1
|
Willford JA, Kaufman JM. Through a teratological lens: A narrative review of exposure to stress and drugs of abuse during pregnancy on neurodevelopment. Neurotoxicol Teratol 2024; 105:107384. [PMID: 39187031 DOI: 10.1016/j.ntt.2024.107384] [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: 02/19/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
Teratological research shows that both prenatal stress and prenatal substance exposure have a significant impact on neurodevelopmental outcomes in children. Using human research, the purpose of this narrative review is to explore the degree to which these exposures may represent complex prenatal and postnatal risks for the development of cognition and behavior in children. An understanding of the HPA axis and its function during pregnancy as well as the types and operationalization of prenatal stress provide a context for understanding the direct and indirect mechanisms by which prenatal stress affects brain and behavior development. In turn, prenatal substance exposure studies are evaluated for their importance in understanding variables that indicate a potential interaction with prenatal stress including reactivity to novelty, arousal, and stress reactivity during early childhood. The similarities and differences between prenatal stress exposure and prenatal substance exposure on neurodevelopmental outcomes including arousal and emotion regulation, cognition, behavior, stress reactivity, and risk for psychopathology are summarized. Further considerations for teratological studies of prenatal stress and/or substance exposure include identifying and addressing methodological challenges, embracing the complexity of pre-and postnatal environments in the research, and the importance of incorporating parenting and resilience into future studies.
Collapse
Affiliation(s)
- Jennifer A Willford
- Slippery Rock University, Department of Psychology, 1 Morrow Way, Slippery Rock, PA 16057, United States of America.
| | - Jesse M Kaufman
- Slippery Rock University, Department of Psychology, 1 Morrow Way, Slippery Rock, PA 16057, United States of America
| |
Collapse
|
2
|
Deng S, Guo A, Huang Z, Guan K, Zhu Y, Chan C, Gui J, Song C, Li X. The exploration of neuroinflammatory mechanism by which CRHR2 deficiency induced anxiety disorder. Prog Neuropsychopharmacol Biol Psychiatry 2024; 128:110844. [PMID: 37640149 DOI: 10.1016/j.pnpbp.2023.110844] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/07/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
Inflammation stimulates the hypothalamic-pituitary adrenal (HPA) axis and triggers glial neuroinflammatory phenotypes, which reduces monoamine neurotransmitters by activating indoleamine 2,3-dioxygenase enzyme. These changes can induce psychiatric diseases, including anxiety. Corticotropin releasing hormone receptor 2 (CRHR2) in the HPA axis is involved in the etiology of anxiety. Omega(n)-3 polyunsaturated fatty acids (PUFAs) can attenuate anxiety through anti-inflammation and HPA axis modulation. However, the underlying molecular mechanism by CRHR2 modulates anxiety and its correlation with neuroinflammation remain unclear. Here, we first constructed a crhr2 zebrafish mutant line, and evaluated anxiety-like behaviors, gene expression associated with the HPA axis, neuroinflammatory response, neurotransmitters, and PUFAs profile in crhr2+/+ and crhr2-/- zebrafish. The crhr2 deficiency decreased cortisol levels and up-regulated crhr1 and down-regulated crhb, crhbp, ucn3l and proopiomelanocortin a (pomc a) in zebrafish. Interestingly, a significant increase in the neuroinflammatory markers, translocator protein (TSPO) and the activation of microglia M1 phenotype (CD11b) were found in crhr2-/- zebrafish. As a consequence, the expression of granulocyte-macrophage colony-stimulating factor, pro-inflammatory cytokines vascular endothelial growth factor, and astrocyte A1 phenotype c3 were up-regulated. While microglia anti-inflammatory phenotype (CD206), central anti-inflammatory cytokine interleukin-4, arginase-1, and transforming growth factor-β were downregulated. In parallel, crhr2-deficient zebrafish showed an upregulation of vdac1 protein, a TSPO ligand, and its downstream caspase-3. Furthermore, 5-HT/5-HIAA ratio was decreased and n-3 PUFAs deficiency was identified in crhr2-/- zebrafish. In conclusion, anxiety-like behavior displayed by crhr2-deficient zebrafish may be caused by the HPA axis dysfunction and enhanced neuroinflammation, which resulted in n-3 PUFAs and monoamine neurotransmitter reductions.
Collapse
Affiliation(s)
- Shuyi Deng
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Anqi Guo
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China
| | - Zhengwei Huang
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China
| | - Kaiyu Guan
- Wenzhou Seventh People's Hospital, Wenzhou, Zhejiang 325000, China
| | - Ya Zhu
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Cheekai Chan
- College of Science and Technology, Wenzhou-Kean University, Zhejiang 325000, China
| | - Jianfang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Cai Song
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Xi Li
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
3
|
Panda SP, Kesharwani A, Singh GD, Prasanth D, Vatchavai BR, Kumari PVK, Panda SK, Mallick SP. Impose of KNDy/GnRH neural circuit in PCOS, ageing, cancer and Alzheimer's disease: StAR actions in prevention of neuroendocrine dysfunction. Ageing Res Rev 2023; 92:102086. [PMID: 37821047 DOI: 10.1016/j.arr.2023.102086] [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: 08/22/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The Kisspeptin1 (KISS1)/neurokinin B (NKB)/Dynorphin (Dyn) [KNDy] neurons in the hypothalamus regulate the reproduction stage in human beings and rodents. KNDy neurons co-expressed all KISS1, NKB, and Dyn peptides, and hence commonly regarded as KISS1 neurons. KNDy neurons contribute to the "GnRH pulse generator" and are implicated in the regulation of pulsatile GnRH release. The estradiol (E2)-estrogen receptor (ER) interactions over GnRH neurons in the hypothalamus cause nitric oxide (NO) discharge, in addition to presynaptic GABA and glutamate discharge from respective neurons. The released GABA and glutamate facilitate the activity of GnRH neurons via GABAA-R and AMPA/kainate-R. The KISS1 stimulates MAPK/ERK1/2 signaling and cause the release of Ca2+ from intracellular store, which contribute to neuroendocrine function, increase apoptosis and decrease cell proliferation and metastasis. The ageing in women deteriorates KISS1/KISS1R interaction in the hypothalamus which causes lower levels of GnRH. Because examining the human brain is so challenging, decades of clinical research have failed to find the causes of KNDy/GnRH dysfunction. The KISS1/KISS1R interactions in the brain have a neuroprotective effect against Alzheimer's disease (AD). These findings modulate the pathophysiological role of the KNDy/GnRH neural network in polycystic ovarian syndrome (PCOS) associated with ageing and, its protective role in cancer and AD. This review concludes with protecting effect of the steroid-derived acute regulatory enzyme (StAR) against neurotoxicity in the hippocampus, and hypothalamus, and these measures are fundamental for delaying ageing with PCOS. StAR could serve as novel diagnostic marker and therapeutic target for the most prevalent hormone-sensitive breast cancers (BCs).
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | | | - Dsnbk Prasanth
- KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, Andhrapradesh, India
| | - Bhaskara Raju Vatchavai
- Sri Vasavi Institute of Pharmaceutical Sciences, Pedatadepalli, Tadepalligudem, Andhrapradesh, India
| | - P V Kamala Kumari
- Vignan Institute of Pharmaceutical Technology, Duvvada, Visakhapatnam, Andhrapradesh, India
| | | | | |
Collapse
|
4
|
García-Marín LM, Reyes-Pérez P, Diaz-Torres S, Medina-Rivera A, Martin NG, Mitchell BL, Rentería ME. Shared molecular genetic factors influence subcortical brain morphometry and Parkinson's disease risk. NPJ Parkinsons Dis 2023; 9:73. [PMID: 37164954 PMCID: PMC10172359 DOI: 10.1038/s41531-023-00515-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/28/2023] [Indexed: 05/12/2023] Open
Abstract
Parkinson's disease (PD) is a late-onset and genetically complex neurodegenerative disorder. Here we sought to identify genes and molecular pathways underlying the associations between PD and the volume of ten brain structures measured through magnetic resonance imaging (MRI) scans. We leveraged genome-wide genetic data from several cohorts, including the International Parkinson's Disease Genomics Consortium (IPDG), the UK Biobank, the Adolescent Brain Cognitive Development (ABCD) study, the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE), the Enhancing Neuroimaging Genetics through Meta-Analyses (ENIGMA), and 23andMe. We observed significant positive genetic correlations between PD and intracranial and subcortical brain volumes. Genome-wide association studies (GWAS) - pairwise analyses identified 210 genomic segments with shared aetiology between PD and at least one of these brain structures. Pathway enrichment results highlight potential links with chronic inflammation, the hypothalamic-pituitary-adrenal pathway, mitophagy, disrupted vesicle-trafficking, calcium-dependent, and autophagic pathways. Investigations for putative causal genetic effects suggest that a larger putamen volume could influence PD risk, independently of the potential causal genetic effects of intracranial volume (ICV) on PD. Our findings suggest that genetic variants influencing larger intracranial and subcortical brain volumes, possibly during earlier stages of life, influence the risk of developing PD later in life.
