive disorder (MDD) and vice versa. Current evidence indicates that psychological stress and overeating can induce chronic low-grade inflammation that can interfere with glutamate metabolism in MDD as well as insulin signaling, particularly in the atypical subtype. Here we first review the interactive role of inflammatory processes in the development of MDD, IR and related metabolic disorders. Next, we describe the role of the anterior cingulate cortex in the pathophysiology of MDD and IR-related disorders. Furthermore, we outline how specific clinical features of atypical depression, such as hyperphagia, are more associated with inflammation and IR-related disorders. Finally, we examine the regional specificity of the effects of inflammation on the brain that show an overlap with the functional and morphometric brain patterns activated in MDD and IR-related disorders.
Collapse
Affiliation(s)
- Zümrüt Duygu Sen
- Department of Psychiatry and Psychotherapy, University Tuebingen, Calwerstraße 14, 72076 Tuebingen, Germany; Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany
| | - Lena Vera Danyeli
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Leipziger Str. 44, Building 65, 39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Marie Woelfer
- Clinical Affective Neuroimaging Laboratory (CANLAB), Leipziger Str. 44, Building 65, 39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Femke Lamers
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Oldenaller 1, 1081 HJ Amsterdam, the Netherlands
| | - Gerd Wagner
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany
| | - Thomas Sobanski
- Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, Thueringen-Kliniken "Georgius Agricola" GmbH, Rainweg 68, 07318 Saalfeld, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, University Tuebingen, Calwerstraße 14, 72076 Tuebingen, Germany; Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Leipziger Str. 44, Building 65, 39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany.
| |
Collapse
|
39
|
Fanet H, Capuron L, Castanon N, Calon F, Vancassel S. Tetrahydrobioterin (BH4) Pathway: From Metabolism to Neuropsychiatry. Curr Neuropharmacol 2021; 19:591-609. [PMID: 32744952 PMCID: PMC8573752 DOI: 10.2174/1570159x18666200729103529] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/03/2020] [Accepted: 07/23/2020] [Indexed: 11/22/2022] Open
Abstract
Tetrahydrobipterin (BH4) is a pivotal enzymatic cofactor required for the synthesis of serotonin, dopamine and nitric oxide. BH4 is essential for numerous physiological processes at periphery and central levels, such as vascularization, inflammation, glucose homeostasis, regulation of oxidative stress and neurotransmission. BH4 de novo synthesis involves the sequential activation of three enzymes, the major controlling point being GTP cyclohydrolase I (GCH1). Complementary salvage and recycling pathways ensure that BH4 levels are tightly kept within a physiological range in the body. Even if the way of transport of BH4 and its ability to enter the brain after peripheral administration is still controversial, data showed increased levels in the brain after BH4 treatment. Available evidence shows that GCH1 expression and BH4 synthesis are stimulated by immunological factors, notably pro-inflammatory cytokines. Once produced, BH4 can act as an anti- inflammatory molecule and scavenger of free radicals protecting against oxidative stress. At the same time, BH4 is prone to autoxidation, leading to the release of superoxide radicals contributing to inflammatory processes, and to the production of BH2, an inactive form of BH4, reducing its bioavailability. Alterations in BH4 levels have been documented in many pathological situations, including Alzheimer's disease, Parkinson's disease and depression, in which increased oxidative stress, inflammation and alterations in monoaminergic function are described. This review aims at providing an update of the knowledge about metabolism and the role of BH4 in brain function, from preclinical to clinical studies, addressing some therapeutic implications.
