1
|
Sepúlveda-Cuéllar RD, Soria-Medina DA, Cañedo-Solares I, Gómez-Chávez F, Molina-López LM, Cruz-Martínez MY, Correa D. Controversies and insights into cytokine regulation of neurogenesis and behavior in adult rodents. Front Immunol 2025; 16:1550660. [PMID: 40352932 PMCID: PMC12061686 DOI: 10.3389/fimmu.2025.1550660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
Adult learning, memory, and social interaction partially depend on neurogenesis in two regions: the hippocampus and the subventricular zone. There is evidence that the immune system is important for these processes in pathological situations, but there is no review of its role in non-pathological or near-physiological conditions. Although further research is warranted in this area, some conclusions can be drawn. Intrusive LyC6hi monocytes and autoreactive CD4+ T cells have a positive impact on neurogenesis and behavior, but the latter are deleterious if specific to external antigens. Mildly activated microglia play a crucial role in promoting these processes, by eliminating apoptotic neuronal progenitors and producing low levels of interleukins, which increase if the cells are activated, leading to inhibition of neurogenesis. Chemokines are poorly studied, but progenitor cells and neurons express their receptors, which appear important for migration and maturation. The few works that jointly analyzed neurogenesis and behavior showed congruent effects of immune cells and cytokines. In conclusion, the immune system components -mostly local- seem of utmost importance for the control of behavior under non-pathological conditions.
Collapse
Affiliation(s)
- Rodrigo Daniel Sepúlveda-Cuéllar
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| | - Diego Alberto Soria-Medina
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
- Facultad de Psicología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Irma Cañedo-Solares
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría (INP), Secretaría de Salud, Ciudad de México, Mexico
| | - Fernando Gómez-Chávez
- Laboratorio de Enfermedades Osteoarticulares e Inmunológicas, Sección de Estudios de Posgrado e Investigación, Escuela Nacional de Medicina y Homeopatía (ENMyH), Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| | - Liliana Monserrat Molina-López
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| | - María Yolanda Cruz-Martínez
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| | - Dolores Correa
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| |
Collapse
|
2
|
Taib B, Deme P, Gupta S, Yoo SW, Khuder SS, Hoke A, Li Z, Ahima RS, Haughey NJ. Insulin acts on astrocytes to shift their substrate preference to fatty acids. iScience 2025; 28:111642. [PMID: 40201123 PMCID: PMC11978350 DOI: 10.1016/j.isci.2024.111642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 12/17/2024] [Indexed: 04/10/2025] Open
Abstract
It is increasingly recognized that brain can β-oxidize fatty acids for use as an energy substrate. However, mechanism(s) by which neural cells switch their preference from glucose to fatty acids are not fully elucidated. Here we provide evidence that insulin acts directly on astrocytes to promote the uptake of glucose and fatty acids while modifying their substrate preference through a sequential shift in the expression of genes associated with fatty acid uptake, synthesis, transport, and metabolism. Under these conditions, fatty acids are converted into TCA cycle intermediates to satisfy astrocyte energy demands, allowing pyruvate derived from glucose to be directed toward the production of lactate; a preferred fuel for neurons. This shift in astrocyte energy substrate preference is required for insulin to enhance long-term potentiation in the Schaffer collateral. These findings establish a homeostatic mechanism where insulin promotes LTP by switching the energy substrate preference of astrocytes to fatty acids.
Collapse
Affiliation(s)
- Bouchra Taib
- The Johns Hopkins University School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Baltimore, MD, USA
- Institute of Sport Professions (IMS), Ibn Tofail University, Kenitra, Morocco
| | - Pragney Deme
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | - Sujasha Gupta
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | - Seung Wan Yoo
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | - Saja S. Khuder
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | - Ahmet Hoke
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
- The Johns Hopkins University School of Medicine, Department of Neuromuscular Division, and Merkin Peripheral Neuropathy and Nerve Regeneration Center, Baltimore, MD, USA
| | - Zhigang Li
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | - Rexford S. Ahima
- The Johns Hopkins University School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Baltimore, MD, USA
| | - Norman J. Haughey
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
- The Johns Hopkins University School of Medicine, Department of Psychiatry, Baltimore, MD, USA
- Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
3
|
Kovacs M, Dominguez-Belloso A, Ali-Moussa S, Deczkowska A. Immune control of brain physiology. Nat Rev Immunol 2025:10.1038/s41577-025-01129-6. [PMID: 39890999 DOI: 10.1038/s41577-025-01129-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/03/2025]
Abstract
The peripheral immune system communicates with the brain through complex anatomical routes involving the skull, the brain borders, circumventricular organs and peripheral nerves. These immune-brain communication pathways were classically considered to be dormant under physiological conditions and active only in cases of infection or damage. Yet, peripheral immune cells and signals are key in brain development, function and maintenance. In this Perspective, we propose an alternative framework for understanding the mechanisms of immune-brain communication. During brain development and in homeostasis, these anatomical structures allow selected elements of the peripheral immune system to affect the brain directly or indirectly, within physiological limits. By contrast, in ageing and pathological settings, detrimental peripheral immune signals hijack the existing communication routes or alter their structure. We discuss why a diversity of communication channels is needed and how they work in relation to one another to maintain homeostasis of the brain.
Collapse
Affiliation(s)
- Mariángeles Kovacs
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, Paris, France
| | - Amaia Dominguez-Belloso
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, Paris, France
| | - Samir Ali-Moussa
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, Paris, France
| | - Aleksandra Deczkowska
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, Paris, France.
| |
Collapse
|
4
|
Wang H, Yang J, Sun Z, Nie Y, He Y. Neoprzewaquinone A alters the migration, phagocytosis and energy metabolism of IL-15-induced HMC3 cells. Mol Immunol 2024; 174:11-17. [PMID: 39128414 DOI: 10.1016/j.molimm.2024.07.012] [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: 06/16/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024]
Abstract
Microglia play a major role in the immune defense system of the central nervous system and are activated in many neurological diseases. The immunomodulatory cytokine interleukin (IL)-15 is known to be involved in microglia response and inflammatory factors release. Neoprzewaquinone A (NEO) is an active compound isolated from Salvia miltiorrhiza Bunge. Our previous study has shown that NEO significantly inhibit the proliferation of IL-15-treated Mo7e cells. However, the role of NEO in the structure and function of IL-15-treated human microglial cells (HMC3) remains unclear. Thus, our study aimed to quantitatively analyze the beneficial effects of NEO on HMC3 cells following IL-15 treatment. The cell viability, phagocytosis, migration and energy metabolism were evaluated by Cell Counting Kit-8 (CCK8), scratch assay, pHrodo™ Red Zymosan BioParticles™ Conjugate, and Agilent Seahorse XF Cell Mito Test. Cephalothin (CEP) was selected as a positive drug because it has obvious inhibitory effect on IL-15 and IL-15Rɑ. Our results showed that IL-15 stimulated the proliferation, migration and phagocytosis of HMC3 cells in a time-dependent manner. Interestingly, NEO exhibited significant suppressive effects on these IL-15-induced changes, which were even superior to those observed with the CEP. Moreover, IL-15 treatment did not significantly alter energy metabolism, including glycolysis and mitochondrial respiration. NEO and CEP alone effectively reduced glycolysis, non-mitochondrial respiration, basal respiration, ATP turnover, respiration capacity, and H+ leak in HMC3 cells. Furthermore, NEO displayed a partial regulatory effect on mitochondrial function in IL-15-treated HMC3 cells. Our study confirms the effectively inhibition of NEO on IL-15-induced microglial activation and provides valuable insights into the therapeutic prospects of NEO in neuropsychiatric disorders associated with IL-15 and microglia.
