1
|
Zhu LL, Li LD, Lin XY, Hu J, Wang C, Wang YJ, Zhou QG, Zhang J. Plasma-Derived Small Extracellular Vesicles miR- 182 - 5p Is a Potential Biomarker for Diagnosing Major Depressive Disorder. Mol Neurobiol 2025:10.1007/s12035-025-04948-9. [PMID: 40261603 DOI: 10.1007/s12035-025-04948-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Depression, particularly major depressive disorder (MDD), is a debilitating neuropsychiatric condition characterized by high disability rates, primarily driven by chronic stress and genetic predispositions. Emerging evidence highlights the critical role of microRNAs (miRNAs) in the pathogenesis of depression, with plasma-derived small extracellular vesicles (sEVs) emerging as promising biomarkers. In this study, we collected peripheral blood plasma samples from patients diagnosed with MDD, as assessed by the Hamilton Depression Rating scale, alongside healthy individuals serving as controls. Plasma-derived sEVs were isolated via ultracentrifugation, followed by high-throughput sequencing of miRNAs encapsulated within sEVs, and finally image acquisition and differential expression analysis. Our results revealed a significant elevation of miR-182-5p in plasma-derived sEVs from MDD patients compared to healthy controls, a finding further validated in chronic mild stress (CMS) models. Further analysis suggested that miRNAs encapsulated within sEVs may influence depression onset and progression by modulating hypothalamic-pituitary-adrenal (HPA) axis activity. These findings underscore the potential of miRNAs and their target genes as novel biomarkers, offering improved diagnostic accuracy and therapeutic efficacy for MDD.
Collapse
Affiliation(s)
- Lin-Lin Zhu
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China
| | - Lian-Di Li
- Anhui Institute for Food and Drug Control, 262 North Zhongshan Road, Nanjing, 210009, Jiangsu, China
| | - Xuan-Yu Lin
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China
| | - Jian Hu
- The Second Affiliated Hospital of Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210009, Jiangsu, China
| | - Chun Wang
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yi-Jun Wang
- The Second Affiliated Hospital of Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210009, Jiangsu, China
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China.
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211167, Jiangsu Province, China.
| | - Jing Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China.
| |
Collapse
|
2
|
Jiang Q, Zhang Y, Ye T, Liang X, Lou B. Metabonomics and Transcriptomics Analyses Reveal the Underlying HPA-Axis-Related Mechanisms of Lethality in Larimichthys polyactis Exposed to Underwater Noise Pollution. Int J Mol Sci 2024; 25:12610. [PMID: 39684322 PMCID: PMC11641136 DOI: 10.3390/ijms252312610] [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: 10/21/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The problem of marine noise pollution has a long history. Strong noise (>120 dB re 1 µPa) will affects the growth, development, physiological responses, and behaviors of fish, and also can induce the stress response, posing a mortal threat. Although many studies have reported that underwater noise may affect the survival of fish by disturbing their nervous system and endocrine system, the underlying causes of death due to noise stimulation remain unknown. Therefore, in this study, we used the underwater noise stress models to conduct underwater strong noise (50-125 dB re 1 µPa, 10-22,000 Hz) stress experiments on small yellow croaker for 10 min (short-term noise stress) and 6 days (long-term noise stress). A total of 150 fishes (body weight: 40-60 g; body length: 12-14 cm) were used in this study. Omics (metabolomics and transcriptomics) studies and quantitative analyses of important genes (HPA (hypothalamic-pituitary-adrenal)-axis functional genes) were performed to reveal genetic and metabolic changes in the important tissues associated with the HPA axis (brain, heart, and adrenal gland). Finally, we found that the strong noise pollution can significantly interfere with the expression of HPA-axis functional genes (including corticotropin releasing hormone (CRH), corticotropin releasing hormone receptor 2 (CRHR2), and arginine vasotocin (AVT)), and long-term stimulation can further induce metabolic disorders of the functional tissues (brain, heart, and adrenal gland), posing a lethal threat. Meanwhile, we also found that there were two kinds of death processes, direct death and chronic death, and both were closely related to the duration of stimulation and the regulation of the HPA axis.
Collapse
Affiliation(s)
| | | | | | | | - Bao Lou
- Zhejiang Key Laboratory of Coastal Biological Germplasm Resources Conservation and Utilization, Institute of Hydrobiology, Zhejiang Academy of Agricultural Sciences, Hangzhou 310000, China; (Q.J.); (Y.Z.); (T.Y.); (X.L.)
| |
Collapse
|
3
|
Liu L, Jin YD, Fan YH. Progress in research of corticotropin-releasing hormone receptor 2 in inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2024; 32:742-749. [DOI: 10.11569/wcjd.v32.i10.742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/26/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024] Open
Abstract
Members of the corticotropin-releasing hormone family and their receptors are widely distributed in central and peripheral tissues and are involved in the regulation of the cardiovascular system, metabolism, immune function, and inflammatory response in the body. Corticotropin-releasing hormone receptor 2 (CRHR2), one of specific receptors for corticotropin releasing factor, attenuates stress-induced intestinal hypersensitivity, influences intestinal microbial composition and diversity, has strong anti-inflammatory capacity, and regulates the proliferation, migration, and apoptosis of intestinal epithelial cells, and promotes intestinal mucosal repair. In recent years, studies have shown that the levels of CRHR2 in the colon tissue of patients with inflammatory bowel disease (IBD) are significantly different from those in normal human intestinal tissue, and it has been suggested that CRHR2 may be a potential therapeutic target for IBD. This paper reviews the physiological functions of CRHR2 and its clinical relevance to IBD, with the aim of exploring its specific mechanism of action and potential clinical application in the treatment of IBD, so as to provide a basis for the development of more effective therapeutic means for IBD in the future.
Collapse
Affiliation(s)
- Liu Liu
- Department of Gastroen-terology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Yi-Dan Jin
- Department of Gastroen-terology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Yi-Hong Fan
- Department of Gastroen-terology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
4
|
Perrelli M, Goparaju P, Postolache TT, del Bosque-Plata L, Gragnoli C. Stress and the CRH System, Norepinephrine, Depression, and Type 2 Diabetes. Biomedicines 2024; 12:1187. [PMID: 38927393 PMCID: PMC11200886 DOI: 10.3390/biomedicines12061187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Major depressive disorder (MDD) increases the risk of type 2 diabetes (T2D) by 60% in untreated patients, and hypercortisolism is common in MDD as well as in some patients with T2D. Patients with MDD, despite hypercortisolism, show inappropriately normal levels of corticotropin-releasing hormone (CRH) and plasma adrenocorticotropin (ACTH) in the cerebrospinal fluid, which might implicate impaired negative feedback. Also, a positive feedback loop of the CRH-norepinephrine (NE)-CRH system may be involved in the hypercortisolism of MDD and T2D. Dysfunctional CRH receptor 1 (CRHR1) and CRH receptor 2 (CRHR2), both of which are involved in glucose regulation, may explain hypercortisolism in MDD and T2D, at least in a subgroup of patients. CRHR1 increases glucose-stimulated insulin secretion. Dysfunctional CRHR1 variants can cause hypercortisolism, leading to serotonin dysfunction and depression, which can contribute to hyperglycemia, insulin resistance, and increased visceral fat, all of which are characteristics of T2D. CRHR2 is implicated in glucose homeostasis through the regulation of insulin secretion and gastrointestinal functions, and it stimulates insulin sensitivity at the muscular level. A few studies show a correlation of the CRHR2 gene with depressive disorders. Based on our own research, we have found a linkage and association (i.e., linkage disequilibrium [LD]) of the genes CRHR1 and CRHR2 with MDD and T2D in families with T2D. The correlation of CRHR1 and CRHR2 with MDD appears stronger than that with T2D, and per our hypothesis, MDD may precede the onset of T2D. According to the findings of our analysis, CRHR1 and CRHR2 variants could modify the response to prolonged chronic stress and contribute to high levels of cortisol, increasing the risk of developing MDD, T2D, and the comorbidity MDD-T2D. We report here the potential links of the CRH system, NE, and their roles in MDD and T2D.
Collapse
Affiliation(s)
| | - Pruthvi Goparaju
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE 68124, USA;
| | - Teodor T. Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO 80246, USA
- Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD 21090, USA
| | - Laura del Bosque-Plata
- Nutrigenetics, and Nutrigenomic Laboratory, National Institute of Genomic Medicine, Mexico City 14610, Mexico;
| | - Claudia Gragnoli
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE 68124, USA;
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033, USA
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, 8091 Zürich, Switzerland
- Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, 00197 Rome, Italy
| |
Collapse
|
5
|
Oka A, Hadano S, Ueda MT, Nakagawa S, Komaki G, Ando T. Rare CRHR2 and GRM8 variants identified as candidate factors associated with eating disorders in Japanese patients by whole exome sequencing. Heliyon 2024; 10:e28643. [PMID: 38644811 PMCID: PMC11031761 DOI: 10.1016/j.heliyon.2024.e28643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 04/23/2024] Open
Abstract
Eating disorders (EDs) are a type of psychiatric disorder characterized by pathological eating and related behavior and considered to be highly heritable. The purpose of this study was to explore rare variants expected to display biological functions associated with the etiology of EDs. We performed whole exome sequencing (WES) of affected sib-pairs corresponding to disease subtype through their lifetime and their parents. From those results, rare single nucleotide variants (SNVs) concordant with sib-pairs were extracted and estimated to be most deleterious in the examined families. Two non-synonymous SNVs located on corticotropin-releasing hormone receptor 2 (CRHR2) and glutamate metabotropic receptor 8 (GRM8) were identified as candidate disease susceptibility factors. The SNV of CRHR2 was included within the cholesterol binding motif of the transmembrane helix region, while the SNV of GRM8 was found to contribute to hydrogen bonds for an α-helix structure. CRHR2 plays important roles in the serotoninergic system of dorsal raphe nuclei, which is involved with feeding and stress-coping behavior, whereas GRM8 modulates glutamatergic neurotransmission. Moreover, GRM8 modulates glutamatergic neurotransmission, and is also considered to have effects on dopaminergic and adrenergic neurotransmission. Thus, identification of rare and deleterious variants in this study is expected to increase understanding and treatment of affected individuals. Further investigation regarding the biological function of these variants may provide an opportunity to elucidate the pathogenesis of EDs.
Collapse
Affiliation(s)
- Akira Oka
- Department of Molecular Life Sciences, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
- The Institute of Medical Sciences, Tokai University, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Shinji Hadano
- The Institute of Medical Sciences, Tokai University, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
- Department of Physiology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
- Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Mahoko Takahashi Ueda
- Department of Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo, Tokyo, 113-8510, Japan
| | - So Nakagawa
- Department of Molecular Life Sciences, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
- The Institute of Medical Sciences, Tokai University, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
- Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Gen Komaki
- Faculty of Medical Science, Fukuoka International University of Health and Welfare, Momochihama, Sawara-ku, Fukuoka, 814-0001, Japan
| | - Tetsuya Ando
- Department of Psychosomatic Medicine, Faculty of Medicine, School of Medicine, International University of Health and Welfare, 4-3 Kozunomori, Narita, Chiba, 286-8686, Japan
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8553, Japan
| |
Collapse
|
6
|
Matsoukas MT, Panagiotopoulos V, Karageorgos V, Chrousos GP, Venihaki M, Liapakis G. Structural and Functional Insights into CRF Peptides and Their Receptors. BIOLOGY 2024; 13:120. [PMID: 38392338 PMCID: PMC10886364 DOI: 10.3390/biology13020120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
Corticotropin-releasing factor or hormone (CRF or CRH) and the urocortins regulate a plethora of physiological functions and are involved in many pathophysiological processes. CRF and urocortins belong to the family of CRF peptides (CRF family), which includes sauvagine, urotensin, and many synthetic peptide and non-peptide CRF analogs. Several of the CRF analogs have shown considerable therapeutic potential in the treatment of various diseases. The CRF peptide family act by interacting with two types of plasma membrane proteins, type 1 (CRF1R) and type 2 (CRF2R), which belong to subfamily B1 of the family B G-protein-coupled receptors (GPCRs). This work describes the structure of CRF peptides and their receptors and the activation mechanism of the latter, which is compared with that of other GPCRs. It also discusses recent structural information that rationalizes the selective binding of various ligands to the two CRF receptor types and the activation of receptors by different agonists.
Collapse
Affiliation(s)
- Minos-Timotheos Matsoukas
- Department of Biomedical Engineering, School of Engineering, University of West Attica, 12243 Athens, Greece
| | - Vasilis Panagiotopoulos
- Department of Biomedical Engineering, School of Engineering, University of West Attica, 12243 Athens, Greece
| | - Vlasios Karageorgos
- Department of Pharmacology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine and UNESCO, National and Kapodistrian University of Athens, Livadias 8, 11527 Athens, Greece
| | - Maria Venihaki
- Department of Clinical Chemistry, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - George Liapakis
- Department of Pharmacology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
7
|
Yang X, Geng F. Corticotropin-releasing factor signaling and its potential role in the prefrontal cortex-dependent regulation of anxiety. J Neurosci Res 2023; 101:1781-1794. [PMID: 37592912 DOI: 10.1002/jnr.25238] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/08/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Abstract
A large body of literature has highlighted the significance of the corticotropin-releasing factor (CRF) system in the regulation of neuropsychiatric diseases. Anxiety disorders are among the most common neuropsychiatric disorders. An increasing number of studies have demonstrated that the CRF family mediates and regulates the development and maintenance of anxiety. Thus, the CRF family is considered to be a potential target for the treatment of anxiety disorders. The prefrontal cortex (PFC) plays a role in the occurrence and development of anxiety, and both CRF and CRF-R1 are widely expressed in the PFC. This paper begins by reviewing CRF-related signaling pathways and their different roles in anxiety and related processes. Then, the role of the CRF system in other neuropsychiatric diseases is reviewed and the potential role of PFC CRF signaling in the regulation of anxiety disorders is discussed. Although other signaling pathways are potentially involved in the process of anxiety, CRF in the PFC primarily modulates anxiety disorders through the activation of corticotropin-releasing factor type1 receptors (CRF-R1) and the excitation of the cAMP/PKA signaling pathway. Moreover, the main signaling pathways of CRF involved in sex differentiation in the PFC appear to be different. In summary, this review suggests that the CRF system in the PFC plays a critical role in the occurrence of anxiety. Thus, CRF signaling is of great significance as a potential target for the treatment of stress-related disorders in the future.