Collapse
Affiliation(s)
- H. Fanet
- INRAe, Nutrition and Integrated Neurobiology, UMR 1286, Bordeaux, France
- Université de Bordeaux, Nutrition and Integrated Neurobiology, UMR 1286, Bordeaux, France
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Neurosciences Axis, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- OptiNutriBrain International Associated Laboratory (NurtriNeuro France-INAF Canada), Quebec City, Canada
| | - L. Capuron
- INRAe, Nutrition and Integrated Neurobiology, UMR 1286, Bordeaux, France
- Université de Bordeaux, Nutrition and Integrated Neurobiology, UMR 1286, Bordeaux, France
- OptiNutriBrain International Associated Laboratory (NurtriNeuro France-INAF Canada), Quebec City, Canada
| | - N. Castanon
- INRAe, Nutrition and Integrated Neurobiology, UMR 1286, Bordeaux, France
- Université de Bordeaux, Nutrition and Integrated Neurobiology, UMR 1286, Bordeaux, France
- OptiNutriBrain International Associated Laboratory (NurtriNeuro France-INAF Canada), Quebec City, Canada
| | - F. Calon
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Neurosciences Axis, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- OptiNutriBrain International Associated Laboratory (NurtriNeuro France-INAF Canada), Quebec City, Canada
| | - S. Vancassel
- INRAe, Nutrition and Integrated Neurobiology, UMR 1286, Bordeaux, France
- Université de Bordeaux, Nutrition and Integrated Neurobiology, UMR 1286, Bordeaux, France
- OptiNutriBrain International Associated Laboratory (NurtriNeuro France-INAF Canada), Quebec City, Canada
| |
Collapse
|
40
|
Karshikoff B, Martucci KT, Mackey S. Relationship Between Blood Cytokine Levels, Psychological Comorbidity, and Widespreadness of Pain in Chronic Pelvic Pain. Front Psychiatry 2021; 12:651083. [PMID: 34248700 PMCID: PMC8267576 DOI: 10.3389/fpsyt.2021.651083] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/14/2021] [Indexed: 12/03/2022] Open
Abstract
Background: Low-grade inflammation has been implicated in the etiology of depression, long-term fatigue and chronic pain. TNFα and IL-6 are perhaps the most studied pro-inflammatory cytokines in the field of psychoneuroimmunology. The purpose of our study was to further investigate these relationships in patients with chronic pelvic pain specifically. Using plasma samples from a large, well-described cohort of patients with pelvic pain and healthy controls via the Multidisciplinary Approach to the Study of Chronic Pelvic Pain (MAPP) Research Network, we examined the relationship between TNFα and IL-6 and comorbid psychological symptoms. We also investigated the relationship between IL-8 and GM-CSF, and widespreadness of pain. Methods: We included baseline blood samples in the analyses, 261 patients (148 women) and 110 healthy controls (74 women). Fourteen pro- and anti-inflammatory or regulatory cytokines were analyzed in a Luminex® xMAP® high-sensitivity assay. We used regression models that accounted for known factors associated with the outcome variables to determine the relationship between cytokine levels and clinical measures. Results: There were no statistical differences in cytokine levels between patients and healthy controls when controlling for age. In patients, TNFα was significantly associated with levels of fatigue (p = 0.026), but not with pain intensity or depression. IL-6 was not significantly related to any of the outcome variables. Women with pelvic pain showed a negative relationship between IL-8 and widespreadness of pain, while men did not (p = 0.003). For both sexes, GM-CSF was positively related to widespreadness of pain (p = 0.039). Conclusion: Our results do not suggest low-grade systemic inflammation in chronic pelvic pain. Higher TNFα blood levels were related to higher fatigue ratings, while higher systemic GM-CSF levels predicted more widespread pain. Our study further suggests a potentially protective role of IL-8 with regard to with regard to the widepreadness of pain in the body, at least for women.
Collapse
Affiliation(s)
- Bianka Karshikoff
- Department of Clinical Neuroscience, Karolinska Institute, Solna, Sweden
| | - Katherine T Martucci
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC, United States
| | - Sean Mackey
- Division of Pain Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
41
|
Shilov YE, Miroshnichenko II. [Neopterin as a potential biomarker in neuropsychiatry]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:125-129. [PMID: 33244968 DOI: 10.17116/jnevro2020120101125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The importance of finding predictors or biomarkers of neuropsychiatric pathology, as well as methods of its prevention, treatment and early diagnosis is beyond question. The level of neopterin in body fluids is one of the possible biomarkers. Increased levels of neopterin in biological fluids (e.g., serum, cerebrospinal fluid or urine) are closely associated with various diseases associated with cellular immune response. The data presented in the review indicate the relevance of the study of neopterin concentrations in body fluids in patients with mental illness for the development of diagnostic and prognostic tests or as a pharmacodynamic marker of drug action.
