1
|
Gilani M, Abak N, Saberian M. Genetic-epigenetic-neuropeptide associations in mood and anxiety disorders: Toward personalized medicine. Pharmacol Biochem Behav 2024; 245:173897. [PMID: 39424200 DOI: 10.1016/j.pbb.2024.173897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/29/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Mood and anxiety disorders are complex psychiatric conditions shaped by the multifactorial interplay of genetic, epigenetic, and neuropeptide factors. This review aims to elucidate the intricate interactions among these factors and their potential in advancing personalized medicine. We examine the genetic underpinnings, emphasizing key heritability studies and specific gene associations. The role of epigenetics is discussed, focusing on how environmental factors can modify gene expression and contribute to these disorders. Neuropeptides, including substance P, CRF, AVP, NPY, galanin, and kisspeptin, are evaluated for their involvement in mood regulation and their potential as therapeutic targets. Additionally, we address the emerging role of the gut microbiome in modulating neuropeptide activity and its connection to mood disorders. This review integrates findings from genetic, epigenetic, and neuropeptide research, offering a comprehensive overview of their collective impact on mood and anxiety disorders. By highlighting novel insights and potential clinical applications, we underscore the importance of a multi-omics approach in developing personalized treatment strategies. Future research directions are proposed to address existing knowledge gaps and translate these findings into clinical practice. Our review provides a fresh perspective on the pathophysiology of mood and anxiety disorders, paving the way for more effective and individualized therapies.
Collapse
Affiliation(s)
- Maryam Gilani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Abak
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Saberian
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Martz J, Shelton MA, Langen TJ, Srinivasan S, Seney ML, Kentner AC. Peripubertal antagonism of corticotropin-releasing factor receptor 1 results in sustained, sex-specific changes in behavioral plasticity and the transcriptomic profile of the amygdala. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.607957. [PMID: 39185241 PMCID: PMC11343213 DOI: 10.1101/2024.08.14.607957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Peripuberty is a significant period of neurodevelopment with long-lasting effects on the brain and behavior. Blocking type 1 corticotropin-releasing factor receptors (CRFR1) in neonatal and peripubertal rats attenuates detrimental effects of early-life stress on neural plasticity, behavior, and stress hormone action, long after exposure to the drug has ended. CRFR1 antagonism can also impact neural and behavioral development in the absence of stressful stimuli, suggesting sustained alterations under baseline conditions. To investigate this further, we administered a CRFR1 antagonist (CRFR1a), R121919, to young adolescent male and female rats across 4 days. Following each treatment, rats were tested for locomotion, social behavior, mechanical allodynia, or PPI of the acoustic startle reflex. Acute CRFR1 blockade immediately reduced PPI in peripubertal males, but not females. In adulthood, each assay was repeated without CRFR1a exposure to test for long-term effects of the adolescent treatment, with males continuing to experience deficits in PPI, while females displayed altered locomotion, PPI, and social behavior. The amygdala was collected to measure long-term effects on gene expression in pathways related to neural plasticity and neurodevelopmental disorders. Relative expression of cannabinoid type 1 receptors (CB1R), which mediate sensorimotor and HPA function, was also measured. In the adult amygdala, peripubertal CRFR1a induced alterations in pathways related to neural plasticity and stress in males and lower expression of CB1R protein in females. Understanding how acute exposure to neuropharmacological agents can have sustained impacts on brain and behavior, in the absence of further exposures, has important clinical implications for adolescent psychiatric treatment protocols.
Collapse
Affiliation(s)
- Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| | - Micah A. Shelton
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive Pittsburgh, PA, 15219
| | - Tristen J. Langen
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| | - Sakhi Srinivasan
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| | - Marianne L. Seney
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive Pittsburgh, PA, 15219
| | - Amanda C. Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| |
Collapse
|
3
|
Islam MR, Markatos C, Pirmettis I, Papadopoulos M, Karageorgos V, Liapakis G, Fahmy H. Design, Synthesis, and Biological Evaluations of Novel Thiazolo[4,5-d]pyrimidine Corticotropin Releasing Factor (CRF) Receptor Antagonists as Potential Treatments for Stress Related Disorders and Congenital Adrenal Hyperplasia (CAH). Molecules 2024; 29:3647. [PMID: 39125051 PMCID: PMC11314199 DOI: 10.3390/molecules29153647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Corticotropin-releasing factor (CRF) is a key neuropeptide hormone that is secreted from the hypothalamus. It is the master hormone of the HPA axis, which orchestrates the physiological and behavioral responses to stress. Many disorders, including anxiety, depression, addiction relapse, and others, are related to over-activation of this system. Thus, new molecules that may interfere with CRF receptor binding may be of value to treat neuropsychiatric stress-related disorders. Also, CRF1R antagonists have recently emerged as potential treatment options for congenital adrenal hyperplasia. Previously, several series of CRF1 receptor antagonists were developed by our group. In continuation of our efforts in this direction, herein we report the synthesis and biological evaluation of a new series of CRF1R antagonists. Representative compounds were evaluated for their binding affinities compared to antalarmin. Four compounds (2, 5, 20, and 21) showed log IC50 values of -8.22, -7.95, -8.04, and -7.88, respectively, compared to -7.78 for antalarmin. This result indicates that these four compounds are superior to antalarmin by 2.5, 1.4, 1.7, and 1.25 times, respectively. It is worth mentioning that compound 2, in terms of IC50, is among the best CRF1R antagonists ever developed in the last 40 years. The in silico physicochemical properties of the lead compounds showed good drug-like properties. Thus, further research in this direction may lead to better and safer CRF receptor antagonists that may have clinical applications, particularly for stress-related disorders and the treatment of congenital adrenal hyperplasia.
Collapse
Affiliation(s)
- Md Rabiul Islam
- Department of Pharmaceutical Science, College of Pharmacy & Allied Health Professions, South Dakota State University, Brookings, SD 57007, USA;
| | - Christos Markatos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece; (C.M.); (V.K.); (G.L.)
| | - Ioannis Pirmettis
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, 15310 Athens, Greece; (I.P.); (M.P.)
| | - Minas Papadopoulos
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, 15310 Athens, Greece; (I.P.); (M.P.)
| | - Vlasios Karageorgos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece; (C.M.); (V.K.); (G.L.)
| | - George Liapakis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece; (C.M.); (V.K.); (G.L.)
| | - Hesham Fahmy
- Department of Pharmaceutical Science, College of Pharmacy & Allied Health Professions, South Dakota State University, Brookings, SD 57007, USA;
| |
Collapse
|
4
|
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
|
5
|
Patel VK, Vaishnaw A, Shirbhate E, Kore R, Singh V, Veerasamy R, Rajak H. Cortisol as a Target for Treating Mental Disorders: A Promising Avenue for Therapy. Mini Rev Med Chem 2024; 24:588-600. [PMID: 37861053 DOI: 10.2174/0113895575262104230928042150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/19/2023] [Accepted: 08/05/2023] [Indexed: 10/21/2023]
Abstract
Cortisol, commonly known as the "stress hormone," plays a critical role in the body's response to stress. Elevated cortisol levels have been associated with various mental disorders, including anxiety, depression, and post-traumatic stress disorder. Consequently, researchers have explored cortisol modulation as a promising avenue for treating these conditions. However, the availability of research on cortisol as a therapeutic option for mental disorders is limited, and existing studies employ diverse methodologies and outcome measures. This review article aimed to provide insights into different treatment approaches, both pharmacological and non-pharmacological, which can effectively modulate cortisol levels. Pharmacological interventions involve the use of substances, such as somatostatin analogs, dopamine agonists, corticotropin-releasing hormone antagonists, and cortisol synthesis inhibitors. Additionally, non-pharmacological techniques, including cognitivebehavioral therapy, herbs and supplements, transcranial magnetic stimulation, lifestyle changes, and surgery, have been investigated to reduce cortisol levels. The emerging evidence suggests that cortisol modulation could be a promising treatment option for mental disorders. However, more research is needed to fully understand the effectiveness and safety of these therapies.
Collapse
Affiliation(s)
- Vijay K Patel
- Pushpendra College of Pharmacy, Ambikapur, Surguja 497101, (C.G.), India
| | - Aayush Vaishnaw
- Dr. C.V. Raman Institute of Pharmacy, Dr. C.V. Raman University, Bilaspur, C.G. 495113, India
| | - Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Rakesh Kore
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| |
Collapse
|
6
|
Corticotropin-Releasing Hormone: Biology and Therapeutic Opportunities. BIOLOGY 2022; 11:biology11121785. [PMID: 36552294 PMCID: PMC9775501 DOI: 10.3390/biology11121785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/16/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
In 1981, Wylie Vale, Joachim Spiess, Catherine Rivier, and Jean Rivier reported on the characterization of a 41-amino-acid peptide from ovine hypothalamic extracts with high potency and intrinsic activity stimulating the secretion of adrenocorticotropic hormone and β-endorphin by cultured anterior pituitary cells. With its sequence known, this neuropeptide was determined to be a hormone and consequently named corticotropin-releasing hormone (CRH), although the term corticotropin-releasing factor (CRF) is still used and preferred in some circumstances. Several decades have passed since this seminal contribution that opened a new research era, expanding the understanding of the coding of stress-related processes. The characterization of CRH receptors, the availability of CRH agonists and antagonists, and advanced immunocytochemical staining techniques have provided evidence that CRH plays a role in the regulation of several biological systems. The purpose of this review is to summarize the present knowledge of this 41-amino-acid peptide.
Collapse
|
7
|
Larauche M, Erchegyi J, Miller C, Sim MS, Rivier J, Behan D, Taché Y. Peripheral CRF-R1/CRF-R2 antagonist, astressin C, induces a long-lasting blockade of acute stress-related visceral pain in male and female rats. Peptides 2022; 157:170881. [PMID: 36185037 PMCID: PMC10389693 DOI: 10.1016/j.peptides.2022.170881] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/06/2022] [Accepted: 09/16/2022] [Indexed: 11/15/2022]
Abstract
Peptide CRF antagonists injected peripherally alleviate stress-induced visceral hypersensitivity (SIVH) to colorectal distension (CRD) in rodents. Here we further evaluated the dose and time-dependent inhibitory activity of several long-acting peptide CRF receptor antagonists related to astressin on SIVH, focusing on astressin C (AstC), which previously showed high efficacy on stress-related alterations of HPA axis and gut secretomotor functions. Male and female Sprague-Dawley rats pretreated subcutaneously (SC) with AstC were injected intraperitoneally (IP) with CRF 15 min later. The visceromotor responses (VMR) to graded phasic CRD (10, 20, 40 and 60 mmHg) were monitored at basal, 15 min and up to 1-8 days after pretreatment. Two other astressin analogs, hexanoyl-astressin D (Hex-AstD) and [CαMeVal19,32]-AstC, were also tested. The response to IP CRF was sex-dependent with female rats requiring a higher dose to exhibit visceral hyperalgesia. Pretreatment with AstC (30-1000 µg/kg) resulted in a dose-related inhibition of IP CRF-induced SIVH and diarrhea in both sexes. The highest dose prevented SIVH and diarrhea up to 5-7 days after a single SC injection and was lost on day 7 (females) and day 8 (males) but reinstated after a second injection of AstC on day 8 or 9 respectively. [CαMeVal19,32]-AstC and Hex-AstD (1000 µg/kg in males) also prevented SIVH. These data show the potent long-lasting anti-hyperalgesic effect of AstC in an acute model of SIVH in both male and female rats. This highlights the potential of long-acting peripheral CRF antagonists to treat stress-sensitive irritable bowel syndrome.