Collapse
Affiliation(s)
- Xin Yang
- Department of Physiology, Shantou University Medical College, Shantou, China
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fei Geng
- Department of Physiology, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| |
Collapse
|
8
|
Flaherty SE, Bezy O, Zheng W, Yan D, Li X, Jagarlapudi S, Albuquerque B, Esquejo RM, Peloquin M, Semache M, Mancini A, Kang L, Drujan D, Breitkopf SB, Griffin JD, Jean Beltran PM, Xue L, Stansfield J, Pashos E, Shakey Q, Pehmøller C, Monetti M, Birnbaum MJ, Fortin JP, Wu Z. Chronic UCN2 treatment desensitizes CRHR2 and improves insulin sensitivity. Nat Commun 2023; 14:3953. [PMID: 37402735 PMCID: PMC10319809 DOI: 10.1038/s41467-023-39597-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 06/21/2023] [Indexed: 07/06/2023] Open
Abstract
Urocortin 2 (UCN2) acts as a ligand for the G protein-coupled receptor corticotropin-releasing hormone receptor 2 (CRHR2). UCN2 has been reported to improve or worsen insulin sensitivity and glucose tolerance in vivo. Here we show that acute dosing of UCN2 induces systemic insulin resistance in male mice and skeletal muscle. Inversely, chronic elevation of UCN2 by injection with adenovirus encoding UCN2 resolves metabolic complications, improving glucose tolerance. CRHR2 recruits Gs in response to low concentrations of UCN2, as well as Gi and β-Arrestin at high concentrations of UCN2. Pre-treating cells and skeletal muscle ex vivo with UCN2 leads to internalization of CRHR2, dampened ligand-dependent increases in cAMP, and blunted reductions in insulin signaling. These results provide mechanistic insights into how UCN2 regulates insulin sensitivity and glucose metabolism in skeletal muscle and in vivo. Importantly, a working model was derived from these results that unifies the contradictory metabolic effects of UCN2.
Collapse
Affiliation(s)
- Stephen E Flaherty
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Olivier Bezy
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Wei Zheng
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Dong Yan
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Xiangping Li
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Srinath Jagarlapudi
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Bina Albuquerque
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Ryan M Esquejo
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Matthew Peloquin
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | | | | | - Liya Kang
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Doreen Drujan
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Susanne B Breitkopf
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - John D Griffin
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Pierre M Jean Beltran
- Machine Learning and Computational Sciences, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Liang Xue
- Machine Learning and Computational Sciences, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - John Stansfield
- Biostatistics, Early Clinical Development, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Evanthia Pashos
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Quazi Shakey
- Biomedicine design, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Christian Pehmøller
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Mara Monetti
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Morris J Birnbaum
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Jean-Philippe Fortin
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Zhidan Wu
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA.
| |
Collapse
|
9
|
Al Jowf GI, Ahmed ZT, Reijnders RA, de Nijs L, Eijssen LMT. To Predict, Prevent, and Manage Post-Traumatic Stress Disorder (PTSD): A Review of Pathophysiology, Treatment, and Biomarkers. Int J Mol Sci 2023; 24:ijms24065238. [PMID: 36982313 PMCID: PMC10049301 DOI: 10.3390/ijms24065238] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) can become a chronic and severely disabling condition resulting in a reduced quality of life and increased economic burden. The disorder is directly related to exposure to a traumatic event, e.g., a real or threatened injury, death, or sexual assault. Extensive research has been done on the neurobiological alterations underlying the disorder and its related phenotypes, revealing brain circuit disruption, neurotransmitter dysregulation, and hypothalamic–pituitary–adrenal (HPA) axis dysfunction. Psychotherapy remains the first-line treatment option for PTSD given its good efficacy, although pharmacotherapy can also be used as a stand-alone or in combination with psychotherapy. In order to reduce the prevalence and burden of the disorder, multilevel models of prevention have been developed to detect the disorder as early as possible and to reduce morbidity in those with established diseases. Despite the clinical grounds of diagnosis, attention is increasing to the discovery of reliable biomarkers that can predict susceptibility, aid diagnosis, or monitor treatment. Several potential biomarkers have been linked with pathophysiological changes related to PTSD, encouraging further research to identify actionable targets. This review highlights the current literature regarding the pathophysiology, disease development models, treatment modalities, and preventive models from a public health perspective, and discusses the current state of biomarker research.
Collapse
Affiliation(s)
- Ghazi I. Al Jowf
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Centre, 6200 MD Maastricht, The Netherlands
- Department of Public Health, College of Applied Medical Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
- Correspondence: (G.I.A.J.); (L.M.T.E.)
| | - Ziyad T. Ahmed
- College of Medicine, Sulaiman Al Rajhi University, Al-Bukairyah 52726, Saudi Arabia
| | - Rick A. Reijnders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Centre, 6200 MD Maastricht, The Netherlands
- European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Laurence de Nijs
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Centre, 6200 MD Maastricht, The Netherlands
- European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Lars M. T. Eijssen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Centre, 6200 MD Maastricht, The Netherlands
- European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department of Bioinformatics—BiGCaT, School of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands
- Correspondence: (G.I.A.J.); (L.M.T.E.)
| |
Collapse
|
10
|
Haass-Koffler CL, Francis TC, Gandhi P, Patel R, Naemuddin M, Nielsen CK, Bartlett SE, Bonci A, Vasile S, Hood BL, Suyama E, Hedrick MP, Smith LH, Limpert AS, Roberto M, Cosford NDP, Sheffler DJ. Development and use of a high-throughput screen to identify novel modulators of the corticotropin releasing factor binding protein. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:448-459. [PMID: 36210051 PMCID: PMC9762412 DOI: 10.1016/j.slasd.2022.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/09/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Stress responses are believed to involve corticotropin releasing factor (CRF), its two cognate receptors (CRF1 and CRF2), and the CRF-binding protein (CRFBP). Whereas decades of research has focused on CRF1, the role of CRF2 in the central nervous system (CNS) has not been thoroughly investigated. We have previously reported that CRF2, interacting with a C terminal fragment of CRFBP, CRFBP(10kD), may have a role in the modulation of neuronal activity. However, the mechanism by which CRF interacts with CRFBP(10kD) and CRF2 has not been fully elucidated due to the lack of useful chemical tools to probe CRFBP. METHODS We miniaturized a cell-based assay, where CRFBP(10kD) is fused as a chimera with CRF2, and performed a high-throughput screen (HTS) of 350,000 small molecules to find negative allosteric modulators (NAMs) of the CRFBP(10kD)-CRF2 complex. Hits were confirmed by evaluating activity toward parental HEK293 cells, toward CRF2 in the absence of CRFBP(10kD), and toward CRF1 in vitro. Hits were further characterized in ex vivo electrophysiology assays that target: 1) the CRF1+ neurons in the central nucleus of the amygdala (CeA) of CRF1:GFP mice that express GFP under the CRF1 promoter, and 2) the CRF-induced potentiation of N-methyl-D-aspartic acid receptor (NMDAR)-mediated synaptic transmission in dopamine neurons in the ventral tegmental area (VTA). RESULTS We found that CRFBP(10kD) potentiates CRF-intracellular Ca2+ release specifically via CRF2, indicating that CRFBP may possess excitatory roles in addition to the inhibitory role established by the N-terminal fragment of CRFBP, CRFBP(27kD). We identified novel small molecule CRFBP-CRF2 NAMs that do not alter the CRF1-mediated effects of exogenous CRF but blunt CRF-induced potentiation of NMDAR-mediated synaptic transmission in dopamine neurons in the VTA, an effect mediated by CRF2 and CRFBP. CONCLUSION These results provide the first evidence of specific roles for CRF2 and CRFBP(10kD) in the modulation of neuronal activity and suggest that CRFBP(10kD)-CRF2 NAMs can be further developed for the treatment of stress-related disorders including alcohol and substance use disorders.
Collapse
Affiliation(s)
- Carolina L Haass-Koffler
- Department of Psychiatry and Human Behavior, Alpert Medical School; Department of Behavioral and Social Sciences, School of Public Health; Center for Alcohol and Addiction Studies; Carney Institute for Brain Science, Brown University, Providence RI, United States.
| | - T Chase Francis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States; Intramural Research Program, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Baltimore, MD, United States
| | - Pauravi Gandhi
- The Scripps Research Institute, La Jolla, CA, United States
| | - Reesha Patel
- The Scripps Research Institute, La Jolla, CA, United States
| | - Mohammad Naemuddin
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Carsten K Nielsen
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Selena E Bartlett
- Translational Research Institute, School of Clinical Sciences, Faculty of Health, Queensland University of Technology, Queensland, Australia
| | | | - Stefan Vasile
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Becky L Hood
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Eigo Suyama
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Michael P Hedrick
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Layton H Smith
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Allison S Limpert
- NCI Designated Cancer Center, La Jolla, CA, United States; Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Marisa Roberto
- The Scripps Research Institute, La Jolla, CA, United States
| | - Nicholas D P Cosford
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States; NCI Designated Cancer Center, La Jolla, CA, United States; Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Douglas J Sheffler
- NCI Designated Cancer Center, La Jolla, CA, United States; Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States.
| |
Collapse
|
11
|
Wang XD, Bai HM, Li XL, Zhang LF, Li F, Bai Y, Wu ZY, Liu SQ, Li H. Corticotropin-releasing factor is involved in acute stress-induced analgesia and antipruritus. Brain Behav 2022; 12:e2783. [PMID: 36209489 PMCID: PMC9660402 DOI: 10.1002/brb3.2783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/31/2022] [Accepted: 09/15/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Under the condition of stress, the hypothalamic-pituitary-adrenal axis (HPA axis) is activated and causes the secretion of corticotropin-releasing factor (CRF). Previous studies have demonstrated that CRF is involved in the regulation of pain and itch. Thus, it remains worthy to explore whether the desensitization of pain and itch under high-intensity acute stress (such as high fear and tension) is related to the sharp increase of CRF. METHODS Forced swimming was used to simulate acute stress. ELISA and pharmacological methods were conducted to observe the effects of forced swimming on acute pain or itch and the relationship between blood CRF content and itch or pain behavior. Intracerebroventricular (ICV) administration of CRF was conducted to examine the effects of CRF on acute pain or itch. Intrathecal administration of CRF receptor agonist or antagonist was conducted to examine the receptor mechanisms of the regulatory role of CRF in pain and itch. RESULTS ELISA experiment showed that the serum CRF in mice reached its peak within 5-10 min after acute stress (forced swimming). Behavioral data showed that the scratching behavior induced by itch agents decreased after acute swimming, while the mechanical pain threshold increased significantly. The inhibitory effect of acute stress on pain and itch is mediated by CRF receptor2 (CRFR2). Then, ICV injection of CRF was used to simulate the massive release of CRF under acute stress, and we observed that the scratching behavior induced by histamine or chloroquine was significantly inhibited after ICV injection of CRF. The above effects of CRF are mainly mediated by CRFR2. These results suggest that 5-10 min after acute stress, a large amount of CRF is released into the blood from the hypothalamus, which significantly inhibits acute pain and itch by acting on CRFR2. ICV injection of CRF can replicate the antipruritus effects of acute stress. CONCLUSIONS The present study investigated the mechanism of acute stress-induced analgesia and antipruritus and provided theoretical support for the treatment of pain and itch.
Collapse
Affiliation(s)
- Xiao-Dong Wang
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, China.,Department of Emergency Medicine, Inner Mongolia Armed Police Corps Hospital, Hohhot, China
| | - Hao-Miao Bai
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, China
| | - Xiao-Lan Li
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, China.,Department of Human Anatomy, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Lin-Fang Zhang
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, China.,Department of Anatomy, Medical School of Yan'an University, Yan'an, China
| | - Fei Li
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Zhen-Yu Wu
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, China
| | - Shang-Qing Liu
- Department of Human Anatomy, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Hui Li
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
12
|
Heuer JG, Meyer CM, Baker HE, Geiser A, Lucchesi J, Xu D, Hamang M, Martin JA, Hu C, Roth KD, Thirunavukkarasu K, Alsina-Fernandez J, Ma YL. Pharmacological Evaluation of a Pegylated Urocortin-1 Peptide in Experimental Autoimmune Disease Models. J Pharmacol Exp Ther 2022; 382:287-298. [PMID: 35688476 DOI: 10.1124/jpet.122.001151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/25/2022] [Indexed: 11/22/2022] Open
Abstract
Urocortin-1 (UCN1) is a member of the corticotropin releasing hormone (CRH) family of peptides that acts through CRH-receptor 1 (CRHR1) and CRH-receptor 2 (CRHR2). UCN1 can induce the adrenocorticotropin hormone and downstream glucocorticoids through CRHR1 and promote beneficial metabolic effects through CRHR2. UCN1 has a short half-life and has been shown to improve experimental autoimmune disease. A pegylated UCN1 peptide (PEG-hUCN1) was generated to extend half-life and was tested in multiple experimental autoimmune disease models and in healthy mice to determine effects on corticosterone induction, autoimmune disease, and glucocorticoid induced adverse effects. Cardiovascular effects were also assessed by telemetry. PEG-hUCN1 demonstrated a dose dependent 4-6-fold elevation of serum corticosterone and significantly improved autoimmune disease comparable to prednisolone in several experimental models. In healthy mice, PEG-hUCN1 showed less adverse effects compared with corticosterone treatment. PEG-hUCN1 peptide induced an initial 30% reduction in blood pressure that was followed by a gradual and sustained 30% increase in blood pressure at the highest dose. Additionally, an adeno-associated viral 8 (AAV8) UCN1 was used to assess adverse effects of chronic elevation of UCN1 in wild type and CRHR2 knockout mice. Chronic UCN1 expression by an AAV8 approach in wild type and CRHR2 knockout mice demonstrated an important role of CRHR2 in countering the adverse metabolic effects of elevated corticosterone from UCN1. Our findings demonstrate that PEG-hUCN1 shows profound effects in treating autoimmune disease with an improved safety profile relative to corticosterone and that CRHR2 activity is important in metabolic regulation. SIGNIFICANCE STATEMENT: This study reports the generation and characterization of a pegylated UCN1 peptide and the role of CRHR2 in UCN1-induced metabolic effects. The potency/selectivity, pharmacokinetic properties, pharmacodynamic effects, and efficacy in four autoimmune models and safety profiles are presented. This pegylated UCN1 shows potential for treating autoimmune diseases with reduced adverse effects compared to corticosterone treatment. Continuous exposure to UCN1 through an AAV8 approach demonstrates some glucocorticoid mediated adverse metabolic effects that are exacerbated in the absence of the CRHR2 receptor.