Collapse
Affiliation(s)
- Haixia Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Jian Yang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Yadan Nie
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Yi He
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
5
|
Di Castro MA, Garofalo S, Mormino A, Carbonari L, Di Pietro E, De Felice E, Catalano M, Maggi L, Limatola C. Interleukin-15 alters hippocampal synaptic transmission and impairs episodic memory formation in mice. Brain Behav Immun 2024; 115:652-666. [PMID: 37992787 DOI: 10.1016/j.bbi.2023.11.015] [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: 07/14/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/24/2023] Open
Abstract
Cytokines are potent immunomodulators exerting pleiotropic effects in the central nervous system (CNS). They influence neuronal functions and circuit activities with effects on memory processes and behaviors. Here, we unravel a neuromodulatory activity of interleukin-15 (IL-15) in mouse brain. Acute exposure of hippocampal slices to IL-15 enhances gamma-aminobutyricacid (GABA) release and reduces glutamatergic currents, while chronic treatment with IL-15 increases the frequency of hippocampal miniature inhibitory synaptic transmission and impairs memory formation in the novel object recognition (NOR) test. Moreover, we describe that serotonin is involved in mediating the hippocampal effects of IL-15, because a selective 5-HT3A receptor antagonist prevents the effects on inhibitory neurotransmission and ameliorates mice performance in the NOR test. These findings provide new insights into the modulatory activities of cytokines in the CNS, with implications on behavior.
Collapse
Affiliation(s)
- Maria Amalia Di Castro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Alessandro Mormino
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Laura Carbonari
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Erika Di Pietro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Eleonora De Felice
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Cristina Limatola
- IRCCS Neuromed Via Atinese 18, 86077 Pozzilli, Italy; Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur, Italy.
| |
Collapse
|
6
|
Haddad FL, Patel SV, Doornaert EE, De Oliveira C, Allman BL, Baines KJ, Renaud SJ, Schmid S. Interleukin 15 modulates the effects of poly I:C maternal immune activation on offspring behaviour. Brain Behav Immun Health 2022; 23:100473. [PMID: 35668725 PMCID: PMC9166394 DOI: 10.1016/j.bbih.2022.100473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 01/21/2023] Open
Abstract
Maternal infections during pregnancy are linked with an increased risk for disorders like Autism Spectrum Disorder and schizophrenia in the offspring. Although precise mechanisms are still unclear, clinical and preclinical evidence suggest a strong role for maternal immune activation (MIA) in the neurodevelopmental disruptions caused by maternal infection. Previously, studies using the Polyinosinic:Polycytidylic (Poly I:C) MIA preclinical model showed that cytokines like Interleukin 6 (Il6) are important mediators of MIA's effects. In this study, we hypothesized that Il15 may similarly act as a mediator of Poly I:C MIA, given its role in the antiviral immune response. To test this hypothesis, we induced Poly I:C MIA at gestational day 9.5 in wildtype (WT) and Il15−/− rat dams and tested their offspring in adolescence and adulthood. Poly I:C MIA and Il15 knockout produced both independent and synergistic effects on offspring behaviour. Poly I:C MIA decreased startle reactivity in adult WT offspring but resulted in increased adolescent anxiety and decreased adult locomotor activity in Il15−/− offspring. In addition, Poly I:C MIA led to genotype-independent effects on locomotor activity and prepulse inhibition. Finally, we showed that Il15−/− offspring exhibit distinct phenotypes that were unrelated to Poly I:C MIA including altered startle reactivity, locomotion and signal transduction in the auditory brainstem. Overall, our findings indicate that the lack of Il15 can leave offspring either more or less susceptible to Poly I:C MIA, depending on the phenotype in question. Future studies should examine the contribution of fetal versus maternal Il15 in MIA to determine the precise developmental mechanisms underlying these changes. Poly I:C MIA decreases startle reactivity in adult WT but not Il15−/− offspring. Il15−/− offspring exposed to Poly I:C MIA show altered PPI and open field exploration. Il15−/− rats exhibit distinct behavioural phenotypes independent from MIA.
Collapse
|
7
|
He Y, Bo Q, Mao Z, Yang J, Liu M, Wang H, Kastin AJ, Pan W, Wang C, Sun Z. Reduced Serum Levels of Soluble Interleukin-15 Receptor α in Schizophrenia and Its Relationship to the Excited Phenotype. Front Psychiatry 2022; 13:842003. [PMID: 35356722 PMCID: PMC8959406 DOI: 10.3389/fpsyt.2022.842003] [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: 12/23/2021] [Accepted: 02/16/2022] [Indexed: 12/02/2022] Open
Abstract
Our previous studies documented that interleukin-15 receptor α (IL-15Rα) knockout (KO) mice exhibited hyperactivity, memory impairment, and desperate behavior, which are core features of schizophrenia and depression. Due to the overlapping symptomology and pathogenesis observed for schizophrenia and depression, the present study attempted to determine whether IL-15Rα was associated with the risk of schizophrenia or depression. One hundred fifty-six participants, including 63 schizophrenia patients, 29 depressive patients, and 64 age-matched healthy controls, were enrolled in the study. We investigated the circulating levels of soluble IL-15Rα and analyzed potential links between the IL-15Rα levels and clinical symptoms present in schizophrenia or depressive patients. We observed reduced serum IL-15Rα levels in schizophrenia patients, but not depressive patients compared with controls. Moreover, a significant negative association was observed between the circulating IL-15Rα levels and excited phenotypes in the schizophrenia patients. The IL-15Rα KO mice displayed pronounced pre-pulse inhibition impairment, which was a typical symptom of schizophrenia. Interestingly, the IL-15Rα KO mice exhibited a remarkable elevation in the startle amplitude in the startle reflex test compared to wild type mice. These results demonstrated that serum levels of soluble IL-15Rα were reduced in schizophrenia and highlighted the relationship of IL-15Rα and the excited phenotype in schizophrenia patients and mice.
