1
|
Huang Q, Zhang H, Ren L, Zeng J, Wen Y, Shu H, Gong G. Effect of oxytocin nasal spray pretreatment on postoperative acute stress disorder in patients with accidental traumatic fracture: a single-centre prospective randomised controlled clinical trial study protocol. BMJ Open 2025; 15:e090167. [PMID: 39809565 PMCID: PMC11751854 DOI: 10.1136/bmjopen-2024-090167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/01/2024] [Indexed: 01/16/2025] Open
Abstract
INTRODUCTION Patients undergoing surgical procedures are often prone to developing acute stress disorder (ASD) postoperatively. Presently, oxytocin nasal spray has shown significant potential in the treatment of stress-related neuropsychiatric diseases. However, there are few reports on the use of oxytocin nasal spray in postoperative ASD, a condition that can potentially develop into a high-risk factor for post-traumatic stress disorder. This study aims to investigate the effect of pretreatment with oxytocin nasal spray on postoperative ASD in patients with accidental trauma and fracture to provide new clinical insights for the prevention of postoperative ASD. METHODS AND ANALYSIS This study is a single-centre, double-blind, randomised controlled clinical trial. The trial aims to recruit 328 patients with accidental traumatic fractures who underwent surgical treatment. Participants will be randomly categorised into two groups: a control group (0.9% normal saline nasal spray, 1 mL) and an oxytocin group (oxytocin nasal spray, 1 mL/40 IU) at a ratio of 1:1 using the random number table method. The primary outcome is the incidence of ASD on postoperative days 1-3. Secondary outcomes include patient resilience, anxiety, depression and pain scores on postoperative days 1-3. The exploratory results include the concentrations of stress response indicators such as malondialdehyde, cortisol and superoxide dismutase in the saliva before the first intervention and on days 1-3 after surgery. ETHICS AND DISSEMINATION The trial was approved by the clinical research ethics committee of the General Hospital of the Western Theater Command (identifier: 2024EC3-ky014). The findings of this trial will be disseminated in a peer-reviewed journal and in national or international paediatric research to guide future practice. TRIAL REGISTRATION NUMBER ChiCTR2400082612.
Collapse
Affiliation(s)
- Qingqing Huang
- Department of Anesthesiology, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Hengdi Zhang
- Department of Ophthalmology, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Ling Ren
- Department of Anesthesiology, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Jingzheng Zeng
- Department of Anesthesiology, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yi Wen
- Department of General Surgery, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Haifeng Shu
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
- Department of Neurosurgery, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Gu Gong
- Department of Anesthesiology, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Rojanaratha T, Tienthai P, Woradulayapinij W, Yimsoo T, Boonkanokwong V, Ritthidej GC. Preparation, physicochemical characterization, ex vivo, and in vivo evaluations of asiatic acid-loaded solid lipid nanoparticles formulated with natural waxes for nose-to-brain delivery. Eur J Pharm Sci 2024; 203:106935. [PMID: 39414172 DOI: 10.1016/j.ejps.2024.106935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Asiatic acid (AA) has neuroprotective potential for prevention and treatment of Alzheimer's disease. Natural waxes with various ratios of Tween 80 and Span 80 or soybean lecithin were formulated to obtain AA-loaded solid lipid nanoparticles (AA-SLN) to improve direct nose to brain transport. Optimal AA-SLN had particle size below 200 nm with uniform size distribution and zeta potential of nearly -30 mV indicating a low risk of particle aggregation. Formulation with rice bran wax, Tween 80, and soybean lecithin (AA-RwS100) showed the highest entrapment efficiency and yield of >98 % while in vitro AA release of AA-SLN was linearly up to 48 h For ex vivo permeation, confocal laser scanning microscopy (CLSM) and histopathological studies on porcine olfactory mucosa (OM) and respiratory mucosa (RM), AA-SLN showed significantly higher permeation across OM than RM (p < 0.05) up to 6 h and AA-RwS100 also showed the highest amount of drug permeated as confirmed by CLSM results. Although AA-SLN showed non-significantly lower permeation than AA solution (AA-SOL) (p > 0.05), no epithelial and mucosal structure damages were observed in OM treated with AA-RwS100 and RM treated with all AA-SLNs indicating safety for nasal administration while AA-SOL showed significant damage to both OM and RM. In addition, in vivo brain distribution study by fluorescence imaging using Rhodamine (R6g) showed higher brain distribution after intranasal administration of R6g-loaded solid lipid nanoparticles (R6g-SLN) than R6g solution (R6g-SOL) and intravenous administration of R6g-SLN, and R6g-RwS100 also showed the highest brain accumulation at 8 h post administration.
Collapse
Affiliation(s)
- Tissana Rojanaratha
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Wang Mai, Pathum Wan, Bangkok 10330, Thailand
| | - Paisan Tienthai
- Department of Anatomy, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Warunya Woradulayapinij
- Thammasat University Research Unit in Mechanisms of Drug Action and Molecular Imaging, Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani 12120, Thailand
| | - Thunyatorn Yimsoo
- Thammasat University Research Unit in Mechanisms of Drug Action and Molecular Imaging, Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani 12120, Thailand; Laboratory Animal Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani 12120, Thailand
| | - Veerakiet Boonkanokwong
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Wang Mai, Pathum Wan, Bangkok 10330, Thailand.