Collapse
Affiliation(s)
- Luis M García-Marín
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
- Laboratorio Internacional de Investigación del Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México.
| | - Paula Reyes-Pérez
- Laboratorio Internacional de Investigación del Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Santiago Diaz-Torres
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Alejandra Medina-Rivera
- Laboratorio Internacional de Investigación del Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Nicholas G Martin
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Brittany L Mitchell
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Miguel E Rentería
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
5
|
Kwon M, Lee JH, Yoon Y, Pleasure SJ, Yoon K. The CRHR1/CREB/REST signaling cascade regulates mammalian embryonic neural stem cell properties. EMBO Rep 2023; 24:e55313. [PMID: 36413000 PMCID: PMC9900344 DOI: 10.15252/embr.202255313] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/31/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
Growing evidence suggests that the corticotropin-releasing hormone (CRH) signaling pathway, mainly known as a critical initiator of humoral stress responses, has a role in normal neuronal physiology. However, despite the evidence of CRH receptor (CRHR) expression in the embryonic ventricular zone, the exact functions of CRH signaling in embryonic brain development have not yet been fully determined. In this study, we show that CRHR1 is required for the maintenance of neural stem cell properties, as assessed by in vitro neurosphere assays and cell distribution in the embryonic cortical layers following in utero electroporation. Identifying the underlying molecular mechanisms of CRHR1 action, we find that CRHR1 functions are accomplished through the increasing expression of the master transcription factor REST. Furthermore, luciferase reporter and chromatin immunoprecipitation assays reveal that CRHR1-induced CREB activity is responsible for increased REST expression at the transcriptional level. Taken together, these findings indicate that the CRHR1/CREB/REST signaling cascade plays an important role downstream of CRH in the regulation of neural stem cells during embryonic brain development.
Collapse
Affiliation(s)
- Mookwang Kwon
- Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwonSouth Korea
| | - Ju Hyun Lee
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwonSouth Korea
| | - Youngik Yoon
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwonSouth Korea
| | - Samuel J Pleasure
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Keejung Yoon
- Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwonSouth Korea
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwonSouth Korea
| |
Collapse
|
6
|
Leschik J, Gentile A, Cicek C, Péron S, Tevosian M, Beer A, Radyushkin K, Bludau A, Ebner K, Neumann I, Singewald N, Berninger B, Lessmann V, Lutz B. Brain-derived neurotrophic factor expression in serotonergic neurons improves stress resilience and promotes adult hippocampal neurogenesis. Prog Neurobiol 2022; 217:102333. [PMID: 35872219 DOI: 10.1016/j.pneurobio.2022.102333] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 06/24/2022] [Accepted: 07/19/2022] [Indexed: 11/19/2022]
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) stimulates adult neurogenesis, but also influences structural plasticity and function of serotonergic neurons. Both, BDNF/TrkB signaling and the serotonergic system modulate behavioral responses to stress and can lead to pathological states when dysregulated. The two systems have been shown to mediate the therapeutic effect of antidepressant drugs and to regulate hippocampal neurogenesis. To elucidate the interplay of both systems at cellular and behavioral levels, we generated a transgenic mouse line that overexpresses BDNF in serotonergic neurons in an inducible manner. Besides displaying enhanced hippocampus-dependent contextual learning, transgenic mice were less affected by chronic social defeat stress (CSDS) compared to wild-type animals. In parallel, we observed enhanced serotonergic axonal sprouting in the dentate gyrus and increased neural stem/progenitor cell proliferation, which was uniformly distributed along the dorsoventral axis of the hippocampus. In the forced swim test, BDNF-overexpressing mice behaved similarly as wild-type mice treated with the antidepressant fluoxetine. Our data suggest that BDNF released from serotonergic projections exerts this effect partly by enhancing adult neurogenesis. Furthermore, independently of the genotype, enhanced neurogenesis positively correlated with the social interaction time after the CSDS, a measure for stress resilience.
Collapse
Affiliation(s)
- Julia Leschik
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany.
| | - Antonietta Gentile
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Department of Systems Medicine, Tor Vergata University, Rome 00183, Italy
| | - Cigdem Cicek
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Faculty of Medicine, Department of Medical Biochemistry, Hacettepe University, 06100 Ankara, Turkey; Faculty of Medicine, Department of Medical Biochemistry, Yuksek Ihtisas University, 06520 Ankara, Turkey
| | - Sophie Péron
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Institute of Psychiatry, Psychology & Neuroscience, Centre for Developmental Neurobiology, King's College London, London SE11UL, United Kingdom
| | - Margaryta Tevosian
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| | - Annika Beer
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| | | | - Anna Bludau
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg 93053, Germany
| | - Karl Ebner
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, Leopold Franzens University Innsbruck, Innsbruck 6020, Austria
| | - Inga Neumann
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg 93053, Germany
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, Leopold Franzens University Innsbruck, Innsbruck 6020, Austria
| | - Benedikt Berninger
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Institute of Psychiatry, Psychology & Neuroscience, Centre for Developmental Neurobiology, King's College London, London SE11UL, United Kingdom; Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE11UL, United Kingdom
| | - Volkmar Lessmann
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39120, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| |
Collapse
|
7
|
Roque A, Valles Méndez KM, Ruiz R, Pineda E, Lajud N. Early life stress induces a transient increase in hippocampal corticotropin-releasing hormone in rat neonates that precedes the effects on hypothalamic neuropeptides. Eur J Neurosci 2022; 55:2108-2121. [PMID: 33745155 DOI: 10.1111/ejn.15193] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
Early life stress (ELS) programs hypothalamus-pituitary-adrenal (HPA) axis activity and affects synaptic plasticity and cognitive performance in adults; however, the effects of ELS during the temporal window of vulnerability are poorly understood. This study aimed to thoroughly characterize the effects of ELS in the form of periodic maternal separation (MS180) during the time of exposure to stress. Hippocampal corticotropin-releasing hormone (CRH) gene expression and baseline HPA axis activity were analyzed at postnatal (P) days 6, 12, 15, and 21, and in adulthood (P75); these factors were correlated with plasticity markers and adult behavior. Our results indicate that MS180 induces an increase in hippocampal CRH expression at P9, P12, and P15, whereas an increase in hypothalamic CRH expression was observed from P12 to P21. Increased arginine-vasopressin expression and corticosterone levels were observed only at P21. Moreover, MS180 caused transient alterations in hypothalamic synaptophysin expression during early life. As adults, MS180 rats showed a passive coping strategy in the forced swimming test, cognitive impairments in the object location test, increased hypothalamic CRH expression, and decreased oxytocin (OXT) expression. Spearman's analysis indicated that cognitive impairments correlated with CRH and OXT expression. In conclusion, our data indicate that MS180 induces a transient increase in hippocampal CRH expression in neonates that precedes the effects on hypothalamic neuropeptides, confirming the role of increased CRH during the temporal window of vulnerability as a mediator of some of the detrimental effects of ELS on brain development and adult behavior.
Collapse
Affiliation(s)
- Angélica Roque
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Kinberli Marcela Valles Méndez
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Roberto Ruiz
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Edel Pineda
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Naima Lajud
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| |
Collapse
|
8
|
Kassotaki I, Valsamakis G, Mastorakos G, Grammatopoulos DK. Placental CRH as a Signal of Pregnancy Adversity and Impact on Fetal Neurodevelopment. Front Endocrinol (Lausanne) 2021; 12:714214. [PMID: 34408727 PMCID: PMC8366286 DOI: 10.3389/fendo.2021.714214] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/09/2021] [Indexed: 11/13/2022] Open
Abstract
Early life is a period of considerable plasticity and vulnerability and insults during that period can disrupt the homeostatic equilibrium of the developing organism, resulting in adverse developmental programming and enhanced susceptibility to disease. Fetal exposure to prenatal stress can impede optimum brain development and deranged mother's hypothalamic-pituitary-adrenal axis (HPA axis) stress responses can alter the neurodevelopmental trajectories of the offspring. Corticotropin-releasing hormone (CRH) and glucocorticoids, regulate fetal neurogenesis and while CRH exerts neuroprotective actions, increased levels of stress hormones have been associated with fetal brain structural alterations such as reduced cortical volume, impoverishment of neuronal density in the limbic brain areas and alterations in neuronal circuitry, synaptic plasticity, neurotransmission and G-protein coupled receptor (GPCR) signalling. Emerging evidence highlight the role of epigenetic changes in fetal brain programming, as stress-induced methylation of genes encoding molecules that are implicated in HPA axis and major neurodevelopmental processes. These serve as molecular memories and have been associated with long term modifications of the offspring's stress regulatory system and increased susceptibility to psychosomatic disorders later in life. This review summarises our current understanding on the roles of CRH and other mediators of stress responses on fetal neurodevelopment.