Collapse
Affiliation(s)
- Yi He
- Beijing Key Laboratory of Mental Disorders, The National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Qijing Bo
- Beijing Key Laboratory of Mental Disorders, The National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Zhen Mao
- Beijing Key Laboratory of Mental Disorders, The National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Jian Yang
- Beijing Key Laboratory of Mental Disorders, The National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Min Liu
- Beijing Key Laboratory of Mental Disorders, The National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Haixia Wang
- Beijing Key Laboratory of Mental Disorders, The National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Abba J Kastin
- Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Weihong Pan
- BioPotentials Consult, Sedona, AZ, United States
| | - Chuanyue Wang
- Beijing Key Laboratory of Mental Disorders, The National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, The National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
He Y, Ge L, Tong F, Zheng P, Yang J, Zhou J, Sun Z, Wang H, Yang S, Li Y, Yu Y. Metabolic responses in the cortex and hippocampus induced by Il-15rα mutation. Mol Omics 2022; 18:865-872. [DOI: 10.1039/d2mo00105e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Metabolomics showed distinct metabolic phenotypes of the different brain regions related to the IL-15 system, enhancing our understanding of the IL-15 system and its interactions with neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yi He
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China
| | - Lijun Ge
- Liyuan Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Fang Tong
- Department of Physiology and Biochemistry, School of Fundamental Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jian Yang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Jingjing Zhou
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zuoli Sun
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China
| | - Haixia Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China
| | - Shun Yang
- Department of General Surgery, Yantian District People's Hospital, Shenzhen, 518081, China
| | - Yifan Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuxin Yu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
9
|
Martinuzzi E, Barbosa S, Courtet P, Olié E, Guillaume S, Ibrahim EC, Daoudlarian D, Davidovic L, Glaichenhaus N, Belzeaux R. Blood cytokines differentiate bipolar disorder and major depressive disorder during a major depressive episode: Initial discovery and independent sample replication. Brain Behav Immun Health 2021; 13:100232. [PMID: 34589747 PMCID: PMC8474674 DOI: 10.1016/j.bbih.2021.100232] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 01/02/2023] Open
Abstract
Bipolar disorder (BD) diagnosis currently relies on assessment of clinical symptoms, mainly retrospective and subject to memory bias. BD is often misdiagnosed as Major Depressive Disorder (MDD) resulting in ineffective treatment and worsened clinical outcome. The primary purpose of this study was to identify blood biomarkers that discriminate MDD from BD patients when in a depressed state. We have used clinical data and serum samples from two independent naturalistic cohorts of patients with a Major Depressive Episode (MDE) who fulfilled the criteria of either BD or MDD at inclusion. The discovery and replication cohorts consisted of 462 and 133 patients respectively. Patients were clinically assessed using standard diagnostic interviews, and clinical variables including current treatments were recorded. Blood was collected and serum assessed for levels of 31 cytokines using a sensitive multiplex assay. A penalized logistic regression model combined with nonparametric bootstrap was subsequently used to identify cytokines associated with BD. Interleukin (IL)-6, IL-10, IL-15, IL-27 and C-X-C ligand chemokine (CXCL)-10 were positively associated with BD in the discovery cohort. Of the five cytokines identified as discriminant features in the discovery cohort, IL-10, IL-15 and IL-27 were also positively associated with BD in the replication cohort therefore providing an external validation to our finding. Should our results be validated in a prospective cohort, they could provide new insights into the pathophysiological mechanisms of mood disorders.
Collapse
Affiliation(s)
- Emanuela Martinuzzi
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Clinical Research Unit, Valbonne, France
| | - Susana Barbosa
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Clinical Research Unit, Valbonne, France
| | - Philippe Courtet
- Centre Hospitalier Universitaire de Montpellier, Institut National de la Santé et de la Recherche Médicale, Ho^pital Lapeyronie, Department of Emergency Psychiatry and Acute Care, Montpellier, France
| | - Emilie Olié
- Centre Hospitalier Universitaire de Montpellier, Institut National de la Santé et de la Recherche Médicale, Ho^pital Lapeyronie, Department of Emergency Psychiatry and Acute Care, Montpellier, France
| | - Sébastien Guillaume
- Centre Hospitalier Universitaire de Montpellier, Institut National de la Santé et de la Recherche Médicale, Ho^pital Lapeyronie, Department of Emergency Psychiatry and Acute Care, Montpellier, France
| | | | - Douglas Daoudlarian
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Clinical Research Unit, Valbonne, France
| | - Laetitia Davidovic
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Clinical Research Unit, Valbonne, France
| | - Nicolas Glaichenhaus
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Clinical Research Unit, Valbonne, France.,Fondation FondaMental, France
| | - Raoul Belzeaux
- Aix Marseille Univ, CNRS, Inst Neurosci Timone, Marseille, France.,Assistance Publique Hôpitaux de Marseille, Department of Psychiatry, Marseille, France.,Fondation FondaMental, France
| |
Collapse
|
10
|
He Y, Yu Y, Li Y, Duan W, Sun Z, Yang J, Kastin AJ, Pan W, Zhang Y, Wang K. Phenotypic Resemblance to Neuropsychiatric Disorder and Altered mRNA Profiles in Cortex and Hippocampus Underlying IL15Rα Knockout. Front Neurosci 2021; 14:582279. [PMID: 33613171 PMCID: PMC7887313 DOI: 10.3389/fnins.2020.582279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/23/2020] [Indexed: 01/05/2023] Open
Abstract
Background Previous studies of the functions of IL15Rα have been limited to immune activities and skeletal muscle development. Immunological factors have been identified as one of the multiple causes of psychosis, and neurological symptoms have been described in IL15Rα knockout (KO) mice. Seeking to explore possible mechanisms for this in the IL15Rα-/- mouse brain, we analyzed gene expression patterns in the cortex and hippocampus using the RNA-seq technique. Methods IL15Rα KO mice were generated and littermate wildtype (WT) mice were used as a control group. A Y-maze was used to assess behavior differences between the two groups. The cortex and hippocampus of 3-month-old male mice were prepared and RNA-seq and transcriptome analysis were performed by gene set enrichment analysis (GSEA). Results Compared with the WT group, IL15Rα KO animals showed higher speed in the novel arm and more entrance frequency in the old arm in the Y-maze experiment. GSEA indicated that 18 pathways were downregulated and 13 pathways upregulated in both cortex and hippocampus from the GO, KEGG, and Hallmark gene sets. The downregulated pathways formed three clusters: respiratory chain and electron transport, regulation of steroid process, and skeletal muscle development. Conclusion IL15Rα KO mice exhibit altered expression of multiple pathways, which could affect many functions of the brain. Lipid biosynthesis and metabolism in the central nervous system (CNS) should be investigated to provide insights into the effect of IL15Rα on psychosis in this murine model.
Collapse
Affiliation(s)
- Yi He
- The National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yuxin Yu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanan Li
- Department of Gastrointestinal Surgery, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weicheng Duan
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zuoli Sun
- The National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Jian Yang
- The National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Abba J Kastin
- Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Weihong Pan
- BioPotentials Consult, Sedona, AZ, United States
| | - Yan Zhang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kang Wang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Barbosa S, Khalfallah O, Forhan A, Galera C, Heude B, Glaichenhaus N, Davidovic L. Serum cytokines associated with behavior: A cross-sectional study in 5-year-old children. Brain Behav Immun 2020; 87:377-387. [PMID: 31923553 DOI: 10.1016/j.bbi.2020.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/23/2019] [Accepted: 01/05/2020] [Indexed: 12/22/2022] Open
Abstract
Nearly 10% of 5-year-old children experience social, emotional or behavioral problems and are at increased risk of developing mental disorders later in life. While animal and human studies have demonstrated that cytokines can regulate brain functions, it is unclear whether individual cytokines are associated with specific behavioral dimensions in population-based pediatric samples. Here, we used data and biological samples from 786 mother-child pairs participating to the French national mother-child cohort EDEN. At the age of 5, children were assessed for behavioral difficulties using the Strengths and Difficulties Questionnaire (SDQ) and had their serum collected. Serum samples were analyzed for levels of well-characterized effector or regulatory cytokines. We then used a penalized logistic regression method (Elastic Net), to investigate associations between serum levels of cytokines and each of the five SDQ-assessed behavioral dimensions after adjustment for relevant covariates and confounders, including psychosocial variables. We found that interleukin (IL)-6, IL-7, and IL-15 were associated with increased odds of problems in prosocial behavior, emotions, and peer relationships, respectively. In contrast, eight cytokines were associated with decreased odds of problems in one dimension: IL-8, IL-10, and IL-17A with emotional problems, Tumor Necrosis Factor (TNF)-α with conduct problems, C-C motif chemokine Ligand (CCL)2 with hyperactivity/inattention, C-X-C motif chemokine Ligand (CXCL)10 with peer problems, and CCL3 and IL-16 with abnormal prosocial behavior. Without implying causation, these associations support the notion that cytokines regulate brain functions and behavior and provide a rationale for launching longitudinal studies.