| | - Garnpimol C Ritthidej
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Wang Mai, Pathum Wan, Bangkok 10330, Thailand; Queen Saovabha Memorial Institute, Thai Red Cross Society, Bangkok 10330, Thailand
| |
Collapse
|
3
|
Pantouli F, Pujol CN, Derieux C, Fonteneau M, Pellissier LP, Marsol C, Karpenko J, Bonnet D, Hibert M, Bailey A, Le Merrer J, Becker JAJ. Acute, chronic and conditioned effects of intranasal oxytocin in the mu-opioid receptor knockout mouse model of autism: Social context matters. Neuropsychopharmacology 2024; 49:1934-1946. [PMID: 39020142 PMCID: PMC11473707 DOI: 10.1038/s41386-024-01915-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/29/2024] [Accepted: 06/24/2024] [Indexed: 07/19/2024]
Abstract
Autism Spectrum Disorders (ASD) are neurodevelopmental disorders whose diagnosis relies on deficient social interaction and communication together with repetitive behaviours. Multiple studies have highlighted the potential of oxytocin (OT) to ameliorate behavioural abnormalities in animal models and subjects with ASD. Clinical trials, however, yielded disappointing results. Our study aimed at assessing the behavioural effects of different regimens of OT administration in the Oprm1 null mouse model of ASD. We assessed the effects of intranasal OT injected once at different doses (0.15, 0.3, and 0.6 IU) and time points (5, 15, and 30 min) following administration, or chronically, on ASD-related behaviours (social interaction and preference, stereotypies, anxiety, nociception) in Oprm1+/+ and Oprm1-/- mice. We then tested whether pairing intranasal OT injection with social experience would influence its outcome on ASD-like symptoms, and measured gene expression in the reward/social circuit. Acute intranasal OT at 0.3 IU improved social behaviour in Oprm1-/- mice 5 min after administration, with limited effects on non-social behaviours. Chronic (8-17 days) OT maintained rescuing effects in Oprm1 null mice but was deleterious in wild-type mice. Finally, improvements in the social behaviour of Oprm1-/- mice were greater and longer lasting when OT was administered in a social context. Under these conditions, the expression of OT and vasopressin receptor genes, as well as marker genes of striatal projection neurons, was suppressed. We detected no sex difference in OT effects. Our results highlight the importance of considering dosage and social context when evaluating the effects of OT treatment in ASD.
Collapse
Affiliation(s)
- Fani Pantouli
- INRAE, CNRS, Université de Tours, Inserm, PRC, 37380, Nouzilly, France
- Florida Research & Innovation Center, Cleveland Clinic, 9801 SW Discovery Way, Port St. Lucie, FL, 34987, USA
- Pharmacology section, Institute of Medical and Biomedical Education, St George's University of London, London, SW17 ORE, UK
| | - Camille N Pujol
- INRAE, CNRS, Université de Tours, Inserm, PRC, 37380, Nouzilly, France
- Department of Psychiatry, Strasbourg University Hospital, 67091, Strasbourg, France
| | - Cécile Derieux
- INRAE, CNRS, Université de Tours, Inserm, PRC, 37380, Nouzilly, France
| | - Mathieu Fonteneau
- UMR1253, iBrain, Université de Tours, Inserm, CNRS, Faculté des Sciences et Techniques, Parc de Grandmont, 37200, Tours, France
| | | | - Claire Marsol
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg, 74 route du Rhin, 67412, Illkirch, France
| | - Julie Karpenko
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg, 74 route du Rhin, 67412, Illkirch, France
| | - Dominique Bonnet
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg, 74 route du Rhin, 67412, Illkirch, France
| | - Marcel Hibert
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg, 74 route du Rhin, 67412, Illkirch, France
| | - Alexis Bailey
- Pharmacology section, Institute of Medical and Biomedical Education, St George's University of London, London, SW17 ORE, UK
| | - Julie Le Merrer
- INRAE, CNRS, Université de Tours, Inserm, PRC, 37380, Nouzilly, France.
- UMR1253, iBrain, Université de Tours, Inserm, CNRS, Faculté des Sciences et Techniques, Parc de Grandmont, 37200, Tours, France.
| | - Jerome A J Becker
- INRAE, CNRS, Université de Tours, Inserm, PRC, 37380, Nouzilly, France.
- UMR1253, iBrain, Université de Tours, Inserm, CNRS, Faculté des Sciences et Techniques, Parc de Grandmont, 37200, Tours, France.
| |
Collapse
|
4
|
Ding S, Liu Y, Tao H, Zhao Y, Zeng H, Han Y, Wang S, Chen Z, Tang Y, Guo W. Chronic intranasal oxytocin alleviates cognitive impairment and reverses oxytocin signaling upregulation in MK801-induced mice. Psychoneuroendocrinology 2024; 168:107138. [PMID: 39068687 DOI: 10.1016/j.psyneuen.2024.107138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/03/2024] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVE Cognitive impairment, especially impaired social cognition, is largely responsible for the deterioration of the social life of patients with schizophrenia (SZ). Oxytocin (OT) is a neuropeptide that offers promising therapy for SZ. This study aimed to explore whether OT could affect dizocilpine (MK801)-induced cognitive impairment and to investigate the effect of exogenous OT on the endogenous OT system in the hippocampus. METHODS The SZ mouse model was established by repeated administration of dizocilpine [MK801, 0.6 mg/kg, intraperitoneal (i.p.)], and then OT (6-60 μg/kg, intranasal) or risperidone (0.3 mg/kg, i.p.) was administered to explore the effect of OT on cognitive impairment. RESULTS OT at a dose of 6 μg/kg alleviated MK801-induced hyperactivity, sociability impairment, and spatial memory impairment. OT at a dose of 20 or 60 μg/kg attenuated the hyperactivity and social novelty impairment. In MK801-injected mice, the compensatory upregulation of OT mRNA in the hippocampus was reversed by three OT doses, whereas 60 μg/kg OT reversed the compensatory upregulation of CD38 protein expression. CONCLUSION OT alleviated cognitive impairment in the SZ mouse model to varying degrees, reversing the compensatory upregulation of OT signaling in the hippocampus.