Collapse
Affiliation(s)
- Yu E Shilov
- Mental Health Research Center, Moscow, Russia
| | | |
Collapse
|
42
|
The anti-inflammatory role of SSRI and SNRI in the treatment of depression: a review of human and rodent research studies. Inflammopharmacology 2020; 29:75-90. [PMID: 33164143 DOI: 10.1007/s10787-020-00777-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022]
Abstract
RATIONALE Depression has the topmost prevalence of all psychiatric diseases. It is characterized by a high recurrence rate, disability, and numerous and mostly unclear pathogenic mechanisms. Besides the monoamine or the neurotrophic hypothesis of depression, the inflammatory mechanism has begun to be supported by more and more evidence. At the same time, the current knowledge about the standard treatment of choice, the selective serotonin reuptake inhibitors (SSRIs) and serotonin and noradrenaline reuptake inhibitors (SNRIs), is expanding rapidly, adding more features to the initial ones. OBJECTIVES This review summarizes the in vivo anti-inflammatory effects of SSRIs and SNRIs in the treatment of depression and outlines the particular mechanisms of these effects for each drug separately. In addition, we provide an overview of the inflammation-related theory of depression and the underlying mechanisms. RESULTS SSRIs and SNRIs decrease the neuroinflammation through multiple mechanisms including the reduction of blood or tissue cytokines or regulating complex inflammatory pathways: nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), inflammasomes, Toll-like receptor 4 (TLR4), peroxisome proliferator-activated receptor gamma (PPARγ). Also, SSRIs and SNRIs show these effects in association with an antidepressant action. CONCLUSIONS SSRIs and SNRIs have an anti-neuroinflammatory role which might contribute the antidepressant effect.
Collapse
|
43
|
Coudert P, Rubat Coudert C. Les traitements de la dépression. ACTUALITES PHARMACEUTIQUES 2020. [DOI: 10.1016/j.actpha.2020.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Mir HD, Milman A, Monnoye M, Douard V, Philippe C, Aubert A, Castanon N, Vancassel S, Guérineau NC, Naudon L, Rabot S. The gut microbiota metabolite indole increases emotional responses and adrenal medulla activity in chronically stressed male mice. Psychoneuroendocrinology 2020; 119:104750. [PMID: 32569990 DOI: 10.1016/j.psyneuen.2020.104750] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/16/2020] [Accepted: 05/30/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS The gut microbiota produces metabolites that are an integral part of the metabolome and, as such, of the host physiology. Changes in gut microbiota metabolism could therefore contribute to pathophysiological processes. We showed previously that a chronic and moderate overproduction of indole from tryptophan in male individuals of the highly stress-sensitive F344 rat strain induced anxiety-like and helplessness behaviors. The aim of the present study was to extend the scope of these findings by investigating whether emotional behaviors of male mice that are moderately stress-sensitive but chronically exposed to environmental stressors would also be affected by indole. METHODS We colonized germ-free male C3H/HeN mice with a wild-type indole-producing Escherichia coli strain, or with the non-indole producing mutant. Gnotobiotic mice were subjected to an unpredictable chronic mild stress procedure, then to a set of tests aimed at assessing anxiety-like (novelty and elevated plus maze tests) and depression-like behaviors (coat state, splash, nesting, tail suspension and sucrose tests). Results of the individual tests were aggregated into a common z-score to estimate the overall emotional response to chronic mild stress and chronic indole production. We also carried out biochemical and molecular analyses in gut mucosa, plasma, brain hippocampus and striatum, and adrenal glands, to examine biological correlates that are usually associated with stress, anxiety and depression. RESULTS Chronic mild stress caused coat state degradation and anhedonia in both indole-producing and non-indole producing mice, but it did not influence behaviors in the other tests. Chronic indole production did not influence mice behavior when tests were considered individually, but it increased the overall emotionality z-score, specifically in mice under chronic mild stress. Interestingly, in the same mice, indole induced a dramatic increase of the expression of the adrenomedullary Pnmt gene, which is involved in catecholamine biosynthesis. By contrast, systemic tryptophan bioavailability, brain serotonin and dopamine levels and turnover, as well as expression of gut and brain genes involved in cytokine production and tryptophan metabolism along the serotonin and kynurenine pathways, remained similar in all mice. CONCLUSIONS Chronic indole production by the gut microbiota increased the vulnerability of male mice to the adverse effects of chronic mild stress on emotional behaviors. It also targeted catecholamine biosynthetic pathway of the adrenal medulla, which plays a pivotal role in body's physiological adaptation to stressful events. Future studies will aim to investigate the action mechanisms responsible for these effects.