Collapse
Affiliation(s)
- Ifigeneia Kassotaki
- Department of Internal Medicine, 2nd Internal Medicine Clinic, Venizeleio Pananeio General Hospital, Heraklion, Greece
| | - Georgios Valsamakis
- Second University Department of Obs and Gynae, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Translational Medicine, Warwick Medical School, Coventry, United Kingdom
| | - George Mastorakos
- Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris K. Grammatopoulos
- Translational Medicine, Warwick Medical School, Coventry, United Kingdom
- Institute of Precision Diagnostics and Translational Medicine, Pathology, University Hospitals Coventry and Warwickshire (UHCW) NHS Trust, Coventry, United Kingdom
| |
Collapse
|
9
|
Ortega VA, Mercer EM, Giesbrecht GF, Arrieta MC. Evolutionary Significance of the Neuroendocrine Stress Axis on Vertebrate Immunity and the Influence of the Microbiome on Early-Life Stress Regulation and Health Outcomes. Front Microbiol 2021; 12:634539. [PMID: 33897639 PMCID: PMC8058197 DOI: 10.3389/fmicb.2021.634539] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Stress is broadly defined as the non-specific biological response to changes in homeostatic demands and is mediated by the evolutionarily conserved neuroendocrine networks of the hypothalamus-pituitary-adrenal (HPA) axis and the sympathetic nervous system. Activation of these networks results in transient release of glucocorticoids (cortisol) and catecholamines (epinephrine) into circulation, as well as activation of sympathetic fibers innervating end organs. These interventions thus regulate numerous physiological processes, including energy metabolism, cardiovascular physiology, and immunity, thereby adapting to cope with the perceived stressors. The developmental trajectory of the stress-axis is influenced by a number of factors, including the gut microbiome, which is the community of microbes that colonizes the gastrointestinal tract immediately following birth. The gut microbiome communicates with the brain through the production of metabolites and microbially derived signals, which are essential to human stress response network development. Ecological perturbations to the gut microbiome during early life may result in the alteration of signals implicated in developmental programming during this critical window, predisposing individuals to numerous diseases later in life. The vulnerability of stress response networks to maladaptive development has been exemplified through animal models determining a causal role for gut microbial ecosystems in HPA axis activity, stress reactivity, and brain development. In this review, we explore the evolutionary significance of the stress-axis system for health maintenance and review recent findings that connect early-life microbiome disturbances to alterations in the development of stress response networks.
Collapse
Affiliation(s)
- Van A Ortega
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, Cumming School of Medicine, Health Sciences Centre, University of Calgary, Calgary, AB, Canada
| | - Emily M Mercer
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, Cumming School of Medicine, Health Sciences Centre, University of Calgary, Calgary, AB, Canada.,Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| | - Gerald F Giesbrecht
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada.,Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada.,Owerko Centre, The Alberta Children's Hospital Research Institute, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, Cumming School of Medicine, Health Sciences Centre, University of Calgary, Calgary, AB, Canada.,Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
10
|
Bourin M. Neurogenesis and Neuroplasticity in Major Depression: Its Therapeutic Implication. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1305:157-173. [PMID: 33834400 DOI: 10.1007/978-981-33-6044-0_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The neurochemical model of depression, based on monoaminergic theories, does not allow on its own to understand the mechanism of action of antidepressants. This approach does not explain the gap between the immediate biochemical modulations induced by antidepressants and the time required for their clinical action. Several hypotheses have been developed to try to explain more precisely the action of these molecules, each of them involving mechanisms of receptor regulation. At the same time, data on the neuroanatomy of depression converge toward the existence of specific lesions of this pathology. This chapter aims to provide an overview of recent advances in understanding the mechanisms of neural plasticity involved in pathophysiology depression and in its treatment.
Collapse
Affiliation(s)
- Michel Bourin
- Neurobiology of Mood Disorders, University of Nantes, Nantes, France.
| |
Collapse
|
11
|
Abstract
In a number of adult tissues, Nestin-positive stem cells/progenitors have been identified and shown to be involved in maintenance and remodeling. Various studies have shown that under stressful conditions, quiescent Nestin-positive progenitor cells are activated. Thereby, they migrate to their target location and differentiate into mature cells. In the current paper, we discuss if Nestin-positive progenitors in the hippocampus and adrenal gland belong to unique cell populations that are responsive to stress. Furthermore, we speculate about the mechanism behind their activation and the clinical importance of this stress-response.
Collapse
Affiliation(s)
- Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Diabetes and Nutritional Sciences Division, King's College London, London, UK
| | - Ilona Berger
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
12
|
Identifying Risk Genes and Interpreting Pathogenesis for Parkinson's Disease by a Multiomics Analysis. Genes (Basel) 2020; 11:genes11091100. [PMID: 32967142 PMCID: PMC7563773 DOI: 10.3390/genes11091100] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified tens of genetic variants associated with Parkinson’s disease (PD). Nevertheless, the genes or DNA elements that affect traits through these genetic variations are usually undiscovered. This study was the first to combine meta-analysis GWAS data and expression data to identify PD risk genes. Four known genes, CRHR1, KANSL1, NSF and LRRC37A, and two new risk genes, STX4 and BST1, were identified. Among them, CRHR1 is a known drug target, indicating that hydrocortisone may become a potential drug for the treatment of PD. Furthermore, the potential pathogenesis of CRHR1 and LRRC37A was explored by applying DNA methylation (DNAm) data, indicating a pathogenesis whereby the effect of a genetic variant on PD is mediated by genetic regulation of transcription through DNAm. Overall, this research identified the risk genes and pathogenesis that affect PD through genetic variants, which has significance for the diagnosis and treatment of PD.
Collapse
|
13
|
CRH Promotes the Neurogenic Activity of Neural Stem Cells in the Adult Hippocampus. Cell Rep 2020; 29:932-945.e7. [PMID: 31644914 DOI: 10.1016/j.celrep.2019.09.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/17/2018] [Accepted: 09/12/2019] [Indexed: 02/07/2023] Open
Abstract
Local cues in the adult neurogenic niches dynamically regulate homeostasis in neural stem cells, whereas their identity and associated molecular mechanisms remain poorly understood. Here, we show that corticotropin-releasing hormone (CRH), the major mediator of mammalian stress response and a key neuromodulator in the adult brain, is necessary for hippocampal neural stem cell (hiNSC) activity under physiological conditions. In particular, we demonstrate functionality of the CRH/CRH receptor (CRHR) system in mouse hiNSCs and conserved expression in humans. Most important, we show that genetic deficiency of CRH impairs hippocampal neurogenesis, affects spatial memory, and compromises hiNSCs' responsiveness to environmental stimuli. These deficits have been partially restored by virus-mediated CRH expression. Additionally, we provide evidence that local disruption of the CRH/CRHR system reduces neurogenesis, while exposure of adult hiNSCs to CRH promotes neurogenic activity via BMP4 suppression. Our findings suggest a critical role of CRH in adult neurogenesis, independently of its stress-related systemic function.
Collapse
|
14
|
Vasconcelos M, Stein DJ, Gallas-Lopes M, Landau L, de Almeida RMM. Corticotropin-releasing factor receptor signaling and modulation: implications for stress response and resilience. TRENDS IN PSYCHIATRY AND PSYCHOTHERAPY 2020; 42:195-206. [PMID: 32696892 DOI: 10.1590/2237-6089-2018-0027] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/25/2019] [Indexed: 11/22/2022]
Abstract
Introduction In addition to their role in regulation of the hypothalamic-pituitary-adrenal-axis, corticotropin-releasing factor (CRF) and its related peptides, the urocortins, are important mediators of physiological and pathophysiological processes of the central nervous, cardiovascular, gastrointestinal, immune, endocrine, reproductive, and skin systems. Altered regulation of CRF-mediated adaptive responses to various stressful stimuli disrupts healthy function and might confer vulnerability to several disorders, including depression and anxiety. Methodology This narrative review was conducted through search and analysis of studies retrieved from online databases using a snowball method. Results This review covers aspects beginning with the discovery of CRF, CRF binding protein and their actions via interaction with CRF receptors type 1 and type 2. These are surface plasma membrane receptors, activation of which is associated with conformational changes and interaction with a variety of G-proteins and signaling pathways. We also reviewed the pharmacology and mechanisms of the receptor signaling modulatory activity of these receptors. Conclusion This review compiles and presents knowledge regarding the CRFergic system, including CRF related peptides, CRF binding protein, and CRF receptors, as well as some evidence that is potentially indicative of the biological roles of these entities in several physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Mailton Vasconcelos
- Instituto de Psicologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Dirson J Stein
- Hospital de Clínicas de Porto Alegre, UFRGS, Porto Alegre, RS, Brazil
| | - Matheus Gallas-Lopes
- Instituto de Psicologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luane Landau
- Instituto de Psicologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rosa Maria M de Almeida
- Instituto de Psicologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
15
|
Williams TA, Bernier NJ. Corticotropin-releasing factor protects against ammonia neurotoxicity in isolated larval zebrafish brains. J Exp Biol 2020; 223:jeb211540. [PMID: 31988165 DOI: 10.1242/jeb.211540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/20/2020] [Indexed: 08/26/2023]
Abstract
The physiological roles of corticotropin-releasing factor (CRF) have recently been extended to cytoprotection. Here, to determine whether CRF is neuroprotective in fish, the effects of CRF against high environmental ammonia (HEA)-mediated neurogenic impairment and cell death were investigated in zebrafish. In vivo, exposure of 1 day post-fertilization (dpf) embryos to HEA only reduced the expression of the determined neuron marker neurod1 In contrast, in 5 dpf larvae, HEA increased the expression of nes and sox2, neural progenitor cell markers, and reduced the expression of neurog1, gfap and mbpa, proneuronal cell, radial glia and oligodendrocyte markers, respectively, and neurod1 The N-methyl-d-aspartate (NMDA) receptor inhibitor MK801 rescued the HEA-induced reduction in neurod1 in 5 dpf larvae but did not affect the HEA-induced transcriptional changes in other neural cell types, suggesting that hyperactivation of NMDA receptors specifically contributes to the deleterious effects of HEA in determined neurons. As observed in vivo, HEA exposure elicited marked changes in the expression of cell type-specific markers in isolated 5 dpf larval brains. The addition of CRF reversed the in vitro effects of HEA on neurod1 expression and prevented an HEA-induced increase in cell death. Finally, the protective effects of CRF against HEA-mediated neurogenic impairment and cell death were prevented by the CRF type 1 receptor selective antagonist antalarmin. Together, these results provide novel evidence that HEA has developmental time- and cell type-specific neurotoxic effects, that NMDA receptor hyperactivation contributes to HEA-mediated impairment of determined neurons, and that CRF has neuroprotective properties in the larval zebrafish brain.