Collapse
Affiliation(s)
- Shan Ding
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yong Liu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Huai Tao
- Department of Biochemistry and Molecular Biology, Hunan University of Chinese Medicine, Changsha, China
| | - Yuxu Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hongtao Zeng
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yiding Han
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shichen Wang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiheng Chen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yamei Tang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Wenbin Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
5
|
Shi CN, Wu XM, Gao YZ, Ma DQ, Yang JJ, Ji MH. Oxytocin attenuates neuroinflammation-induced anxiety through restoration of excitation and inhibition balance in the anterior cingulate cortex in mice. J Affect Disord 2024; 362:341-355. [PMID: 38821372 DOI: 10.1016/j.jad.2024.05.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/28/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Accumulative evidence suggested that the oxytocin system plays a role in socio-emotional disorders, although its role in neuroinflammation-induced anxiety remains unclear. METHOD In the present study, anxiety-like behavior was induced in cohorts of animals through repeated lipopolysaccharide (LPS, 0.5 mg/kg, daily, Escherichia coli O55:B5) i.p. injections for seven consecutive days. These different cohorts were subsequently used for anxiety-like behavior assessment with open field test, elevated plus maze, and novelty-suppressed feeding test or for electrophysiology (EEG) recordings of miniature excitatory postsynaptic currents (mEPSCs), miniature inhibitory postsynaptic currents (mIPSCs), or local field potential (LFP) in vivo or ex vivo settings. Samples of the anterior cingulate cortex (ACC) from some cohorts were harvested to conduct immunostaining or western blotting analysis of oxytocin, oxytocin receptor, CamkII, GABA, vGAT, vGLUT2, and c-fos. The dendritic spine density was assessed by Golgi-Cox staining. RESULTS Repeated LPS injections induced anxiety-like behavior with concurrent decreases of oxytocin, vGLUT2, mEPSC, dendritic spine, c-fos, membrane excitability, and EEG beta and gamma oscillations, but increased oxytocin receptor and vGAT expressions in the ACC; all these changes were ameliorated by oxytocin intranasal or local brain (via cannula) administration. CONCLUSION Taken together, our data suggested that oxytocin system may be a therapeutic target for developing treatment to tackle neuroinflammation-induced anxiety.
Collapse
Affiliation(s)
- Cui-Na Shi
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Miao Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Zhu Gao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Da-Qing Ma
- Department of Anesthesiology, Perioperative and Systems Medicine laboratory, National Clinical Research Center for Child Health, Children's hospital, Zhejiang University School of Medicine, Hangzhou, China; Division of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Mu-Huo Ji
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
6
|
Arda DB, Tunç KC, Bozkurt MF, Bora ES, Çiğel A, Erbaş O. Intranasal Insulin Eases Autism in Rats via GDF-15 and Anti-Inflammatory Pathways. Curr Issues Mol Biol 2024; 46:10530-10544. [PMID: 39329976 PMCID: PMC11431515 DOI: 10.3390/cimb46090624] [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: 08/24/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
In rat models, it is well-documented that chronic administration of propionic acid (PPA) leads to autism-like behaviors. Although the intranasal (IN) insulin approach is predominantly recognized for its effects on food restriction, it has also been shown to enhance cognitive memory by influencing various proteins, modulating anti-inflammatory pathways in the brain, and reducing signaling molecules such as interleukins. This study seeks to explore the potential therapeutic benefits of IN insulin in a rat model of autism induced by PPA. Thirty male Wistar albino rats were categorized into three cohorts: the control group, the PPA-induced autism (250 mg/kg/day intraperitoneal PPA dosage for five days) group, treated with saline via IN, and the PPA-induced autism group, treated with 25 U/kg/day (250 µL/kg/day) insulin via IN. All treatments were administered for 15 days. After behavioral testing, all animals were euthanized, and brain tissue and blood samples were collected for histopathological and biochemical assessments. Following insulin administration, a substantial reduction in autism symptoms was observed in all three social behavior tests conducted on the rats. Moreover, insulin exhibited noteworthy capabilities in decreasing brain MDA, IL-2, IL-17, and TNF-α levels within autism models. Additionally, there is a notable elevation in the brain nerve growth factor level (p < 0.05) and GDF-15 (p < 0.05). The assessment of cell counts within the hippocampal region and cerebellum revealed that insulin displayed effects in decreasing glial cells and inducing a significant augmentation in cell types such as the Purkinje and Pyramidal cells. The administration of insulin via IN exhibits alleviating effects on autism-like behavioral, biochemical, and histopathological alterations induced by PPA in rats. Insulin-dependent protective effects show anti-inflammatory, anti-oxidative, and neuroprotective roles of insulin admitted nasally.
Collapse
Affiliation(s)
- Duygu Burcu Arda
- Department of Pediatrics, Taksim Research and Training Hospital, Istanbul 34365, Türkiye;
| | - Kerem Can Tunç
- Department of Biology, Faculty of Science, Adnan Menderes University, Aydın 09010, Türkiye;
| | - Mehmet Fatih Bozkurt
- Department of Pathology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyon 03100, Türkiye;
| | - Ejder Saylav Bora
- Department of Emergency Medicine, Faculty of Medicine, Izmir Katip Çelebi University, Izmir 35150, Türkiye
| | - Ayşe Çiğel
- Department of Physiology, Faculty of Medicine, Izmir Democracy University, Izmir 35150, Türkiye;
| | - Oytun Erbaş
- Department of Physiology, Faculty of Medicine, Demiroğlu Bilim University, Istanbul 34381, Türkiye;
| |
Collapse
|
7
|
Tanaka A, Kiriyama A, Sano A, Changung C, Katsumi H, Yamamoto A, Furubayashi T. Left-Right Difference in Brain Pharmacokinetics Following Nasal Administration Via One-Site Nostrils. J Pharm Sci 2024; 113:2633-2640. [PMID: 38734208 DOI: 10.1016/j.xphs.2024.05.003] [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: 11/10/2023] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024]
Abstract
The olfactory and trigeminal pathways are direct delivery pathways between the nose and brain. To determine the effect of direct delivery on drug distribution in the brain, two model drugs with different physical properties, antipyrine (ANP), with high membrane permeability, and ranitidine (RNT), with low membrane permeability, were selected. For ANP, direct delivery from the nose to the brain was observed only in the olfactory bulb beside the nasal cavity, with a direct transport percentage (DTP) of approximately 45 %, whereas in the frontal and occipital brains, the contribution from the systemic circulation to the brain was observed as the primary route of brain distribution. No significant variations were observed in the pharmacokinetics of ANP in the left and right brain, whereas RNT was distributed in all brain regions with a DTP of > 95 %. The closer the brain region is to the nasal cavity, the higher the DTP. Furthermore, the left brain, the same nostril site (left nostril) of administration, had a larger level of drug delivery than the right brain. These findings imply that the influence of the administered nostril site differs based on the physicochemical properties and amount of the drug.