Collapse
Affiliation(s)
- Muriel Larauche
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, West Los Angeles, CA, USA.
| | | | | | - Myung Shin Sim
- Department of Medicine, Statistic Core, UCLA, Los Angeles, CA, USA
| | - Jean Rivier
- Sentia Medical Sciences, Inc., San Diego, CA, USA
| | | | - Yvette Taché
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, West Los Angeles, CA, USA
| |
Collapse
|
8
|
Holley D, Fox AS. The central extended amygdala guides survival-relevant tradeoffs: Implications for understanding common psychiatric disorders. Neurosci Biobehav Rev 2022; 142:104879. [PMID: 36115597 PMCID: PMC11178236 DOI: 10.1016/j.neubiorev.2022.104879] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 10/31/2022]
Abstract
To thrive in challenging environments, individuals must pursue rewards while avoiding threats. Extensive studies in animals and humans have identified the central extended amygdala (EAc)-which includes the central nucleus of the amygdala (Ce) and bed nucleus of the stria terminalis (BST)-as a conserved substrate for defensive behavior. These studies suggest the EAc influences defensive responding and assembles fearful and anxious states. This has led to the proliferation of a view that the EAc is fundamentally a defensive substrate. Yet mechanistic work in animals has implicated the EAc in numerous appetitive and consummatory processes, yielding fresh insights into the microcircuitry of survival- and emotion-relevant response selection. Coupled with the EAc's centrality in a conserved network of brain regions that encode multisensory environmental and interoceptive information, these findings suggest a broader role for the EAc as an arbiter of survival- and emotion-relevant tradeoffs for action selection. Determining how the EAc optimizes these tradeoffs promises to improve our understanding of common psychiatric illnesses such as anxiety, depression, alcohol- and substance-use disorders, and anhedonia.
Collapse
Affiliation(s)
- Dan Holley
- Department of Psychology and the California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Andrew S Fox
- Department of Psychology and the California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
9
|
The Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) System of the Central Amygdala Mediates the Detrimental Effects of Chronic Social Defeat Stress in Rats. eNeuro 2022; 9:ENEURO.0260-22.2022. [PMID: 36566434 PMCID: PMC9506682 DOI: 10.1523/eneuro.0260-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/28/2022] [Accepted: 08/12/2022] [Indexed: 01/21/2023] Open
Abstract
Many psychiatric diseases stem from an inability to cope with stressful events, as chronic stressors can precipitate or exacerbate psychopathologies. The neurobiological mechanisms underlying the response to chronic stress and the resulting anxiety states remain poorly understood. Stress neuropeptides in the extended amygdala circuitry mediate the behavioral response to stress, and hyperactivity of these systems has been hypothesized to be responsible for the emergence of persistent negative outcomes and for the pathogenesis of anxiety-related and trauma-related disorders. Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor PAC1R are highly expressed within the central amygdala (CeA) and play a key role in stress regulation. Here, we used chronic social defeat stress (CSDS), a clinically relevant model of psychosocial stress that produces robust maladaptive behaviors in rodents. We found that 10 days of CSDS cause a significant increase in PACAP levels selectively in the CeA of rats, as well as an increase in PAC1R mRNA. Using a viral vector strategy, we found that PAC1R knock-down in the CeA attenuates the CSDS-induced body weight loss and prevents the CSDS-induced increase in anxiety-like behavior. Notably, CSDS animals display reduced basal corticosterone (CORT) levels and PAC1R knock-down in CeA further reduce them. Finally, the CeA PAC1R knock-down blocks the increase in corticotropin-releasing factor (CRF) immunoreactivity induced by CSDS in CeA. Our findings support the notion that the persistent activation of the PACAP-PAC1R system in the CeA mediates the behavioral outcomes of chronic psychosocial stress independently of the hypothalamic-pituitary-adrenal axis, perhaps via the recruitment of the CRF system.
Collapse
|
10
|
Scaffold Repurposing Reveals New Nanomolar Phosphodiesterase Type 5 (PDE5) Inhibitors Based on Pyridopyrazinone Scaffold: Investigation of In Vitro and In Silico Properties. Pharmaceutics 2022; 14:pharmaceutics14091954. [PMID: 36145702 PMCID: PMC9501832 DOI: 10.3390/pharmaceutics14091954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Inhibition of PDE5 results in elevation of cGMP leading to vascular relaxation and reduction in the systemic blood pressure. Therefore, PDE5 inhibitors are used as antihypertensive and antianginal agents in addition to their major use as male erectile dysfunction treatments. Previously, we developed a novel series of 34 pyridopyrazinone derivatives as anticancer agents (series A–H). Herein, a multi-step in silico approach was preliminary conducted to evaluate the predicted PDE5 inhibitory activity, followed by an in vitro biological evaluation over the enzymatic level and a detailed SAR study. The designed 2D-QSAR model which was carried out to predict the IC50 of the tested compounds revealed series B, D, E and G with nanomolar range of IC50 values (6.00–81.56 nM). A further docking simulation model was performed to investigate the binding modes within the active site of PDE5. Interestingly, most of the tested compounds showed almost the same binding modes of that of reported PDE5 inhibitors. To validate the in silico results, an in vitro enzymatic assay over PDE5 enzyme was performed for a number of the promising candidates with different substitutions. Both series E and G exhibited a potent inhibitory activity (IC50 = 18.13–41.41 nM). Compound 11b (series G, oxadiazole-based derivatives with terminal 4-NO2 substituted phenyl ring and rigid linker) was the most potent analogue with IC50 value of 18.13 nM. Structure–activity relationship (SAR) data attained for various substitutions were rationalized. Furthermore, a molecular dynamic simulation gave insights into the inhibitory activity of the most active compound (11b). Accordingly, this report presents a successful scaffold repurposing approach that reveals compound 11b as a highly potent nanomolar PDE5 inhibitor worthy of further investigation.
Collapse
|
11
|
Rodriguez L, Kirson D, Wolfe SA, Patel RR, Varodayan FP, Snyder AE, Gandhi PJ, Khom S, Vlkolinsky R, Bajo M, Roberto M. Alcohol Dependence Induces CRF Sensitivity in Female Central Amygdala GABA Synapses. Int J Mol Sci 2022; 23:7842. [PMID: 35887190 PMCID: PMC9318832 DOI: 10.3390/ijms23147842] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 02/05/2023] Open
Abstract
Alcohol use disorder (AUD) is a chronically relapsing disease characterized by loss of control in seeking and consuming alcohol (ethanol) driven by the recruitment of brain stress systems. However, AUD differs among the sexes: men are more likely to develop AUD, but women progress from casual to binge drinking and heavy alcohol use more quickly. The central amygdala (CeA) is a hub of stress and anxiety, with corticotropin-releasing factor (CRF)-CRF1 receptor and Gamma-Aminobutyric Acid (GABA)-ergic signaling dysregulation occurring in alcohol-dependent male rodents. However, we recently showed that GABAergic synapses in female rats are less sensitive to the acute effects of ethanol. Here, we used patch-clamp electrophysiology to examine the effects of alcohol dependence on the CRF modulation of rat CeA GABAergic transmission of both sexes. We found that GABAergic synapses of naïve female rats were unresponsive to CRF application compared to males, although alcohol dependence induced a similar CRF responsivity in both sexes. In situ hybridization revealed that females had fewer CeA neurons containing mRNA for the CRF1 receptor (Crhr1) than males, but in dependence, the percentage of Crhr1-expressing neurons in females increased, unlike in males. Overall, our data provide evidence for sexually dimorphic CeA CRF system effects on GABAergic synapses in dependence.
Collapse
Affiliation(s)
- Larry Rodriguez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
| | - Dean Kirson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
- Department of Pharmacology, Addiction Science, and Toxicology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sarah A. Wolfe
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
| | - Reesha R. Patel
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
| | - Florence P. Varodayan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY 13902, USA
| | - Angela E. Snyder
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
| | - Pauravi J. Gandhi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
| | - Sophia Khom
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
- Department of Pharmaceutical Sciences, University of Vienna Josef-Holaubek-Platz 2, A-1090 Vienna, Austria
| | - Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (L.R.); (S.A.W.); (R.R.P.); (F.P.V.); (A.E.S.); (P.J.G.); (S.K.); (R.V.); (M.B.)
| |
Collapse
|
12
|
Intrinsically disordered proteins and proteins with intrinsically disordered regions in neurodegenerative diseases. Biophys Rev 2022; 14:679-707. [DOI: 10.1007/s12551-022-00968-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/28/2022] [Indexed: 12/14/2022] Open
|
13
|
Patel H. The role of the lateral septum in neuropsychiatric disease. J Neurosci Res 2022; 100:1422-1437. [PMID: 35443088 DOI: 10.1002/jnr.25052] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 02/22/2022] [Accepted: 03/23/2022] [Indexed: 12/25/2022]
Abstract
The lateral septum (LS) is a structure in the midline of the brain that is interconnected with areas associated with stress and feeding. This review highlights the role of the LS in anxiety, depression, and eating disorders and their comorbidity. There is a prevailing view that the LS is anxiolytic. This review finds that the LS is both anxiolytic and anxiogenic. Furthermore, the LS can promote and inhibit feeding. Given these shared roles, the LS represents a common site for the comorbidity of neuropsychiatric disorders, and therefore a potential pharmacological target. This is crucial since currently available treatments are not always effective. Corticotrophin-releasing factor 2 antagonists are potential drugs for the treatment of anxiety and anorexia and require further research. Furthermore, other drugs currently in trials for binge eating, such as alpha-adrenergic agonists, may in fact promote food intake. It is hoped that the advancements in chemo- and optogenetic techniques will allow future studies to profile the specific neural connections of the LS and their function. This information could facilitate our understanding of the underlying mechanisms, and therefore pharmacological targets, of these psychiatric conditions.