Collapse
Affiliation(s)
- Tegan A Williams
- Department of Integrative Biology, University of Guelph, Guelph, ON, Canada N1G 2W1
| | - Nicholas J Bernier
- Department of Integrative Biology, University of Guelph, Guelph, ON, Canada N1G 2W1
| |
Collapse
|
16
|
Solomon ER, Caldwell KK, Allan AM. Developmental arsenic exposure is associated with sex differences in the epigenetic regulation of stress genes in the adult mouse frontal cortex. Toxicol Appl Pharmacol 2020; 391:114920. [PMID: 32061746 PMCID: PMC7948303 DOI: 10.1016/j.taap.2020.114920] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 11/15/2022]
Affiliation(s)
- Elizabeth R Solomon
- Department of Neurosciences, School of Medicine, University of New Mexico HSC, MSC08 4740, 1 University of New Mexico, Albuquerque, NM 87131-0001, United States
| | - Kevin K Caldwell
- Department of Neurosciences, School of Medicine, University of New Mexico HSC, MSC08 4740, 1 University of New Mexico, Albuquerque, NM 87131-0001, United States
| | - Andrea M Allan
- Department of Neurosciences, School of Medicine, University of New Mexico HSC, MSC08 4740, 1 University of New Mexico, Albuquerque, NM 87131-0001, United States.
| |
Collapse
|
17
|
Lavin KM, Sealfon SC, McDonald MLN, Roberts BM, Wilk K, Nair VD, Ge Y, Lakshman Kumar P, Windham ST, Bamman MM. Skeletal muscle transcriptional networks linked to type I myofiber grouping in Parkinson's disease. J Appl Physiol (1985) 2020; 128:229-240. [PMID: 31829804 PMCID: PMC7052589 DOI: 10.1152/japplphysiol.00702.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/20/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder impacting cognition, movement, and quality of life in >10 million individuals worldwide. We recently characterized and quantified a skeletal muscle pathology in PD represented by exaggerated type I myofiber grouping presumed to result from denervation-reinnervation processes. Our previous findings indicated that impaired neuromuscular junction integrity may be involved in type I grouping, which is associated with excessive motor unit activation during weight-bearing tasks. In this study, we performed transcriptional profiling to test the hypothesis that type I grouping severity would link to distinct gene expression networks. We generated transcriptome-wide poly(A) RNA-Seq data from skeletal muscle of individuals with PD [n = 12 (9 men, 3 women); 67 ± 2 yr], age- and sex-matched older adults (n = 12; 68 ± 2 yr), and sex-matched young adults (n = 12; 30 ± 1 yr). Differentially expressed genes were evaluated across cohorts. Weighted gene correlation network analysis (WGCNA) was performed to identify gene networks most correlated with indicators of abnormal type I grouping. Among coexpression networks mapping to phenotypes pathologically increased in PD muscle, one network was highly significantly correlated to type I myofiber group size and another to percentage of type I myofibers found in groups. Annotation of coexpressed networks revealed that type I grouping is associated with altered expression of genes involved in neural development, postsynaptic signaling, cell cycle regulation and cell survival, protein and energy metabolism, inflammation/immunity, and posttranscriptional regulation (microRNAs). These transcriptomic findings suggest that skeletal muscle may play an active role in signaling to promote myofiber survival, reinnervation, and remodeling, perhaps to an extreme in PD.NEW & NOTEWORTHY Despite our awareness of the impact of Parkinson's disease (PD) on motor function for over two centuries, limited attention has focused on skeletal muscle. We previously identified type I myofiber grouping, a novel indicator of muscle dysfunction in PD, presumably a result of heightened rates of denervation/reinnervation. Using transcriptional profiling to identify networks associated with this phenotype, we provide insight into potential mechanistic roles of skeletal muscle in signaling to promote its survival in PD.
Collapse
Affiliation(s)
- Kaleen M Lavin
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stuart C Sealfon
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Merry-Lynn N McDonald
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Brandon M Roberts
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Katarzyna Wilk
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Venugopalan D Nair
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Preeti Lakshman Kumar
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Samuel T Windham
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Marcas M Bamman
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Geriatric Research, Education, and Clinical Center, Department of Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
18
|
Melanoma and autoimmunity: spontaneous regressions as a possible model for new therapeutic approaches. Melanoma Res 2019; 29:231-236. [PMID: 30615013 DOI: 10.1097/cmr.0000000000000573] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Until now, malignancy has been considered a cellular problem represented by the perturbed (uncontrolled) division of the cells associated with invasion and metastasis. Contrary to this classical approach, a new perspective suggests that cancerous disease is, in fact, a supracellular problem represented by inadequate evolution of complex supracellular processes (embryogenesis, development, regeneration, etc.). Such complex processes would be disconnected from the real needs of the body, inducing unnecessary or even dangerous events such as an exacerbated rate of the cell division, angiogenesis, immunosuppression (specific to embryogenesis and melanoma), invasion (mediated by trophoblastic/placental factors in melanoma), and migration (specific to neural crest cells, which generate melanocytes - the most common origin for melanoma). As a result, a correct and comprehensive interpretation of cancer (causes, evolution, therapy, and prevention) should be conducted from a supracellular perspective. After presenting the supracellular perspective, this article further investigates the favorable evolution of malignant melanoma in two distinct situations: in patients receiving no therapy and in patients treated with immune-checkpoint inhibitors. In patients receiving no therapy, spontaneous regressions of melanoma could be the result of several autoimmune reactions (inducing not only melanoma regression but also vitiligo, an autoimmune event frequently associated with melanoma). Patients treated with immune-checkpoint inhibitors develop similar autoimmune reactions, which are clearly correlated with better therapeutic results. The best example is vitiligo, which is considered a positive prognostic factor for patients receiving immune-checkpoint inhibitors. This finding indicates that immune-checkpoint inhibitors induce distinct types of autoimmune events, some corresponding to specific favorable autoimmune mechanisms (favoring tumor regression) and others to common unfavorable adverse reactions (which should be avoided or minimized). In conclusion, the spectrum of autoimmune reactions induced by immune-checkpoint inhibitors should be restricted in the near future to only these specific favorable autoimmune mechanisms. In this way, the unnecessary autoimmune reactions/autoaggressions could be avoided (a better quality of life), and treatment specificity and efficiency should increase (a higher response rate for melanoma therapy).
Collapse
|
19
|
Jiang Y, Peng T, Gaur U, Silva M, Little P, Chen Z, Qiu W, Zhang Y, Zheng W. Role of Corticotropin Releasing Factor in the Neuroimmune Mechanisms of Depression: Examination of Current Pharmaceutical and Herbal Therapies. Front Cell Neurosci 2019; 13:290. [PMID: 31312123 PMCID: PMC6614517 DOI: 10.3389/fncel.2019.00290] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
Abstract
Approximately 3% of the world population suffers from depression, which is one of the most common form of mental disorder. Recent findings suggest that an interaction between the nervous system and immune system might be behind the pathophysiology of various neurological and psychiatric disorders, including depression. Neuropeptides have been shown to play a major role in mediating response to stress and inducing immune activation or suppression. Corticotropin releasing factor (CRF) is a major regulator of the hypothalamic pituitary adrenal (HPA) axis response. CRF is a stress-related neuropeptide whose dysregulation has been associated with depression. In this review, we summarized the role of CRF in the neuroimmune mechanisms of depression, and the potential therapeutic effects of Chinese herbal medicines (CHM) as well as other agents. Studying the network of CRF and immune responses will help to enhance our understanding of the pathogenesis of depression. Additionally, targeting this important network may aid in developing novel treatments for this debilitating psychiatric disorder.