Collapse
Affiliation(s)
- Akiko Tanaka
- Laboratory of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-machi 4-19-1, Higashinada, Kobe 658-8558, Japan.
| | - Akiko Kiriyama
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kodo, Kyotanabe, Kyoto 610-0395, Japan
| | - Ayaka Sano
- Laboratory of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-machi 4-19-1, Higashinada, Kobe 658-8558, Japan
| | - Cho Changung
- Laboratory of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-machi 4-19-1, Higashinada, Kobe 658-8558, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina, Kyoto 607-8414, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina, Kyoto 607-8414, Japan
| | - Tomoyuki Furubayashi
- Laboratory of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-machi 4-19-1, Higashinada, Kobe 658-8558, Japan
| |
Collapse
|
8
|
Farokhi E, Alaofi AL, Prasasty VD, Stephanie F, Laksitorini MD, Kuczera K, Siahaan TJ. Mechanism of the blood-brain barrier modulation by cadherin peptides. EXPLORATION OF DRUG SCIENCE 2024; 2:322-338. [PMID: 39118806 PMCID: PMC11309765 DOI: 10.37349/eds.2024.00049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/21/2024] [Indexed: 08/10/2024]
Abstract
Aim This study was aimed at finding the binding site on the human E-cadherin for Ala-Asp-Thr Cyclic 5 (ADTC5), ADTC7, and ADTC9 peptides as blood-brain barrier modulator (BBBM) for determining their mechanism of action in modulating the blood-brain barrier (BBB). Methods ADTC7 and ADTC9 were derivatives of ADTC5 where the Val6 residue in ADTC5 was replaced by Glu6 and Tyr6 residues, respectively. The binding properties of ADTC5, ADTC7, and ADTC9 to the extracellular-1 (EC1) domain of E-cadherin were evaluated using chemical shift perturbation (CSP) method in the two dimensional (2D) 1H-15N-heteronuclear single quantum coherence (HSQC) nuclear magnetic resonance (NMR) spectroscopy. Molecular docking experiments were used to determine the binding sites of these peptides to the EC1 domain of E-cadherin. Results This study indicates that ADTC5 has the highest binding affinity to the EC1 domain of E-cadherin compared to ADTC7 and ADTC9, suggesting the importance of the Val6 residue as shown in our previous in vitro study. All three peptides have a similar binding site at the hydrophobic binding pocket where the domain swapping occurs. ADTC5 has a higher overlapping binding site with ADTC7 than that of ADTC9. Binding of ADTC5 on the EC1 domain influences the conformation of the EC1 C-terminal tail. Conclusions These peptides bind the domain swapping region of the EC1 domain to inhibit the trans-cadherin interaction that creates intercellular junction modulation to increase the BBB paracellular porosity.
Collapse
Affiliation(s)
- Elinaz Farokhi
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: Analytical Department, Johnson & Johnson, San Diego, CA 92123, USA
| | - Ahmed L. Alaofi
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Vivitri D. Prasasty
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Filia Stephanie
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
| | - Marlyn D. Laksitorini
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: School of Pharmacy, University of Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Krzysztof Kuczera
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66047, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
| |
Collapse
|
9
|
Liu S, He Y, Yin J, Zhu Q, Liao C, Jiang G. Neurotoxicities induced by micro/nanoplastics: A review focusing on the risks of neurological diseases. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:134054. [PMID: 38503214 DOI: 10.1016/j.jhazmat.2024.134054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/21/2024]
Abstract
Pollution of micro/nano-plastics (MPs/NPs) is ubiquitously prevalent in the environment, leading to an unavoidable exposure of the human body. Despite the protection of the blood-brain barrier, MPs/NPs can be transferred and accumulated in the brain, which subsequently exert negative effects on the brain. Nevertheless, the potential neurodevelopmental and/or neurodegenerative risks of MPs/NPs remain largely unexplored. In this review, we provide a systematic overview of recent studies related to the neurotoxicity of MPs/NPs. It covers the environmental hazards and human exposure pathways, translocation and distribution into the brain, the neurotoxic effects, and the possible mechanisms of environmental MPs/NPs. MPs/NPs are widely found in different environment matrices, including air, water, soil, and human food. Ambient MPs/NPs can enter the human body by ingestion, inhalation and dermal contact, then be transferred into the brain via the blood circulation and nerve pathways. When MPs/NPs are present in the brain, they can initiate a series of molecular or cellular reactions that may harm the blood-brain barrier, cause oxidative stress, trigger inflammatory responses, affect acetylcholinesterase activity, lead to mitochondrial dysfunction, and impair autophagy. This can result in abnormal protein folding, loss of neurons, disruptions in neurotransmitters, and unusual behaviours, ultimately contributing to the initiation and progression of neurodegenerative changes and neurodevelopmental abnormalities. Key challenges and further research directions are also proposed in this review as more studies are needed to focus on the potential neurotoxicity of MPs/NPs under realistic conditions.