Collapse
Affiliation(s)
- Josef G Heuer
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Catalina M Meyer
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Hana E Baker
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Andrea Geiser
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Jonathan Lucchesi
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Daniel Xu
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Matthew Hamang
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Jennifer A Martin
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Charlie Hu
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Kenneth D Roth
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Kannan Thirunavukkarasu
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Jorge Alsina-Fernandez
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Yanfei L Ma
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| |
Collapse
|
13
|
Amin M, Ott J, Gordon D, Wu R, Postolache TT, Vergare M, Gragnoli C. Comorbidity of Novel CRHR2 Gene Variants in Type 2 Diabetes and Depression. Int J Mol Sci 2022; 23:9819. [PMID: 36077219 PMCID: PMC9456299 DOI: 10.3390/ijms23179819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 12/18/2022] Open
Abstract
The corticotropin-releasing hormone receptor 2 (CRHR2) gene encodes CRHR2, contributing to the hypothalamic-pituitary-adrenal stress response and to hyperglycemia and insulin resistance. CRHR2-/- mice are hypersensitive to stress, and the CRHR2 locus has been linked to type 2 diabetes and depression. While CRHR2 variants confer risk for mood disorders, MDD, and type 2 diabetes, they have not been investigated in familial T2D and MDD. In 212 Italian families with type 2 diabetes and depression, we tested 17 CRHR2 single nucleotide polymorphisms (SNPs), using two-point parametric-linkage and linkage-disequilibrium (i.e., association) analysis (models: dominant-complete-penetrance-D1, dominant-incomplete-penetrance-D2, recessive-complete-penetrance-R1, recessive-incomplete-penetrance-R2). We detected novel linkage/linkage-disequilibrium/association to/with depression (3 SNPs/D1, 2 SNPs/D2, 3 SNPs/R1, 3 SNPs/R2) and type 2 diabetes (3 SNPs/D1, 2 SNPs/D2, 2 SNPs/R1, 1 SNP/R2). All detected risk variants are novel. Two depression-risk variants within one linkage-disequilibrium block replicate each other. Two independent novel SNPs were comorbid while the most significant conferred either depression- or type 2 diabetes-risk. Although the families were primarily ascertained for type 2 diabetes, depression-risk variants showed higher significance than type 2 diabetes-risk variants, implying CRHR2 has a stronger role in depression-risk than type 2 diabetes-risk. In silico analysis predicted variants' dysfunction. CRHR2 is for the first time linked to/in linkage-disequilibrium/association with depression-type 2 diabetes comorbidity and may underlie the shared genetic pathogenesis via pleiotropy.
Collapse
Affiliation(s)
- Mutaz Amin
- Institut National de la Santé et de la Recherche Médicale (INSERM), US14-Orphanet, 75014 Paris, France
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Jurg Ott
- Laboratory of Statistical Genetics, Rockefeller University, New York, NY 10065, USA
| | - Derek Gordon
- Department of Genetics, Rutgers University, Piscataway, NJ 08854, USA
| | - Rongling Wu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033, USA
- Departments of Statistics, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Teodor T. Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Denver, CO 80246, USA
- Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO 80246, USA
- Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD 21090, USA
| | - Michael Vergare
- Department of Psychiatry and Human Behavior, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Claudia Gragnoli
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033, USA
- Division of Endocrinology, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE 68124, USA
- Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, 00197 Rome, Italy
| |
Collapse
|
14
|
Featherstone RE, Gifford RL, Crown LM, Amirfathi F, Alaniz JP, Yi J, Tran A, Adomian D, Schwenk A, Melnychenko O, Duval C, Parekh K, Lee DJ, Siegel SJ. Early life social instability stress causes lasting cognitive decrement and elevated hippocampal stress-related gene expression. Exp Neurol 2022; 354:114099. [PMID: 35490720 DOI: 10.1016/j.expneurol.2022.114099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/07/2022] [Accepted: 04/24/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Early life stress may have profound effects on brain health, yielding both short- and long-term cognitive or psychiatric impairment. Early life Social Instability Stress (SIS) in rodents has been used to model the effects of early chronic human stress. While many studies have assessed acute and short-term responses to this stressor, less attention has been paid to the lasting effects of early life stress in rodents. METHODS The current study utilized SIS in young mice to assess the impact of early life adversity over the lifespan. Mice were assessed in adulthood between the ages of 18 to 66 weeks for changes in behaviors associated with anxiety, affect, sociability, aggression, motivation, and recognition memory. Additionally, mice were assessed for changes in glucocorticoid level and hippocampal mRNA expression in a subset of genes that display alterations in humans following exposure to stress (CRHR1, CRHR2, FKBP5, SLC6A4). RESULTS Mice exposed to early SIS showed disrupted memory and increased hippocampal expression of FKBP5, CRHR2 and SLC6A4 mRNA compared to non-stressed mice. Importantly, there was a significant association between increased FKBP5 and CRHR2 with reduced recognition memory. Additionally, mice exposed to SIS showed increased responding on a progressive ratio schedule of reinforcement, indicating that reduction in memory performance was not mediated by decreased effort. CONCLUSIONS Ecologically-relevant social stress in mice causes long-term decrements in recognition memory, possibly mediated by persistent changes in moderators of the stress cascade. Additionally, animals exposed to early life stress showed increased motivation for reward, which may contribute to a host of hedonic seeking behaviors throughout life. These data suggest that SIS can be used to evaluate therapeutic interventions to attenuate or reverse lasting effects of early life adversity.
Collapse
Affiliation(s)
- Robert E Featherstone
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Raymond L Gifford
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Lindsey M Crown
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Felix Amirfathi
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Jon P Alaniz
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Janice Yi
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - AiVi Tran
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Derrick Adomian
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Andrew Schwenk
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Olya Melnychenko
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Christina Duval
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Krishna Parekh
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America
| | - Darrin J Lee
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America; Department of Neurosurgery, Keck School of Medicine, University of Southern California, 1200 North State St., Suite 3300, Los Angeles, CA 90033, United States of America
| | - Steven J Siegel
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, United States of America.
| |
Collapse
|
15
|
Lautherbach N, Gonçalves DAP, Silveira WA, Paula-Gomes S, Valentim RR, Zanon NM, Pereira MG, Miyabara EH, Navegantes LCC, Kettelhut IC. Urocortin 2 promotes hypertrophy and enhances skeletal muscle function through cAMP and insulin/IGF-1 signaling pathways. Mol Metab 2022; 60:101492. [PMID: 35390501 PMCID: PMC9035725 DOI: 10.1016/j.molmet.2022.101492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/27/2022] [Accepted: 03/29/2022] [Indexed: 11/28/2022] Open
Abstract
Objective Although it is well established that urocortin 2 (Ucn2), a peptide member of the corticotrophin releasing factor (CRF) family, and its specific corticotrophin-releasing factor 2 receptor (CRF2R) are highly expressed in skeletal muscle, the role of this peptide in the regulation of skeletal muscle mass and protein metabolism remains elusive. Methods To elucidate the mechanisms how Ucn2 directly controls protein metabolism in skeletal muscles of normal mice, we carried out genetic tools, physiological and molecular analyses of muscles in vivo and in vitro. Results Here, we demonstrated that Ucn2 overexpression activated cAMP signaling and promoted an expressive muscle hypertrophy associated with higher rates of protein synthesis and activation of Akt/mTOR and ERK1/2 signaling pathways. Furthermore, Ucn2 induced a decrease in mRNA levels of atrogin-1 and in autophagic flux inferred by an increase in the protein content of LC3-I, LC3-II and p62. Accordingly, Ucn2 reduced both the transcriptional activity of FoxO in vivo and the overall protein degradation in vitro through an inhibition of lysosomal proteolytic activity. In addition, we demonstrated that Ucn2 induced a fast-to-slow fiber type shift and improved fatigue muscle resistance, an effect that was completely blocked in muscles co-transfected with mitogen-activated protein kinase phosphatase 1 (MKP-1), but not with dominant-negative Akt mutant (Aktmt). Conclusions These data suggest that Ucn2 triggers an anabolic and anti-catabolic response in skeletal muscle of normal mice probably through the activation of cAMP cascade and participation of Akt and ERK1/2 signaling. These findings open new perspectives in the development of therapeutic strategies to cope with the loss of muscle mass. Ucn2 overexpression promotes muscle growth due to an increase in protein synthesis. Ucn2 inhibits FoxO activity and autophagic-lysosomal system. Ucn2-induced skeletal muscle phenotype is dependent on Akt and ERK1/2. Ucn2 induces a fast-to-slow fiber type shift and improves fatigue resistance. The increase in muscle fatigue resistance is dependent on ERK1/2.
Collapse
Affiliation(s)
- Natalia Lautherbach
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Biochemistry/Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Dawit A P Gonçalves
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Wilian A Silveira
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Biochemistry, Pharmacology and Physiology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil.
| | - Sílvia Paula-Gomes
- Department of Biochemistry/Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil.
| | - Rafael Rossi Valentim
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Neuza M Zanon
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Marcelo G Pereira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Elen H Miyabara
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Luiz C C Navegantes
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Isis C Kettelhut
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Biochemistry/Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
16
|
Bruce JK, Burns GL, Sinn Soh W, Nair PM, Sherwin S, Fan K, Dowling LR, Goggins BJ, Koloski N, Potter M, Bollipo S, Foster R, Gan LT, Veysey M, Philpott DJ, Girardin SE, Holtmann G, Kaiko GE, Walker MM, Talley NJ, Keely S. Defects in NLRP6, autophagy and goblet cell homeostasis are associated with reduced duodenal CRH receptor 2 expression in patients with functional dyspepsia. Brain Behav Immun 2022; 101:335-345. [PMID: 35093492 DOI: 10.1016/j.bbi.2022.01.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/09/2021] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
Functional dyspepsia (FD) affects up to 15% of the population and is characterised by recurring upper gastrointestinal (GI) symptoms occurring in the absence of clinically identifiable pathology. Psychological stress is a key factor associated with the onset of FD and locally acting hypothalamic-pituitary-adrenal (HPA) axis hormones have been implicated in GI motility and barrier dysfunction. Recent pre-clinical work has identified mechanistic pathways linking corticotropin-releasing hormone (CRH) with the innate epithelial immune protein NLRP6, an inflammasome that has been shown to regulate GI mucus secretion. We recruited twelve FD patients and twelve healthy individuals to examine whether dysregulation of hypothalamic-pituitary adrenal (HPA) axis hormones and altered NLRP6 pathways were evident in the duodenal mucosa. Protein expression was assessed by immunoblot and immunohistochemistry in D2 duodenal biopsies. Plasma HPA axis hormones were assayed by ELISA and enteroid and colorectal cancer cell line cultures were used to verify function. FD patients exhibited reduced duodenal CRH-receptor 2, compared to non-GI disease controls, indicating a dysregulation of duodenal HPA signalling. The loss of CRH-receptor 2 correlated with reduced NLRP6 expression and autophagy function, processes critical for maintaining goblet cell homeostasis. In accordance, duodenal goblet cell numbers and mucin exocytosis was reduced in FD patients compared to controls. In vitro studies demonstrated that CRH could reduce NLRP6 in duodenal spheroids and promote mucus secretion in the HT29-MTX-E12 cell line. In conclusion, FD patients exhibit defects in the NLRP6-autophagy axis with decreased goblet cell function that may drive symptoms of disease. These features correlated with loss of CRH receptor 2 and may be driven by dysregulation of HPA signalling in the duodenum of FD patients.