Collapse
Affiliation(s)
- Yizhou Jiang
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Tangming Peng
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
| | - Uma Gaur
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Marta Silva
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Peter Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, Australia
| | - Zhong Chen
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, China
| | - Wei Qiu
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yandong Zhang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Wenhua Zheng
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
20
|
Stress-inducible-stem cells: a new view on endocrine, metabolic and mental disease? Mol Psychiatry 2019; 24:2-9. [PMID: 30242231 PMCID: PMC6755998 DOI: 10.1038/s41380-018-0244-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 07/25/2018] [Indexed: 02/08/2023]
|
21
|
Chatuphonprasert W, Jarukamjorn K, Ellinger I. Physiology and Pathophysiology of Steroid Biosynthesis, Transport and Metabolism in the Human Placenta. Front Pharmacol 2018; 9:1027. [PMID: 30258364 PMCID: PMC6144938 DOI: 10.3389/fphar.2018.01027] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/24/2018] [Indexed: 12/11/2022] Open
Abstract
The steroid hormones progestagens, estrogens, androgens, and glucocorticoids as well as their precursor cholesterol are required for successful establishment and maintenance of pregnancy and proper development of the fetus. The human placenta forms at the interface of maternal and fetal circulation. It participates in biosynthesis and metabolism of steroids as well as their regulated exchange between maternal and fetal compartment. This review outlines the mechanisms of human placental handling of steroid compounds. Cholesterol is transported from mother to offspring involving lipoprotein receptors such as low-density lipoprotein receptor (LDLR) and scavenger receptor class B type I (SRB1) as well as ATP-binding cassette (ABC)-transporters, ABCA1 and ABCG1. Additionally, cholesterol is also a precursor for placental progesterone and estrogen synthesis. Hormone synthesis is predominantly performed by members of the cytochrome P-450 (CYP) enzyme family including CYP11A1 or CYP19A1 and hydroxysteroid dehydrogenases (HSDs) such as 3β-HSD and 17β-HSD. Placental estrogen synthesis requires delivery of sulfate-conjugated precursor molecules from fetal and maternal serum. Placental uptake of these precursors is mediated by members of the solute carrier (SLC) family including sodium-dependent organic anion transporter (SOAT), organic anion transporter 4 (OAT4), and organic anion transporting polypeptide 2B1 (OATP2B1). Maternal-fetal glucocorticoid transport has to be tightly regulated in order to ensure healthy fetal growth and development. For that purpose, the placenta expresses the enzymes 11β-HSD 1 and 2 as well as the transporter ABCB1. This article also summarizes the impact of diverse compounds and diseases on the expression level and activity of the involved transporters, receptors, and metabolizing enzymes and concludes that the regulatory mechanisms changing the physiological to a pathophysiological state are barely explored. The structure and the cellular composition of the human placental barrier are introduced. While steroid production, metabolism and transport in the placental syncytiotrophoblast have been explored for decades, few information is available for the role of placental-fetal endothelial cells in these processes. With regard to placental structure and function, significant differences exist between species. To further decipher physiologic pathways and their pathologic alterations in placental steroid handling, proper model systems are mandatory.
Collapse
Affiliation(s)
- Waranya Chatuphonprasert
- Pathophysiology of the Placenta, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Faculty of Medicine, Mahasarakham University, Maha Sarakham, Thailand
| | - Kanokwan Jarukamjorn
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Isabella Ellinger
- Pathophysiology of the Placenta, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Alderman SL, Leishman EM, Fuzzen MLM, Bernier NJ. Corticotropin-releasing factor regulates caspase-3 and may protect developing zebrafish from stress-induced apoptosis. Gen Comp Endocrinol 2018; 265:207-213. [PMID: 29807032 DOI: 10.1016/j.ygcen.2018.05.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 02/06/2023]
Abstract
The corticotropin-releasing factor (CRF) system is expressed in the earliest stages of zebrafish development, long before its canonical function in the endocrine stress response is realized, and yet its function during embryogenesis is unknown. We tested the hypothesis that CRF protects embryos from stress-induced apoptosis. Here we confirm that a 1 h heat shock applied at either 6 h post-fertilization (hpf) or 30 hpf elicits an increase in caspase-3 activity, a key effector of apoptosis. Temporal changes in the expression of crf and its binding protein (crf-bp) during recovery from heat shock indicate that the CRF system is responsive to stressors experienced as early as gastrulation. Next, we heat shocked embryos that were microinjected with crf mRNA, and showed that caspase-3 induction is significantly reduced in embryos that overexpress CRF relative to control embryos. In addition, incubating embryos in the presence of the CRF receptor type 1 (CRF-R1) antagonist, antalarmin, during recovery from heat shock significantly increased caspase-3 activity, suggesting that CRF regulates caspase-3 via a CRF-R1-dependent pathway. Finally, we show that most heat shock-induced mortality occurred during the first hour of recovery, long before a significant increase in caspase-3 activity was detected. Indeed, the delayed caspase-3 induction coincided with a mortality plateau, and neither CRF overexpression nor antalarmin treatment altered heat shock induced mortality, supporting previous in vitro evidence that CRF-mediated cytoprotection occurs through the slow and tightly controlled apoptotic pathway. This study provides novel in vivo evidence that CRF regulates stress-induced apoptosis in a vertebrate model species, and demonstrates for the first time a function for the CRF system in early development that precedes its role in the endocrine stress response.
Collapse
Affiliation(s)
- Sarah L Alderman
- Department of Integrative Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1 Canada.
| | - Emily M Leishman
- Department of Integrative Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1 Canada
| | - Meghan L M Fuzzen
- Department of Integrative Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1 Canada
| | - Nicholas J Bernier
- Department of Integrative Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1 Canada
| |
Collapse
|
23
|
Abstract
OBJECTIVES The main aims of this paper are to review and evaluate the neurobiology of the depressive syndrome from a neurodevelopmental perspective. METHODS An English language literature search was performed using PubMed. RESULTS Depression is a complex syndrome that involves anatomical and functional changes that have an early origin in brain development. In subjects with genetic risk for depression, early stress factors are able to mediate not only the genetic risk but also gene expression. There is evidence that endocrine and immune interactions have an important impact on monoamine function and that the altered monoamine signalling observed in the depressive syndrome has a neuro-endocrino-immunological origin early in the development. CONCLUSIONS Neurodevelopment is a key aspect to understand the whole neurobiology of depression.
Collapse
Affiliation(s)
- Juan M Lima-Ojeda
- a Department of Psychiatry and Psychotherapy , University of Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- a Department of Psychiatry and Psychotherapy , University of Regensburg, Regensburg, Germany
| | - Thomas C Baghai
- a Department of Psychiatry and Psychotherapy , University of Regensburg, Regensburg, Germany
| |
Collapse
|
24
|
Justice NJ. The relationship between stress and Alzheimer's disease. Neurobiol Stress 2018; 8:127-133. [PMID: 29888308 PMCID: PMC5991350 DOI: 10.1016/j.ynstr.2018.04.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/03/2018] [Accepted: 04/19/2018] [Indexed: 12/04/2022] Open
Abstract
Stress is critically involved in the development and progression of disease. From the stress of undergoing treatments to facing your own mortality, the physiological processes that stress drives have a serious detrimental effect on the ability to heal, cope and maintain a positive quality of life. This is becoming increasingly clear in the case of neurodegenerative diseases. Neurodegenerative diseases involve the devastating loss of cognitive and motor function which is stressful in itself, but can also disrupt neural circuits that mediate stress responses. Disrupting these circuits produces aberrant emotional and aggressive behavior that causes long-term care to be especially difficult. In addition, added stress drives progression of the disease and can exacerbate symptoms. In this review, I describe how neural and endocrine pathways activated by stress interact with ongoing neurodegenerative disease from both a clinical and experimental perspective.
Collapse
Affiliation(s)
- Nicholas J. Justice
- Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, TX, 77030, USA
| |
Collapse
|
25
|
Davis EG, Keller J, Hallmayer J, Pankow HR, Murphy GM, Gotlib IH, Schatzberg AF. Corticotropin-releasing factor 1 receptor haplotype and cognitive features of major depression. Transl Psychiatry 2018; 8:5. [PMID: 29317606 PMCID: PMC5802461 DOI: 10.1038/s41398-017-0051-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/13/2017] [Indexed: 12/22/2022] Open
Abstract
Corticotropin-releasing factor signaling through CRF receptor type 1 (CRF1) has been shown to contribute to learning and memory function. A haplotype of alleles T-A-T in a set of common polymorphisms in the gene encoding for CRF1 (CRHR1) has been associated with both depression vulnerability and alterations in cognitive functioning. The present study investigated the relations between the TAT haplotype and specific symptoms of depression, self-reported ruminative behaviors, and neuropsychological performance on a learning and memory task. Participants were adults with major depression with and without psychotic features (N = 406). Associations were examined between TAT haplotype and endorsement of depression symptoms from diagnostic interviews, scores on the rumination response scale (RRS), and verbal memory performance on the California Verbal Learning Test-II (CVLT-II). All analyses included depression subtype, age, and sex as covariates; CVLT-II analyses also included evening cortisol levels. Across the entire sample, carriers of more copies of the TAT haplotype reported greater endorsement of the symptom describing difficulty concentrating and making decisions. In separate subsamples, TAT homozygotes had higher rumination scores on the RRS, both brooding and reflection subscales, and more TAT copies were associated with poorer CVLT-II performance in both total learning and free recall trials. These data demonstrate that the CRHR1 TAT haplotype is associated with cognitive features of depression including difficulty with decision-making, higher rumination, and poorer learning and memory. It will be important in future research to identify the specific molecular mechanisms for CRF1 signaling that contribute to depression-related cognitive dysfunction.