Collapse
Affiliation(s)
- Shuang Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinling He
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Jia Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Qingqing Zhu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunyang Liao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
10
|
Tsukamoto H, Olesen ND, Petersen LG, Suga T, Sørensen H, Nielsen HB, Ogoh S, Secher NH, Hashimoto T. Circulating Plasma Oxytocin Level Is Elevated by High-Intensity Interval Exercise in Men. Med Sci Sports Exerc 2024; 56:927-932. [PMID: 38115226 DOI: 10.1249/mss.0000000000003360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
PURPOSE We evaluated whether repeated high-intensity interval exercise (HIIE) influences plasma oxytocin (OT) concentration in healthy men, and, given that OT is mainly synthesized in the hypothalamus, we assessed the concentration difference between the arterial (OT ART ) versus the internal jugular venous OT concentration (OT IJV ). Additionally, we hypothesized that an increase in cerebral OT release and the circulating concentration would be augmented by repeated HIIE. METHODS Fourteen healthy men (age = 24 ± 2 yr; mean ± SD) performed two identical bouts of HIIE. These HIIE bouts included a warm-up at 50%-60% maximal workload ( Wmax ) for 5 min followed by four bouts of exercise at 80%-90% Wmax for 4 min interspersed by exercise at 50%-60% Wmax for 3 min. The HIIE bouts were separated by 60 min of rest. OT was evaluated in blood through radial artery and internal jugular vein catheterization. RESULTS Both HIIE bouts increased both OT ART (median [IQR], from 3.9 [3.4-5.4] to 5.3 [4.4-6.3] ng·mL -1 in the first HIIE, P < 0.01) and OT IJV (from 4.6 [3.4-4.8] to 5.9 [4.3-8.2] ng·mL -1 , P < 0.01), but OT ART-IJV was unaffected (from -0.24 [-1.16 to 1.08] to 0.04 [-0.88 to 0.78] ng·mL -1 , P = 1.00). The increased OT levels were similar in the first and second HIIE bouts (OT ARTP = 0.25, OT IJVP = 0.36). CONCLUSIONS Despite no change in the cerebral OT release via the internal jugular vein, circulating OT increases during HIIE regardless of the accumulated exercise volume, indicating that OT may play role as one of the exerkines.
Collapse
Affiliation(s)
| | | | | | - Tadashi Suga
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, Shiga, JAPAN
| | - Henrik Sørensen
- Department of Anesthesia, Rigshospitalet, Department of Clinical Medicine, University of Copenhagen, Copenhagen, DENMARK
| | | | - Shigehiko Ogoh
- Department of Biomedical Engineering, Toyo University, Saitama, JAPAN
| | - Niels H Secher
- Department of Anesthesia, Rigshospitalet, Department of Clinical Medicine, University of Copenhagen, Copenhagen, DENMARK
| | | |
Collapse
|
11
|
Han SW, Choi J, Ryu KY. Recent progress and future directions of the research on nanoplastic-induced neurotoxicity. Neural Regen Res 2024; 19:331-335. [PMID: 37488886 PMCID: PMC10503636 DOI: 10.4103/1673-5374.379016] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/13/2023] [Accepted: 05/15/2023] [Indexed: 07/26/2023] Open
Abstract
Many types of plastic products, including polystyrene, have long been used in commercial and industrial applications. Microplastics and nanoplastics, plastic particles derived from these plastic products, are emerging as environmental pollutants that can pose health risks to a wide variety of living organisms, including humans. However, it is not well understood how microplastics and nanoplastics affect cellular functions and induce stress responses. Humans can be exposed to polystyrene-microplastics and polystyrene-nanoplastics through ingestion, inhalation, or skin contact. Most ingested plastics are excreted from the body, but inhaled plastics may accumulate in the lungs and can even reach the brain via the nose-to-brain route. Small-sized polystyrene-nanoplastics can enter cells by endocytosis, accumulate in the cytoplasm, and cause various cellular stresses, such as inflammation with increased pro-inflammatory cytokine production, oxidative stress with generation of reactive oxygen species, and mitochondrial dysfunction. They induce autophagy activation and autophagosome formation, but autophagic flux may be impaired due to lysosomal dysfunction. Unless permanently exposed to polystyrene-nanoplastics, they can be removed from cells by exocytosis and subsequently restore cellular function. However, neurons are very susceptible to this type of stress, thus even acute exposure can lead to neurodegeneration without recovery. This review focuses specifically on recent advances in research on polystyrene-nanoplastic-induced cytotoxicity and neurotoxicity. Furthermore, in this review, based on mechanistic studies of polystyrene-nanoplastics at the cellular level other than neurons, future directions for overcoming the negative effects of polystyrene-nanoplastics on neurons were suggested.
Collapse
Affiliation(s)
- Seung-Woo Han
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, Seoul, South Korea
| | - Kwon-Yul Ryu
- Department of Life Science, University of Seoul, Seoul, South Korea
| |
Collapse
|
12
|
Zhan S, Qi Z, Cai F, Gao Z, Xie J, Hu J. Oxytocin neurons mediate stress-induced social memory impairment. Curr Biol 2024; 34:36-45.e4. [PMID: 38103551 DOI: 10.1016/j.cub.2023.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 10/27/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023]
Abstract
Oxytocin has long been thought to play a substantial role in social behaviors, such as social attachment and parenting behavior. However, how oxytocin neurons respond to social and non-social stimuli is largely unknown, especially in high temporal resolution. Here, we recorded the in vivo real-time responses of oxytocin neurons in the paraventricular nucleus of the hypothalamus (PVN) in freely behaving mice. Our results revealed that oxytocin neurons were activated more significantly by stressors than social stimuli. The activation of oxytocin neurons was precisely correlated with struggling behavior during stress. Furthermore, we found that oxytocin mediated stress-induced social memory impairment. Our results reveal an important role of PVN oxytocin neurons in stress-induced social amnesia.