Collapse
|
14
|
Lichlyter DA, Krumm ZA, Golde TA, Doré S. Role of CRF and the hypothalamic-pituitary-adrenal axis in stroke: revisiting temporal considerations and targeting a new generation of therapeutics. FEBS J 2022; 290:1986-2010. [PMID: 35108458 DOI: 10.1111/febs.16380] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/10/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022]
Abstract
Ischaemic neurovascular stroke represents a leading cause of death in the developed world. Preclinical and human epidemiological evidence implicates the corticotropin-releasing factor (CRF) family of neuropeptides as mediators of acute neurovascular injury pathology. Preclinical investigations of the role of CRF, CRF receptors and CRF-dependent activation of the hypothalamic-pituitary-adrenal (HPA) axis have pointed toward a tissue-specific and temporal relationship between activation of these pathways and physiological outcomes. Based on the literature, the major phases of ischaemic stroke aetiology may be separated into an acute phase in which CRF and anti-inflammatory stress signalling are beneficial and a chronic phase in which these contribute to neural degeneration, toxicity and apoptotic signalling. Significant gaps in knowledge remain regarding the pathway, temporality and systemic impact of CRF signalling and stress biology in neurovascular injury progression. Heterogeneity among experimental designs poses a challenge to defining the apparent reciprocal relationship between neurological injury and stress metabolism. Despite these challenges, it is our opinion that the elucidated temporality may be best matched with an antibody against CRF with a half-life of days to weeks as opposed to minutes to hours as with small-molecule CRF receptor antagonists. This state-of-the-art review will take a multipronged approach to explore the expected potential benefit of a CRF antibody by modulating CRF and corticotropin-releasing factor receptor 1 signalling, glucocorticoids and autonomic nervous system activity. Additionally, this review compares the modulation of CRF and HPA axis activity in neuropsychiatric diseases and their counterpart outcomes post-stroke and assess lessons learned from antibody therapies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Daniel A Lichlyter
- Department of Anesthesiology, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Zachary A Krumm
- Department of Neuroscience, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Todd A Golde
- Department of Neuroscience, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,Departments of Neurology, Psychiatry, Pharmaceutics, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
15
|
Lv Y, Wen J, Fang Y, Zhang H, Zhang J. Corticotropin-releasing factor receptor 1 (CRF-R1) antagonists: Promising agents to prevent visceral hypersensitivity in irritable bowel syndrome. Peptides 2022; 147:170705. [PMID: 34822913 DOI: 10.1016/j.peptides.2021.170705] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 11/17/2022]
Abstract
Corticotropin-releasing factor (CRF) is a 41-amino acid polypeptide that coordinates the endocrine system, autonomic nervous system, immune system, and physiological behavior. CRF is a signaling regulator in the neuro-endocrine-immune (NEI) network that mediates visceral hypersensitivity. Rodent models to simulate changes in intestinal motility similar to those reported in the irritable bowel syndrome (IBS), demonstrate that the CRF receptor 1 (CRF-R1) mediates intestinal hypersensitivity under many conditions. However, the translation of preclinical studies into clinical trials has not been successful possibly due to the lack of sufficient understanding of the multiple variants of CRF-R1 and CRF-R1 antagonists. Investigating the sites of action of central and peripheral CRF is critical for accelerating the translation from preclinical to clinical studies.
Collapse
Affiliation(s)
- Yuanxia Lv
- School of Pharmacy, North Sichuan Medical College, Nanchong City, China.
| | - Jing Wen
- School of Pharmacy, North Sichuan Medical College, Nanchong City, China.
| | - Yingying Fang
- School of Pharmacy, North Sichuan Medical College, Nanchong City, China.
| | - Haoyuan Zhang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong City, China.
| | - Jianwu Zhang
- School of Pharmacy, North Sichuan Medical College, Nanchong City, China.
| |
Collapse
|
16
|
Design, synthesis, structural optimization, SAR, in silico prediction of physicochemical properties and pharmacological evaluation of novel & potent thiazolo[4,5-d]pyrimidine corticotropin releasing factor (CRF) receptor antagonists. Eur J Pharm Sci 2021; 169:106084. [PMID: 34856350 DOI: 10.1016/j.ejps.2021.106084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/12/2021] [Accepted: 11/26/2021] [Indexed: 11/21/2022]
Abstract
Corticotropin-releasing factor (CRF) is a 41-amino-acid neuropeptide secreted from the hypothalamus and is the main regulator of the hypothalamus-pituitary-adrenocortical (HPA) axis. CRF is the master hormone which modulates physiological and behavioral responses to stress. Many disorders including anxiety, depression, addictive disorders and others are related to over activation of the CRF system. This suggests that new molecules which can interfere with CRF binding to its receptors may be potential candidates for neuropsychiatric drugs to treat stress-related disorders. Previously, three series of pyrimidine and fused pyrimidine CRF1 receptor antagonists were synthesized by our group and specific binding assays, competitive binding studies and determination of the ability to antagonize the agonist-stimulated accumulation of cAMP (the second messenger for CRF receptors) were reported. In continuation of our efforts in this direction, in the current manuscript, we report the synthesis & biological evaluation of a new series of CRF1 receptor antagonists. Seven compounds showed promising binding affinity with the best two compounds (compounds 6 & 43) displaying a superior binding affinity to all of our previous compounds. Compounds 6 & 43 have only 4 times and 2 times less binding affinity than the standard CRF antagonist antalarmin, respectively. Thus, our two best lead compounds (compound 6 & 43) can be considered potent CRF receptor antagonists with binding affinity of 41.0 & 19.2 nM versus 9.7 nM for antalarmin.
Collapse
|
17
|
Ding Y, Wei Z, Yan H, Guo W. Efficacy of Treatments Targeting Hypothalamic-Pituitary-Adrenal Systems for Major Depressive Disorder: A Meta-Analysis. Front Pharmacol 2021; 12:732157. [PMID: 34566653 PMCID: PMC8461240 DOI: 10.3389/fphar.2021.732157] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
Abnormal hypothalamic-pituitary-adrenal (HPA) axis has been implicated in major depressive disorder (MDD). A number of studies have attempted to use HPA-modulating medications to treat depression. However, their results are inconsistent. The efficacy of these drugs for MDD remains uncertain. The aims of this meta-analysis were to determine the effect and safety profile of HPA-targeting medications for MDD. World of Science and PubMed databases were comprehensively searched up to March 2021. All randomized controlled trials (RCTs) and open-label trials exploring antiglucocorticoid and related medications in patients with depression were included. Standardized mean differences (SMDs) and risk ratios (RRs) with 95% confidence intervals (CIs) were calculated for continuous or dichotomous outcomes, respectively. In the meta-analysis, we identified 16 RCTs and seven open-label studies that included 2972 subjects. Pooling the change data that assessed the efficacy across all included HPA-targeting medications for depression showed a significant difference between interventions and controls with very small heterogeneity after influence analysis (SMD = 0.138, 95%CI = 0.052, 0.224, p = 0.002; I2 = 20.7%, p = 0.212). No obvious publication bias was observed (p = 0.127). Effectiveness remained significant in patients with MDD (SMD = 0.136, 95%CI = 0.049, 0.223, p = 0.002). Subgroup analysis showed a significant difference favoring mifepristone and vasopressin 1B (V1B) receptor antagonist treatment. Adverse events were reported by 14 studies and our analysis of high-quality studies showed a significant difference in favor of controls (RR = 1.283, 95%CI = 1.134, 1.452, p = 0). Our study suggested that patients with MDD may benefit from mifepristone and V1B receptor antagonist treatments that have tolerable side effects. HPA-based medications are promising for depression treatment. However, additional high-quality RCTs, including head-to-head trials, are needed. Systematic Review Registration:https://www.crd.york.ac.uk/PROSPERO/, identifier registration number: CRD42021247279
Collapse
Affiliation(s)
- Yudan Ding
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zirou Wei
- Mental Health Center, The Second Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Haohao Yan
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wenbin Guo
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
18
|
Elgiushy HR, Abou-Taleb NA, Holz GG, Chepurny OG, Pirmettis I, Kakabakos S, Karageorgos V, Liapakis G, Albohy A, Abouzid KAM, Hammad SF. Synthesis, in vitro biological investigation, and molecular dynamics simulations of thiazolopyrimidine based compounds as corticotrophin releasing factor receptor-1 antagonists. Bioorg Chem 2021; 114:105079. [PMID: 34174633 PMCID: PMC8387444 DOI: 10.1016/j.bioorg.2021.105079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 11/23/2022]
Abstract
Corticotrophin releasing factor receptor-1 (CRFR1) is a potential target for treatment of depression and anxiety through modifying stress response. A series of new thiazolo[4,5-d]pyrimidine derivatives were designed, prepared and biologically evaluated as potential CRFR1 antagonists. Four compounds produced more than fifty percent inhibition in the [125I]-Tyr0-sauvagine specific binding assay. Assessment of binding affinities revealed that compound (3-(2,4-dimethoxyphenyl)-7-(dipropylamino)-5-methylthiazolo[4,5-d]pyrimidin-2(3H)-one) 8c was the best candidate with highest binding affinity (Ki = 32.1 nM). Further evaluation showed the ability of compound 8c to inhibit CRF induced cAMP accumulation in a dose response manner. Docking and molecular dynamics simulations were used to investigate potential binding modes of synthesized compounds as well as the stability of 8c-CRFR1 complex. These studies suggest similar allosteric binding of 8c compared to that of the co-crystalized ligand CP-376395 in 4K5Y pdb file.