Collapse
Affiliation(s)
- Elena Goetz Davis
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA.
| | - Jennifer Keller
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Joachim Hallmayer
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Heather Ryan Pankow
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Greer M. Murphy
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Ian H. Gotlib
- 0000000419368956grid.168010.eDepartment of Psychology, Stanford University, Stanford, USA
| | - Alan F. Schatzberg
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
26
|
Mehrshad A, Shahraki M, Ehteshamfar S. Local Administration of Methylprednisolone Laden Hydrogel Enhances Functional Recovery of Transected Sciatic Nerve in Rat. Bull Emerg Trauma 2017; 5:231-239. [PMID: 29177169 DOI: 10.18869/acadpub.beat.5.4.509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Objective To determine the effects of methylprednisolone-laden hydrogel loaded into a chitosan conduit on the functional recovery of peripheral nerve using a rat sciatic nerve regeneration model was assessed. Methods 10-mm sciatic nerve defect was bridged using a chitosan conduit (CHIT/CGP-Hydrogel) filled with CGP-hydrogel. In authograft group (AUTO) a segment of sciatic nerve was transected and reimplanted reversely. In methylprednisolone treated group (CHIT/MP) the conduit was filled with methylprednisolone-laden CGP-hydrogel. The regenerated fibers were studied within 16 weeks after surgery. Results The behavioral, functional and electrophysiological studies confirmed faster recovery of the regenerated axons in methylprednisolone treated group compared to CHIT/Hydrogel group (p<0.05). The mean ratios of gastrocnemius muscles weight were measured. There was statistically significant difference between the muscle weight ratios of CHIT/MP and CHIT/Hydrogel groups (p<0.05). Morphometric indices of regenerated fibers showed number and diameter of the myelinated fibers were significantly higher in CHIT/MP than in CHIT/Hydrogel group. Conclusion Methylprednisolone-laden hydrogel when loaded in a chitosan conduit resulted in improvement of functional recovery and quantitative morphometric indices of sciatic nerve.
Collapse
Affiliation(s)
- Ali Mehrshad
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Mohammad Shahraki
- Department Clinical Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Shahin Ehteshamfar
- Department of Internal Medicine and Clinical Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| |
Collapse
|
27
|
Howland MA, Sandman CA, Glynn LM. Developmental origins of the human hypothalamic-pituitary-adrenal axis. Expert Rev Endocrinol Metab 2017; 12:321-339. [PMID: 30058893 PMCID: PMC6334849 DOI: 10.1080/17446651.2017.1356222] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The developmental origins of disease or fetal programming model predicts that intrauterine exposures have life long consequences for physical and psychological health. Prenatal programming of the fetal hypothalamic-pituitary-adrenal (HPA) axis is proposed as a primary mechanism by which early experiences are linked to later disease risk. Areas covered: This review describes the development of the fetal HPA axis, which is determined by an intricately timed cascade of endocrine events during gestation and is regulated by an integrated maternal-placental-fetal steroidogenic unit. Mechanisms by which stress-induced elevations in hormones of maternal, fetal, or placental origin influence the structure and function of the emerging fetal HPA axis are discussed. Recent prospective studies documenting persisting associations between prenatal stress exposures and altered postnatal HPA axis function are summarized, with effects observed beginning in infancy into adulthood. Expert commentary: The results of these studies are synthesized, and potential moderating factors are discussed. Promising areas of further research highlighted include epigenetic mechanisms and interactions between pre and postnatal influences.
Collapse
Affiliation(s)
- Mariann A. Howland
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA
| | - Curt A. Sandman
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA
| | - Laura M. Glynn
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA
- Department of Psychology, Chapman University, Orange, CA, USA
| |
Collapse
|
28
|
Curran MM, Sandman CA, Poggi Davis E, Glynn LM, Baram TZ. Abnormal dendritic maturation of developing cortical neurons exposed to corticotropin releasing hormone (CRH): Insights into effects of prenatal adversity? PLoS One 2017; 12:e0180311. [PMID: 28658297 PMCID: PMC5489219 DOI: 10.1371/journal.pone.0180311] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/13/2017] [Indexed: 01/17/2023] Open
Abstract
Corticotropin releasing hormone (CRH) produced by the hypothalamus initiates the hypothalamic-pituitary-adrenal (HPA) axis, which regulates the body's stress response. CRH levels typically are undetectable in human plasma, but during pregnancy the primate placenta synthesizes and releases large amounts of CRH into both maternal and fetal circulations. Notably, placental CRH synthesis increases in response to maternal stress signals. There is evidence that human fetal exposure to high concentrations of placental CRH is associated with behavioral consequences during infancy and into childhood, however the direct effects on of the peptide on the human brain are unknown. In this study, we used a rodent model to test the plausibility that CRH has direct effects on the developing cortex. Because chronic exposure to CRH reduces dendritic branching in hippocampal neurons, we tested the hypothesis that exposure to CRH would provoke impoverishment of dendritic trees in cortical neurons. This might be reflected in humans as cortical thinning. We grew developing cortical neurons in primary cultures in the presence of graded concentrations of CRH. We then employed Sholl analyses to measure dendritic branching and total dendritic length of treated cells. A seven-day exposure to increasing levels of CRH led to a significant, dose-dependent impoverishment of the branching of pyramidal-like cortical neurons. These results are consistent with the hypothesis that, rather than merely being a marker of prenatal stress, CRH directly decreases dendritic branching. Because dendrites comprise a large portion of cortical volume these findings might underlie reduced cortical thickness and could contribute to the behavioral consequences observed in children exposed to high levels of CRH in utero.
Collapse
Affiliation(s)
- Megan M. Curran
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, California, United States of America
| | - Curt A. Sandman
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, California, United States of America
| | - Elysia Poggi Davis
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, California, United States of America
- Department of Psychology, University of Denver, Denver, Colorado, United States of America
| | - Laura M. Glynn
- Department of Psychology, Chapman University, Orange, CA, United States of America
| | - Tallie Z. Baram
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, California, United States of America
- Department of Pediatrics, University of California Irvine, Irvine, California, United States of America
- Department of Neurology, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
29
|
Inda C, Bonfiglio JJ, Dos Santos Claro PA, Senin SA, Armando NG, Deussing JM, Silberstein S. cAMP-dependent cell differentiation triggered by activated CRHR1 in hippocampal neuronal cells. Sci Rep 2017; 7:1944. [PMID: 28512295 PMCID: PMC5434020 DOI: 10.1038/s41598-017-02021-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/04/2017] [Indexed: 12/18/2022] Open
Abstract
Corticotropin-releasing hormone receptor 1 (CRHR1) activates the atypical soluble adenylyl cyclase (sAC) in addition to transmembrane adenylyl cyclases (tmACs). Both cAMP sources were shown to be required for the phosphorylation of ERK1/2 triggered by activated G protein coupled receptor (GPCR) CRHR1 in neuronal and neuroendocrine contexts. Here, we show that activated CRHR1 promotes growth arrest and neurite elongation in neuronal hippocampal cells (HT22-CRHR1 cells). By characterising CRHR1 signalling mechanisms involved in the neuritogenic effect, we demonstrate that neurite outgrowth in HT22-CRHR1 cells takes place by a sAC-dependent, ERK1/2-independent signalling cascade. Both tmACs and sAC are involved in corticotropin-releasing hormone (CRH)-mediated CREB phosphorylation and c-fos induction, but only sAC-generated cAMP pools are critical for the neuritogenic effect of CRH, further highlighting the engagement of two sources of cAMP downstream of the activation of a GPCR, and reinforcing the notion that restricted cAMP microdomains may regulate independent cellular processes.
Collapse
Affiliation(s)
- Carolina Inda
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina.,DFBMC, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan José Bonfiglio
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina.,Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Paula A Dos Santos Claro
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina.,DFBMC, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sergio A Senin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Natalia G Armando
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Jan M Deussing
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, Molecular Neurogenetics, Munich, Germany
| | - Susana Silberstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina. .,DFBMC, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
30
|
Alessio N, Özcan S, Tatsumi K, Murat A, Peluso G, Dezawa M, Galderisi U. The secretome of MUSE cells contains factors that may play a role in regulation of stemness, apoptosis and immunomodulation. Cell Cycle 2016; 16:33-44. [PMID: 27463232 DOI: 10.1080/15384101.2016.1211215] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are a heterogeneous population, which contain several cell phenotypes: mesenchymal stem cells, progenitor cells, fibroblasts and other type of cells. Previously, we identified unique stem cells that we named multilineage-differentiating stress enduring (Muse) cells as one to several percent of MSCs of the bone marrow, adipose tissue and dermis. Among different cell populations in MSCs, Muse cells, positive for pluripotent surface marker SSEA-3, may represent cells responsible for pluripotent-like property of MSCs, since they express pluripotency genes, able to differentiated into triploblastic cells from a single cells and are self-renewable. MSCs release biologically active factors that have profound effects on local cellular dynamics. A thorough examination of MSC secretome seems essential for understanding the physiological functions exerted by these cells in our organism and also for rational cellular therapy design. In this setting, studies on secretome of Muse cells may shed light on pathways that are associated with their specific features. Our findings evidenced that secretomes of MSCs and Muse cells contain factors that regulate extracellular matrix remodeling, ox-redox activities and immune system. Muse cells appear to secrete factors that may preserve their stem cell features, allow survival under stress conditions and may contribute to their immunomodulation capacity. In detail, the proteins belonging to protein kinase A signaling, FXR/RXR activation and LXR/RXR activation pathways may play a role in regulation of Muse stem cell features. These last 2 pathways together with proteins associated with antigen presentation pathway and coagulation system may play a role in immunomodulation.