Collapse
Affiliation(s)
- Susana Barbosa
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Olfa Khalfallah
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Anne Forhan
- Université de Paris, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Centre de Recherche en Épidémiologie et Statistiques, Paris, France
| | - Cédric Galera
- University Bordeaux Segalen, Charles Perrens Hospital, Child and Adolescent Psychiatry Department, Bordeaux, France
| | - Barbara Heude
- Université de Paris, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Centre de Recherche en Épidémiologie et Statistiques, Paris, France
| | - Nicolas Glaichenhaus
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Laetitia Davidovic
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.
| |
Collapse
|
12
|
Mayagoitia K, Shin SD, Rubini M, Siebold L, Wilson CG, Bellinger DL, Figueroa JD, Soriano S. Short-term exposure to dietary cholesterol is associated with downregulation of interleukin-15, reduced thigmotaxis and memory impairment in mice. Behav Brain Res 2020; 393:112779. [PMID: 32585301 DOI: 10.1016/j.bbr.2020.112779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/15/2020] [Accepted: 06/16/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition associated with loss of memory function, depression and anxiety. The etiology of AD is poorly understood, but both cholesterol dyshomeostasis and dysregulation of the immune system are contributing factors. Current evidence is consistent with a detrimental effect of excess cholesterol on neuroinflammation, both in mouse models of memory loss and in dementia in humans. However, whether the impact of cholesterol on neuroinflammation occurs early and contributes to pathogenesis of the disease or simply reflects a pleiotropic impact at advanced stages of disease is unclear. To explore this question, we measured, in 9-13 week-old mice, cognitive status and changes in brain inflammatory mediators in response to a short-term high-cholesterol diet. We hypothesized that short-term exposure to excess dietary cholesterol would alter the early inflammatory responses associated with cognitive and/or behavioral impairment. We report that short-term exposure to a high-cholesterol diet led to decreased thigmotaxis and short-term spatial memory impairment without affecting long-term recognition memory. Furthermore, cognitive and behavioral phenotypes in these mice were associated with a reduction in interleukin-15 levels in the absence of changes in other inflammatory mediators. Our findings indicate that interleukin-15 may play a role in early stages of cognitive impairment secondary to hypercholesterolemia. Consequently, optimization of interleukin-15 signaling may be a viable effective cognitive therapy in the population susceptible to developing dementia due to risk factors associated with cholesterol dysregulation.
Collapse
Affiliation(s)
- Karina Mayagoitia
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Sam D Shin
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Marsilio Rubini
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Lorraine Siebold
- Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Christopher G Wilson
- Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Denise L Bellinger
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Johnny D Figueroa
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda CA, USA
| | - Salvador Soriano
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA, USA.
| |
Collapse
|
13
|
Pan Y, Wang Z, Zhang G, Guo J, Zhu X, Zhou J, Zhang Z, Sun Z, Yang J, Kastin AJ, Pan W, Wu X, Zhang J, Wang X, Wang C, He Y. Schizophrenia Patient Shows a Rare Interleukin 15 Receptor alpha Variant Disrupting Signal Transduction. Curr Mol Med 2019; 19:560-569. [PMID: 31244423 DOI: 10.2174/1566524019666190617172054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/25/2019] [Accepted: 05/28/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Schizophrenia is a complex and debilitating mental disorder with strong heritability. Its pathogenesis involves immune dysregulation. Interleukin 15 and interleukin 15 receptor alpha(IL-15Rα) are classical immune molecules. They also help maintain normal brain function, leading to our hypothesis that IL-15Rα gene(IL- 15RA) variants contribute to the pathogenesis of schizophrenia. OBJECTIVE We determine whether the genetic variants of IL-15RA are associated with the development and progression of schizophrenia and whether IL-15RA single nucleotide polymorphism(SNP) plays a key role in downstream signaling transduction. METHODS AND RESULTS We sequenced IL-15RA exon from 132 Chinese schizophrenic patients and identified a rare variant(rs528238821) in a patient diagnosed with catatonic schizophrenia and ankylosing spondylitis(AS). We overexpressed this missense variant in cells driven by pBI-CMV vector. The cells showed attenuated STAT3 phosphorylation in response to interleukin15. CONCLUSION IL-15RA mutation is rare in schizophrenic patients but interfered with IL- 15Rα intracellular signal transduction. Given the similarity of symptoms of catatonic schizophrenia and the known phenotype of IL-15Rα knockout mice, gene variation might offer diagnostic value for sub-types of schizophrenia.
Collapse
Affiliation(s)
- Yanli Pan
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zhimin Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Guangping Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Junhua Guo
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Xuequan Zhu
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Jia Zhou
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zhenrong Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zuoli Sun
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Jian Yang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Abba J Kastin
- Pennington Biomedical Research Center, Baton Rouge, LA70808, United States
| | - Weihong Pan
- BioPotentials Consulting, Sedona, AZ 86351, United States
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Material Medical, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianliang Zhang
- Department of Neurobiology, Capital Medical University, Beijing 100069, China.,Beijing Institute for Brain Disorders, Beijing 100069, China.,Beijing Center of Neural Regeneration and Repair, Beijing 100069, China
| | - Xiaomin Wang
- Beijing Institute for Brain Disorders, Beijing 100069, China.,Department of Physiology and Pathologic Physiology, Capital Medical University, Beijing 100069, China.,Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Chuanyue Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Yi He
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
14
|
Pérez-Sánchez G, Becerril-Villanueva E, Arreola R, Martínez-Levy G, Hernández-Gutiérrez ME, Velasco-Velásquez MA, Alvarez-Herrera S, Cruz-Fuentes C, Palacios L, de la Peña F, Pavón L. Inflammatory Profiles in Depressed Adolescents Treated with Fluoxetine: An 8-Week Follow-up Open Study. Mediators Inflamm 2018; 2018:4074051. [PMID: 30662368 PMCID: PMC6312587 DOI: 10.1155/2018/4074051] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/30/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
Changes in cytokine levels in major depression and during treatment have been reported in adults. However, few studies have examined cytokine levels in an adolescent sample despite this being a common age of onset. Methods. We measured proinflammatory (IL-2, IFN-γ, IL-1β, TNF-α, IL-6, IL-12, and IL-15) and anti-inflammatory (IL-4, IL-5, IL-13, IL-1Ra, and IL-10) cytokine serum levels in 22 adolescents with major depression and 18 healthy volunteers. Cytokines were measured by multiplex bead-based immunoassays at baseline, and 4 and 8 weeks after commencement of fluoxetine administration in the clinical group. Results. Compared to healthy volunteers, adolescents with major depression at baseline showed significant increases in all pro- and anti-inflammatory cytokines, except IL-1Ra and IL-10. Significant changes were observed in fluoxetine treatment compared to baseline: proinflammatory cytokines IFN-γ, IL-1β, TNF-α, IL-6, IL-12, and IL-15 were decreased only at week 4 whereas IL-2 was increased only at week 8; anti-inflammatory cytokines IL-4 and IL-5 were increased at week 8 while IL-1Ra was reduced only at week 4. There were no significant correlations between cytokine levels and symptomatic improvement in HDRS. Discussion. The results suggest a significant interplay between cytokine levels, the depressive state, and the stage of treatment with an SSRI. To the best of our knowledge, this is the first report in depressed adolescents with elevated IL-12, IL-13, and IL-15 levels. Further studies are necessary to clarify the role and mechanisms of altered cytokine levels in the pathogenesis and physiopathology of major depressive disorder.