Collapse
Affiliation(s)
- Hossam R Elgiushy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Ain Helwan 11795, Cairo, Egypt; Basic and Applied Sciences Institute, Egypt-Japan University of Science and Technology (E-JUST), New Borg El-Arab City, 21934 Alexandria, Egypt
| | - Nageh A Abou-Taleb
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Ain Helwan 11795, Cairo, Egypt
| | - George G Holz
- Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Oleg G Chepurny
- Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Ioannis Pirmettis
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Sotirios Kakabakos
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Vlasios Karageorgos
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, 71003, Heraklion, Crete, Greece
| | - George Liapakis
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, 71003, Heraklion, Crete, Greece
| | - Amgad Albohy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Suez Desert Road, Cairo 11837, Egypt
| | - Khaled A M Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia 11566, Cairo, Egypt; Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt.
| | - Sherif F Hammad
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Ain Helwan 11795, Cairo, Egypt; Basic and Applied Sciences Institute, Egypt-Japan University of Science and Technology (E-JUST), New Borg El-Arab City, 21934 Alexandria, Egypt.
| |
Collapse
|
19
|
Matzeu A, Martin-Fardon R. Blockade of corticotropin-releasing factor receptor 1 in the central amygdala prevents cocaine-seeking behaviour induced by orexin-A administered to the posterior paraventricular nucleus of the thalamus in male rats. J Psychiatry Neurosci 2021; 46:E459-E471. [PMID: 34318655 PMCID: PMC8519495 DOI: 10.1503/jpn.200213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Orexin-A (OrxA) administration in the posterior paraventricular nucleus of the thalamus (pPVT) reinstates extinguished cocaine-seeking behaviour following extended access to the drug (a model of dependence). The pPVT receives and integrates information associated with emotionally salient events and sends excitatory inputs to brain regions involved in the expression of emotional states, such as those driving cocaine-seeking behaviour (i.e., the nucleus accumbens, the central nucleus of the amygdala [CeA], the basolateral amygdala, the bed nucleus of the stria terminalis [BNST] and the prefrontal cortex). METHODS We monitored the activation pattern of these regions (measured by Fos) during cocaine-seeking induced by OrxA administered to the pPVT. The BNST and CeA emerged as being selectively activated. To test whether the functionality of these regions was pivotal during OrxA-induced cocaine-seeking behaviour, we transiently inactivated these regions concomitantly with OrxA administration to the pPVT. We then tested the participation of corticotropin-releasing factor receptors (CRF1) in the CeA during OrxA-induced cocaine-seeking using the CRF1 antagonist CP154526. RESULTS We observed selective activation of the CeA and BNST during cocaine-seeking induced by OrxA administered to the pPVT, but only transient inactivation of the CeA prevented cocaine-seeking behaviour. Administration of CP154526 to the CeA prevented OrxAinduced cocaine-seeking behaviour. LIMITATIONS The use of only male rats could have been a limitation. Other limitations could have been the use of an indirect approach to test the hypothesis that administration of OrxA to the pPVT drives cocaine-seeking via CRF1 signalling in the CeA, and a lack of analysis of the participation of CeA subregions. CONCLUSION Cocaine-seeking behaviour induced by OrxA administered to the pPVT is driven by activation of the CeA via CRF1 signalling.
Collapse
Affiliation(s)
- Alessandra Matzeu
- From The Scripps Research Institute, La Jolla, California, USA (Matzeu, Martin-Fardon)
| | - Rémi Martin-Fardon
- From The Scripps Research Institute, La Jolla, California, USA (Matzeu, Martin-Fardon)
| |
Collapse
|
20
|
Kumar H, Das R, Choithramani A, Gupta A, Khude D, Bothra G, Shard A. Efficient Green Protocols for the Preparation of Pyrazolopyrimidines. ChemistrySelect 2021. [DOI: 10.1002/slct.202101298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Hansal Kumar
- Dept. of Medicinal Chemistry National Institute of Pharmaceutical Education and Research-Ahmedabad Opposite Air force Station, Palaj Gandhinagar Gujarat 382355 India
| | - Rudradip Das
- Dept. of Medicinal Chemistry National Institute of Pharmaceutical Education and Research-Ahmedabad Opposite Air force Station, Palaj Gandhinagar Gujarat 382355 India
| | - Asmita Choithramani
- Dept. of Medicinal Chemistry National Institute of Pharmaceutical Education and Research-Ahmedabad Opposite Air force Station, Palaj Gandhinagar Gujarat 382355 India
| | - Astha Gupta
- Dept. of Medicinal Chemistry National Institute of Pharmaceutical Education and Research-Ahmedabad Opposite Air force Station, Palaj Gandhinagar Gujarat 382355 India
| | - Datta Khude
- Dept. of Medicinal Chemistry National Institute of Pharmaceutical Education and Research-Ahmedabad Opposite Air force Station, Palaj Gandhinagar Gujarat 382355 India
| | - Gourav Bothra
- Dept. of Medicinal Chemistry National Institute of Pharmaceutical Education and Research-Ahmedabad Opposite Air force Station, Palaj Gandhinagar Gujarat 382355 India
| | - Amit Shard
- Dept. of Medicinal Chemistry National Institute of Pharmaceutical Education and Research-Ahmedabad Opposite Air force Station, Palaj Gandhinagar Gujarat 382355 India
| |
Collapse
|
21
|
Tasma Z, Wills P, Hay DL, Walker CS. Agonist bias and agonist-dependent antagonism at corticotrophin releasing factor receptors. Pharmacol Res Perspect 2021; 8:e00595. [PMID: 32529807 PMCID: PMC7290078 DOI: 10.1002/prp2.595] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 01/14/2023] Open
Abstract
The corticotropin-releasing factor (CRF) receptors represent potential drug targets for the treatment of anxiety, stress, and other disorders. However, it is not known if endogenous CRF receptor agonists display biased signaling, how effective CRF receptor antagonists are at blocking different agonists and signaling pathways or how receptor activity-modifying proteins (RAMPs) effect these processes. This study aimed to address this by investigating agonist and antagonist action at CRF1 and CRF2 receptors. We used CRF1 and CRF2 receptor transfected Cos7 cells to assess the ability of CRF and urocortin (UCN) peptides to activate cAMP, inositol monophosphate (IP1 ), and extracellular signal-regulated kinase 1/2 signaling and determined the ability of antagonists to block agonist-stimulated cAMP and IP1 accumulation. The ability of RAMPs to interact with CRF receptors was also examined. At the CRF1 receptor, CRF and UCN1 activated signaling in the same manner. However, at the CRF2 receptor, UCN1 and UCN2 displayed similar signaling profiles, whereas CRF and UCN3 displayed bias away from IP1 accumulation over cAMP. The antagonist potency was dependent on the receptor, agonist, and signaling pathway. CRF1 and CRF2 receptors had no effect on RAMP1 or RAMP2 surface expression. The presence of biased agonism and agonist-dependent antagonism at the CRF receptors offers new avenues for developing drugs tailored to activate a specific signaling pathway or block a specific agonist. Our findings suggest that the already complex CRF receptor pharmacology may be underappreciated and requires further investigation.
Collapse
Affiliation(s)
- Zoe Tasma
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Peter Wills
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Debbie L Hay
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Christopher S Walker
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Abstract
Alcohol dependence is a chronically relapsing disorder characterized by compulsive drug-seeking and drug-taking, loss of control in limiting intake, and the emergence of a withdrawal syndrome in the absence of the drug. Accumulating evidence suggests an important role for synaptic transmission in the central nucleus of the amygdala (CeA) in mediating alcohol-related behaviors and neuroadaptive mechanisms associated with alcohol dependence. Acute alcohol facilitates γ-aminobutyric acid (GABA)ergic transmission in the CeA via both pre- and postsynaptic mechanisms, and chronic alcohol increases baseline GABAergic transmission. Acute alcohol inhibits glutamatergic transmission via effects at N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the CeA, whereas chronic alcohol up-regulates NMDA receptor (NMDAR)-mediated transmission. Pro- (e.g., corticotropin-releasing factor [CRF]) and antistress (e.g., nociceptin/orphanin FQ, oxytocin) neuropeptides affect alcohol- and anxiety-related behaviors, and also alter the alcohol-induced effects on CeA neurotransmission. Alcohol dependence produces plasticity in these neuropeptide systems, reflecting a recruitment of those systems during the transition to alcohol dependence.
Collapse
Affiliation(s)
- Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Dean Kirson
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Sophia Khom
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
23
|
Wang Q, Dwivedi Y. Advances in novel molecular targets for antidepressants. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110041. [PMID: 32682872 PMCID: PMC7484229 DOI: 10.1016/j.pnpbp.2020.110041] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/24/2020] [Accepted: 07/12/2020] [Indexed: 12/18/2022]
Abstract
Depression is the most common psychiatric illness affecting numerous people world-wide. The currently available antidepressant treatment presents low response and remission rates. Thus, new effective antidepressants need to be developed or discovered. Aiming to give an overview of novel possible antidepressant drug targets, we summarized the molecular targets of antidepressants and the underlying neurobiology of depression. We have also addressed the multidimensional perspectives on the progress in the psychopharmacological treatment of depression and on the new potential approaches with effective drug discovery.
Collapse
Affiliation(s)
- Qingzhong Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
24
|
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
|
25
|
Discovery of a stable tripeptide targeting the N-domain of CRF1 receptor. Amino Acids 2020; 52:1337-1351. [PMID: 32996057 DOI: 10.1007/s00726-020-02895-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 09/19/2020] [Indexed: 10/23/2022]
Abstract
The corticotropin-releasing factor (CRF) and its CRF1 receptor (CRF1R) play a central role in the maintenance of homeostasis. Malfunctioning of the CRF/CRF1R unit is associated with several disorders, such as anxiety and depression. Non-peptide CRF1R-selective antagonists have been shown to exert anxiolytic and antidepressant effects on experimental animals. However, none of them is in clinical use today because of several side effects, thus demonstrating the need for the development of other more suitable CRF1R antagonists. In an effort to develop novel CRF1R antagonists we designed, synthesized and chemically characterized two tripeptide analogues of CRF, namely (R)-LMI and (S)-LMI, having their Leu either in R (or D) or in S (or L) configuration, respectively. Their design was based on the crystal structure of the N-extracellular domain (N-domain) of CRF1R/CRF complex, using a relevant array of computational methods. Experimental evaluation of the stability of synthetic peptides in human plasma has revealed that (R)-LMI is proteolytically more stable than (S)-LMI. Based on this finding, (R)-LMI was selected for pharmacological characterization. We have found that (R)-LMI is a CRF antagonist, inhibiting (1) the CRF-stimulated accumulation of cAMP in HEK 293 cells expressing the CRF1R, (2) the production of interleukins by adipocytes and (3) the proliferation rate of RAW 264.7 cells. (R)-LMI likely blocked agonist actions by interacting with the N-domain of CRF1R as suggested by data using a constitutively active chimera of CRF1R. We propose that (R)-LMI can be used as an optimal lead compound in the rational design of novel CRF antagonists.
Collapse
|
26
|
Dwyer JB, Aftab A, Radhakrishnan R, Widge A, Rodriguez CI, Carpenter LL, Nemeroff CB, McDonald WM, Kalin NH. Hormonal Treatments for Major Depressive Disorder: State of the Art. Am J Psychiatry 2020; 177:686-705. [PMID: 32456504 PMCID: PMC7841732 DOI: 10.1176/appi.ajp.2020.19080848] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Major depressive disorder is a common psychiatric disorder associated with marked suffering, morbidity, mortality, and cost. The World Health Organization projects that by 2030, major depression will be the leading cause of disease burden worldwide. While numerous treatments for major depression exist, many patients do not respond adequately to traditional antidepressants. Thus, more effective treatments for major depression are needed, and targeting certain hormonal systems is a conceptually based approach that has shown promise in the treatment of this disorder. A number of hormones and hormone-manipulating compounds have been evaluated as monotherapies or adjunctive treatments for major depression, with therapeutic actions attributable not only to the modulation of endocrine systems in the periphery but also to the CNS effects of hormones on non-endocrine brain circuitry. The authors describe the physiology of the hypothalamic-pituitary-adrenal (HPA), hypothalamic-pituitary thyroid (HPT), and hypothalamic-pituitary-gonadal (HPG) axes and review the evidence for selected hormone-based interventions for the treatment of depression in order to provide an update on the state of this field for clinicians and researchers. The review focuses on the HPA axis-based interventions of corticotropin-releasing factor antagonists and the glucocorticoid receptor antagonist mifepristone, the HPT axis-based treatments of thyroid hormones (T3 and T4), and the HPG axis-based treatments of estrogen replacement therapy, the progesterone derivative allopregnanolone, and testosterone. While some treatments have largely failed to translate from preclinical studies, others have shown promising initial results and represent active fields of study in the search for novel effective treatments for major depression.