Collapse
Affiliation(s)
- Nicola Alessio
- a Department of Experimental Medicine , Biotechnology and Molecular Biology Section, Second University of Naples , Naples , Italy
| | - Servet Özcan
- b Genome and Stem Cell Center (GENKOK), Erciyes University , Kayseri , Turkey.,c Graduate School of Health Sciences, Erciyes Universty , Kayseri , Turkey
| | - Kazuki Tatsumi
- d Department of Stem Cell Biology and Histology , Tohoku University Graduate School of Medicine , Sendai , Japan.,e Tohoku Laboratory Non-clinical Research Division, Clio, Inc. , Sendai , Japan
| | - Ayşegül Murat
- c Graduate School of Health Sciences, Erciyes Universty , Kayseri , Turkey
| | | | - Mari Dezawa
- e Tohoku Laboratory Non-clinical Research Division, Clio, Inc. , Sendai , Japan
| | - Umberto Galderisi
- a Department of Experimental Medicine , Biotechnology and Molecular Biology Section, Second University of Naples , Naples , Italy.,b Genome and Stem Cell Center (GENKOK), Erciyes University , Kayseri , Turkey.,g Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University , Philadelphia , PA , USA
| |
Collapse
|
31
|
Howland MA, Sandman CA, Glynn LM, Crippen C, Davis EP. Fetal exposure to placental corticotropin-releasing hormone is associated with child self-reported internalizing symptoms. Psychoneuroendocrinology 2016; 67:10-7. [PMID: 26855003 PMCID: PMC4808336 DOI: 10.1016/j.psyneuen.2016.01.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Fetal exposure to maternal prenatal stress hormones such as cortisol exerts influences on the developing nervous system that persist and include risk for internalizing symptoms later in life. Placental corticotropin-releasing hormone (pCRH) is a feto-placental stress signal that also shapes fetal neurodevelopment and may be a more direct indicator of the fetal experience than maternal stress hormones. The programming effects of pCRH on child development are unknown. The current investigation examined associations between prenatal maternal and placental stress hormone exposures (maternal cortisol and pCRH) and child self-reported internalizing symptoms at age 5. METHOD Maternal plasma cortisol and pCRH levels were measured at 15, 19, 25, 31, and 36 weeks' gestation in a sample of 83 women and their 91 children (8 sibling pairs from separate pregnancies), who were born full-term. Child self-reported internalizing symptoms at age 5 were obtained using scales of the Berkeley Puppet Interview. RESULTS Placental CRH profiles (including elevations in mid-gestation) were associated with higher levels of internalizing symptoms at age 5. This effect was not explained by critical prenatal or postnatal influences, including obstetric risk, concurrent maternal psychological state, and family socio-economic status. Prenatal maternal cortisol was not significantly associated with child self-reported internalizing symptoms. CONCLUSIONS Findings suggest that elevated exposures to the feto-placental stress signal pCRH exert programming effects on the developing fetal central nervous system, with lasting consequences for child mental health.
Collapse
|
32
|
Dogra S, Sona C, Kumar A, Yadav PN. Epigenetic regulation of G protein coupled receptor signaling and its implications in psychiatric disorders. Int J Biochem Cell Biol 2016; 77:226-39. [PMID: 27046448 DOI: 10.1016/j.biocel.2016.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 12/21/2022]
Abstract
G protein-coupled receptors (GPCRs) act as a relay center through which extracellular signals, in the form of neurotransmitters or therapeutics, are converted into an intracellular response, which ultimately shapes the overall response at the tissue and behavioral level. Remarkably in similar ways, epigenetic mechanisms also modulate the expression pattern of a large number of genes in response to the dynamic environment inside and outside of the body, and consequently overall response. Emerging evidences from the pharmacogenomics and preclinical studies clearly suggest that these two distinct mechanisms criss-cross each other in several neurological disorders. At one hand such cross-talks between two distinct mechanisms make disease etiology more challenging to understand, while on the other hand if dealt appropriately, such situations might provide an opportunity to find novel druggable target and strategy for the treatment of complex diseases. In this review article, we have summarized and highlighted the main findings that tie epigenetic mechanisms to GPCR mediated signaling in the pathophysiology of central nervous system (CNS) disorders, including depression, addiction and pain.
Collapse
Affiliation(s)
- Shalini Dogra
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Chandan Sona
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Ajeet Kumar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Prem N Yadav
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India.
| |
Collapse
|
33
|
The Role of Hypothalamic Neuropeptides in Neurogenesis and Neuritogenesis. Neural Plast 2016; 2016:3276383. [PMID: 26881105 PMCID: PMC4737468 DOI: 10.1155/2016/3276383] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/18/2015] [Accepted: 11/22/2015] [Indexed: 01/23/2023] Open
Abstract
The hypothalamus is a source of neural progenitor cells which give rise to different populations of specialized and differentiated cells during brain development. Newly formed neurons in the hypothalamus can synthesize and release various neuropeptides. Although term neuropeptide recently undergoes redefinition, small-size hypothalamic neuropeptides remain major signaling molecules mediating short- and long-term effects on brain development. They represent important factors in neurite growth and formation of neural circuits. There is evidence suggesting that the newly generated hypothalamic neurons may be involved in regulation of metabolism, energy balance, body weight, and social behavior as well. Here we review recent data on the role of hypothalamic neuropeptides in adult neurogenesis and neuritogenesis with special emphasis on the development of food intake and social behavior related brain circuits.
Collapse
|
34
|
Bronson SL, Bale TL. The Placenta as a Mediator of Stress Effects on Neurodevelopmental Reprogramming. Neuropsychopharmacology 2016; 41:207-18. [PMID: 26250599 PMCID: PMC4677129 DOI: 10.1038/npp.2015.231] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/10/2015] [Accepted: 07/30/2015] [Indexed: 02/07/2023]
Abstract
Adversity experienced during gestation is a predictor of lifetime neuropsychiatric disease susceptibility. Specifically, maternal stress during pregnancy predisposes offspring to sex-biased neurodevelopmental disorders, including schizophrenia, attention deficit/hyperactivity disorder, and autism spectrum disorders. Animal models have demonstrated disease-relevant endophenotypes in prenatally stressed offspring and have provided unique insight into potential programmatic mechanisms. The placenta has a critical role in the deleterious and sex-specific effects of maternal stress and other fetal exposures on the developing brain. Stress-induced perturbations of the maternal milieu are conveyed to the embryo via the placenta, the maternal-fetal intermediary responsible for maintaining intrauterine homeostasis. Disruption of vital placental functions can have a significant impact on fetal development, including the brain, outcomes that are largely sex-specific. Here we review the novel involvement of the placenta in the transmission of the maternal adverse environment and effects on the developing brain.
Collapse
Affiliation(s)
- Stefanie L Bronson
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Tracy L Bale
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| |
Collapse
|
35
|
Stehouwer JS, Birnbaum MS, Voll RJ, Owens MJ, Plott SJ, Bourke CH, Wassef MA, Kilts CD, Goodman MM. Synthesis, F-18 radiolabeling, and microPET evaluation of 3-(2,4-dichlorophenyl)-N-alkyl-N-fluoroalkyl-2,5-dimethylpyrazolo[1,5-a]pyrimidin-7-amines as ligands of the corticotropin-releasing factor type-1 (CRF1) receptor. Bioorg Med Chem 2015; 23:4286-4302. [PMID: 26145817 DOI: 10.1016/j.bmc.2015.06.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/04/2015] [Accepted: 06/12/2015] [Indexed: 12/28/2022]
Abstract
A series of 3-(2,4-dichlorophenyl)-N-alkyl-N-fluoroalkyl-2,5-dimethylpyrazolo[1,5-a]pyrimidin-7-amines were synthesized and evaluated as potential positron emission tomography (PET) tracers for the corticotropin-releasing factor type-1 (CRF1) receptor. Compounds 27, 28, 29, and 30 all displayed high binding affinity (⩽1.2 nM) to the CRF1 receptor when assessed by in vitro competition binding assays at 23 °C, whereas a decrease in affinity (⩾10-fold) was observed with compound 26. The logP7.4 values of [(18)F]26-[(18)F]29 were in the range of ∼2.2-2.8 and microPET evaluation of [(18)F]26-[(18)F]29 in an anesthetized male cynomolgus monkey demonstrated brain penetrance, but specific binding was not sufficient enough to differentiate regions of high CRF1 receptor density from regions of low CRF1 receptor density. Radioactivity uptake in the skull, and sphenoid bone and/or sphenoid sinus during studies with [(18)F]28, [(18)F]28-d8, and [(18)F]29 was attributed to a combination of [(18)F]fluoride generated by metabolic defluorination of the radiotracer and binding of intact radiotracer to CRF1 receptors expressed on mast cells in the bone marrow. Uptake of [(18)F]26 and [(18)F]27 in the skull and sphenoid region was rapid but then steadily washed out which suggests that this behavior was the result of binding to CRF1 receptors expressed on mast cells in the bone marrow with no contribution from [(18)F]fluoride.