Collapse
Affiliation(s)
- Shulu Zhan
- School of Life Science and Technology, ShanghaiTech University, 393 Huaxia Middle Road, Shanghai 201210, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Institute of Neuroscience, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenhua Qi
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Fang Cai
- School of Life Science and Technology, ShanghaiTech University, 393 Huaxia Middle Road, Shanghai 201210, China
| | - Zilong Gao
- Chinese Institute for Brain Research, Beijing (CIBR), Bldg. 3, No. 9, YIKE Rd, Zhongguancun Life Science Park, Changping District, Beijing 102206, China.
| | - Jingdun Xie
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China.
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, 393 Huaxia Middle Road, Shanghai 201210, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
13
|
Hatakawa Y. [Delivery of JAL-TA9 to the brain by nasal application]. Nihon Yakurigaku Zasshi 2024; 159:396-401. [PMID: 39496416 DOI: 10.1254/fpj.24075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
We have reported on two Catalytides (Catalytic peptides), JAL-TA9 (YKGSGFRMI) and ANA-TA9 (SKGQAYRMI). Both peptides belong to the Tob/BTG family proteins and cleave Aβ42. Although Catalytides must be delivered to the brain parenchyma to treat Alzheimer's disease, the blood-brain barrier (BBB) limits their entry into the brain from the systemic circulation. Thus, we evaluated the direct route of ANA-TA9 from the nasal cavity to the brain to bypass the BBB. In this study, we present our findings on JAL-TA9. Animal studies using rats and mice clarified that the plasma clearance of JAL-TA9 was more rapid than its in vitro degradation in plasma, whole blood, and cerebrospinal fluid (CSF). After nasal administration of JAL-TA9, brain concentrations were significantly higher than after intraperitoneal administration, despite much lower plasma concentration. This observation strongly suggests direct delivery of JAL-TA9 to the brain from the nasal cavity. Similar findings were observed for its transport to CSF after nasal and intravenous administration. The concentration of JAL-TA9 in the olfactory bulb peaked at 5 min, while those in the frontal brain peaked at 30 min and in the occipital brain at 60 min. In conclusion, JAL-TA9 was efficiently delivered to the brain by nasal application compared to other routes.
Collapse
Affiliation(s)
- Yusuke Hatakawa
- Department of Bio-analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
14
|
Patharapankal EJ, Ajiboye AL, Mattern C, Trivedi V. Nose-to-Brain (N2B) Delivery: An Alternative Route for the Delivery of Biologics in the Management and Treatment of Central Nervous System Disorders. Pharmaceutics 2023; 16:66. [PMID: 38258077 PMCID: PMC10818989 DOI: 10.3390/pharmaceutics16010066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
In recent years, there have been a growing number of small and large molecules that could be used to treat diseases of the central nervous system (CNS). Nose-to-brain delivery can be a potential option for the direct transport of molecules from the nasal cavity to different brain areas. This review aims to provide a compilation of current approaches regarding drug delivery to the CNS via the nose, with a focus on biologics. The review also includes a discussion on the key benefits of nasal delivery as a promising alternative route for drug administration and the involved pathways or mechanisms. This article reviews how the application of various auxiliary agents, such as permeation enhancers, mucolytics, in situ gelling/mucoadhesive agents, enzyme inhibitors, and polymeric and lipid-based systems, can promote the delivery of large molecules in the CNS. The article also includes a discussion on the current state of intranasal formulation development and summarizes the biologics currently in clinical trials. It was noted that significant progress has been made in this field, and these are currently being applied to successfully transport large molecules to the CNS via the nose. However, a deep mechanistic understanding of this route, along with the intimate knowledge of various excipients and their interactions with the drug and nasal physiology, is still necessary to bring us one step closer to developing effective formulations for nasal-brain drug delivery.
Collapse
Affiliation(s)
- Elizabeth J. Patharapankal
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, Canterbury ME4 4TB, UK; (E.J.P.); (A.L.A.)
| | - Adejumoke Lara Ajiboye
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, Canterbury ME4 4TB, UK; (E.J.P.); (A.L.A.)
| | | | - Vivek Trivedi
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, Canterbury ME4 4TB, UK; (E.J.P.); (A.L.A.)
| |
Collapse
|
15
|
Schwinghamer K, Siahaan TJ. Enhancing Antibody Exposure in the Central Nervous System: Mechanisms of Uptake, Clearance, and Strategies for Improved Brain Delivery. JOURNAL OF NANOTHERANOSTICS 2023; 4:463-479. [PMID: 39897432 PMCID: PMC11784990 DOI: 10.3390/jnt4040020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
Abstract
Antibodies (mAbs) are attractive molecules for their application as a diagnostic and therapeutic agent for diseases of the central nervous system (CNS). mAbs can be generated to have high affinity and specificity to target molecules in the CNS. Unfortunately, only a very small number of mAbs have been specifically developed and approved for neurological indications. This is primarily attributed to their low exposure within the CNS, hindering their ability to reach and effectively engage their potential targets in the brain. This review discusses aspects of various barriers such as the blood-brain barrier (BBB) and blood-cerebrospinal fluid (CSF) barrier (BCSFB) that regulate the entry and clearance of mAbs into and from the brain. The roles of the glymphatic system on brain exposure and clearance are being described. We also discuss the proposed mechanisms of the uptake of mAbs into the brain and for clearance. Finally, several methods of enhancing the exposure of mAbs in the CNS were discussed, including receptor-mediated transcytosis, osmotic BBB opening, focused ultrasound (FUS), BBB-modulating peptides, and enhancement of mAb brain retention.