Collapse
Affiliation(s)
- Gilberto Pérez-Sánchez
- Laboratory of Psychoimmunology, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| | - Enrique Becerril-Villanueva
- Laboratory of Psychoimmunology, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| | - Rodrigo Arreola
- Psychiatric Genetics Department, Clinical Research Branch, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| | - Gabriela Martínez-Levy
- Psychiatric Genetics Department, Clinical Research Branch, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| | - María Eugenia Hernández-Gutiérrez
- Neuropharmacology Laboratory, Clinical Research Branch, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| | - Marco A. Velasco-Velásquez
- School of Medicine, National Autonomous University of México, Ciudad Universitaria, 04510 México City, Mexico
| | - Samantha Alvarez-Herrera
- Laboratory of Psychoimmunology, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| | - Carlos Cruz-Fuentes
- Psychiatric Genetics Department, Clinical Research Branch, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| | - Lino Palacios
- Adolescent clinic, Clinical Services, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| | - Francisco de la Peña
- Adolescent clinic, Clinical Services, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| | - Lenin Pavón
- Laboratory of Psychoimmunology, National Institute of Psychiatry Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 México City, Mexico
| |
Collapse
|
15
|
A systematic review on the association between inflammatory genes and cognitive decline in non-demented elderly individuals. Eur Neuropsychopharmacol 2017; 27:568-588. [PMID: 26718789 DOI: 10.1016/j.euroneuro.2015.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/27/2015] [Accepted: 12/01/2015] [Indexed: 01/15/2023]
Abstract
Cognitive impairment, or decline, is not only a feature of Alzheimer׳s disease and other forms of dementia but also normal ageing. Abundant evidence from epidemiological studies points towards perturbed inflammatory mechanisms in aged individuals, though the cause-effect nature of this apparent relationship is difficult to establish. Genetic association studies focusing on polymorphism in and around inflammatory genes represent a viable approach to establish whether inflammatory mechanisms might play a causal role in cognitive decline, whilst also enabling the identification of specific genes potentially influencing specific cognitive facets. Thus, here we provide a review of published genetic association studies investigating inflammatory genes in the context of cognitive decline in elderly, non-demented, samples. Numerous candidate gene association studies have been performed to date, focusing almost exclusively on genes encoding major cytokines. Some of these studies report significant cognitive domain-specific associations implicating Interleukin 1β (IL1β) (rs16944), Tumour Necrosis Factor α (TNFα) (rs1800629) and C-reactive protein (CRP) in various domains of cognitive function. However, the majority of these studies are lacking in statistical power and have other methodological limitations, suggesting some of them may have yielded false positive results. Genome-wide association studies have implicated less direct and less obvious regulators of inflammatory processes (i.e., PDE7A, HS3ST4, SPOCK3), indicating that a shift away from the major cytokine-encoding genes in future studies will be important. Furthermore, better cohesion across studies with regards to the cognitive test batteries administered to participants along with the continued application of longitudinal designs will be vital.
Collapse
|
16
|
Niu T, Li J, Wang J, Ma JZ, Li MD. Identification of Novel Signal Transduction, Immune Function, and Oxidative Stress Genes and Pathways by Topiramate for Treatment of Methamphetamine Dependence Based on Secondary Outcomes. Front Psychiatry 2017; 8:271. [PMID: 29321746 PMCID: PMC5733474 DOI: 10.3389/fpsyt.2017.00271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/20/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Topiramate (TPM) is suggested to be a promising medication for treatment of methamphetamine (METH) dependence, but the molecular basis remains to be elucidated. METHODS Among 140 METH-dependent participants randomly assigned to receive either TPM (N = 69) or placebo (N = 71) in a previously conducted randomized controlled trial, 50 TPM- and 49 placebo-treated participants had a total 212 RNA samples available at baseline, week 8, and week 12 time points. Following our primary analysis of gene expression data, we reanalyzed the microarray expression data based on a latent class analysis of binary secondary outcomes during weeks 1-12 that provided a classification of 21 responders and 31 non-responders with consistent responses at both time points. RESULTS Based on secondary outcomes, 1,381, 576, 905, and 711 differentially expressed genes at nominal P values < 0.05 were identified in responders versus non-responders for week 8 TPM, week 8 placebo, week 12 TPM, and week 12 placebo groups, respectively. Among 1,381 genes identified in week 8 TPM responders, 359 genes were identified in both week 8 and week 12 TPM groups, of which 300 genes were exclusively detected in TPM responders. Of them, 32 genes had nominal P values < 5 × 10-3 at either week 8 or week 12 and false discovery rates < 0.15 at both time points with consistent directions of gene expression changes, which include GABARAPL1, GPR155, and IL15RA in GABA receptor signaling that represent direct targets for TPM. Analyses of these 300 genes revealed 7 enriched pathways belonging to neuronal function/synaptic plasticity, signal transduction, inflammation/immune function, and oxidative stress response categories. No pathways were enriched for 72 genes exclusively detected in both week 8 and week 12 placebo groups. CONCLUSION This secondary analysis study of gene expression data from a TPM clinical trial not only yielded consistent results with those of primary analysis but also identified additional new genes and pathways on TPM response to METH addiction.
Collapse
Affiliation(s)
- Tianhua Niu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jingjing Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Ju Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Jennie Z Ma
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, China.,Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, United States
| |
Collapse
|
17
|
Patidar M, Yadav N, Dalai SK. Interleukin 15: A key cytokine for immunotherapy. Cytokine Growth Factor Rev 2016; 31:49-59. [PMID: 27325459 DOI: 10.1016/j.cytogfr.2016.06.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/20/2016] [Accepted: 06/03/2016] [Indexed: 12/20/2022]
Abstract
Interleukin (IL)-15, a member of the immunoregulatory cytokines family, is a pluripotent molecule with therapeutic potential. It is predominantly expressed by the myeloid cells, as well as other cell types. IL-15 serves multiple functions including dictating T cell response, regulating tissue repair and B cell homing, modulating inflammation, and activating NK cells. Among cytokines, IL-15 is unique because of its wide expression, tightly regulated secretion, trans-presentation, and therapeutic potential. IL-15 has been investigated for its therapeutic potential for the induction and maintenance of T cell responses. In addition, IL-15 can be targeted by antibody- or mutant IL-15 therapy to reduce inflammation. Its multifaceted biological applications are crucial in immunotherapy. In this article, we review the functions, expression, and regulation of IL-15 for designing an improved IL-15-based therapy targeting the IL-15 signaling pathway.