Collapse
Affiliation(s)
| | | | | | - Alik Widge
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis
| | - Carolyn I. Rodriguez
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and VA Palo Alto Health Care System, Palo Alto, Calif
| | - Linda L. Carpenter
- Department of Psychiatry and Human Behavior, Butler Hospital, Brown University, Providence, R.I
| | | | - William M. McDonald
- Department of Psychiatry and Human Behavior, Emory University School of Medicine, Atlanta
| | - Ned H. Kalin
- Department of Psychiatry, University of Wisconsin–Madison
| | -
- Child Study Center and Department of Radiology and Biomedical Imaging, Yale University, New Haven, Conn. (Dwyer); Department of Psychiatry, Case Western Reserve University, Cleveland, and Northcoast Behavioral Healthcare Hospital, Northfield, Ohio (Aftab); Yale School of Medicine, New Haven, Conn. (Radhakrishnan); Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis (Widge); Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and VA Palo Alto Health Care System, Palo Alto, Calif. (Rodriguez); Department of Psychiatry and Human Behavior, Butler Hospital, Brown University, Providence, R.I. (Carpenter); Department of Psychiatry, University of Texas at Austin (Nemeroff); Department of Psychiatry and Human Behavior, Emory University School of Medicine, Atlanta (McDonald); and Department of Psychiatry, University of Wisconsin-Madison (Kalin)
| |
Collapse
|
27
|
Seiglie MP, Huang L, Cottone P, Sabino V. Role of the PACAP system of the extended amygdala in the acoustic startle response in rats. Neuropharmacology 2019; 160:107761. [PMID: 31493466 PMCID: PMC6842120 DOI: 10.1016/j.neuropharm.2019.107761] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/27/2019] [Accepted: 09/03/2019] [Indexed: 11/19/2022]
Abstract
Anxiety-related disorders are the most prevalent mental disorders in the world and they are characterized by abnormal responses to stressors. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide highly expressed in the extended amygdala, a brain macrostructure involved in the response to threat that includes the central nucleus of the amygdala (CeA) and the bed nucleus of the stria terminalis (BNST). The aim of this series of experiments was to systematically elucidate the role of the PACAP system of the CeA and BNST under both control, unstressed conditions and after the presentation of a stressor in rats. For this purpose, we used the acoustic startle response (ASR), an unconscious response to sudden acoustic stimuli sensitive to changes in stress which can be used as an operationalization of the hypervigilance present in anxiety- and trauma-related disorders. We found that infusion of PACAP, but not the related peptide vasoactive intestinal peptide (VIP), into either the CeA or the BNST causes a dose-dependent increase in ASR. In addition, while infusion of the antagonist PACAP(6-38) into either the CeA or the BNST does not affect ASR in non-stressed conditions, it prevents the sensitization of ASR induced by an acute footshock stress. Finally, we found that footshock stress induces a significant increase in PACAP, but not VIP, levels in both of these brain areas. Altogether, these data show that the PACAP system of the extended amygdala contributes to stress-induced hyperarousal and suggest it as a potential novel target for the treatment of stress-related disorders.
Collapse
Affiliation(s)
- Mariel P Seiglie
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Lillian Huang
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
28
|
Canet G, Hernandez C, Zussy C, Chevallier N, Desrumaux C, Givalois L. Is AD a Stress-Related Disorder? Focus on the HPA Axis and Its Promising Therapeutic Targets. Front Aging Neurosci 2019; 11:269. [PMID: 31611783 PMCID: PMC6776918 DOI: 10.3389/fnagi.2019.00269] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/18/2019] [Indexed: 01/04/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that has important health and economic impacts in the elderly. Despite a better understanding of the molecular mechanisms leading to the appearance of major pathological hallmarks (senile plaques and neurofibrillary tangles), effective treatments are still lacking. Sporadic AD forms (98% of all cases) are multifactorial, and a panoply of risk factors have been identified. While the major risk factor is aging, growing evidence suggests that chronic stress or stress-related disorders increase the probability to develop AD. An early dysregulation of the hypothalamic-pituitary-adrenal axis (HPA axis or stress axis) has been observed in patients. The direct consequence of such perturbation is an oversecretion of glucocorticoids (GC) associated with an impairment of its receptors (glucocorticoid receptors, GR). These steroids hormones easily penetrate the brain and act in synergy with excitatory amino acids. An overexposure could be highly toxic in limbic structures (prefrontal cortex and hippocampus) and contribute in the cognitive decline occurring in AD. GC and GR dysregulations seem to be involved in lots of functions disturbed in AD and a vicious cycle appears, where AD induces HPA axis dysregulation, which in turn potentiates the pathology. This review article presents some preclinical and clinical studies focusing on the HPA axis hormones and their receptors to fight AD. Due to its primordial role in the maintenance of homeostasis, the HPA axis appears as a key-actor in the etiology of AD and a prime target to tackle AD by offering multiple angles of action.
Collapse
Affiliation(s)
- Geoffrey Canet
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory (MMDN), INSERM, U1198, Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz) Team, EPHE, University of Montpellier, Paris, France
| | - Célia Hernandez
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory (MMDN), INSERM, U1198, Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz) Team, EPHE, University of Montpellier, Paris, France
| | - Charleine Zussy
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory (MMDN), INSERM, U1198, Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz) Team, EPHE, University of Montpellier, Paris, France
| | - Nathalie Chevallier
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory (MMDN), INSERM, U1198, Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz) Team, EPHE, University of Montpellier, Paris, France
| | - Catherine Desrumaux
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory (MMDN), INSERM, U1198, Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz) Team, EPHE, University of Montpellier, Paris, France
| | - Laurent Givalois
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory (MMDN), INSERM, U1198, Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz) Team, EPHE, University of Montpellier, Paris, France
| |
Collapse
|
29
|
Sartori SB, Singewald N. Novel pharmacological targets in drug development for the treatment of anxiety and anxiety-related disorders. Pharmacol Ther 2019; 204:107402. [PMID: 31470029 DOI: 10.1016/j.pharmthera.2019.107402] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022]
Abstract
Current medication for anxiety disorders is suboptimal in terms of efficiency and tolerability, highlighting the need for improved drug treatments. In this review an overview of drugs being studied in different phases of clinical trials for their potential in the treatment of fear-, anxiety- and trauma-related disorders is presented. One strategy followed in drug development is refining and improving compounds interacting with existing anxiolytic drug targets, such as serotonergic and prototypical GABAergic benzodiazepines. A more innovative approach involves the search for compounds with novel mechanisms of anxiolytic action using the growing knowledge base concerning the relevant neurocircuitries and neurobiological mechanisms underlying pathological fear and anxiety. The target systems evaluated in clinical trials include glutamate, endocannabinoid and neuropeptide systems, as well as ion channels and targets derived from phytochemicals. Examples of promising novel candidates currently in clinical development for generalised anxiety disorder, social anxiety disorder, panic disorder, obsessive compulsive disorder or post-traumatic stress disorder include ketamine, riluzole, xenon with one common pharmacological action of modulation of glutamatergic neurotransmission, as well as the neurosteroid aloradine. Finally, compounds such as D-cycloserine, MDMA, L-DOPA and cannabinoids have shown efficacy in enhancing fear-extinction learning in humans. They are thus investigated in clinical trials as an augmentative strategy for speeding up and enhancing the long-term effectiveness of exposure-based psychotherapy, which could render chronic anxiolytic drug treatment dispensable for many patients. These efforts are indicative of a rekindled interest and renewed optimism in the anxiety drug discovery field, after decades of relative stagnation.
Collapse
Affiliation(s)
- Simone B Sartori
- Institute of Pharmacy, Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck (CMBI), Leopold Franzens University Innsbruck, Innsbruck, Austria
| | - Nicolas Singewald
- Institute of Pharmacy, Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck (CMBI), Leopold Franzens University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
30
|
Sakellaris S, Matsoukas MT, Karageorgos V, Poulaki S, Kuppast B, Margioris A, Venihaki M, Fahmy H, Liapakis G. Selective antagonism of CRF1 receptor by a substituted pyrimidine. Hormones (Athens) 2019; 18:215-221. [PMID: 30980254 DOI: 10.1007/s42000-019-00105-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 03/20/2019] [Indexed: 01/13/2023]
Abstract
The corticotrophin-releasing factor (CRF) and its type 1 receptor (CRF1R) regulate the hypothalamic-pituitary-adrenal axis, as well as other systems, thus playing a crucial role in the maintenance of homeostasis. Non-peptide CRF1R-selective antagonists exert therapeutic effects on experimental animals with abnormal regulation of their homeostatic mechanisms. However, none of them is as yet in clinical use. In an effort to develop novel small non-peptide CRF1R-selective antagonists, we have synthesized a series of substituted pyrimidines described in a previous study. These small molecules bind to CRF1R, with analog 3 having the highest affinity. Characteristic structural features of analog 3 are a N,N-bis(methoxyethyl)amino group at position 6 and a methyl in the alkythiol group at position 5. Based on the binding profile of analog 3, we selected it in the present study for further pharmacological characterization. The results of this study suggest that analog 3 is a potent CRF1R-selective antagonist, blocking the ability of sauvagine, a CRF-related peptide, to stimulate cAMP accumulation in HEK 293 cells via activation of CRF1R, but not via CRF2R. Moreover, analog 3 blocked sauvagine to stimulate the proliferation of macrophages, further supporting its antagonistic properties. We have also constructed molecular models of CRF1R to examine the interactions of this receptor with analog 3 and antalarmin, a prototype CRF1R-selective non-peptide antagonist, which lacks the characteristic structural features of analog 3. Our data facilitate the design of novel non-peptide CRF1R antagonists for clinical use.