Collapse
Affiliation(s)
- Jessica K Bruce
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Grace L Burns
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Wai Sinn Soh
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Prema M Nair
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Simonne Sherwin
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - KeNing Fan
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Laura R Dowling
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Bridie J Goggins
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Natasha Koloski
- School of Medicine & Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; Department of Gastroenterology, John Hunter Hospital, Newcastle, New South Wales, Australia; Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, and Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Michael Potter
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Medicine & Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia
| | - Steven Bollipo
- Department of Gastroenterology, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Robert Foster
- Department of Gastroenterology, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Lay T Gan
- Department of Gastroenterology, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Martin Veysey
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Medicine & Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Stephen E Girardin
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Gerald Holtmann
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, and Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Gerard E Kaiko
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Marjorie M Walker
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Medicine & Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia
| | - Nicholas J Talley
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Medicine & Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia
| | - Simon Keely
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia; NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
17
|
Mori Y, Tsuchihira A, Yoshida T, Yoshida S, Fujiuchi A, Ohmi M, Isogai Y, Sakaguchi T, Eguchi S, Tsuda T, Kato K, Ohashi K, Ouchi N, Park HM, Murohara T, Takefuji M. Corticotropin releasing hormone receptor 2 antagonist, RQ-00490721, for the prevention of pressure overload-induced cardiac dysfunction. Biomed Pharmacother 2021; 146:112566. [PMID: 34954642 DOI: 10.1016/j.biopha.2021.112566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/14/2021] [Accepted: 12/19/2021] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND G protein-coupled receptors (GPCRs) regulate the pathological and physiological functions of the heart. GPCR antagonists are widely used in the treatment of chronic heart failure. Despite therapeutic advances in the treatments for cardiovascular diseases, heart failure is a major clinical health problem, with significant mortality and morbidity. Corticotropin releasing hormone receptor 2 (CRHR2) is highly expressed in cardiomyocytes, and cardiomyocyte-specific deletion of the genes encoding CRHR2 suppresses pressure overload-induced cardiac dysfunction. This suggests that the negative modulation of CRHR2 may prevent the progression of heart failure. However, there are no systemic drugs against CRHR2. FINDINGS We developed a novel, oral, small molecule antagonist of CRHR2, RQ-00490721, to investigate the inhibition of CRHR2 as a potential therapeutic approach for the treatment of heart failure. In vitro, RQ-00490721 decreased CRHR2 agonist-induced 3', 5'-cyclic adenosine monophosphate (cAMP) production. In vivo, RQ-00490721 showed sufficient oral absorption and better distribution to peripheral organs than to the central nervous system. Oral administration of RQ-00490721 inhibited the CRHR2 agonist-induced phosphorylation of cAMP-response element binding protein (CREB) in the heart, which regulates a transcription activator involved in heart failure. RQ-00490721 administration was not found to affect basal heart function in mice but protected them from pressure overload-induced cardiac dysfunction. INTERPRETATION Our results suggest that RQ-00490721 is a promising agent for use in the treatment of chronic heart failure.
Collapse
Affiliation(s)
- Yu Mori
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | | | - Tatsuya Yoshida
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Satoya Yoshida
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Akiyoshi Fujiuchi
- Discovery Research, RaQualia Pharma Inc., Nagoya, Japan; RaQualia Pharma Industry-Academia Collaborative Research Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Masashi Ohmi
- Discovery Research, RaQualia Pharma Inc., Nagoya, Japan
| | - Yumi Isogai
- Discovery Research, RaQualia Pharma Inc., Nagoya, Japan
| | - Teruhiro Sakaguchi
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Shunsuke Eguchi
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Takuma Tsuda
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Noriyuki Ouchi
- Department of Molecular Medicine and Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Hyi-Man Park
- Discovery Research, RaQualia Pharma Inc., Nagoya, Japan; RaQualia Pharma Industry-Academia Collaborative Research Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan.
| |
Collapse
|
18
|
Ivanova D, Li XF, McIntyre C, Liu Y, Kong L, O’Byrne KT. Urocortin3 in the Posterodorsal Medial Amygdala Mediates Stress-induced Suppression of LH Pulsatility in Female Mice. Endocrinology 2021; 162:6383454. [PMID: 34618891 PMCID: PMC8547342 DOI: 10.1210/endocr/bqab206] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Indexed: 01/09/2023]
Abstract
Psychosocial stress disrupts reproduction and interferes with pulsatile LH secretion. The posterodorsal medial amygdala (MePD) is an upstream modulator of the reproductive axis and stress. Corticotropin-releasing factor type 2 receptors (CRFR2s) are activated in the presence of psychosocial stress together with increased expression of the CRFR2 ligand Urocortin3 (Ucn3) in the MePD of rodents. We investigate whether Ucn3 signalling in the MePD is involved in mediating the suppressive effect of psychosocial stress on LH pulsatility. First, we administered Ucn3 into the MePD and monitored the effect on LH pulses in ovariectomized mice. Next, we delivered Astressin2B, a selective CRFR2 antagonist, intra-MePD in the presence of predator odor, 2,4,5-trimethylthiazole (TMT) and examined the effect on LH pulses. Subsequently, we virally infected Ucn3-cre-tdTomato mice with inhibitory designer receptor exclusively activated by designer drugs (DREADDs) targeting MePD Ucn3 neurons while exposing mice to TMT or restraint stress and examined the effect on LH pulsatility as well as corticosterone release. Administration of Ucn3 into the MePD dose-dependently inhibited LH pulses and administration of Astressin2B blocked the suppressive effect of TMT on LH pulsatility. Additionally, DREADDs inhibition of MePD Ucn3 neurons blocked TMT and restraint stress-induced inhibition of LH pulses and corticosterone release. These results demonstrate for the first time that Ucn3 neurons in the MePD mediate psychosocial stress-induced suppression of the GnRH pulse generator and corticosterone secretion. Ucn3 signalling in the MePD plays a role in modulating the hypothalamic-pituitary-gonadal and hypothalamic-pituitary-adrenal axes, and this brain locus may represent a nodal center in the interaction between the reproductive and stress axes.
Collapse
Affiliation(s)
- Deyana Ivanova
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College, London SE1 1UL, UK
- Correspondence: Deyana Ivanova, PhD, Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Science and Medicine, King’s College London, 2.92W Hodgkin Building, Guy’s Campus, London SE1 1UL, UK.
| | - Xiao-Feng Li
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College, London SE1 1UL, UK
| | - Caitlin McIntyre
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College, London SE1 1UL, UK
| | - Yali Liu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, People’s Republic of China
| | - Lingsi Kong
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College, London SE1 1UL, UK
| | - Kevin T O’Byrne
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College, London SE1 1UL, UK
- Correspondence: Kevin T. O’Byrne, Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Science and Medicine, King’s College London, 2.92W Hodgkin Building, Guy’s Campus, London SE1 1UL, UK.
| |
Collapse
|
19
|
Phumsatitpong C, Wagenmaker ER, Moenter SM. Neuroendocrine interactions of the stress and reproductive axes. Front Neuroendocrinol 2021; 63:100928. [PMID: 34171353 PMCID: PMC8605987 DOI: 10.1016/j.yfrne.2021.100928] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 01/27/2023]
Abstract
Reproduction is controlled by a sequential regulation of the hypothalamo-pituitary-gonadal (HPG) axis. The HPG axis integrates multiple inputs to maintain proper reproductive functions. It has long been demonstrated that stress alters fertility. Nonetheless, the central mechanisms of how stress interacts with the reproductive system are not fully understood. One of the major pathways that is activated during the stress response is the hypothalamo-pituitary-adrenal (HPA) axis. In this review, we discuss several aspects of the interactions between these two neuroendocrine systems to offer insights to mechanisms of how the HPA and HPG axes interact. We have also included discussions of other systems, for example GABA-producing neurons, where they are informative to the overall picture of stress effects on reproduction.
Collapse
Affiliation(s)
- Chayarndorn Phumsatitpong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Elizabeth R Wagenmaker
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
20
|
Kovács DK, Farkas N, Soós A, Hegyi P, Kelava L, Eitmann S, Schekk A, Molnár Z, Erőss B, Balaskó M. Assessment of clinical data on urocortins and their therapeutic potential in cardiovascular diseases: A systematic review and meta-analysis. Clin Transl Sci 2021; 14:2461-2473. [PMID: 34378854 PMCID: PMC8604231 DOI: 10.1111/cts.13114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 05/26/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) and cardiovascular diseases present public health challenges. Although great progress was achieved in their treatment, there is continuous need for new therapies. Urocortins of the corticotropin neuropeptide family were reported to exert beneficial effects in animal models of HF and cardiovascular diseases. We aimed to assess the available clinical evidence on the potential role of urocortins in HF and other cardiovascular diseases. We explored MEDLINE, Embase, CENTRAL, and Scopus databases. Twenty‐seven studies were included in the qualitative and 15 studies (2005 patients) in the quantitative syntheses. Available data allowed us to meta‐analyze the blood pressure (BP) lowering and heart rate (HR) increasing effects of urocortin 2 in HF with reduced ejection fraction. We applied meta‐regression to explore the association between left ventricular ejection fraction and serum urocortin 1 and urocortin 2 levels. Short‐term urocortin 2 infusion decreased mean arterial pressure in chronic HF with reduced ejection fraction (mean difference = −9.161 mmHg, 95% confidence interval [CI] −12.661 to −5.660 mmHg, p < 0.001). Such infusions increased HR mildly (mean difference = 5.629 beats/min, 95% CI 1.612 to 9.646 beats/min, p = 0.006). Although some studies reported increased urocortin 1 and urocortin 2 levels in HF with growing severity, our meta‐regressions failed to confirm associations between blood urocortin levels and left ventricular ejection fraction. Clinical evidence confirms short‐term BP lowering effects of urocortin 2, whereas individual studies report additional beneficial effects. Further clinical investigations are necessary to confirm the latter and the long‐term value of these peptides in cardiovascular diseases. Review protocol: CRD42020163203.
Collapse
Affiliation(s)
- Dóra K Kovács
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Nelli Farkas
- Institute of Bioanalysis, Medical School, University of Pécs, Pécs, Hungary
| | - Alexandra Soós
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Hegyi
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary.,Department of Translational Medicine, First Department of Internal Medicine, University of Pécs, Pécs, Hungary.,MTA-SZTE Translational Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Leonardo Kelava
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Szimonetta Eitmann
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Anna Schekk
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Zsolt Molnár
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Bálint Erőss
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Márta Balaskó
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
21
|
Cano G, Hernan SL, Sved AF. Centrally Projecting Edinger-Westphal Nucleus in the Control of Sympathetic Outflow and Energy Homeostasis. Brain Sci 2021; 11:1005. [PMID: 34439626 PMCID: PMC8392615 DOI: 10.3390/brainsci11081005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
The centrally projecting Edinger-Westphal nucleus (EWcp) is a midbrain neuronal group, adjacent but segregated from the preganglionic Edinger-Westphal nucleus that projects to the ciliary ganglion. The EWcp plays a crucial role in stress responses and in maintaining energy homeostasis under conditions that require an adjustment of energy expenditure, by virtue of modulating heart rate and blood pressure, thermogenesis, food intake, and fat and glucose metabolism. This modulation is ultimately mediated by changes in the sympathetic outflow to several effector organs, including the adrenal gland, heart, kidneys, brown and white adipose tissues and pancreas, in response to environmental conditions and the animal's energy state, providing for appropriate energy utilization. Classic neuroanatomical studies have shown that the EWcp receives inputs from forebrain regions involved in these functions and projects to presympathetic neuronal populations in the brainstem. Transneuronal tracing with pseudorabies virus has demonstrated that the EWcp is connected polysynaptically with central circuits that provide sympathetic innervation to all these effector organs that are critical for stress responses and energy homeostasis. We propose that EWcp integrates multimodal signals (stress, thermal, metabolic, endocrine, etc.) and modulates the sympathetic output simultaneously to multiple effector organs to maintain energy homeostasis under different conditions that require adjustments of energy demands.
Collapse
Affiliation(s)
- Georgina Cano
- Department of Neuroscience, A210 Langley Hall, University of Pittsburgh, Pittsburgh, PA 15260, USA; (S.L.H.); (A.F.S.)
| | | | | |
Collapse
|
22
|
Vandael D, Wierda K, Vints K, Baatsen P, De Groef L, Moons L, Rybakin V, Gounko NV. Corticotropin-releasing factor induces functional and structural synaptic remodelling in acute stress. Transl Psychiatry 2021; 11:378. [PMID: 34234103 PMCID: PMC8263770 DOI: 10.1038/s41398-021-01497-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
Biological responses to stress are complex and highly conserved. Corticotropin-releasing factor (CRF) plays a central role in regulating these lifesaving physiological responses to stress. We show that, in mice, CRF rapidly changes Schaffer Collateral (SC) input into hippocampal CA1 pyramidal cells (PC) by modulating both functional and structural aspects of these synapses. Host exposure to acute stress, in vivo CRF injection, and ex vivo CRF application all result in fast de novo formation and remodeling of existing dendritic spines. Functionally, CRF leads to a rapid increase in synaptic strength of SC input into CA1 neurons, e.g., increase in spontaneous neurotransmitter release, paired-pulse facilitation, and repetitive excitability and improves synaptic plasticity: long-term potentiation (LTP) and long-term depression (LTD). In line with the changes in synaptic function, CRF increases the number of presynaptic vesicles, induces redistribution of vesicles towards the active zone, increases active zone size, and improves the alignment of the pre- and postsynaptic compartments. Therefore, CRF rapidly enhances synaptic communication in the hippocampus, potentially playing a crucial role in the enhanced memory consolidation in acute stress.
Collapse
Affiliation(s)
- Dorien Vandael
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49,, box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49,, box 602, 3000, Leuven, Belgium
| | - Keimpe Wierda
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49,, box 602, 3000, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Electrophysiology Expertise Unit, O&N5 Herestraat 49, 3000, Leuven, Belgium
| | - Katlijn Vints
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49,, box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49,, box 602, 3000, Leuven, Belgium
| | - Pieter Baatsen
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49,, box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49,, box 602, 3000, Leuven, Belgium
| | - Lies De Groef
- KU Leuven Faculty of Science, Department of Biology, Laboratory of Neural Circuit Development and Regeneration, Naamsestraat 61, 3000, Leuven, Belgium
| | - Lieve Moons
- KU Leuven Faculty of Science, Department of Biology, Laboratory of Neural Circuit Development and Regeneration, Naamsestraat 61, 3000, Leuven, Belgium
| | - Vasily Rybakin
- National University of Singapore, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and Immunology Program, 5 Science Drive 2, Blk MD4, 117545, Singapore, Singapore
| | - Natalia V Gounko
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49,, box 602, 3000, Leuven, Belgium.