Collapse
Affiliation(s)
- Manoj Patidar
- Institute of Science, Nirma University, Ahmedabad 382481, India.
| | - Naveen Yadav
- Institute of Science, Nirma University, Ahmedabad 382481, India.
| | - Sarat K Dalai
- Institute of Science, Nirma University, Ahmedabad 382481, India.
| |
Collapse
|
18
|
Panek M, Jonakowski M, Zioło J, Pietras T, Wieteska Ł, Małachowska B, Mokros Ł, Szemraj J, Kuna P. Identification of Relationships Between Interleukin 15 mRNA and Brain-Derived Neurotrophic Factor II mRNA Levels With Formal Components of Temperament in Asthmatic Patients. Mol Neurobiol 2016; 54:1733-1744. [PMID: 26874516 DOI: 10.1007/s12035-016-9768-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/01/2016] [Indexed: 11/30/2022]
Abstract
Asthma is a chronic inflammatory and heterogeneous disease developing mostly through allergic inflammation, which modifies the expression of various cytokines and neurotrophins. Previous studies suggest the involvement of interleukin (IL)-15 in the regulation of immune response in asthma. Brain-derived neurotrophic factor (BDNF) II plays an important role as a regulator of development and survival of neurons as well as maintenance of their physiological activity. Chronic stress associated with asthma and elevated IL-15 mRNA and BDNFII mRNA levels may affect the mood and a subjective sensation of dyspnoea-inducing anxiety. Psychopathological variables and numerous cytokine/neurotrophin interactions influence the formation of temperament and strategies of coping with stress. The aim of the study was to identify the role of IL-15 mRNA and BDNFII mRNA expressions and their effect on components of temperament and strategies of coping with stress in asthmatics. A total of 352 subjects (176 healthy volunteers and 176 asthmatic patients) participated in the study. The Formal Characteristic of Behaviour-Temperament Inventory (FCB-TI), Coping Inventory for Stressful Situations (CISS), Beck Depression Inventory, State-Trait Anxiety Inventory, and Borg Rating of Perceived Exertion (RPE) Scale were applied in all the subjects. The expression of IL-15 and BDNFII gene was measured using quantitative real-time polymerase chain reaction (qRT-PCR). Different levels of IL-15 and BDNFII expressions between healthy volunteers and patients were revealed in the study. IL-15 enhanced the BDNFII mRNA expression among patients with bronchial asthma. The depression level negatively correlated with the BDNFII mRNA expression. This neurotrophin modified the temperament variable. BDNFII significantly affected (proportional relationship) the level of briskness in asthmatic patients. BDNFII might influence the level and style of coping with stress (emotion-oriented style). This hypothesis requires further studies on protein functional models. The obtained data confirms the role of IL-15 and BDNFII in the pathomechanisms of depression and formation of selected traits defining the temperament in asthmatics.
Collapse
Affiliation(s)
- Michał Panek
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, 22 Kopcinskiego St., 90-153, Lodz, Poland.
| | - Mateusz Jonakowski
- Students Research Group at the Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, 22 Kopcinskiego St., 90-153, Lodz, Poland
| | - Jan Zioło
- Students Research Group at the Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, 22 Kopcinskiego St., 90-153, Lodz, Poland
| | - Tadeusz Pietras
- Department of Pneumology and Allergology, Medical University of Lodz, 22 Kopcinskiego St., 90-153, Lodz, Poland
| | - Łukasz Wieteska
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland
| | - Beata Małachowska
- Department of Pediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, 36/50 Sporna St., 91-738, Lodz, Poland
| | - Łukasz Mokros
- Students Research Group at the Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, 22 Kopcinskiego St., 90-153, Lodz, Poland.,Department of Pneumology and Allergology, Medical University of Lodz, 22 Kopcinskiego St., 90-153, Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland
| | - Piotr Kuna
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, 22 Kopcinskiego St., 90-153, Lodz, Poland
| |
Collapse
|
19
|
Bortell N, Morsey B, Basova L, Fox HS, Marcondes MCG. Phenotypic changes in the brain of SIV-infected macaques exposed to methamphetamine parallel macrophage activation patterns induced by the common gamma-chain cytokine system. Front Microbiol 2015; 6:900. [PMID: 26441851 PMCID: PMC4568411 DOI: 10.3389/fmicb.2015.00900] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/17/2015] [Indexed: 12/12/2022] Open
Abstract
One factor in the development of neuroAIDS is the increase in the migration of pro-inflammatory CD8 T cells across the blood–brain barrier. Typically these cells are involved with keeping the viral load down. However, the persistence of above average numbers of CD8 T cells in the brain, not necessarily specific to viral peptides, is facilitated by the upregulation of IL15 from astrocytes, in the absence of IL2, in the brain environment. Both IL15 and IL2 are common gamma chain (γc) cytokines. Here, using the non-human primate model of neuroAIDS, we have demonstrated that exposure to methamphetamine, a powerful illicit drug that has been associated with HIV exposure and neuroAIDS severity, can cause an increase in molecules of the γc system. Among these molecules, IL15, which is upregulated in astrocytes by methamphetamine, and that induces the proliferation of T cells, may also be involved in driving an inflammatory phenotype in innate immune cells of the brain. Therefore, methamphetamine and IL15 may be critical in the development and aggravation of central nervous system immune-mediated inflammatory pathology in HIV-infected drug abusers.
Collapse
Affiliation(s)
- Nikki Bortell
- Department of Molecular and Cellular Neurosciences, The Scripps Research Institute La Jolla, CA, USA
| | - Brenda Morsey
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center Omaha, NE, USA
| | - Liana Basova
- Department of Molecular and Cellular Neurosciences, The Scripps Research Institute La Jolla, CA, USA
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center Omaha, NE, USA
| | | |
Collapse
|
20
|
Arisi GM. Nervous and immune systems signals and connections: cytokines in hippocampus physiology and pathology. Epilepsy Behav 2014; 38:43-7. [PMID: 24534466 DOI: 10.1016/j.yebeh.2014.01.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 01/20/2014] [Accepted: 01/22/2014] [Indexed: 02/07/2023]
Abstract
Signaling through secretion of small molecules is a hallmark of both nervous and immune systems. The scope and influence of the intense message exchange between these two complex systems are only now becoming objects of scientific inquiry. Both neurotransmitters and cytokines affect their target cells through surface receptors and also by other molecular mechanisms. Cytokine receptors are present in neurons and glial cell populations in discrete brain regions. This review firstly focuses on the role of cytokines in hippocampal physiological processes, such as memory and learning, and secondly on the pathological involvement of cytokines in diseases like depression and epilepsy. Interleukin-1β is necessary for long-term potentiation (LTP) maintenance in the hippocampus. On the other hand, interleukin-6 has a negative regulatory role in long-term memory acquisition. Astrocyte-secreted tumor necrosis factor plays a role in synaptic strength by increasing surface translocation of glutamate AMPA receptors, and the chemokine CXCL12 can silence the tonic activity of Cajal-Retzius neurons in the hippocampus. Manifold increased concentrations of interleukin-10, interferon-γ, ICAM1, CCL2, and CCL4 are observed in the hippocampi of patients with temporal lobe epilepsy. A contemporary view of the role of cytokines as neuromodulators is emerging from studies in humans and manipulations of experimental animals. Despite the accumulating evidence of the role of cytokines on nervous system physiology and pathology, it is important not to exaggerate its relevance.