Collapse
Affiliation(s)
- Stelios Sakellaris
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | | | - Vlasios Karageorgos
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | - Smaragda Poulaki
- Department of Clinical Chemistry, School of Medicine, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | - Bhimanna Kuppast
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD, 57007, USA
| | - Andrew Margioris
- Department of Clinical Chemistry, School of Medicine, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | - Maria Venihaki
- Department of Clinical Chemistry, School of Medicine, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | - Hesham Fahmy
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD, 57007, USA
| | - George Liapakis
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, Heraklion, 71003, Crete, Greece.
| |
Collapse
|
31
|
Blackburn TP. Depressive disorders: Treatment failures and poor prognosis over the last 50 years. Pharmacol Res Perspect 2019; 7:e00472. [PMID: 31065377 PMCID: PMC6498411 DOI: 10.1002/prp2.472] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/11/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Depression like many diseases is pleiotropic but unlike cancer and Alzheimer's disease for example, is still largely stigmatized and falls into the dark shadows of human illness. The failure of depression to be in the spotlight for successful treatment options is inherent in the complexity of the disease(s), flawed clinical diagnosis, overgeneralization of the illness, inadequate and biased clinical trial design, restrictive and biased inclusion/exclusion criteria, lack of approved/robust biomarkers, expensive imaging technology along with few advances in neurobiological hypotheses in decades. Clinical trial studies summitted to the regulatory agencies (FDA/EMA) for approval, have continually failed to show significant differences between active and placebo. For decades, we have acknowledged this failure, despite vigorous debated by all stakeholders to provide adequate answers to this escalating problem, with only a few new antidepressants approved in the last 20 years with equivocal efficacy, little improvement in side effects or onset of efficacy. It is also clear that funding and initiatives for mental illness lags far behind other life-treating diseases. Thus, it is no surprise we have not achieved much success in the last 50 years in treating depression, but we are accountable for the many failures and suboptimal treatment. This review will therefore critically address where we have failed and how future advances in medical science offers a glimmer of light for the patient and aid our future understanding of the neurobiology and pathophysiology of the disease, enabling transformative therapies for the treatment of depressive disorders.
Collapse
|
32
|
Szlenk CT, Gc JB, Natesan S. Does the Lipid Bilayer Orchestrate Access and Binding of Ligands to Transmembrane Orthosteric/Allosteric Sites of G Protein-Coupled Receptors? Mol Pharmacol 2019; 96:527-541. [PMID: 30967440 DOI: 10.1124/mol.118.115113] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/03/2019] [Indexed: 01/08/2023] Open
Abstract
The ligand-binding sites of many G protein-coupled receptors (GPCRs) are situated around and deeply embedded within the central pocket formed by their seven transmembrane-spanning α-helical domains. Generally, these binding sites are assumed accessible to endogenous ligands from the aqueous phase. Recent advances in the structural biology of GPCRs, along with biophysical and computational studies, suggest that amphiphilic and lipophilic molecules may gain access to these receptors by first partitioning into the membrane and then reaching the binding site via lateral diffusion through the lipid bilayer. In addition, several crystal structures of class A and class B GPCRs bound to their ligands offer unprecedented details on the existence of lipid-facing allosteric binding sites outside the transmembrane helices that can only be reached via lipid pathways. The highly organized structure of the lipid bilayer may direct lipophilic or amphiphilic drugs to a specific depth within the bilayer, changing local concentration of the drug near the binding site and affecting its binding kinetics. Additionally, the constraints of the lipid bilayer, including its composition and biophysical properties, may play a critical role in "pre-organizing" ligand molecules in an optimal orientation and conformation to facilitate receptor binding. Despite its clear involvement in molecular recognition processes, the critical role of the membrane in binding ligands to lipid-exposed transmembrane binding sites remains poorly understood and warrants comprehensive investigation. Understanding the mechanistic basis of the structure-membrane interaction relationship of drugs will not only provide useful insights about receptor binding kinetics but will also enhance our ability to take advantage of the apparent membrane contributions when designing drugs that target transmembrane proteins with improved efficacy and safety. In this minireview, we summarize recent structural and computational studies on membrane contributions to binding processes, elucidating both lipid pathways of ligand access and binding mechanisms for several orthosteric and allosteric ligands of class A and class B GPCRs.
Collapse
Affiliation(s)
- Christopher T Szlenk
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Jeevan B Gc
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Senthil Natesan
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| |
Collapse
|
33
|
Miles OW, Maren S. Role of the Bed Nucleus of the Stria Terminalis in PTSD: Insights From Preclinical Models. Front Behav Neurosci 2019; 13:68. [PMID: 31024271 PMCID: PMC6461014 DOI: 10.3389/fnbeh.2019.00068] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/18/2019] [Indexed: 12/18/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) afflicts approximately 8% of the United States population and represents a significant public health burden, but the underlying neural mechanisms of this and other anxiety- and stressor-related disorders are largely unknown. Within the last few decades, several preclinical models of PSTD have been developed to help elucidate the mechanisms underlying dysregulated fear states. One brain area that has emerged as a critical mediator of stress-related behavioral processing in both clinical and laboratory settings is the bed nucleus of the stria terminalis (BNST). The BNST is interconnected with essential emotional processing regions, including prefrontal cortex, hippocampus and amygdala. It is activated by stressor exposure and undergoes neurochemical and morphological alterations as a result of stressor exposure. Stress-related neuro-peptides including corticotropin-releasing factor (CRF) and pituitary adenylate cyclase activating peptide (PACAP) are also abundant in the BNST, further implicating an involvement of BNST in stress responses. Behaviorally, the BNST is critical for acquisition and expression of fear and is well positioned to regulate fear relapse after periods of extinction. Here, we consider the role of the BNST in stress and memory processes in the context of preclinical models of PTSD.
Collapse
Affiliation(s)
- Olivia W. Miles
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | | |
Collapse
|
34
|
White MR, Graziano MJ, Sanderson TP. Toxicity of Pexacerfont, a Corticotropin-Releasing Factor Type 1 Receptor Antagonist, in Rats and Dogs. Int J Toxicol 2019; 38:110-120. [DOI: 10.1177/1091581819827501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pexacerfont is a corticotropin-releasing factor subtype 1 receptor antagonist that was developed for the treatment of anxiety- and stress-related disorders. This report describes the results of repeat-dose oral toxicity studies in rats (3 and 6 months) and dogs (3 months and 1 year). Pexacerfont was well tolerated in all of these studies at exposures equal to or greater than areas under the curve in humans (clinical dose of 100 mg). Microscopic changes in the liver (hepatocellular hypertrophy), thyroid glands (hypertrophy/hyperplasia and adenomas of follicular cells), and pituitary (hypertrophy/hyperplasia and vacuolation of thyrotrophs) were only observed in rats and were considered adaptive changes in response to hepatic enzyme induction and subsequent alterations in serum thyroid hormone levels. Evidence for hepatic enzyme induction in dogs was limited to increased liver weights and reduced thyroxine (T4) levels. Mammary gland hyperplasia and altered female estrous cycling were only observed in rats, whereas adverse testicular effects (consistent with minimal to moderate degeneration of the germinal epithelium) were only noted following chronic dosing in dogs. The testicular effects were reversible changes with exposure margins of 8× at the no observed adverse effect level. It is not clear whether the changes in mammary gland, estrous cycling, and testes represent secondary hormonal changes due to perturbation of the hypothalamic–pituitary–adrenal axis or are off-target effects. In conclusion, the results of chronic toxicity studies in rats and dogs show that pexacerfont has an acceptable safety profile to support further clinical testing.
Collapse
|
35
|
Tache Y, Larauche M, Yuan PQ, Million M. Brain and Gut CRF Signaling: Biological Actions and Role in the Gastrointestinal Tract. Curr Mol Pharmacol 2018; 11:51-71. [PMID: 28240194 DOI: 10.2174/1874467210666170224095741] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/16/2016] [Accepted: 08/03/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Corticotropin-releasing factor (CRF) pathways coordinate behavioral, endocrine, autonomic and visceral responses to stress. Convergent anatomical, molecular, pharmacological and functional experimental evidence supports a key role of brain CRF receptor (CRF-R) signaling in stress-related alterations of gastrointestinal functions. These include the inhibition of gastric acid secretion and gastric-small intestinal transit, stimulation of colonic enteric nervous system and secretorymotor function, increase intestinal permeability, and visceral hypersensitivity. Brain sites of CRF actions to alter gut motility encompass the paraventricular nucleus of the hypothalamus, locus coeruleus complex and the dorsal motor nucleus while those modulating visceral pain are localized in the hippocampus and central amygdala. Brain CRF actions are mediated through the autonomic nervous system (decreased gastric vagal and increased sacral parasympathetic and sympathetic activities). The activation of brain CRF-R2 subtype inhibits gastric motor function while CRF-R1 stimulates colonic secretomotor function and induces visceral hypersensitivity. CRF signaling is also located within the gut where CRF-R1 activates colonic myenteric neurons, mucosal cells secreting serotonin, mucus, prostaglandin E2, induces mast cell degranulation, enhances mucosal permeability and propulsive motor functions and induces visceral hyperalgesia in animals and humans. CRF-R1 antagonists prevent CRF- and stressrelated gut alterations in rodents while not influencing basal state. DISCUSSION These preclinical studies contrast with the limited clinical positive outcome of CRF-R1 antagonists to alleviate stress-sensitive functional bowel diseases such as irritable bowel syndrome. CONCLUSION The translational potential of CRF-R1 antagonists in gut diseases will require additional studies directed to novel anti-CRF therapies and the neurobiology of brain-gut interactions under chronic stress.
Collapse
Affiliation(s)
- Yvette Tache
- CURE/Digestive Diseases Research Center, G Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073. United States
| | - Muriel Larauche
- CURE/Digestive Diseases Research Center, G Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073. United States
| | - Pu-Qing Yuan
- CURE/Digestive Diseases Research Center, G Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073. United States
| | - Mulugeta Million
- CURE/Digestive Diseases Research Center, G Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073. United States
| |
Collapse
|
36
|
Nicholson JR, Sommer B. The research domain criteria framework in drug discovery for neuropsychiatric diseases: focus on negative valence. Brain Neurosci Adv 2018; 2:2398212818804030. [PMID: 32166151 PMCID: PMC7058263 DOI: 10.1177/2398212818804030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/24/2018] [Indexed: 12/22/2022] Open
Abstract
Drug discovery, particularly in the field of central nervous system, has had very limited success in the last few decades. A likely contributor is the poor translation between preclinical and clinical phases. The Research Domain Criteria of the National Institutes of Mental Health is a framework which aims to identify new ways of classifying mental illnesses that are based on observable behaviour and neurobiological measures, and to provide a guiding and evolving framework to improve the translation from preclinical to clinical research. At the core of the Research Domain Criteria approach is the assumption that the dimensional constructs described can be assessed across different units of analysis, thus enabling a more precise quantitative understanding of their neurobiological underpinnings, increasing the likelihood of identifying new and effective therapeutic approaches. In the present review, we discuss how the Research Domain Criteria can be applied to drug discovery with the domain Negative Valence, construct Potential Threat (‘Anxiety’) as an example. We will discuss the evidence supporting the utility of the Research Domain Criteria approach and evaluate how close we are to achieving a common thread of translational research from gene to self-report.