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49,, box 602, 3000, Leuven, Belgium.
| |
Collapse
|
23
|
Malta MB, Martins J, Novaes LS, Dos Santos NB, Sita L, Camarini R, Scavone C, Bittencourt J, Munhoz CD. Norepinephrine and Glucocorticoids Modulate Chronic Unpredictable Stress-Induced Increase in the Type 2 CRF and Glucocorticoid Receptors in Brain Structures Related to the HPA Axis Activation. Mol Neurobiol 2021; 58:4871-4885. [PMID: 34213722 DOI: 10.1007/s12035-021-02470-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/22/2021] [Indexed: 12/26/2022]
Abstract
The stress response is multifactorial and enrolls circuitries to build a coordinated reaction, leading to behavioral, endocrine, and autonomic changes. These changes are mainly related to the hypothalamus-pituitary-adrenal (HPA) axis activation and the organism's integrity. However, when self-regulation is ineffective, stress becomes harmful and predisposes the organism to pathologies. The chronic unpredictable stress (CUS) is a widely used experimental model since it induces physiological and behavioral changes and better mimics the stressors variability encountered in daily life. Corticotropin-releasing factor (CRF) and glucocorticoids (GCs) are deeply implicated in the CUS-induced physiological and behavioral changes. Nonetheless, the CUS modulation of CRF receptors and GR and the norepinephrine role in extra-hypothalamic brain areas were not well explored. Here, we show that 14 days of CUS induced a long-lasting HPA axis hyperactivity evidenced by plasmatic corticosterone increase and adrenal gland hypertrophy, which was dependent on both GCs and NE release induced by each stress session. CUS also increased CRF2 mRNA expression and GR protein levels in fundamental brain structures related to HPA regulation and behavior, such as the lateral septal nucleus intermedia part (LSI), ventromedial hypothalamic nucleus (VMH), and central nucleus of the amygdala (CeA). We also showed that NE participates in the CUS-induced increase in CRF2 and GR levels in the LSI, reinforcing the locus coeruleus (LC) involvement in the HPA axis modulation. Despite the CUS-induced molecular changes in essential areas related to anxiety-like behavior, this phenotype was not observed in CUS animals 24 h after the last stress session.
Collapse
Affiliation(s)
- Marilia B Malta
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Joelcimar Martins
- Central of Facilities, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, 05508-000, Brazil
| | - Leonardo S Novaes
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Nilton B Dos Santos
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Luciane Sita
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, 05508-000, Brazil
| | - Rosana Camarini
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Jackson Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, 05508-000, Brazil.,Center for Neurosciences and Behavior, Institute of Psychology, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Carolina D Munhoz
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
24
|
Antunes C, Da Silva JD, Guerra-Gomes S, Alves ND, Ferreira F, Loureiro-Campos E, Branco MR, Sousa N, Reik W, Pinto L, Marques CJ. Tet3 ablation in adult brain neurons increases anxiety-like behavior and regulates cognitive function in mice. Mol Psychiatry 2021; 26:1445-1457. [PMID: 32103150 DOI: 10.1038/s41380-020-0695-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 01/16/2020] [Accepted: 02/18/2020] [Indexed: 01/25/2023]
Abstract
TET3 is a member of the ten-eleven translocation (TET) family of enzymes which oxidize 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC). Tet3 is highly expressed in the brain, where 5hmC levels are most abundant. In adult mice, we observed that TET3 is present in mature neurons and oligodendrocytes but is absent in astrocytes. To investigate the function of TET3 in adult postmitotic neurons, we crossed Tet3 floxed mice with a neuronal Cre-expressing mouse line, Camk2a-CreERT2, obtaining a Tet3 conditional KO (cKO) mouse line. Ablation of Tet3 in adult mature neurons resulted in increased anxiety-like behavior with concomitant hypercorticalism, and impaired hippocampal-dependent spatial orientation. Transcriptome and gene-specific expression analysis of the hippocampus showed dysregulation of genes involved in glucocorticoid signaling pathway (HPA axis) in the ventral hippocampus, whereas upregulation of immediate early genes was observed in both dorsal and ventral hippocampal areas. In addition, Tet3 cKO mice exhibit increased dendritic spine maturation in the ventral CA1 hippocampal subregion. Based on these observations, we suggest that TET3 is involved in molecular alterations that govern hippocampal-dependent functions. These results reveal a critical role for epigenetic modifications in modulating brain functions, opening new insights into the molecular basis of neurological disorders.
Collapse
Affiliation(s)
- Cláudia Antunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Jorge D Da Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Sónia Guerra-Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Nuno D Alves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Fábio Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Eduardo Loureiro-Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Miguel R Branco
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Wolf Reik
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.,The Wellcome Trust Sanger Institute, Cambridge, CB10 1SA, UK
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.
| | - C Joana Marques
- Department of Genetics, Faculty of Medicine, University of Porto (FMUP), 4200-319, Porto, Portugal. .,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
25
|
Horii-Hayashi N, Nomoto K, Endo N, Yamanaka A, Kikusui T, Nishi M. Hypothalamic perifornical Urocortin-3 neurons modulate defensive responses to a potential threat stimulus. iScience 2021; 24:101908. [PMID: 33385113 PMCID: PMC7770982 DOI: 10.1016/j.isci.2020.101908] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 08/31/2020] [Accepted: 12/03/2020] [Indexed: 11/13/2022] Open
Abstract
Defensive behaviors are evolved responses to threat stimuli, and a potential threat elicits risk assessment (RA) behavior. However, neural mechanisms underlying RA behavior are hardly understood. Urocortin-3 (Ucn3) is a member of corticotropin-releasing factor peptide family and here, we report that Ucn3 neurons in the hypothalamic perifornical area (PeFA) are involved in RA of a novel object, a potential threat stimulus, in mice. Histological and in vivo fiber photometry studies revealed that the activity of PeFA Ucn3 neurons was associated with novel object investigation involving the stretch-attend posture, a behavioral marker for RA. Chemogenetic activation of these neurons increased RA and burying behaviors toward a novel object without affecting anxiety and corticosterone levels. Ablation of these neurons caused the abnormal behaviors of gnawing and direct contacts with novel objects, especially in a home-cage. These results suggest that PeFA Ucn3 neurons modulate defensive responses to a potential threat stimulus.
Collapse
Affiliation(s)
- Noriko Horii-Hayashi
- Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara 643-8521, Japan
| | - Kensaku Nomoto
- Companion Animal Research Laboratory, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa 252–5201, Japan
- Department of Physiology, Dokkyo Medical University, Mibu, Tochigi, 321-0293, Japan
| | - Nozomi Endo
- Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara 643-8521, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Takefumi Kikusui
- Companion Animal Research Laboratory, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa 252–5201, Japan
| | - Mayumi Nishi
- Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara 643-8521, Japan
| |
Collapse
|
26
|
Augustine-Rauch K, Liaw JJ, Graziano M. T4-mediated rescue of aortic malformations in hypothyroid rats indicates maternal thyroid status can affect great vessel development. Toxicol Appl Pharmacol 2020; 411:115367. [PMID: 33340518 DOI: 10.1016/j.taap.2020.115367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 11/30/2022]
Abstract
Pexacerfont is a corticotrophin-releasing factor subtype 1 receptor (CRF-1) antagonist developed for potential treatment of anxiety and stress-related disorders. In male rats, pexacerfont caused hepatic enzyme induction leading to increased thyroxine (T4) clearance. When administered to pregnant rats on gestation day 6 to 15, pexacerfont at 300 mg/kg/day (30× mean AUC in humans at 100 mg/day) produced similar effects on thyroid homeostasis with serum T4 and thyroid-stimulating hormone levels that were 0.3-0.5× and 3.3-3.7× of controls, respectively. At this dose, fetuses of pexacerfont-treated dams presented findings associated with maternal hypothyroidism including growth retardation and increased skeletal alterations. Additionally, there were unexpected great vessel malformations that were mostly derived from the 4th pharyngeal arch artery in 5 (4.3%) fetuses from 3 (15.8%) litters. The etiology was unclear whether the vascular malformations were related to insufficient thyroid hormones or another mechanism. To better understand this relationship, pregnant rats were implanted with a subcutaneous L-thyroxine pellet designed to provide a sustained release of T4 throughout organogenesis in rat embryos (GD 6 to 15; the dosing period of pexacerfont). T4 supplementation produced a near euthyroid state in pexacerfont-treated dams and completely prevented the fetal vascular malformations. These results suggest maternal T4 levels during organogenesis may have a role in great vessel morphogenesis associated with patterning and/or regression of pharyngeal arch arteries. Although previous clinical reports have speculated a potential relationship between thyroid hormone homeostasis and early cardiovascular development, this is the first report to experimentally demonstrate this relationship in great vessel morphogenesis.
Collapse
Affiliation(s)
| | - Jiin-Jia Liaw
- Non Clinical Safety, Bristol Myers Squibb, New Brunswick, NJ, USA
| | - Michael Graziano
- Non Clinical Safety, Bristol Myers Squibb, New Brunswick, NJ, USA
| |
Collapse
|
27
|
Chrissobolis S, Luu AN, Waldschmidt RA, Yoakum ME, D'Souza MS. Targeting the renin angiotensin system for the treatment of anxiety and depression. Pharmacol Biochem Behav 2020; 199:173063. [PMID: 33115635 DOI: 10.1016/j.pbb.2020.173063] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/13/2020] [Accepted: 10/22/2020] [Indexed: 12/27/2022]
Abstract
Emotional disorders like anxiety and depression are responsible for considerable morbidity and mortality all over the world. Several antidepressant and anxiolytic medications are available for the treatment of anxiety and depression. However, a significant number of patients either do not respond to these medications or respond inadequately. Hence, there is a need to identify novel targets for the treatment of anxiety and depression. In this review we focus on the renin angiotensin system (RAS) as a potential target for the treatment of these disorders. We review work that has evaluated the effects of various compounds targeting the RAS on anxiety- and depression-like behaviors. Further, we suggest future work that must be carried out to fully exploit the RAS for the treatment of anxiety and depression. The RAS provides an attractive target for both the identification of novel anxiolytic and antidepressant medications and/or for enhancing the efficacy of currently available medications used for the treatment of anxiety and depression.
Collapse
Affiliation(s)
- Sophocles Chrissobolis
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH 45810, United States of America
| | - Anh N Luu
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH 45810, United States of America
| | - Ryan A Waldschmidt
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH 45810, United States of America
| | - Madison E Yoakum
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH 45810, United States of America
| | - Manoranjan S D'Souza
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH 45810, United States of America.
| |
Collapse
|
28
|
Rilett KC, Luo OD, McVey-Neufeld KA, MacKenzie RN, Foster JA. Loss of T cells influences sex differences in stress-related gene expression. J Neuroimmunol 2020; 343:577213. [PMID: 32278229 DOI: 10.1016/j.jneuroim.2020.577213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/05/2020] [Accepted: 03/07/2020] [Indexed: 12/12/2022]
Abstract
Deficiencies in the adaptive immune system have been linked to anxiety-like behaviours and stress reactivity. Mice lacking T lymphocytes through knockout of the T cell receptor (TCR) β and δ chains were compared to wild type C57Bl/6 mice. Central stress circuitry gene expression was assessed following repeated restraint stress. TCRβ-/-δ-/- mice showed an increased baseline plasma corticosterone and exaggerated changes in stress-related gene expression after repeated restraint stress. Sexual dimorphic stress responses were observed in wild-type C57Bl/6 mice but not in TCRβ-/-δ-/- mice. These data suggest that T cell-brain interactions influence sex-differences in CNS stress circuitry and stress reactivity.
Collapse
Affiliation(s)
- Kelly C Rilett
- Department of Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, ON, Canada
| | - Owen D Luo
- Department of Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, ON, Canada.
| | - Karen-Anne McVey-Neufeld
- Department of Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, ON, Canada.
| | - Robyn N MacKenzie
- Department of Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, ON, Canada
| | - Jane A Foster
- Department of Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
29
|
Simpson S, Shankar K, Kimbrough A, George O. Role of corticotropin-releasing factor in alcohol and nicotine addiction. Brain Res 2020; 1740:146850. [PMID: 32330519 DOI: 10.1016/j.brainres.2020.146850] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/31/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022]
Abstract
The two most prevalent substance use disorders involve alcohol and nicotine, which are often co-abused. Robust preclinical and translational evidence indicates that individuals initiate drug use for the acute rewarding effects of the substance. The development of negative emotional states is key for the transition from recreational use to substance use disorders as subjects seek the substance to obtain relief from the negative emotional states of acute withdrawal and protracted abstinence. The neuropeptide corticotropin-releasing factor (CRF) is a major regulator of the brain stress system and key in the development of negative affective states. The present review examines the role of CRF in preclinical models of alcohol and nicotine abuse and explores links between CRF and anxiety-like, dysphoria-like, and other negative affective states. Finally, the present review discusses preclinical models of nicotine and alcohol use with regard to the CRF system, advances in molecular and genetic manipulations of CRF, and the importance of examining both males and females in this field of research.