Collapse
Affiliation(s)
- Hayatte-Dounia Mir
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France.
| | - Alexandre Milman
- IGF, Univ. Montpellier, CNRS, INSERM, 34000 Montpellier, France.
| | - Magali Monnoye
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France.
| | - Véronique Douard
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France.
| | - Catherine Philippe
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France.
| | - Agnès Aubert
- Université de Bordeaux, INRAE, UMR NutriNeurO, 33000 Bordeaux, France.
| | - Nathalie Castanon
- Université de Bordeaux, INRAE, UMR NutriNeurO, 33000 Bordeaux, France.
| | - Sylvie Vancassel
- Université de Bordeaux, INRAE, UMR NutriNeurO, 33000 Bordeaux, France.
| | | | - Laurent Naudon
- Université Paris-Saclay, INRAE, AgroParisTech, CNRS, Micalis Institute, 78350 Jouy-en-Josas, France.
| | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France.
| |
Collapse
|
45
|
Zlatopolskiy BD, Endepols H, Krasikova RN, Fedorova OS, Ermert J, Neumaier B. 11C- and 18F-labelled tryptophans as PET-tracers for imaging of altered tryptophan metabolism in age-associated disorders. RUSSIAN CHEMICAL REVIEWS 2020. [DOI: 10.1070/rcr4954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The ageing of the world’s population is the result of increased life expectancy observed in almost all countries throughout the world. Consequently, a rising tide of ageing-associated disorders, like cancer and neurodegenerative diseases, represents one of the main global challenges of the 21st century. The ability of mankind to overcome these challenges is directly dependent on the capability to develop novel methods for therapy and diagnosis of age-associated diseases. One hallmark of age-related pathologies is an altered tryptophan metabolism. Numerous pathological processes including neurodegenerative and neurological diseases like epilepsy, Parkinson’s and Alzheimer’s diseases, cancer and diabetes exhibit marked changes in tryptophan metabolism. Visualization of key processes of tryptophan metabolic pathways, especially using positron emission tomography (PET) and related hybrid methods like PET/CT and PET/MRI, can be exploited to early detect the aforementioned disorders with considerable accuracy, allowing appropriate and timely treatment of patients. Here we review the published 11C- and 18F-labelled tryptophans with respect to the production and also preclinical and clinical evaluation as PET-tracers for visualization of different branches of tryptophan metabolism.
The bibliography includes 159 references.
Collapse
|
46
|
Russo MA, Georgius P, Pires AS, Heng B, Allwright M, Guennewig B, Santarelli DM, Bailey D, Fiore NT, Tan VX, Latini A, Guillemin GJ, Austin PJ. Novel immune biomarkers in complex regional pain syndrome. J Neuroimmunol 2020; 347:577330. [PMID: 32731051 DOI: 10.1016/j.jneuroim.2020.577330] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023]
Abstract
We investigated serum levels of 29 cytokines and immune-activated kynurenine and tetrahydrobiopterin pathway metabolites in 15 complex regional pain syndrome (CRPS) subjects and 14 healthy controls. Significant reductions in interleukin-37 and tryptophan were found in CRPS subjects, along with positive correlations between kynurenine/tryptophan ratio and TNF-α levels with kinesiophobia, tetrahydrobiopterin levels with McGill pain score, sRAGE, and xanthurenic acid and neopterin levels with depression, anxiety and stress scores. Using machine learning, we identified a set of binary variables, including IL-37 and GM-CSF, capable of distinguishing controls from established CRPS subjects. These results suggest possible involvement of various inflammatory markers in CRPS pathogenesis.
Collapse
Affiliation(s)
- Marc A Russo
- Hunter Pain Specialists, 91 Chatham Street, Broadmeadow, NSW, 2292, Australia; Genesis Research Services, 220 Denison St, Broadmeadow, NSW, 2292, Australia
| | - Peter Georgius
- Pain Rehab, Suite 4 Noosa Central, 6 Bottlebrush Avenue, Sunshine Coast, QLD, 4567, Australia
| | - Ananda Staats Pires
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia; Laboratório de Bioenergética e Estresse Oxidativo, LABOX; Departamento de Bioquímica, CCB; Universidade Federal de Santa Catarina; Florianópolis / SC, Brazil
| | - Benjamin Heng
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia
| | - Michael Allwright
- ForeFront, Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | - Boris Guennewig
- ForeFront, Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | | | - Dominic Bailey
- Genesis Research Services, 220 Denison St, Broadmeadow, NSW, 2292, Australia
| | - Nathan T Fiore
- Discipline of Anatomy & Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| | - Vanessa X Tan
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia
| | - Alexandra Latini
- Laboratório de Bioenergética e Estresse Oxidativo, LABOX; Departamento de Bioquímica, CCB; Universidade Federal de Santa Catarina; Florianópolis / SC, Brazil
| | - Gilles J Guillemin
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia
| | - Paul J Austin
- Discipline of Anatomy & Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| |
Collapse
|
47
|
Swann OG, Kilpatrick M, Breslin M, Oddy WH. Dietary fiber and its associations with depression and inflammation. Nutr Rev 2020; 78:394-411. [PMID: 31750916 DOI: 10.1093/nutrit/nuz072] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dietary fiber is a crucial component of a healthy diet, with benefits that can be attributed to processes in the gut microbiota and the resulting by-products. Observational studies support associations between dietary fiber intake and depression and inflammation, but the potential mechanisms are poorly understood. This review examines evidence of the effects of dietary fiber on depression and inflammation and considers plausible mechanisms linking dietary fiber and depression, including microbiota-driven modification of gene expression and increased production of neurotransmitters. Additionally, inflammation may mediate the relationship between dietary fiber intake and depression. A high-fiber diet potentially lowers inflammation by modifying both the pH and the permeability of the gut. The resultant reduction in inflammatory compounds may alter neurotransmitter concentrations to reduce symptoms of depression. Further research into the link between dietary fiber intake and inflammation and depression is essential, as findings could potentially provide guidance for improvement in or prevention of inflammatory and depressive disorders.