Collapse
Affiliation(s)
- Janet R Nicholson
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Bernd Sommer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
37
|
Montagud-Romero S, Blanco-Gandía MC, Reguilón MD, Ferrer-Pérez C, Ballestín R, Miñarro J, Rodríguez-Arias M. Social defeat stress: Mechanisms underlying the increase in rewarding effects of drugs of abuse. Eur J Neurosci 2018; 48:2948-2970. [PMID: 30144331 DOI: 10.1111/ejn.14127] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/11/2018] [Accepted: 08/14/2018] [Indexed: 12/27/2022]
Abstract
Social interaction is known to be the main source of stress in human beings, which explains the translational importance of this research in animals. Evidence reported over the last decade has revealed that, when exposed to social defeat experiences (brief episodes of social confrontations during adolescence and adulthood), the rodent brain undergoes remodeling and functional modifications, which in turn lead to an increase in the rewarding and reinstating effects of different drugs of abuse. The mechanisms by which social stress cause changes in the brain and behavior are unknown, and so the objective of this review is to contemplate how social defeat stress induces long-lasting consequences that modify the reward system. First of all, we will describe the most characteristic results of the short- and long-term consequences of social defeat stress on the rewarding effects of drugs of abuse such as psychostimulants and alcohol. Secondly, and throughout the review, we will carefully assess the neurobiological mechanisms underlying these effects, including changes in the dopaminergic system, corticotrophin releasing factor signaling, epigenetic modifications and the neuroinflammatory response. To conclude, we will consider the advantages and disadvantages and the translational value of the social defeat stress model, and will discuss challenges and future directions.
Collapse
Affiliation(s)
- Sandra Montagud-Romero
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | | | - Marina D Reguilón
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Carmen Ferrer-Pérez
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Raul Ballestín
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Jose Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| |
Collapse
|
38
|
Pituitary Adenylate Cyclase-Activating Peptide (PACAP) Signaling and the Dark Side of Addiction. J Mol Neurosci 2018; 68:453-464. [PMID: 30074172 DOI: 10.1007/s12031-018-1147-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/27/2018] [Indexed: 12/15/2022]
Abstract
While addiction to drugs of abuse represents a significant health problem worldwide, the behavioral and neural mechanisms that underlie addiction and relapse are largely unclear. The concept of the dark side of addiction, developed and explored by George Koob and colleagues, describes a systematic decrease in reward-related processing following drug self-administration and subsequent recruitment of anti-reward (i.e., stress) systems. Indeed, the activation of central nervous system (CNS) stress-response systems by drugs of abuse is contributory not only to mood and anxiety-related disorders but critical to both the maintenance of addiction and relapse following abstinence. In both human and animal studies, compounds that activate the bed nucleus of the stria terminalis (BNST) have roles in stress-related behaviors and addiction processes. The activation of pituitary adenylate cyclase-activating peptide (PACAP) systems in the BNST mediates many consequences of chronic stressor exposure that may engage in part downstream corticotropin-releasing hormone (CRH) signaling. Similar to footshock stress, the BNST administration of PACAP or the PAC1 receptor-specific agonist maxadilan can facilitate relapse following extinction of cocaine-seeking behavior. Further, in the same paradigm, the footshock-induced relapse could be attenuated following BNST pretreatment with PAC1 receptor antagonist PACAP6-38, implicating PACAP systems as critical components underlying stress-induced reinstatement. In congruence with previous work, the PAC1 receptor internalization and endosomal MEK/ERK signaling appear contributory mechanisms to the addiction processes. The studies offer new insights and approaches to addiction and relapse therapeutics.
Collapse
|
39
|
Abstract
Neuropeptides are the largest class of intercellular signaling molecules, contributing to a wide variety of physiological processes. Neuropeptide receptors are therapeutic targets for a broad range of drugs, including medications to treat pain, addiction, sleep disorders, and nausea. In addition to >100 peptides with known functions, many peptides have been identified in mammalian brain for which the cognate receptors have not been identified. Similarly, dozens of "orphan" G protein-coupled receptors have been identified in the mammalian genome. While it would seem straightforward to match the orphan peptides and receptors, this is not always easily accomplished. In this review we focus on peptides named PEN and big LEN, which are among the most abundant neuropeptides in mouse brain, and their recently identified receptors: GPR83 and GPR171. These receptors are co-expressed in some brain regions and are able to interact. Because PEN and big LEN are produced from the same precursor protein and co-secreted, the interaction of GPR83 and GPR171 is physiologically relevant. In addition to interactions of these two peptides/receptors, PEN and LEN are co-localized with neuropeptide Y and Agouti-related peptide in neurons that regulate feeding. In this review, using these peptide receptors as an example, we highlight the multiple modes of regulation of receptors and present the emerging view that neuropeptides function combinatorially to generate a network of signaling messages. The complexity of neuropeptides, receptors, and their signaling pathways is important to consider both in the initial deorphanization of peptides and receptors, and in the subsequent development of therapeutic applications.
Collapse
Affiliation(s)
- Lloyd D Fricker
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, United States
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
40
|
Dong H, Keegan JM, Hong E, Gallardo C, Montalvo-Ortiz J, Wang B, Rice KC, Csernansky J. Corticotrophin releasing factor receptor 1 antagonists prevent chronic stress-induced behavioral changes and synapse loss in aged rats. Psychoneuroendocrinology 2018; 90:92-101. [PMID: 29477954 PMCID: PMC5864558 DOI: 10.1016/j.psyneuen.2018.02.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 01/25/2018] [Accepted: 02/14/2018] [Indexed: 11/24/2022]
Abstract
Mounting evidence suggests that chronic stress can alter brain structure and function and promote the development of neuropsychiatric disorders, such as depression and Alzheimer's disease. Although the results of several studies have indicated that aged brains are more vulnerable to chronic stress, it remains unknown whether antagonists of a key stress regulator, the corticotrophin releasing factor receptor 1 (CRF1), can prevent stress-induced anxiety and memory deficits in animal models. In this study, we evaluated the potential benefits of two CRF1 antagonists, R121919 and antalarmin, for preventing stress-induced anxiety-related behavioral and memory deficits and neurodegeneration in aged rats. We stressed rats using isolation-restraint for 3 months starting from the 18 months of age. Subsets of animals were administrated either R121919 or antalarmin through food chow for 3 months, followed by a series of behavioral, biochemical and morphological analyses. We found that stressed aged rats displayed body weight losses and increased corticosterone levels, as well as anxiety-related behaviors and memory deficits. Additionally, chronic stress induced a loss of cortical dendritic spines and synapses. However, R121919 and antalarmin both prevented stress-induced behavioral changes including anxiety-related behaviors and memory deficits and prevented synapse loss, perhaps through reversing HPA axis dysfunction. These results suggest that CRF1 antagonists may hold promise as a potential therapy for preventing stress-induced anxiety and memory deficits in aged individuals.
Collapse
Affiliation(s)
- Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA.
| | - Jack M Keegan
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Ellie Hong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Christopher Gallardo
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Janitza Montalvo-Ortiz
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Becky Wang
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Kenner C. Rice
- National Institute on Drug Abuse, and National Institute Alcohol Abuse and Alcoholism Intramural Research Program, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - John Csernansky
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| |
Collapse
|
41
|
Langenecker SA, Crane NA, Jenkins LM, Phan KL, Klumpp H. Pathways to Neuroprediction: Opportunities and challenges to prediction of treatment response in depression. Curr Behav Neurosci Rep 2018; 5:48-60. [PMID: 29892518 PMCID: PMC5992916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
PURPOSE OF REVIEW We set out to review the current state of science in neuroprediction, using biological measures of brain function, with task based fMRI to prospectively predict response to a variety of treatments. RECENT FINDINGS Task-based fMRI neuroprediction studies are balanced between whole brain and ROI specific analyses. The predominant tasks are emotion processing, with ROIs based upon amygdala and subgenual anterior cingulate gyrus, both within the salience and emotion network. A rapidly emerging new area of neuroprediction is of disease course and illness recurrence. Concerns include use of open-label and single arm studies, lack of consideration of placebo effects, unbalanced adjustments for multiple comparisons (over focus on type I error), small sample sizes, unreported effect sizes, overreliance on ROI studies. SUMMARY There is a need to adjust neuroprediction study reporting so that greater coherence can facilitate meta analyses, and increased funding for more multiarm studies in neuroprediction.
Collapse
|
42
|
Karageorgos V, Venihaki M, Sakellaris S, Pardalos M, Kontakis G, Matsoukas MT, Gravanis A, Margioris A, Liapakis G. Current understanding of the structure and function of family B GPCRs to design novel drugs. Hormones (Athens) 2018; 17:45-59. [PMID: 29858864 DOI: 10.1007/s42000-018-0009-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 01/25/2018] [Indexed: 01/10/2023]
Abstract
Family B of G-protein-coupled receptors (GPCRs) and their ligands play a central role in a number of homeostatic mechanisms in the endocrine, gastrointestinal, skeletal, immune, cardiovascular and central nervous systems. Alterations in family B GPCR-regulated homeostatic mechanisms may cause a variety of potentially life-threatening conditions, signifying the necessity to develop novel ligands targeting these receptors. Obtaining structural and functional information on family B GPCRs will accelerate the development of novel drugs to target these receptors. Family B GPCRs are proteins that span the plasma membrane seven times, thus forming seven transmembrane domains (TM1-TM7) which are connected to each other by three extracellular (EL) and three intracellular (IL) loops. In addition, these receptors have a long extracellular N-domain and an intracellular C-tail. The upper parts of the TMs and ELs form the J-domain of receptors. The C-terminal region of peptides first binds to the N-domain of receptors. This 'first-step' interaction orients the N-terminal region of peptides towards the J-domain of receptors, thus resulting in a 'second-step' of ligand-receptor interaction that activates the receptor. Activation-associated structural changes of receptors are transmitted through TMs to their intracellular regions and are responsible for their interaction with the G proteins and activation of the latter, thus resulting in a biological effect. This review summarizes the current information regarding the structure and function of family B GPCRs and their physiological and pathophysiological roles.