Collapse
Affiliation(s)
- Sierra Simpson
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States; Department of Neuroscience, Scripps Research, La Jolla, CA 92037, United States
| | - Kokila Shankar
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States; Department of Neuroscience, Scripps Research, La Jolla, CA 92037, United States
| | - Adam Kimbrough
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States
| | - Olivier George
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States.
| |
Collapse
|
30
|
Qi J, Zhang X, Li Y, Xu S, Wang M, Chen H, Tang N, Wang S, Wang B, Chen D, Zhou B, Li Z. The suppression effects of feeding and mechanisms in CRF system of animals. Gene 2020; 733:144363. [PMID: 31935510 DOI: 10.1016/j.gene.2020.144363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 01/27/2023]
Abstract
CRF system is comprised of 4 homologous lineages, 2 main receptors (CRF-R1 and CRF-R2), and a binding protein CRF-BP. The homologous lineages are corticotropin-releasing factor (CRF), urotensin I (UI)/sauvagine (SVG)/urocortin 1 (UCN1), urocortin 2 (UCN2), and urocortin 3 (UCN3), and UI, SVG, UCN1 are orthologous genes. CRF system genes are widely distributed in the brain and gastrointestinal tract, which may relate to feeding regulation. According the research progress about CRF system on mammals and non-mammals, this paper summarized the discovery, structure, tissue distribution, appetite regulation and mechanism of CRF system in animals, which can provide the reference for further research and production of feeding regulation and growth in mammals and fish species.
Collapse
Affiliation(s)
- Jinwen Qi
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, 156# Gaozhuang Bridge Community, Yibin, Sichuan, China
| | - Xin Zhang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China; The Key Laboratory of Mariculture, Ministry of Education, Fisheries College, Ocean University of China, 5# Yushan Road, Qingdao, Shandong, China
| | - Ya Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China
| | - Shaoqi Xu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China
| | - Mei Wang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China
| | - Hu Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China
| | - Ni Tang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China
| | - Shuyao Wang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China
| | - Bin Wang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China
| | - Defang Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China
| | - Bo Zhou
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, 156# Gaozhuang Bridge Community, Yibin, Sichuan, China.
| | - Zhiqiong Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211# Huimin Road, Chengdu, Sichuan, China.
| |
Collapse
|
31
|
Corticotropin-Releasing Factor Family: A Stress Hormone-Receptor System's Emerging Role in Mediating Sex-Specific Signaling. Cells 2020; 9:cells9040839. [PMID: 32244319 PMCID: PMC7226788 DOI: 10.3390/cells9040839] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/25/2020] [Accepted: 03/28/2020] [Indexed: 12/14/2022] Open
Abstract
No organ in the body is impervious to the effects of stress, and a coordinated response from all organs is essential to deal with stressors. A dysregulated stress response that fails to bring systems back to homeostasis leads to compromised function and ultimately a diseased state. The components of the corticotropin-releasing factor (CRF) family, an ancient and evolutionarily conserved stress hormone-receptor system, helps both initiate stress responses and bring systems back to homeostasis once the stressors are removed. The mammalian CRF family comprises of four known agonists, CRF and urocortins (UCN1–3), and two known G protein-coupled receptors (GPCRs), CRF1 and CRF2. Evolutionarily, precursors of CRF- and urocortin-like peptides and their receptors were involved in osmoregulation/diuretic functions, in addition to nutrient sensing. Both CRF and UCN1 peptide hormones as well as their receptors appeared after a duplication event nearly 400 million years ago. All four agonists and both CRF receptors show sex-specific changes in expression and/or function, and single nucleotide polymorphisms are associated with a plethora of human diseases. CRF receptors harbor N-terminal cleavable peptide sequences, conferring biased ligand properties. CRF receptors have the ability to heteromerize with each other as well as with other GPCRs. Taken together, CRF receptors and their agonists due to their versatile functional adaptability mediate nuanced responses and are uniquely positioned to orchestrate sex-specific signaling and function in several tissues.
Collapse
|
32
|
The blockade of corticotropin-releasing factor 1 receptor attenuates anxiety-related symptoms and hypothalamus-pituitary-adrenal axis reactivity in mice with mild traumatic brain injury. Behav Pharmacol 2020; 30:220-228. [PMID: 30883392 DOI: 10.1097/fbp.0000000000000450] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Recent studies have shown that mild traumatic brain injury (mTBI) is associated with higher risk for anxiety-related disorders. Dysregulation in the hypothalamus-pituitary-adrenal (HPA) axis following mTBI has been proposed to be involved in the development of neurobehavioral abnormalities; however, the underlying mechanisms are largely unknown. The aim of this study was to determine whether the corticotropin-releasing-factor-1 (CRF-1) receptor is involved in the regulation of anxiety-related symptoms in a mouse model of mTBI. Animals with or without mTBI received intracerebroventricular injections of a CRF-1 receptor agonist (CRF; 0.01 nmol/mouse) or antagonist (antalarmin; 1 µg/mouse) for 5 days, and then the animals were subjected to anxiety tests (light-dark box and zero maze). The levels of adrenocorticotropic hormone and corticosterone, the most important markers of HPA axis, were also measured after behavioral tests. Our results indicated that mTBI-induced anxiety-related symptoms in mice through increased levels of adrenocorticotropic hormone and corticosterone, showing HPA axis hyperactivity. Interestingly, activation of CRF receptor by a subthreshold dose of CRF resulted in significant increases in anxiety-like behaviors and HPA axis response to stress, whereas blockade of CRF receptors by a subthreshold dose of antalarmin decreased anxiety-related symptoms and HPA axis response to stress in mTBI-induced mice. Collectively, these findings suggest that the CRF-1 receptor plays an important role in the regulation of anxiety-related behaviors following mTBI induction in mice and support the hypothesis that blockade of the CRF-1 receptor may be a promising therapeutic target for anxiety-related disorders in patients with TBI.
Collapse
|
33
|
Gupta SK, Patel SK, Tomar MS, Singh SK, Mesharam MK, Krishnamurthy S. Long-term exposure of 2450 MHz electromagnetic radiation induces stress and anxiety like behavior in rats. Neurochem Int 2019; 128:1-13. [DOI: 10.1016/j.neuint.2019.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/20/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
|
34
|
Pandey GN, Rizavi HS, Bhaumik R, Ren X. Increased protein and mRNA expression of corticotropin-releasing factor (CRF), decreased CRF receptors and CRF binding protein in specific postmortem brain areas of teenage suicide subjects. Psychoneuroendocrinology 2019; 106:233-243. [PMID: 31005044 PMCID: PMC7061258 DOI: 10.1016/j.psyneuen.2019.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/07/2019] [Accepted: 04/10/2019] [Indexed: 10/27/2022]
Abstract
Overactivity of hypothalamic-pituitary-adrenal (HPA) axis function has been implicated in depression and suicidal behavior. This is based on the observation of an abnormal dexamethasone (DEX) and DEX-adrenocorticotropic hormone (ACTH) test in patients with depression and suicidal behavior. Recently, some studies have also found abnormalities of glucocorticoid receptors (GR), mineralocorticoid receptors (MR), corticotropin releasing factor (CRF), CRF receptors (CRF-R) and CRF binding protein (CRF-BP) in depressed and suicidal patients. Some investigators have also observed increased levels of CRF in the cerebrospinal fluid (CSF) and altered levels of MR, GR and CRF in the postmortem brain of depressed and suicidal subjects. We have earlier reported decreased protein and mRNA expression of GR and GILZ, a chaperone protein, in the postmortem brain of teenage suicide subjects. We have further studied CRF and its receptors in different areas of the postmortem brain of suicide subjects, i.e., the prefrontal cortex (PFC), hippocampus (HIPPO), subiculum and amygdala (AMY) from teenage suicide subjects. The CRF and its receptors were determined in the PFC (Brodmann area 9), HIPPO, subiculum and different amygdaloid nuclei from 24 normal control subjects and 24 teenage suicide subjects. Protein expression of CRF, its receptors and CRF-BP was determined by immunolabeling using the Western blot technique and mRNA expression was determined by real-time PCR (qPCR) technique. We found that the mRNA levels of CRF were significantly increased in the PFC, in the central amygdaloid nucleus (CeAMY) and in the subiculum. mRNA levels of CRF-R1 and CRF-BP were significantly decreased in the PFC. We did not find any changes in the HIPPO of any of the CRF components we studied. When we compared the protein expression of CRF components we found that CRF was significantly increased and CRF-R1, CRF-R2 and CRF-BP significantly decreased in the PFC. On the other hand, there were no changes in the protein expression of CRF components in the HIPPO. Our results in the postmortem brain suggest that, as found by clinical studies in the CSF, there are significant alterations of CRF and its receptors in the postmortem brain of teenage suicide subjects. These alterations of CRF and its components were region-specific, as changes were not generally observed in the HIPPO.
Collapse
Affiliation(s)
- Ghanshyam N. Pandey
- Corresponding Author: Ghanshyam N. Pandey, Ph.D., University of Illinois at Chicago, 1601 West Taylor Street, Chicago, IL 60612, USA, Phone (312) 413-4540, Fax: (312) 413-4547,
| | | | | | | |
Collapse
|
35
|
Fischl MJ, Ueberfuhr MA, Drexl M, Pagella S, Sinclair JL, Alexandrova O, Deussing JM, Kopp-Scheinpflug C. Urocortin 3 signalling in the auditory brainstem aids recovery of hearing after reversible noise-induced threshold shift. J Physiol 2019; 597:4341-4355. [PMID: 31270820 PMCID: PMC6852351 DOI: 10.1113/jp278132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/03/2019] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Ongoing, moderate noise exposure does not instantly damage the auditory system but may cause lasting deficits, such as elevated thresholds and accelerated ageing of the auditory system. The neuromodulatory peptide urocortin-3 (UCN3) is involved in the body's recovery from a stress response, and is also expressed in the cochlea and the auditory brainstem. Lack of UCN3 facilitates age-induced hearing loss and causes permanently elevated auditory thresholds following a single 2 h noise exposure at moderate intensities. Outer hair cell function in mice lacking UCN3 is unaffected, so that the observed auditory deficits are most likely due to inner hair cell function or central mechanisms. Highly specific, rather than ubiquitous, expression of UCN3 in the brain renders it a promising candidate for designing drugs to ameliorate stress-related auditory deficits, including recovery from acoustic trauma. ABSTRACT Environmental acoustic noise is omnipresent in our modern society, with sound levels that are considered non-damaging still causing long-lasting or permanent changes in the auditory system. The small neuromodulatory peptide urocortin-3 (UCN3) is the endogenous ligand for corticotropin-releasing factor receptor type 2 and together they are known to play an important role in stress recovery. UCN3 expression has been observed in the auditory brainstem, but its role remains unclear. Here we describe the detailed distribution of UCN3 expression in the murine auditory brainstem and provide evidence that UCN3 is expressed in the synaptic region of inner hair cells in the cochlea. We also show that mice with deficient UCN3 signalling experience premature ageing of the auditory system starting at an age of 4.7 months with significantly elevated thresholds of auditory brainstem responses (ABRs) compared to age-matched wild-type mice. Following a single, 2 h exposure to moderate (84 or 94 dB SPL) noise, UCN3-deficient mice exhibited significantly larger shifts in ABR thresholds combined with maladaptive recovery. In wild-type mice, the same noise exposure did not cause lasting changes to auditory thresholds. The presence of UCN3-expressing neurons throughout the auditory brainstem and the predisposition to hearing loss caused by preventing its normal expression suggests UCN3 as an important neuromodulatory peptide in the auditory system's response to loud sounds.
Collapse
Affiliation(s)
- Matthew J Fischl
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany
| | - Margarete A Ueberfuhr
- German Center for Vertigo and Balance Disorders, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University, Munich, Germany
| | - Markus Drexl
- German Center for Vertigo and Balance Disorders, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Sara Pagella
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University, Munich, Germany
| | - James L Sinclair
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany
| | - Olga Alexandrova
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany
| | - Jan M Deussing
- Max Planck Institute of Psychiatry, Molecular Neurogenetics, Munich, Germany
| | - Conny Kopp-Scheinpflug
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
36
|
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: 4.2] [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
|
37
|
Borg ML, Massart J, Schönke M, De Castro Barbosa T, Guo L, Wade M, Alsina-Fernandez J, Miles R, Ryan A, Bauer S, Coskun T, O'Farrell E, Niemeier EM, Chibalin AV, Krook A, Karlsson HK, Brozinick JT, Zierath JR. Modified UCN2 Peptide Acts as an Insulin Sensitizer in Skeletal Muscle of Obese Mice. Diabetes 2019; 68:1403-1414. [PMID: 31010957 DOI: 10.2337/db18-1237] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/08/2019] [Indexed: 11/13/2022]
Abstract
The neuropeptide urocortin 2 (UCN2) and its receptor corticotropin-releasing hormone receptor 2 (CRHR2) are highly expressed in skeletal muscle and play a role in regulating energy balance and glucose metabolism. We investigated a modified UCN2 peptide as a potential therapeutic agent for the treatment of obesity and insulin resistance, with a specific focus on skeletal muscle. High-fat-fed mice (C57BL/6J) were injected daily with a PEGylated UCN2 peptide (compound A) at 0.3 mg/kg subcutaneously for 14 days. Compound A reduced body weight, food intake, whole-body fat mass, and intramuscular triglycerides compared with vehicle-treated controls. Furthermore, whole-body glucose tolerance was improved by compound A treatment, with increased insulin-stimulated Akt phosphorylation at Ser473 and Thr308 in skeletal muscle, concomitant with increased glucose transport into extensor digitorum longus and gastrocnemius muscle. Mechanistically, this is linked to a direct effect on skeletal muscle because ex vivo exposure of soleus muscle from chow-fed lean mice to compound A increased glucose transport and insulin signaling. Moreover, exposure of GLUT4-Myc-labeled L6 myoblasts to compound A increased GLUT4 trafficking. Our results demonstrate that modified UCN2 peptides may be efficacious in the treatment of type 2 diabetes by acting as an insulin sensitizer in skeletal muscle.