Collapse
Affiliation(s)
- Gabriel Maisonnave Arisi
- Neurobiology Laboratory, Department of Physiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil.
| |
Collapse
|
21
|
Abstract
Interleukin (IL)-15 is a ubiquitously expressed cytokine existing in both intracellular and secretory forms. Here we review the expression, regulation, and functions of IL15 and its receptors in the brain. IL15 receptors show robust upregulation after neuroinflammation, suggesting a major role of IL15 signaling in cerebral function. Involvement of the IL15 system in neuropsychiatric behavior is reflected by the effects of IL15, IL15Rα, and IL2Rγ deletions on neurobehavior and neurotransmitters, the effects of IL15 treatment on neuronal activity, and the potential role of IL15 in neuroplasticity/neurogenesis. The results show that IL15 modulates GABA and serotonin transmission. This may underlie deficits in mood (depressive-like behavior and decreased normal anxiety) and memory, as well as activity level, sleep, and thermoregulation. Although IL15 has only a low level of permeation across the blood-brain barrier, peripheral IL15 is able to activate multiple signaling pathways in neurons widely distributed in CNS regions. The effects of IL15 in "preventing" neuropsychiatric symptoms in normal mice implicate a potential therapeutic role of this polypeptide cytokine.
Collapse
|
22
|
Chang SD, Liang K. Roles of hippocampal GABAA and muscarinic receptors in consolidation of context memory and context–shock association in contextual fear conditioning: A double dissociation study. Neurobiol Learn Mem 2012; 98:17-24. [DOI: 10.1016/j.nlm.2012.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 04/06/2012] [Accepted: 04/12/2012] [Indexed: 11/26/2022]
|
23
|
Gómez-Nicola D, Valle-Argos B, Pallas-Bazarra N, Nieto-Sampedro M. Interleukin-15 regulates proliferation and self-renewal of adult neural stem cells. Mol Biol Cell 2011; 22:1960-70. [PMID: 21508317 PMCID: PMC3113763 DOI: 10.1091/mbc.e11-01-0053] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The role of IL-15 in the regulation of neural stem cell biology appears as a key mechanism in the control of adult neurogenesis, with direct implications for the development of pathologies with a neuroimmune component. The impact of inflammation is crucial for the regulation of the biology of neural stem cells (NSCs). Interleukin-15 (IL-15) appears as a likely candidate for regulating neurogenesis, based on its well-known mitogenic properties. We show here that NSCs of the subventricular zone (SVZ) express IL-15, which regulates NSC proliferation, as evidenced by the study of IL-15−/− mice and the effects of acute IL-15 administration, coupled to 5-bromo-2′-deoxyuridine/5-ethynyl-2′-deoxyuridine dual-pulse labeling. Moreover, IL-15 regulates NSC differentiation, its deficiency leading to an impaired generation of neuroblasts in the SVZ–rostral migratory stream axis, recoverable through the action of exogenous IL-15. IL-15 expressed in cultured NSCs is linked to self-renewal, proliferation, and differentiation. IL-15–/– NSCs presented deficient proliferation and self-renewal, as evidenced in proliferation and colony-forming assays and the analysis of cell cycle–regulatory proteins. Moreover, IL-15–deficient NSCs were more prone to differentiate than wild-type NSCs, not affecting the cell population balance. Lack of IL-15 led to a defective activation of the JAK/STAT and ERK pathways, key for the regulation of proliferation and differentiation of NSCs. The results show that IL-15 is a key regulator of neurogenesis in the adult and is essential to understanding diseases with an inflammatory component.
Collapse
Affiliation(s)
- Diego Gómez-Nicola
- Functional and Systems Neurobiology Department, Cajal Institute (CSIC), Madrid, Spain.
| | | | | | | |
Collapse
|
24
|
Wu X, Hsuchou H, Kastin AJ, He Y, Khan RS, Stone KP, Cash MS, Pan W. Interleukin-15 affects serotonin system and exerts antidepressive effects through IL15Rα receptor. Psychoneuroendocrinology 2011; 36:266-78. [PMID: 20724079 PMCID: PMC3015024 DOI: 10.1016/j.psyneuen.2010.07.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 07/18/2010] [Accepted: 07/21/2010] [Indexed: 01/23/2023]
Abstract
Contrary to the reduction of depressive-like behavior observed in several strains of cytokine receptor knockout mice, mice lacking the specific receptor for interleukin (IL)-15 showed increased immobility in tail suspension and modified forced swimming tests. There was also a reduction in social interactions. The hippocampus of the IL15Rα knockout mice had decreased mRNA for 5-HT(1A), increased mRNA for 5-HT(2C), and region-specific changes of serotonin reuptake transporter (SERT) immunoreactivity. Fluoxetine (the classic antidepressant Prozac, which inhibits 5-HT(2C) and SERT) reduced the immobility of the IL15Rα knockout mice in comparison with their pretreatment baseline. Together with the unchanged performance of the IL15Rα knockout mice on the rotarod, this response to fluoxetine indicates that the immobility reflects depression. Wildtype mice responded to IL15 treatment with improvement of immobility induced by forced swimming, whereas the knockout mice failed to respond. Thus, the cognate IL15 receptor is necessary for the antidepressive activity of IL15. In ex vivo studies, IL15 decreased synaptosomal uptake of 5-HT, and modulated the expression of 5-HT(2C) and SERT in cultured neurons in a dose- and time-dependent manner. Thus, the effect of IL15 on serotonin transmission may underlie the depressive-like behavior of IL15Rα knockout mice. We speculate that IL15 is essential to maintain neurochemical homeostasis and thereby plays a role in preventing neuropsychiatric symptoms.
Collapse
MESH Headings
- Animals
- Antidepressive Agents/pharmacology
- Cells, Cultured
- Depression/genetics
- Depression/metabolism
- Depression/pathology
- Depression/prevention & control
- Dose-Response Relationship, Drug
- Drug Evaluation, Preclinical
- Fluoxetine/pharmacology
- Freezing Reaction, Cataleptic/drug effects
- Freezing Reaction, Cataleptic/physiology
- Interleukin-15/pharmacology
- Interleukin-15/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Nervous System/drug effects
- Nervous System/metabolism
- Receptors, Interleukin-15/agonists
- Receptors, Interleukin-15/genetics
- Receptors, Interleukin-15/metabolism
- Receptors, Interleukin-15/physiology
- Serotonin/metabolism
- Synaptic Transmission/drug effects
- Synaptic Transmission/genetics
- Synaptic Transmission/physiology
- Synaptosomes/drug effects
- Synaptosomes/metabolism
- Time Factors
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Weihong Pan
- Corresponding author: Weihong Pan, MD, PhD, Blood-Brain Barrier Group, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA, Tel. 1-225-763-2707, Fax 1-225-763-0261, , Web: http://labs.pbrc.edu/bloodbrainbarrier
| |
Collapse
|
25
|
Stone KP, Kastin AJ, Hsuchou H, Yu C, Pan W. Rapid endocytosis of interleukin-15 by cerebral endothelia. J Neurochem 2011; 116:544-53. [PMID: 21155807 DOI: 10.1111/j.1471-4159.2010.07142.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Interleukin (IL)-15 receptors are present in the cerebral endothelia composing the blood-brain barrier where they show robust up-regulation by neuroinflammation. To determine how IL15 receptor subunits participate in the endocytosis and intracellular trafficking of IL15, we performed confocal microscopic imaging and radioactive tracer uptake assays in primary brain microvessel endothelial cells and related cell lines transfected with modulatory molecules. By immunostaining and co-localization studies with organelle markers, we showed that IL15 was rapidly endocytosed via lipid rafts and was directed to diverse intracellular pathways. During the course of intracellular trafficking, Alexa dye-conjugated IL15 was partially co-localized with both the specific receptor IL15Rα and the co-receptor IL2Rγ. However, deletion of one of the receptor subunits had only a minor effect in slowing IL15 uptake when primary brain microvessel endothelial cells from the receptor knockout mice were compared with those from wildtype mice. IL15 was trafficked to early, recycling, and late endosomes, to the Golgi, and to lysosomes. The diffuse distribution suggests that IL15 activates multiple endothelial signaling events.