Collapse
Affiliation(s)
- Olivia G Swann
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Michelle Kilpatrick
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Monique Breslin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Wendy H Oddy
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
48
|
Bazinet RP, Metherel AH, Chen CT, Shaikh SR, Nadjar A, Joffre C, Layé S. Brain eicosapentaenoic acid metabolism as a lead for novel therapeutics in major depression. Brain Behav Immun 2020; 85:21-28. [PMID: 31278982 DOI: 10.1016/j.bbi.2019.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
The results of several meta-analyses suggest that eicosapentaenoic acid (EPA) supplementation is therapeutic in managing the symptoms of major depression. It was previously assumed that because EPA is extremely low in the brain it did not cross the blood-brain barrier and any therapeutic effects it exerted would be via the periphery. However, more recent studies have established that EPA does enter the brain, but is rapidly metabolised following entry. While EPA does not accumulate within the brain, it is present in microglia and homeostatic mechanisms may regulate its esterification to phospholipids that serve important roles in cell signaling. Furthermore, a variety of signaling molecules from EPA have been described in the periphery and they have the potential to exert effects within the brain. If EPA is confirmed to be therapeutic in major depression as a result of adequately powered randomized clinical trials, future research on brain EPA metabolism could lead to the discovery of novel targets for treating or preventing major depression.
Collapse
Affiliation(s)
- Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Adam H Metherel
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Chuck T Chen
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, North Bethesda, MD 20852, United States
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Agnes Nadjar
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - Corinne Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - Sophie Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| |
Collapse
|
49
|
Cui Y, Cao K, Lin H, Cui S, Shen C, Wen W, Mo H, Dong Z, Bai S, Yang L, Shi Y, Zhang R. Early-Life Stress Induces Depression-Like Behavior and Synaptic-Plasticity Changes in a Maternal Separation Rat Model: Gender Difference and Metabolomics Study. Front Pharmacol 2020; 11:102. [PMID: 32174832 PMCID: PMC7055479 DOI: 10.3389/fphar.2020.00102] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/28/2020] [Indexed: 12/18/2022] Open
Abstract
More than 300 million people suffer from depressive disorders globally. People under early-life stress (ELS) are reportedly vulnerable to depression in their adulthood, and synaptic plasticity can be the molecular mechanism underlying such depression. Herein, we simulated ELS by using a maternal separation (MS) model and evaluated the behavior of Sprague-Dawley (SD) rats in adulthood through behavioral examination, including sucrose preference, forced swimming, and open-field tests. The behavior tests showed that SD rats in the MS group were more susceptible to depression- and anxiety-like behaviors than did the non-MS (NMS) group. Nissl staining analysis indicated a significant reduction in the number of neurons at the prefrontal cortex and hippocampus, including the CA1, CA2, CA3, and DG regions of SD rats in the MS group. Immunohistochemistry results showed that the percentages of synaptophysin-positive area in the prefrontal cortex and hippocampus (including the CA1, CA2, CA3, and DG regions) slice of the MS group significantly decreased compared with those of the NMS group. Western blot analysis was used to assess synaptic-plasticity protein markers, including postsynaptic density 95, synaptophysin, and growth-associated binding protein 43 protein expression in the cortex and hippocampus. Results showed that the expression levels of these three proteins in the MS group were significantly lower than those in the NMS group. LC-MS/MS analysis revealed no significant differences in the peak areas of sex hormones and their metabolites, including estradiol, testosterone, androstenedione, estrone, estriol, and 5β-dihydrotestosterone. Through the application of nontargeted metabolomics to the overall analysis of differential metabolites, pathway-enrichment results showed the importance of arginine and proline metabolism; pantothenate and CoA biosyntheses; glutathione metabolism; and the phenylalanine, tyrosine, and tryptophan biosynthesis pathways. In summary, the MS model caused adult SD rats to be susceptible to depression, which may regulate synaptic plasticity through arginine and proline metabolism; pantothenate and CoA biosyntheses; glutathione metabolism; and phenylalanine, tyrosine, and tryptophan biosyntheses.