Collapse
Affiliation(s)
- Melissa L Borg
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Julie Massart
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Milena Schönke
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Thais De Castro Barbosa
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Lili Guo
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Mark Wade
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | | | - Rebecca Miles
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Andrew Ryan
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Steve Bauer
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Tamer Coskun
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Elizabeth O'Farrell
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Evan M Niemeier
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Håkan K Karlsson
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Joseph T Brozinick
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
38
|
Differential and temporal expression of corticotropin releasing hormone and its receptors in the nucleus of the hippocampal commissure and paraventricular nucleus during the stress response in chickens (Gallus gallus). Brain Res 2019; 1714:1-7. [DOI: 10.1016/j.brainres.2019.02.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/06/2019] [Accepted: 02/14/2019] [Indexed: 12/21/2022]
|
39
|
Yang LZ, Chen Y. Research on the Effects of the Chronic Treatment With Different Doses of Urocortin 2 in Heart Failure Rats. Dose Response 2019; 17:1559325819860018. [PMID: 31263386 PMCID: PMC6595674 DOI: 10.1177/1559325819860018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/29/2019] [Accepted: 06/04/2019] [Indexed: 12/19/2022] Open
Abstract
Corticotropin-releasing factor (CRF) receptor type 2 (CRF2) exists in both cardiomyocytes and neurocytes. The purpose of this research was to explore if chronic treatment with urocortin 2 (UCN2), a CRF2 receptor agonist, at different doses can improve prognosis and regulate the expression of CRF2 receptor and calcium handling proteins without any adverse effects on behavior in heart failure. Heart failure was established in Sprague-Dawley rats and was confirmed by echocardiography. Heart failure rats were injected intraperitoneally with UCN2 (5, 10, or 20 µg·kg−1·d−1) for 30 days. Survival rate, cardiac function, expressions of cardiac CRF2 receptor, RyR2, SERCA2, and hypothalamic and hippocampal c-FOS, CRF receptor type 1 (CRF1) and CRF2 receptor were determined. Behavior was evaluated by Morris Water-Maze and Open-Field tests. Results showed that chronic peripheral UCN2 treatment improved survival rate in a dose–response manner and increased cardiac function and expression of CRF2 receptor and SERCA2 in heart failure, especially at the high dosage. Moreover, cellular-fos (c-FOS), CRF1 receptor, and CRF2 receptor expressions of both hypothalamic and hippocampal tissues were significantly increased in high dosage group. Furthermore, the behavior tests suggested that chronic UCN2 treatment did not exacerbate stress/anxiety-like behavior in HF. In conclusion, chronic peripheral treatment with UCN2 increases survival in a dose–response manner in heart failure rats without inducing stress/anxiety-like behavior.
Collapse
Affiliation(s)
- Li-Zhen Yang
- Division of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Chen
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
40
|
de Miguel E, Vekovischeva O, Elsilä LV, Panhelainen A, Kankuri E, Aitta-Aho T, Korpi ER. Conditioned Aversion and Neuroplasticity Induced by a Superagonist of Extrasynaptic GABA A Receptors: Correlation With Activation of the Oval BNST Neurons and CRF Mechanisms. Front Mol Neurosci 2019; 12:130. [PMID: 31178693 PMCID: PMC6543524 DOI: 10.3389/fnmol.2019.00130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/03/2019] [Indexed: 11/13/2022] Open
Abstract
THIP (gaboxadol), a superagonist of the δ subunit-containing extrasynaptic GABAA receptors, produces persistent neuroplasticity in dopamine (DA) neurons of the ventral tegmental area (VTA), similarly to rewarding drugs of abuse. However, unlike them THIP lacks abuse potential and induces conditioned place aversion in mice. The mechanism underlying the aversive effects of THIP remains elusive. Here, we show that mild aversive effects of THIP were detected 2 h after administration likely reflecting an anxiety-like state with increased corticosterone release and with central recruitment of corticotropin-releasing factor corticotropin-releasing factor receptor 1 (CRF1) receptors. A detailed immunohistochemical c-Fos expression mapping for THIP-activated brain areas revealed a correlation between the activation of CRF-expressing neurons in the oval nucleus of the bed nuclei of stria terminalis and THIP-induced aversive effects. In addition, the neuroplasticity of mesolimbic DA system (24 h after administration) and conditioned place aversion by THIP after four daily acute sessions were dependent on extrasynaptic GABAA receptors (abolished in δ-GABAA receptor knockout mice) and activation of the CRF1 receptors (abolished in wildtype mice by a CRF1 receptor antagonist). A selective THIP-induced activation of CRF-expressing neurons in the oval part of the bed nucleus of stria terminalis may constitute a novel mechanism for inducing plasticity in a population of VTA DA neurons and aversive behavioral states.
Collapse
Affiliation(s)
- Elena de Miguel
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olga Vekovischeva
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Lauri V Elsilä
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anne Panhelainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Teemu Aitta-Aho
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
41
|
Huang G, Cao X, Li Y, Zhou C, Li L, Wang K, Li H, Yu P, Jin Y, Gao L. Gene expression profile of the hippocampus of rats subjected to traumatic brain injury. J Cell Biochem 2019; 120:15776-15789. [PMID: 31074048 DOI: 10.1002/jcb.28848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 01/02/2019] [Accepted: 01/07/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Guo‐Hui Huang
- Department of Neurosurgery Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai China
| | - Xiang‐Yuan Cao
- Department of Neurosurgery Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai China
| | - Yuan‐Yuan Li
- Department of Endocrinology Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Cheng‐Cheng Zhou
- Department of Neurosurgery Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai China
| | - Lei Li
- Department of Neurosurgery Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai China
| | - Ke Wang
- Department of Neurosurgery Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai China
| | - Hong Li
- Department of Neurosurgery Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai China
| | - Peng Yu
- Department of Neurosurgery Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai China
| | - Yi Jin
- Department of Neurosurgery Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai China
| | - Liang Gao
- Department of Neurosurgery Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai China
| |
Collapse
|
42
|
Bagosi Z, Csabafi K, Karasz G, Jászberényi M, Földesi I, Siska A, Szabó G, Telegdy G. The effects of the urocortins on the hypothalamic-pituitary-adrenal axis - similarities and discordancies between rats and mice. Peptides 2019; 112:1-13. [PMID: 30414887 DOI: 10.1016/j.peptides.2018.10.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/16/2018] [Accepted: 10/30/2018] [Indexed: 11/16/2022]
Abstract
The urocortins (Ucn I, Ucn II and Ucn III) are structural analogues of corticotropin-releasing factor (CRF). The aim of our present experiments was to compare the effects of the urocortins on the hypothalamic-pituitary-adrenal (HPA) axis in rats and mice, including the hypothalamic adrenocorticotropic hormone (ACTH) secretagogues, such as CRF and arginine vasopressin (AVP). Therefore, male CFLP mice and male Wistar rats were injected intracerebroventricularly (icv) with 0.5, 1, 2 and 5 μg/2 μl of Ucn I, Ucn II or Ucn III. After 30 min the animals were decapitated, and then, hypothalamic CRF and AVP concentrations and plasma ACTH and corticosterone (CORT) levels were measured. All measurements were performed by enzyme-linked immunosorbent assays (ELISA), except that of the plasma CORT level, which was determined by chemofluorescent assay. Ucn I increased significantly the hypothalamic CRF and AVP concentrations in both rats and mice. Ucn II and Ucn III influenced significantly only the hypothalamic CRF concentration in rats, without affecting the hypothalamic AVP concentration. In contrast, Ucn II and Ucn III increased significantly only the hypothalamic AVP concentration in mice, without affecting the hypothalamic CRF concentration. The hypothalamic changes were reflected more or less accurately by changes of the plasma ACTH and CORT levels. The present experiments demonstrate that the urocortins regulate the HPA axis centrally via modulation of the hypothalamic ACTH secretagogues and that there are some similarities and discordancies between rats and mice regarding this regulation.
Collapse
Affiliation(s)
- Zsolt Bagosi
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, Hungary.
| | - Krisztina Csabafi
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, Hungary
| | - Gergely Karasz
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, Hungary
| | - Miklós Jászberényi
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, Hungary
| | - Imre Földesi
- Institute of Laboratory Medicine, Faculty of Medicine, University of Szeged, Hungary
| | - Andrea Siska
- Institute of Laboratory Medicine, Faculty of Medicine, University of Szeged, Hungary
| | - Gyula Szabó
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, Hungary
| | - Gyula Telegdy
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, Hungary
| |
Collapse
|
43
|
Hagiwara SI, Hasdemir B, Heyman MB, Chang L, Bhargava A. Plasma Corticotropin-Releasing Factor Receptors and B7-2⁺ Extracellular Vesicles in Blood Correlate with Irritable Bowel Syndrome Disease Severity. Cells 2019; 8:cells8020101. [PMID: 30704133 PMCID: PMC6406316 DOI: 10.3390/cells8020101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/10/2019] [Accepted: 01/27/2019] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are composed of bilayer membranes that are released by different cell types and are present in bodily fluids, such as blood, urine, and bile. EVs are thought to play a key role in intracellular communication. Based on their size and density, EVs are classified into small, medium, or large EVs. Cargo composition in EVs reflects physiological changes in health and disease. Patients with irritable bowel syndrome (IBS) exhibit visceral hypersensitivity and mood disorders. Stressful episodes often precede disease symptoms in IBS patients. Stress-induced symptoms include, but are not limited to, abdominal pain and mood swings. Perceived stress responses are mediated by two known G protein-coupled receptors (GPCRs), corticotropin-releasing factor receptor 1 and 2 (CRFRs). CRFRs belong to the Class B secretin receptor family of GPCRs. Here, we show that CRFRs were present in human and murine plasma, and in EVs purified from mouse serum. CRFRs were present in plasma from IBS patients and healthy controls. EVs secreted from immune cells influence both adaptive and innate immune responses via exchange of EVs between different immune cell types. B7-2 (CD86), a plasma membrane antigen-presenting protein, is present on EVs secreted from dendritic, B-, and mast cells, whereas CD9 is present on EVs secreted from dendritic and intestinal epithelial cells. We found that plasma CRFR levels positively correlated with B7-2+ EVs (R = 0.8597, p < 0.0001), but no association was seen with CD9+ EVs. Plasma CRFRs expression negatively correlated with IBS severity scores. Our data suggests that plasma EVs from immune cells carry CRFRs as cargos and influence cell-cell communication in health and disease.
Collapse
Affiliation(s)
- Shin-Ichiro Hagiwara
- The Osher Center for Integrative Medicine, University of California, San Francisco, CA 94143, USA.
| | - Burcu Hasdemir
- The Osher Center for Integrative Medicine, University of California, San Francisco, CA 94143, USA.
- Department of OBGYN, University of California, San Francisco, CA 94143-0556, USA.
| | - Melvin B Heyman
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA.
| | - Lin Chang
- G. Oppenheimer Center for Neurobiology of Stress and Resilience Vatche and Tamar Manoukian Division of Digestive Diseases David Geffen School of Medicine, University of California, Los Angeles, CA 90095-7378, USA.
| | - Aditi Bhargava
- The Osher Center for Integrative Medicine, University of California, San Francisco, CA 94143, USA.
- Department of OBGYN, University of California, San Francisco, CA 94143-0556, USA.
| |
Collapse
|
44
|
Parra-Mercado GK, Fuentes-Gonzalez AM, Hernandez-Aranda J, Diaz-Coranguez M, Dautzenberg FM, Catt KJ, Hauger RL, Olivares-Reyes JA. CRF 1 Receptor Signaling via the ERK1/2-MAP and Akt Kinase Cascades: Roles of Src, EGF Receptor, and PI3-Kinase Mechanisms. Front Endocrinol (Lausanne) 2019; 10:869. [PMID: 31920979 PMCID: PMC6921279 DOI: 10.3389/fendo.2019.00869] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/27/2019] [Indexed: 12/28/2022] Open
Abstract
In the present study, we determined the cellular regulators of ERK1/2 and Akt signaling pathways in response to human CRF1 receptor (CRF1R) activation in transfected COS-7 cells. We found that Pertussis Toxin (PTX) treatment or sequestering Gβγ reduced CRF1R-mediated activation of ERK1/2, suggesting the involvement of a Gi-linked cascade. Neither Gs/PKA nor Gq/PKC were associated with ERK1/2 activation. Besides, CRF induced EGF receptor (EGFR) phosphorylation at Tyr1068, and selective inhibition of EGFR kinase activity by AG1478 strongly inhibited the CRF1R-mediated phosphorylation of ERK1/2, indicating the participation of EGFR transactivation. Furthermore, CRF-induced ERK1/2 phosphorylation was not altered by pretreatment with batimastat, GM6001, or an HB-EGF antibody indicating that metalloproteinase processing of HB-EGF ligands is not required for the CRF-mediated EGFR transactivation. We also observed that CRF induced Src and PYK2 phosphorylation in a Gβγ-dependent manner. Additionally, using the specific Src kinase inhibitor PP2 and the dominant-negative-SrcYF-KM, it was revealed that CRF-stimulated ERK1/2 phosphorylation depends on Src activation. PP2 also blocked the effect of CRF on Src and EGFR (Tyr845) phosphorylation, further demonstrating the centrality of Src. We identified the formation of a protein complex consisting of CRF1R, Src, and EGFR facilitates EGFR transactivation and CRF1R-mediated signaling. CRF stimulated Akt phosphorylation, which was dependent on Gi/βγ subunits, and Src activation, however, was only slightly dependent on EGFR transactivation. Moreover, PI3K inhibitors were able to inhibit not only the CRF-induced phosphorylation of Akt, as expected, but also ERK1/2 activation by CRF suggesting a PI3K dependency in the CRF1R ERK signaling. Finally, CRF-stimulated ERK1/2 activation was similar in the wild-type CRF1R and the phosphorylation-deficient CRF1R-Δ386 mutant, which has impaired agonist-dependent β-arrestin-2 recruitment; however, this situation may have resulted from the low β-arrestin expression in the COS-7 cells. When β-arrestin-2 was overexpressed in COS-7 cells, CRF-stimulated ERK1/2 phosphorylation was markedly upregulated. These findings indicate that on the base of a constitutive CRF1R/EGFR interaction, the Gi/βγ subunits upstream activation of Src, PYK2, PI3K, and transactivation of the EGFR are required for CRF1R signaling via the ERK1/2-MAP kinase pathway. In contrast, Akt activation via CRF1R is mediated by the Src/PI3K pathway with little contribution of EGFR transactivation.