Collapse
Affiliation(s)
- Kelly Schwinghamer
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66046, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66046, USA
| |
Collapse
|
16
|
Schimmer J, Patwell R, Küppers S, Grinevich V. The Relationship Between Oxytocin and Alcohol Dependence. Curr Top Behav Neurosci 2023. [PMID: 37697074 DOI: 10.1007/7854_2023_444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
The hypothalamic neuropeptide oxytocin (OT) is well known for its prosocial, anxiolytic, and ameliorating effects on various psychiatric conditions, including alcohol use disorder (AUD). In this chapter, we will first introduce the basic neurophysiology of the OT system and its interaction with other neuromodulatory and neurotransmitter systems in the brain. Next, we provide an overview over the current state of research examining the effects of acute and chronic alcohol exposure on the OT system as well as the effects of OT system manipulation on alcohol-related behaviors in rodents and humans. In rodent models of AUD, OT has been repeatedly shown to reduce ethanol consumption, particularly in models of acute alcohol exposure. In humans however, the results of OT administration on alcohol-related behaviors are promising but not yet conclusive. Therefore, we further discuss several physiological and methodological limitations to the effective application of OT in the clinic and how they may be mitigated by the application of synthetic OT receptor (OTR) agonists. Finally, we discuss the potential efficacy of cutting-edge pharmacology and gene therapies designed to specifically enhance endogenous OT release and thereby rescue deficient expression of OT in the brains of patients with severe forms of AUD and other incurable mental disorders.
Collapse
Affiliation(s)
- Jonas Schimmer
- Department of Neuropeptide Research in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Ryan Patwell
- Department of Neuropeptide Research in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Stephanie Küppers
- Department of Neuropeptide Research in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
17
|
Potretzke S, Zhang Y, Li J, Fecteau KM, Erikson DW, Hibert M, Ryabinin AE. Male-selective effects of oxytocin agonism on alcohol intake: behavioral assessment in socially housed prairie voles and involvement of RAGE. Neuropsychopharmacology 2023; 48:920-928. [PMID: 36369481 PMCID: PMC10156683 DOI: 10.1038/s41386-022-01490-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022]
Abstract
Targeting the oxytocin (OXT) peptide system has emerged as a promising new approach for the treatment of alcohol use disorder (AUD). However, further advancements in this development depend on properly modeling various complex social aspects of AUD and its treatment. Here we examined behavioral and molecular underpinnings of OXT receptor (OXTR) agonism in prairie voles, a rodent species with demonstrated translational validity for neurobiological mechanisms regulating social affiliations. To further improve translational validity of these studies, we examined effects of intranasal (IN) OXT administration in male and female prairie voles socially housed in the presence of untreated cagemates. IN OXT selectively inhibited alcohol drinking in male, but not female, animals. Further, we confirmed that exogenously administered OXT penetrates the prairie vole brain and showed that Receptor for Advanced Glycation End-products assists this penetration after IN, but not intraperitoneal (IP), OXT administration. Finally, we demonstrated that IP administration of LIT-001, a small-molecule OXTR agonist, inhibits alcohol intake in male, but not female, prairie voles socially housed in the presence of untreated cagemates. Taken together, results of this study support the promise of selectively targeting OXTR for individualized treatment of AUD.
Collapse
Affiliation(s)
- Sheena Potretzke
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Yangmiao Zhang
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Ju Li
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kristopher M Fecteau
- Endocrine Technologies Core, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - David W Erikson
- Endocrine Technologies Core, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Marcel Hibert
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg, 74 Route du Rhin, F-67412, Illkirch, France
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
18
|
Dergacheva O, Polotsky VY, Mendelowitz D. Oxytocin mediated excitation of hypoglossal motoneurons: implications for treating obstructive sleep apnea. Sleep 2023; 46:zsad009. [PMID: 36846973 PMCID: PMC10091096 DOI: 10.1093/sleep/zsad009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/21/2022] [Indexed: 03/01/2023] Open
Abstract
Clinical studies have shown that oxytocin administered intranasally (IN) decreased the incidence and duration of obstructive events in patients with obstructive sleep apnea (OSA). Although the mechanisms by which oxytocin promotes these beneficial effects are unknown, one possible target of oxytocin could be the excitation of tongue-projecting hypoglossal motoneurons in the medulla, that exert central control of upper airway patency. This study tested the hypothesis that IN oxytocin enhances tongue muscle activity via the excitation of hypoglossal motoneurons projecting to tongue protrudor muscles (PMNs). To test this hypothesis we performed in vivo and in vitro electrophysiological studies in C57BL6/J mice as well as fluorescent imaging studies in transgenic mice in which neurons that express oxytocin receptors co-express fluorescent protein. IN oxytocin significantly increased the amplitude of inspiratory-related tongue muscle activity. This effect was abolished by severing the medial branch of hypoglossal nerve that innervates PMNs of the tongue. Oxytocin receptor-positive neurons were more prevalent in the population of PMNs than in retractor-projecting hypoglossal motoneurons (RMNs). Oxytocin administration increased action potential firing in PMNs, but had no significant effect on firing activity in RMNs. In conclusion, IN oxytocin stimulates respiratory-relating tongue muscle activity likely acting on central hypoglossal motoneurons that provide tongue protrusion and upper airway opening. This mechanism may play a role in oxytocin-induced reductions in upper airway obstructions in patients with OSA.