Collapse
Affiliation(s)
- Kirsten P Stone
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | | | | | | | | |
Collapse
|
26
|
Wu X, He Y, Hsuchou H, Kastin AJ, Rood JC, Pan W. Essential role of interleukin-15 receptor in normal anxiety behavior. Brain Behav Immun 2010; 24:1340-6. [PMID: 20600810 PMCID: PMC2949491 DOI: 10.1016/j.bbi.2010.06.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 06/15/2010] [Accepted: 06/22/2010] [Indexed: 10/19/2022] Open
Abstract
The interactions between the cytokine interleukin (IL)-15 and the classical neurotransmitter GABA have been shown in IL15Rα receptor knockout mice by observations of memory deficits and reduction of GABA. To test the hypothesis that IL15 affects anxiety-like behavior, knockout mice without IL15, IL15Rα, or the co-receptor IL2Rγ were subjected to open-field and elevated plus maze tests. All three strains showed reduction of anxiety, with greater changes in the IL15Rα knockout mice than in the IL15 or IL2Rγ knockout mice. This unexpected observation is opposite to the reported increase of anxiety in mice lacking other proinflammatory cytokines or their receptors. The reduced anxiety was not associated with changes in associated serum cytokines. However, Western blotting, qPCR, and immunohistochemistry all showed that IL15Rα knockout mice had mild microgliosis and astrogliosis in the hippocampus. To determine whether this gliosis plays a role in decreasing anxiety, IL15Rα knockout mice were treated with minocycline, but this did not cause a change in open field performance. To determine whether IL15 plays a direct role in anxiety, wildtype mice were treated with IL15 by intraperitoneal injection. This also failed to cause a change in open field behavior under the experimental conditions tested. Thus, IL15Rα is essential for normal anxiety-like behavior, but inhibition of gliosis in the fearless IL15Rα knockout mice or IL15 treatment of normal mice did not acutely modulate behavioral performance as tested.
Collapse
|
27
|
Pan W, Wu X, Kastin AJ, Zhang Y, Hsuchou H, Halberg F, Chatu F, Khan RS, Robert B, Cornelissen-Guillaume GG. Potential protective role of IL15Rα during inflammation. J Mol Neurosci 2010; 43:412-23. [PMID: 20981579 DOI: 10.1007/s12031-010-9459-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Accepted: 09/27/2010] [Indexed: 10/18/2022]
Abstract
We have shown that TNFα specifically activates the interleukin-15 (IL15) system in cerebral endothelial cells composing the blood-brain barrier. To determine the functions of cerebral IL15 signaling in inflammation, we first treated mice with lipopolysaccharide (LPS) and determined the expression of the three receptor subtypes of IL15. Robust time-dependent upregulation occurred in all subunits. We then tested whether IL15Rα knockout (KO) affected the maintenance of body temperature and activity level after a single dose of LPS. Circadian telemetry data were analyzed by the cosinor method. Both wild-type and KO mice had clear 24-h rhythms of basal temperature and activity. KO mice had a significantly higher midline estimating statistic of rhythm (MESOR; approximating 24 h mean) of temperature and delayed 24-h acrophase (peak) of activity than the wild-type mice. LPS disrupted the circadian rhythm of activity more severely in the KO group. Besides a decrease in MESOR and 24-h amplitude of activity after LPS, the KO mice showed a significant reduction of MESOR, amplitude, and changed acrophase for temperature on the second of 2 days. The disrupted circadian rhythm of temperature and activity in the KO mice after LPS suggests that upregulated IL15 receptors may serve a beneficial role to counteract the consequences of neuroinflammation.
Collapse
Affiliation(s)
- Weihong Pan
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wu X, Kastin AJ, Hsuchou H, Pan W. The effects of IL2Rgamma knockout on depression and contextual memory. Behav Brain Res 2010; 213:319-22. [PMID: 20438766 DOI: 10.1016/j.bbr.2010.04.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 04/23/2010] [Accepted: 04/27/2010] [Indexed: 12/29/2022]
Abstract
Interleukin (IL)-2Rgamma shows robust upregulation in neuroinflammatory states associated with clinical depression. We tested the hypothesis that mice lacking IL2Rgamma would have decreased depressive-like behavior. Contrary to this expectation, these knockout mice showed increased immobility in both the Porsolt forced swimming and Nomura water wheel tests. By comparison, the auditory fear conditioning test showed increased retention of contextual freezing. Thus, intact IL2Rgamma combats depressive-like behavior.
Collapse
Affiliation(s)
- Xiaojun Wu
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | | | | | | |
Collapse
|
29
|
Wu X, Pan W, He Y, Hsuchou H, Kastin AJ. Cerebral interleukin-15 shows upregulation and beneficial effects in experimental autoimmune encephalomyelitis. J Neuroimmunol 2010; 223:65-72. [PMID: 20430449 DOI: 10.1016/j.jneuroim.2010.04.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 03/31/2010] [Accepted: 04/01/2010] [Indexed: 01/05/2023]
Abstract
Interleukin (IL)-15 can cross the blood-brain barrier to act on its specific brain receptor (IL15Ralpha) and co-receptors. The important roles of neuronal IL15 and IL15Ralpha in experimental autoimmune encephalomeylitis (EAE) are suggested by the upregulation of IL15Ralpha mRNA in different regions of the brain and spinal cord, and by double-labeling immunohistochemistry showing neuronal localization of IL15 and IL15Ralpha in different neurons. Contrary to expectations, IL15 treatment lessened EAE severity. IL15 knockout mice showed heightened susceptibility to EAE with significantly higher scores that were decreased by treatment with IL15. Thus, IL15 improves this CNS autoimmune disorder as a potential therapeutic agent.
Collapse
MESH Headings
- Adjuvants, Immunologic/biosynthesis
- Adjuvants, Immunologic/deficiency
- Adjuvants, Immunologic/physiology
- Adjuvants, Immunologic/therapeutic use
- Animals
- Brain/immunology
- Brain/metabolism
- Brain/pathology
- Encephalomyelitis, Autoimmune, Experimental/diagnosis
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Female
- Genetic Predisposition to Disease/etiology
- Interleukin-15/biosynthesis
- Interleukin-15/deficiency
- Interleukin-15/physiology
- Interleukin-15/therapeutic use
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neurons/immunology
- Neurons/metabolism
- Neurons/pathology
- Receptors, Interleukin-15/biosynthesis
- Receptors, Interleukin-15/deficiency
- Receptors, Interleukin-15/genetics
- Receptors, Interleukin-15/physiology
- Severity of Illness Index
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Xiaojun Wu
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | | | | | | |
Collapse
|