Collapse
Affiliation(s)
- G. Karina Parra-Mercado
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | - Alma M. Fuentes-Gonzalez
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | - Judith Hernandez-Aranda
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | - Monica Diaz-Coranguez
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | | | - Kevin J. Catt
- Section on Hormonal Regulation, Program on Developmental Endocrinology and Genetics, National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Richard L. Hauger
- Center of Excellence for Stress and Mental Health, VA Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - J. Alberto Olivares-Reyes
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
- *Correspondence: J. Alberto Olivares-Reyes
| |
Collapse
|
45
|
Castañeda Cortés DC, Arias Padilla LF, Langlois VS, Somoza GM, Fernandino JI. The central nervous system acts as a transducer of stress-induced masculinization through corticotropin-releasing hormone B. Development 2019; 146:dev.172866. [DOI: 10.1242/dev.172866] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/21/2019] [Indexed: 01/02/2023]
Abstract
Exposure to environmental stressors, like high temperature (HT), during early development of fish induces sex reversal of genotypic females. Nevertheless, the involvement of the brain in this process is not well clarified. In the present work, we investigated the mRNA levels of corticotropin-releasing hormone b (crhb) and its receptors (crhr1 and crhr2), and found out that they were up-regulated at HT during the critical period of gonadal sex determination in medaka. In order to clarify their roles in sex reversal, biallelic mutants for crhr1 and crhr2 were produced by CRISPR/Cas9 technology. Remarkably, biallelic mutant of both loci (crhr1 and crhr2) did not undergo female-to-male sex reversal upon HT exposition. Inhibition of this process in double crhrs mutants could be successfully rescued through the administration of the downstream effector of the hypothalamic-pituitary interrenal axis, the cortisol. Taken together, these results revealed for the first time the participation of the CNS acting as a transducer of masculinization induced by thermal stress.
Collapse
Affiliation(s)
- D. C. Castañeda Cortés
- Laboratorio de Biología del Desarrollo - Instituto Tecnológico de Chascomús. INTECH (CONICET-UNSAM), Argentina
| | - L. F. Arias Padilla
- Laboratorio de Biología del Desarrollo - Instituto Tecnológico de Chascomús. INTECH (CONICET-UNSAM), Argentina
| | - V. S. Langlois
- Institut national de la recherche scientifique (INRS) - Centre Eau Terre Environnement, Quebec, Canada
| | - G. M. Somoza
- Laboratorio de Ictiofisiología y Acuicultura - INTECH (CONICET-UNSAM), Argentina
| | - J. I. Fernandino
- Laboratorio de Biología del Desarrollo - Instituto Tecnológico de Chascomús. INTECH (CONICET-UNSAM), Argentina
- Institut national de la recherche scientifique (INRS) - Centre Eau Terre Environnement, Quebec, Canada
| |
Collapse
|
46
|
Abstract
The identification of corticotropin-releasing hormone (CRH) has led to the discovery of a growing family of ligands and receptors. CRH receptor 1 (CRHR1) and CRHR2 are mammalian G-protein coupled receptors (GPCRs) with high affinity for CRH and the CRH family of peptides. CRHR1 is predominantly expressed in the brain and plays a vital role in the hypothalamic-pituitary-adrenal (HPA) axis stress responses by secreting adrenal corticotropic hormone (ACTH). CRHR2 is predominantly expressed in the heart, and a CRHR2-specific ligand, urocortin 2 (UCN2), shows positive cardiac chronotropic and inotropic effects through 3´,5´-cyclic adenosine monophosphate (cAMP) signaling in response to CRHR2-mediated Gαs activation in mice and humans. Central administration of the CRH family of peptides increases mean arterial pressure through CRHR1 activation, whereas peripheral administration of the peptides decreases mean arterial pressure through CRHR2 activation. These observations have led to further investigations of CRHR2 as an important and unique GPCR in the physiological and pathological functioning of the cardiovascular (CV) system. Moreover, recent clinical trials demonstrate CRHR2 as a potentially therapeutic target in the treatment of heart failure. We present recent reviews of the role of CRHRs in basic CV physiology and in the pathophysiology of CV diseases.
Collapse
Affiliation(s)
- Mikito Takefuji
- Department of Cardiology, Nagoya University School of Medicine
| | | |
Collapse
|
47
|
Dedic N, Chen A, Deussing JM. The CRF Family of Neuropeptides and their Receptors - Mediators of the Central Stress Response. Curr Mol Pharmacol 2018; 11:4-31. [PMID: 28260504 PMCID: PMC5930453 DOI: 10.2174/1874467210666170302104053] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 11/26/2015] [Accepted: 08/03/2016] [Indexed: 12/12/2022]
Abstract
Background: Dysregulated stress neurocircuits, caused by genetic and/or environmental changes, underlie the development of many neuropsychiatric disorders. Corticotropin-releasing factor (CRF) is the major physiological activator of the hypothalamic-pituitary-adrenal (HPA) axis and conse-quently a primary regulator of the mammalian stress response. Together with its three family members, urocortins (UCNs) 1, 2, and 3, CRF integrates the neuroendocrine, autonomic, metabolic and behavioral responses to stress by activating its cognate receptors CRFR1 and CRFR2. Objective: Here we review the past and current state of the CRF/CRFR field, ranging from pharmacologi-cal studies to genetic mouse models and virus-mediated manipulations. Results: Although it is well established that CRF/CRFR1 signaling mediates aversive responses, includ-ing anxiety and depression-like behaviors, a number of recent studies have challenged this viewpoint by revealing anxiolytic and appetitive properties of specific CRF/CRFR1 circuits. In contrast, the UCN/CRFR2 system is less well understood and may possibly also exert divergent functions on physiol-ogy and behavior depending on the brain region, underlying circuit, and/or experienced stress conditions. Conclusion: A plethora of available genetic tools, including conventional and conditional mouse mutants targeting CRF system components, has greatly advanced our understanding about the endogenous mecha-nisms underlying HPA system regulation and CRF/UCN-related neuronal circuits involved in stress-related behaviors. Yet, the detailed pathways and molecular mechanisms by which the CRF/UCN-system translates negative or positive stimuli into the final, integrated biological response are not completely un-derstood. The utilization of future complementary methodologies, such as cell-type specific Cre-driver lines, viral and optogenetic tools will help to further dissect the function of genetically defined CRF/UCN neurocircuits in the context of adaptive and maladaptive stress responses.
Collapse
Affiliation(s)
- Nina Dedic
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| |
Collapse
|
48
|
Natural and synthetic peptides in the cardiovascular diseases: An update on diagnostic and therapeutic potentials. Arch Biochem Biophys 2018; 662:15-32. [PMID: 30481494 DOI: 10.1016/j.abb.2018.11.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/31/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023]
Abstract
Several peptides play an important role in physiological and pathological conditions into the cardiovascular system. In addition to well-known vasoactive agents such as angiotensin II, endothelin, serotonin or natriuretic peptides, the vasoconstrictor Urotensin-II (Uro-II) and the vasodilators Urocortins (UCNs) and Adrenomedullin (AM) have been implicated in the control of vascular tone and blood pressure as well as in cardiovascular disease states including congestive heart failure, atherosclerosis, coronary artery disease, and pulmonary and systemic hypertension. Therefore these peptides, together with their receptors, become important therapeutic targets in cardiovascular diseases (CVDs). Circulating levels of these agents in the blood are markedly modified in patients with specific CVDs compared with those in healthy patients, becoming also potential biomarkers for these pathologies. This review will provide an overview of current knowledge about the physiological roles of Uro-II, UCN and AM in the cardiovascular system and their implications in cardiovascular diseases. It will further focus on the structural modifications carried out on original peptide sequences in the search of analogues with improved physiochemical properties as well as in the delivery methods. Finally, we have overviewed the possible application of these peptides and/or their precursors as biomarkers of CVDs.
Collapse
|
49
|
Paruthiyil S, Hagiwara SI, Kundassery K, Bhargava A. Sexually dimorphic metabolic responses mediated by CRF 2 receptor during nutritional stress in mice. Biol Sex Differ 2018; 9:49. [PMID: 30400826 PMCID: PMC6218963 DOI: 10.1186/s13293-018-0208-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/21/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Chronic stress is a major contributor in the development of metabolic syndrome and associated diseases, such as diabetes. High-fat diet (HFD) and sex are known modifiers of metabolic parameters. Peptide hormones corticotropin-releasing factor (CRF) and urocortins (UCN) mediate stress responses via activation and feedback to the hypothalamic-pituitary-adrenal (HPA) axis. UCN3 is a marker of pancreatic β-cell differentiation, and UCN2 is known to ameliorate glucose levels in mice rendered diabetic with HFD. CRF receptor 2 (CRF2) is the only known cognate receptor for UCN2/3. Here, we ascertained the role of CRF2 in glucose clearance, insulin sensitivity, and other parameters associated with metabolic syndrome in a mouse model of nutritional stress. METHODS Wild-type (WT) and Crhr2-/- (null) mice of both sexes were fed either normal chow diet or HFD. After 8 weeks, blood glucose levels in response to glucose and insulin challenge were determined. Change in body and fat mass, plasma insulin, and lipid profile were assessed. Histological evaluation of liver sections was performed. RESULTS Here, we show that genotype (Crhr2), sex, and diet were all independent variables in the regulation of blood glucose levels, body and fat mass gain/redistribution, and insulin resistance. Surprisingly, CRF2-deficient mice (Crhr2-/-) male mice showed similarly impaired glucose clearance on HFD and chow. HFD-fed female Crhr2-/- mice redistributed their fat depots that were distinct from wild-type females and male mice on either diet. Blood cholesterol and low-density lipoprotein (LDL) levels were elevated significantly in male Crhr2-/- mice; female Crhr2-/- mice were protected. Male, but not female Crhr2-/- mice developed peripheral insulin resistance. HFD, but not chow-fed wild-type male mice developed hepatic macrovesicular steatosis. In contrast, livers of Crhr2-/- male mice showed microvesicular steatosis on either diet, whereas livers of female mice on this 8-week HFD regimen did not develop steatosis. CONCLUSIONS CRF2 receptor dysregulation is a sexually dimorphic risk factor in development of pre-diabetic and metabolic symptoms.
Collapse
Affiliation(s)
- Sreenivasan Paruthiyil
- Department of Obstetrics and Gynecology, Center for reproductive Sciences, and the Osher Center for Integrative Medicine, University of California San Francisco, 513 Parnassus Ave., HSE1645, Box 0556, San Francisco, CA, 94143-0556, USA
| | - Shin-Ichiro Hagiwara
- Department of Obstetrics and Gynecology, Center for reproductive Sciences, and the Osher Center for Integrative Medicine, University of California San Francisco, 513 Parnassus Ave., HSE1645, Box 0556, San Francisco, CA, 94143-0556, USA
| | - Keshav Kundassery
- Department of Obstetrics and Gynecology, Center for reproductive Sciences, and the Osher Center for Integrative Medicine, University of California San Francisco, 513 Parnassus Ave., HSE1645, Box 0556, San Francisco, CA, 94143-0556, USA
| | - Aditi Bhargava
- Department of Obstetrics and Gynecology, Center for reproductive Sciences, and the Osher Center for Integrative Medicine, University of California San Francisco, 513 Parnassus Ave., HSE1645, Box 0556, San Francisco, CA, 94143-0556, USA.
| |
Collapse
|
50
|
Deussing JM, Chen A. The Corticotropin-Releasing Factor Family: Physiology of the Stress Response. Physiol Rev 2018; 98:2225-2286. [DOI: 10.1152/physrev.00042.2017] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The physiological stress response is responsible for the maintenance of homeostasis in the presence of real or perceived challenges. In this function, the brain activates adaptive responses that involve numerous neural circuits and effector molecules to adapt to the current and future demands. A maladaptive stress response has been linked to the etiology of a variety of disorders, such as anxiety and mood disorders, eating disorders, and the metabolic syndrome. The neuropeptide corticotropin-releasing factor (CRF) and its relatives, the urocortins 1–3, in concert with their receptors (CRFR1, CRFR2), have emerged as central components of the physiological stress response. This central peptidergic system impinges on a broad spectrum of physiological processes that are the basis for successful adaptation and concomitantly integrate autonomic, neuroendocrine, and behavioral stress responses. This review focuses on the physiology of CRF-related peptides and their cognate receptors with the aim of providing a comprehensive up-to-date overview of the field. We describe the major molecular features covering aspects of gene expression and regulation, structural properties, and molecular interactions, as well as mechanisms of signal transduction and their surveillance. In addition, we discuss the large body of published experimental studies focusing on state-of-the-art genetic approaches with high temporal and spatial precision, which collectively aimed to dissect the contribution of CRF-related ligands and receptors to different levels of the stress response. We discuss the controversies in the field and unravel knowledge gaps that might pave the way for future research directions and open up novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jan M. Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|