Collapse
Affiliation(s)
- Yongfei Cui
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kerun Cao
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiyuan Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sainan Cui
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chongkun Shen
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhao Wen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haixin Mo
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhaoyang Dong
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yafei Shi
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
50
|
Almeida PG, Nani JV, Oses JP, Brietzke E, Hayashi MA. Neuroinflammation and glial cell activation in mental disorders. Brain Behav Immun Health 2020; 2:100034. [PMID: 38377429 PMCID: PMC8474594 DOI: 10.1016/j.bbih.2019.100034] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 02/07/2023] Open
Abstract
Mental disorders (MDs) are highly prevalent and potentially debilitating complex disorders which causes remain elusive. Looking into deeper aspects of etiology or pathophysiology underlying these diseases would be highly beneficial, as the scarce knowledge in mechanistic and molecular pathways certainly represents an important limitation. Association between MDs and inflammation/neuroinflammation has been widely discussed and accepted by many, as high levels of pro-inflammatory cytokines were reported in patients with several MDs, such as schizophrenia (SCZ), bipolar disorder (BD) and major depression disorder (MDD), among others. Correlation of pro-inflammatory markers with symptoms intensity was also reported. However, the mechanisms underlying the inflammatory dysfunctions observed in MDs are not fully understood yet. In this context, microglial dysfunction has recently emerged as a possible pivotal player, as during the neuroinflammatory response, microglia can be over-activated, and excessive production of pro-inflammatory cytokines, which can modify the kynurenine and glutamate signaling, is reported. Moreover, microglial activation also results in increased astrocyte activity and consequent glutamate release, which are both toxic to the Central Nervous System (CNS). Also, as a result of increased microglial activation in MDs, products of the kynurenine pathway were shown to be changed, influencing then the dopaminergic, serotonergic, and glutamatergic signaling pathways. Therefore, in the present review, we aim to discuss how neuroinflammation impacts on glutamate and kynurenine signaling pathways, and how they can consequently influence the monoaminergic signaling. The consequent association with MDs main symptoms is also discussed. As such, this work aims to contribute to the field by providing insights into these alternative pathways and by shedding light on potential targets that could improve the strategies for pharmacological intervention and/or treatment protocols to combat the main pharmacologically unmatched symptoms of MDs, as the SCZ.
Collapse
Key Words
- AMPA, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
- APCs, antigen presenting cells
- BBB, blood-brain barrier
- BD, bipolar disorder
- CCL, C–C motif chemokine ligand
- CLRs, C-type lectin receptors
- CNS, central nervous system
- CSF, cerebrospinal fluid
- CXCL, X–C motif chemokine ligand
- Glia
- IDO, indolamine 2,3-dioxygenase
- IFN, interferon
- IL, interleukin
- IRF, interferon regulatory factor
- Inflammation
- KYNA, kynurenic acid
- MD, mental disorders
- MDD, major depression disorder
- MRI, magnetic resonance imaging
- Mental disorders
- Microglial activation
- NF, necrosis factor
- NMDA, N-methyl-D-aspartate
- NMR, nuclear magnetic resonance
- PPI, prepulse inhibition
- PRRs, pattern recognition receptors
- QUIN, quinolinic acid
- SCZ, schizophrenia
- Schizophrenia
- TGF, tumor growth factor
- TLRs, toll-like receptors
- TNF, tumor necrosis factor
- α7-nAchR, alpha 7 nicotinic acetylcholine receptor
Collapse
Affiliation(s)
- Priscila G.C. Almeida
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - João Victor Nani
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Jean Pierre Oses
- Programa Multicêntrico de Pós-Graduação em Bioquímica e Biologia Molecular, Instituto de Biociências, Universidade Federal do Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Elisa Brietzke
- Department of Psychiatry, Queen’s University School of Medicine, Kingston, ON, Canada
| | - Mirian A.F. Hayashi
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| |
Collapse
|