Collapse
Affiliation(s)
- Jeffrey S Stehouwer
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA.
| | - Matthew S Birnbaum
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA
| | - Ronald J Voll
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA
| | - Michael J Owens
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Susan J Plott
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Chase H Bourke
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Michael A Wassef
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA
| | - Clinton D Kilts
- Department of Psychiatry and Behavioral Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mark M Goodman
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| |
Collapse
|
36
|
Koutmani Y, Karalis KP. Neural stem cells respond to stress hormones: distinguishing beneficial from detrimental stress. Front Physiol 2015; 6:77. [PMID: 25814957 PMCID: PMC4356227 DOI: 10.3389/fphys.2015.00077] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/26/2015] [Indexed: 11/13/2022] Open
Abstract
Neural stem cells (NSCs), the progenitors of the nervous system, control distinct, position-specific functions and are critically involved in the maintenance of homeostasis in the brain. The responses of these cells to various stressful stimuli are shaped by genetic, epigenetic, and environmental factors via mechanisms that are age and developmental stage-dependent and still remain, to a great extent, elusive. Increasing evidence advocates for the beneficial impact of the stress response in various settings, complementing the extensive number of studies on the detrimental effects of stress, particularly in the developing brain. In this review, we discuss suggested mechanisms mediating both the beneficial and detrimental effects of stressors on NSC activity across the lifespan. We focus on the specific effects of secreted factors and we propose NSCs as a “sensor,” capable of distinguishing among the different stressors and adapting its functions accordingly. All the above suggest the intriguing hypothesis that NSCs are an important part of the adaptive response to stressors via direct and indirect, specific mechanisms.
Collapse
Affiliation(s)
- Yassemi Koutmani
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece
| | - Katia P Karalis
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece ; Endocrine Division, Children's Hospital, Harvard Medical School Boston, MA, USA
| |
Collapse
|
37
|
Suri D, Vaidya VA. The adaptive and maladaptive continuum of stress responses – a hippocampal perspective. Rev Neurosci 2015; 26:415-42. [DOI: 10.1515/revneuro-2014-0083] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/22/2015] [Indexed: 12/21/2022]
Abstract
AbstractExposure to stressors elicits a spectrum of responses that span from potentially adaptive to maladaptive consequences at the structural, cellular and physiological level. These responses are particularly pronounced in the hippocampus where they also appear to influence hippocampal-dependent cognitive function and emotionality. The factors that influence the nature of stress-evoked consequences include the chronicity, severity, predictability and controllability of the stressors. In addition to adult-onset stress, early life stress also elicits a wide range of structural and functional responses, which often exhibit life-long persistence. However, the outcome of early stress exposure is often contingent on the environment experienced in adulthood, and could either aid in stress coping or could serve to enhance susceptibility to the negative consequences of adult stress. This review comprehensively examines the consequences of adult and early life stressors on the hippocampus, with a focus on their effects on neurogenesis, neuronal survival, structural and synaptic plasticity and hippocampal-dependent behaviors. Further, we discuss potential factors that may tip stress-evoked consequences from being potentially adaptive to largely maladaptive.
Collapse
|
38
|
Spinas E, Saggini A, Kritas S, Cerulli G, Caraffa A, Antinolfi P, Pantalone A, Frydas A, Tei M, Speziali A, Saggini R, Conti P. Obesity, Inflammation and Neurological Alterations. EUR J INFLAMM 2014. [DOI: 10.1177/1721727x1401200302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Inflammation, neurodegeneration, imbalance of neurotransmitter systems, oxidative stress and depression are all risk factors for obesity. There is evidence regarding the cross-talk between adipose tissue and the immune system and obese patients may show an alteration of immune functions with major depression, including immune suppression with reduced T-cell and macrophage activity. Obesity is mediated by inflammatory cells such as lymphocytes, macrophages and mast cells which release pro-inflammatory cytokines and chemokines. Obesity-induced leukocyte infiltrations in adipose tissue cause cytokine/chemokine release and inflammation. Here, we report the relationship between obesity, neurological alterations and inflammation.
Collapse
Affiliation(s)
- E. Spinas
- Department of Surgery and Odontostomatological Sciences, University of Cagliari, Italy
| | - A. Saggini
- Department of Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - S.K. Kritas
- Department of Microbiology and Infectious Diseases, School of Veterinary Medicine, Aristotle University of Thessaloniki, Macedonia, Greece
| | - G. Cerulli
- Nicola's Foundation, Onlus, Arezzo, Italy
| | - A. Caraffa
- Orthopedic Division, University of Perugia, Perugia, Italy
| | - P. Antinolfi
- Orthopedic Division, University of Perugia, Perugia, Italy
| | - A. Pantalone
- Orthopedic Division, University of Chieti-Pescara, Chieti, Italy
| | - A. Frydas
- Aristotelian University, Thessaloniki, Greece
| | - M. Tei
- Nicola's Foundation, Onlus, Arezzo, Italy
| | | | - R. Saggini
- Department of Neurosciences and Imaging, Faculty of Medicine and Surgery, G. d'Annunzio University Chieti-Pescara, Chieti, Italy
| | - P. Conti
- Immunology Division, Medical School, University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
39
|
Rau TF, Kothiwal AS, Rova AR, Brooks DM, Rhoderick JF, Poulsen AJ, Hutchinson J, Poulsen DJ. Administration of low dose methamphetamine 12 h after a severe traumatic brain injury prevents neurological dysfunction and cognitive impairment in rats. Exp Neurol 2013; 253:31-40. [PMID: 24333768 DOI: 10.1016/j.expneurol.2013.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/20/2013] [Accepted: 12/02/2013] [Indexed: 10/25/2022]
Abstract
We recently published data that showed low dose of methamphetamine is neuroprotective when delivered 3 h after a severe traumatic brain injury (TBI). In the current study, we further characterized the neuroprotective potential of methamphetamine by determining the lowest effective dose, maximum therapeutic window, pharmacokinetic profile and gene expression changes associated with treatment. Graded doses of methamphetamine were administered to rats beginning 8 h after severe TBI. We assessed neuroprotection based on neurological severity scores, foot fault assessments, cognitive performance in the Morris water maze, and histopathology. We defined 0.250 mg/kg/h as the lowest effective dose and treatment at 12 h as the therapeutic window following severe TBI. We examined gene expression changes following TBI and methamphetamine treatment to further define the potential molecular mechanisms of neuroprotection and determined that methamphetamine significantly reduced the expression of key pro-inflammatory signals. Pharmacokinetic analysis revealed that a 24-hour intravenous infusion of methamphetamine at a dose of 0.500 mg/kg/h produced a plasma Cmax value of 25.9 ng/ml and a total exposure of 544 ng/ml over a 32 hour time frame. This represents almost half the 24-hour total exposure predicted for a daily oral dose of 25mg in a 70 kg adult human. Thus, we have demonstrated that methamphetamine is neuroprotective when delivered up to 12 h after injury at doses that are compatible with current FDA approved levels.
Collapse
Affiliation(s)
- Thomas F Rau
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Aakriti S Kothiwal
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Annela R Rova
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Diane M Brooks
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Joseph F Rhoderick
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Austin J Poulsen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Jim Hutchinson
- Montana Department of Justice Forensic Science Division, 2679 Palmer Street, Missoula, MT 59808, USA
| | - David J Poulsen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA.
| |
Collapse
|
40
|
Immunolocalization of Kisspeptin Associated with Amyloid-β Deposits in the Pons of an Alzheimer's Disease Patient. JOURNAL OF NEURODEGENERATIVE DISEASES 2013; 2013:879710. [PMID: 26317001 PMCID: PMC4437339 DOI: 10.1155/2013/879710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 04/23/2013] [Accepted: 04/24/2013] [Indexed: 02/03/2023]
Abstract
The pons region of the Alzheimer's disease (AD) brain is one of the last to show amyloid-β (Aβ) deposits and has been suggested to contain neuroprotective compounds. Kisspeptin (KP) is a hormone that activates the hypothalamic-pituitary-gonadal axis and has been suggested to be neuroprotective against Aβ toxicity. The localization of KP, plus the established endogenous neuroprotective compounds corticotropin releasing hormone (CRH) and catalase, in tissue sections from the pons region of a male AD subject has been determined in relation to Aβ deposits. Results showed Aβ deposits also stained with KP, CRH, and catalase antibodies. At high magnification the staining of deposits was either KP or catalase positive, and there was only a limited area of the deposits with KP-catalase colocalization. The CRH does not bind Aβ, whilst both KP and catalase can bind Aβ, suggesting that colocalization in Aβ deposits is not restricted to compounds that directly bind Aβ. The neuroprotective actions of KP, CRH, and catalase were confirmed in vitro, and fibrillar Aβ preparations were shown to stimulate the release of KP in vitro. In conclusion, neuroprotective KP, CRH, and catalase all colocalize with Aβ plaque-like deposits in the pons region from a male AD subject.
Collapse
|