Collapse
Affiliation(s)
- Vlasios Karageorgos
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, 71003, Heraklion, Crete, Greece
| | - Maria Venihaki
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Stelios Sakellaris
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, 71003, Heraklion, Crete, Greece
| | - Michail Pardalos
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, 71003, Heraklion, Crete, Greece
| | - George Kontakis
- Department of Orthopedics, University Hospital of Heraklion, Crete, Greece
| | | | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, 71003, Heraklion, Crete, Greece
| | - Andreas Margioris
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - George Liapakis
- Department of Pharmacology, School of Medicine, University of Crete, Voutes, 71003, Heraklion, Crete, Greece.
| |
Collapse
|
43
|
Ferrer-Pérez C, Reguilón MD, Manzanedo C, Aguilar MA, Miñarro J, Rodríguez-Arias M. Antagonism of corticotropin-releasing factor CRF 1 receptors blocks the enhanced response to cocaine after social stress. Eur J Pharmacol 2018; 823:87-95. [PMID: 29391155 DOI: 10.1016/j.ejphar.2018.01.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 01/26/2018] [Accepted: 01/26/2018] [Indexed: 01/07/2023]
Abstract
Numerous studies have shown that social defeat stress induces an increase in the rewarding effects of cocaine. In this study we have investigated the role played by the main hypothalamic stress hormone, corticotropin-releasing factor (CRF), in the effects that repeated social defeat (RSD) induces in the conditioned rewarding effects and locomotor sensitization induced by cocaine. A total of 220 OF1 mice were divided into experimental groups according to the treatment received before each social defeat: saline, 5 or 10 mg/kg of the nonpeptidic corticotropin-releasing factor CRF1 receptor antagonist CP-154,526, or 15 or 30 µg/kg of the peptidic corticotropin-releasing factor CRF2 receptor antagonist Astressin2-B. Three weeks after the last defeat, conditioned place preference (CPP) induced by 1 mg/kg of cocaine was evaluated. Motor response to 10 mg/kg of cocaine was also studied after a sensitization induction. Blockade of corticotropin-releasing factor CRF1 receptor reversed the increase in cocaine CPP induced by social defeat. Conversely, peripheral corticotropin-releasing factor CRF2 receptor blockade produced similar effects to those observed in socially stressed animals. The effect of RSD on cocaine sensitization was again blocked by the corticotropin-releasing factor CRF1 receptor antagonist, while peripheral CRF2 receptor antagonist did not show effect. Acute administration of Astressin2-B induced an anxiogenic response. Our results confirm that CRF modulates the effects of social stress on reinforcement and sensitization induced by cocaine in contrasting ways. These findings highlight CRF receptors as potential therapeutic targets to be explored by research about stress-related addiction problems.
Collapse
Affiliation(s)
- Carmen Ferrer-Pérez
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Marina D Reguilón
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Carmen Manzanedo
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - M Asunción Aguilar
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - José Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain.
| |
Collapse
|
44
|
Pathways to Neuroprediction: Opportunities and Challenges to Prediction of Treatment Response in Depression. Curr Behav Neurosci Rep 2018. [DOI: 10.1007/s40473-018-0140-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
45
|
Murrough JW, Charney DS. Corticotropin-Releasing Factor Type 1 Receptor Antagonists for Stress-Related Disorders: Time to Call It Quits? Biol Psychiatry 2017; 82:858-860. [PMID: 29129198 DOI: 10.1016/j.biopsych.2017.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 01/08/2023]
Affiliation(s)
- James W Murrough
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Dennis S Charney
- Office of the Dean, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
46
|
Kaye JT, Bradford DE, Magruder KP, Curtin JJ. Probing for Neuroadaptations to Unpredictable Stressors in Addiction: Translational Methods and Emerging Evidence. J Stud Alcohol Drugs 2017; 78:353-371. [PMID: 28499100 DOI: 10.15288/jsad.2017.78.353] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Stressors clearly contribute to addiction etiology and relapse in humans, but our understanding of specific mechanisms remains limited. Rodent models of addiction offer the power, flexibility, and precision necessary to delineate the causal role and specific mechanisms through which stressors influence alcohol and other drug use. This review describes a program of research using startle potentiation to unpredictable stressors that is well positioned to translate between animal models and clinical research with humans on stress neuroadaptations in addiction. This research rests on a solid foundation provided by three separate pillars of evidence from (a) rodent behavioral neuroscience on stress neuroadaptations in addiction, (b) rodent affective neuroscience on startle potentiation, and (c) human addiction and affective science with startle potentiation. Rodent stress neuroadaptation models implicate adaptations in corticotropin-releasing factor and norepinephrine circuits within the central extended amygdala following chronic alcohol and other drug use that mediate anxious behaviors and stress-induced reinstatement among drug-dependent rodents. Basic affective neuroscience indicates that these same neural mechanisms are involved in startle potentiation to unpredictable stressors in particular (vs. predictable stressors). We believe that synthesis of these evidence bases should focus us on the role of unpredictable stressors in addiction etiology and relapse. Startle potentiation in unpredictable stressor tasks is proposed to provide an attractive and flexible test bed to encourage tight translation and reverse translation between animal models and human clinical research on stress neuroadaptations. Experimental therapeutics approaches focused on unpredictable stressors hold high promise to identify, repurpose, or refine pharmacological and psychosocial interventions for addiction.
Collapse
Affiliation(s)
- Jesse T Kaye
- University of Wisconsin-Madison, Madison, Wisconsin
| | | | | | | |
Collapse
|
47
|
Fahradpour M, Keov P, Tognola C, Perez-Santamarina E, McCormick PJ, Ghassempour A, Gruber CW. Cyclotides Isolated from an Ipecac Root Extract Antagonize the Corticotropin Releasing Factor Type 1 Receptor. Front Pharmacol 2017; 8:616. [PMID: 29033832 PMCID: PMC5627009 DOI: 10.3389/fphar.2017.00616] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/23/2017] [Indexed: 12/22/2022] Open
Abstract
Cyclotides are plant derived, cystine-knot stabilized peptides characterized by their natural abundance, sequence variability and structural plasticity. They are abundantly expressed in Rubiaceae, Psychotrieae in particular. Previously the cyclotide kalata B7 was identified to modulate the human oxytocin and vasopressin G protein-coupled receptors (GPCRs), providing molecular validation of the plants' uterotonic properties and further establishing cyclotides as valuable source for GPCR ligand design. In this study we screened a cyclotide extract derived from the root powder of the South American medicinal plant ipecac (Carapichea ipecacuanha) for its GPCR modulating activity of the corticotropin-releasing factor type 1 receptor (CRF1R). We identified and characterized seven novel cyclotides. One cyclotide, caripe 8, isolated from the most active fraction, was further analyzed and found to antagonize the CRF1R. A nanomolar concentration of this cyclotide (260 nM) reduced CRF potency by ∼4.5-fold. In contrast, caripe 8 did not inhibit forskolin-, or vasopressin-stimulated cAMP responses at the vasopressin V2 receptor, suggesting a CRF1R-specific mode-of-action. These results in conjunction with our previous findings establish cyclotides as modulators of both classes A and B GPCRs. Given the diversity of cyclotides, our data point to other cyclotide-GPCR interactions as potentially important sources of drug-like molecules.
Collapse
Affiliation(s)
- Mohsen Fahradpour
- Center for Physiology and Pharmacology, Medical University of ViennaVienna, Austria.,Medicinal Plants and Drugs Research Institute, Shahid Beheshti UniversityTehran, Iran
| | - Peter Keov
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, BrisbaneQLD, Australia
| | - Carlotta Tognola
- Center for Physiology and Pharmacology, Medical University of ViennaVienna, Austria
| | | | - Peter J McCormick
- School of Veterinary Medicine, University of SurreyGuildford, United Kingdom
| | - Alireza Ghassempour
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti UniversityTehran, Iran
| | - Christian W Gruber
- Center for Physiology and Pharmacology, Medical University of ViennaVienna, Austria.,Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, BrisbaneQLD, Australia
| |
Collapse
|
48
|
Teleb M, Kuppast B, Spyridaki K, Liapakis G, Fahmy H. Synthesis of 2-imino and 2-hydrazono thiazolo[4,5- d ]pyrimidines as corticotropin releasing factor (CRF) antagonists. Eur J Med Chem 2017; 138:900-908. [DOI: 10.1016/j.ejmech.2017.07.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/27/2017] [Accepted: 07/11/2017] [Indexed: 12/18/2022]
|
49
|
Roberto M, Spierling SR, Kirson D, Zorrilla EP. Corticotropin-Releasing Factor (CRF) and Addictive Behaviors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:5-51. [PMID: 29056155 PMCID: PMC6155477 DOI: 10.1016/bs.irn.2017.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Drug addiction is a complex disorder that is characterized by compulsivity to seek and take the drug, loss of control in limiting intake of the drug, and emergence of a withdrawal syndrome in the absence of the drug. The transition from casual drug use to dependence is mediated by changes in reward and brain stress functions and has been linked to a shift from positive reinforcement to negative reinforcement. The recruitment of brain stress systems mediates the negative emotional state produced by dependence that drives drug seeking through negative reinforcement mechanisms, defined as the "dark side" of addiction. In this chapter we focus on behavioral and cellular neuropharmacological studies that have implicated brain stress systems (i.e., corticotropin-releasing factor [CRF]) in the transition to addiction and the predominant brain regions involved. We also discuss the implication of CRF recruitment in compulsive eating disorders.
Collapse
Affiliation(s)
- Marisa Roberto
- The Scripps Research Institute, La Jolla, CA, United States.
| | | | - Dean Kirson
- The Scripps Research Institute, La Jolla, CA, United States
| | | |
Collapse
|
50
|
Spierling SR, Zorrilla EP. Don't stress about CRF: assessing the translational failures of CRF 1antagonists. Psychopharmacology (Berl) 2017; 234:1467-1481. [PMID: 28265716 PMCID: PMC5420464 DOI: 10.1007/s00213-017-4556-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/27/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Dr. Athina Markou sought treatments for a common neural substrate shared by depression and drug dependence. Antagonists of corticotropin-releasing factor (CRF) receptors, a target of interest to her, have not reached the clinic despite strong preclinical rationale and sustained translational efforts. METHODS We explore potential causes for the failure of CRF1 antagonists and review recent findings concerning CRF-CRF1 systems in psychopathology. RESULTS Potential causes for negative outcomes include (1) poor safety and efficacy of initial drug candidates due to bad pharmacokinetic and physicochemical properties, (2) specificity problems with preclinical screens, (3) the acute nature of screens vs. late-presenting patients, (4) positive preclinical results limited to certain models and conditions with dynamic CRF-CRF1 activation not homologous to tested patients, (5) repeated CRF1 activation-induced plasticity that reduces the importance of ongoing CRF1 agonist stimulation, and (6) therapeutic silencing which may need to address CRF2 receptor or CRF-binding protein molecules, constitutive CRF1 activity, or molecules that influence agonist-independent activity or to target structural regions other than the allosteric site bound by all drug candidates. We describe potential markers of activation towards individualized treatment, human genetic, and functional data that still implicate CRF1 systems in emotional disturbance, sex differences, and suggestive clinical findings for CRF1 antagonists in food craving and CRF-driven HPA-axis overactivation. CONCLUSION The therapeutic scope of selective CRF1 antagonists now appears narrower than had been hoped. Yet, much remains to be learned about CRF's role in the neurobiology of dysphoria and addiction and the potential for novel anti-CRF therapies therein.
Collapse
Affiliation(s)
- Samantha R Spierling
- Committee on the Neurobiology of Addictive Disorders, SP30-2400, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Eric P Zorrilla
- Committee on the Neurobiology of Addictive Disorders, SP30-2400, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|