Collapse
Affiliation(s)
- Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| |
Collapse
|
19
|
Szewczyk AK, Ulutas S, Aktürk T, Al-Hassany L, Börner C, Cernigliaro F, Kodounis M, Lo Cascio S, Mikolajek D, Onan D, Ragaglini C, Ratti S, Rivera-Mancilla E, Tsanoula S, Villino R, Messlinger K, Maassen Van Den Brink A, de Vries T. Prolactin and oxytocin: potential targets for migraine treatment. J Headache Pain 2023; 24:31. [PMID: 36967387 PMCID: PMC10041814 DOI: 10.1186/s10194-023-01557-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/28/2023] [Indexed: 03/28/2023] Open
Abstract
Migraine is a severe neurovascular disorder of which the pathophysiology is not yet fully understood. Besides the role of inflammatory mediators that interact with the trigeminovascular system, cyclic fluctuations in sex steroid hormones are involved in the sex dimorphism of migraine attacks. In addition, the pituitary-derived hormone prolactin and the hypothalamic neuropeptide oxytocin have been reported to play a modulating role in migraine and contribute to its sex-dependent differences. The current narrative review explores the relationship between these two hormones and the pathophysiology of migraine. We describe the physiological role of prolactin and oxytocin, its relationship to migraine and pain, and potential therapies targeting these hormones or their receptors.In summary, oxytocin and prolactin are involved in nociception in opposite ways. Both operate at peripheral and central levels, however, prolactin has a pronociceptive effect, while oxytocin appears to have an antinociceptive effect. Therefore, migraine treatment targeting prolactin should aim to block its effects using prolactin receptor antagonists or monoclonal antibodies specifically acting at migraine-pain related structures. This action should be local in order to avoid a decrease in prolactin levels throughout the body and associated adverse effects. In contrast, treatment targeting oxytocin should enhance its signalling and antinociceptive effects, for example using intranasal administration of oxytocin, or possibly other oxytocin receptor agonists. Interestingly, the prolactin receptor and oxytocin receptor are co-localized with estrogen receptors as well as calcitonin gene-related peptide and its receptor, providing a positive perspective on the possibilities for an adequate pharmacological treatment of these nociceptive pathways. Nevertheless, many questions remain to be answered. More particularly, there is insufficient data on the role of sex hormones in men and the correct dosing according to sex differences, hormonal changes and comorbidities. The above remains a major challenge for future development.
Collapse
Affiliation(s)
- Anna K Szewczyk
- Doctoral School, Medical University of Lublin, Lublin, Poland
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| | - Samiye Ulutas
- Department of Neurology, Kartal Dr. Lutfi Kirdar Research and Training Hospital, Istanbul, Turkey
| | - Tülin Aktürk
- Department of Neurology, Kartal Dr. Lutfi Kirdar Research and Training Hospital, Istanbul, Turkey
| | - Linda Al-Hassany
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Corinna Börner
- Department of Pediatrics - Dr. von Hauner Children's Hospital, LMU Hospital, Division of Pediatric Neurology and Developmental Medicine, Ludwig-Maximilians Universität München, Lindwurmstr. 4, 80337, Munich, Germany
- LMU Center for Children with Medical Complexity - iSPZ Hauner, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, 80337, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Federica Cernigliaro
- Child Neuropsychiatry Unit Department, Pro.M.I.S.E. "G D'Alessandro, University of Palermo, 90133, Palermo, Italy
| | - Michalis Kodounis
- First Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Salvatore Lo Cascio
- Child Neuropsychiatry Unit Department, Pro.M.I.S.E. "G D'Alessandro, University of Palermo, 90133, Palermo, Italy
| | - David Mikolajek
- Department of Neurology, City Hospital Ostrava, Ostrava, Czech Republic
| | - Dilara Onan
- Spine Health Unit, Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Chiara Ragaglini
- Neuroscience Section, Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, 67100, L'Aquila, Italy
| | - Susanna Ratti
- Neuroscience Section, Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, 67100, L'Aquila, Italy
| | - Eduardo Rivera-Mancilla
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sofia Tsanoula
- Department of Neurology, 401 Military Hospital of Athens, Athens, Greece
| | - Rafael Villino
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Karl Messlinger
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Antoinette Maassen Van Den Brink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tessa de Vries
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
20
|
Kurano T, Kanazawa T, Iioka S, Kondo H, Kosuge Y, Suzuki T. Intranasal Administration of N-acetyl-L-cysteine Combined with Cell-Penetrating Peptide-Modified Polymer Nanomicelles as a Potential Therapeutic Approach for Amyotrophic Lateral Sclerosis. Pharmaceutics 2022; 14:2590. [PMID: 36559085 PMCID: PMC9785447 DOI: 10.3390/pharmaceutics14122590] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Intranasal administration is a promising route for direct drug delivery to the brain; its combination with nanocarriers enhances delivery. We have previously shown that intranasal administration combined with PEG-PCL-Tat (a nanocarrier) efficiently delivers drugs to the brain and exhibits excellent therapeutic efficacy against brain diseases. We aimed to clarify whether intranasal administration combined with PEG-PCL-Tat represents a useful drug delivery system (DDS) for amyotrophic lateral sclerosis (ALS) pharmacotherapy. We used N-acetyl-L-cysteine (NAC) as a model drug with low transferability to the spinal cord and determined the physicochemical properties of NAC/PEG-PCL-Tat. After intranasal administration of NAC/PEG-PCL-Tat, we measured the survival duration of superoxide dismutase-1 G93A mutant transgenic mice (G93A mice), widely used in ALS studies, and quantitatively analyzed the tissue distribution of NAC/PEG-PCL-Tat in ddY mice. The mean particle size and zeta potential of NAC/PEG-PCL-Tat were 294 nm and + 9.29 mV, respectively. Treatment with repeated intranasal administration of NAC/PEG-PCL-Tat considerably prolonged the median survival of G93A mice by 11.5 days compared with that of untreated G93A mice. Moreover, the highest distribution after a single administration of NAC/PEG-PCL-Tat was measured in the spinal cord. These results suggest that intranasal administration combined with PEG-PCL-Tat might represent a useful DDS for ALS therapeutics.
Collapse
Affiliation(s)
- Takumi Kurano
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan
| | - Takanori Kanazawa
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Shizuoka, Japan
| | - Shingo Iioka
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Shizuoka, Japan
| | - Hiromu Kondo
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Shizuoka, Japan
| | - Yasuhiro Kosuge
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan
| | - Toyofumi Suzuki
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan
| |
Collapse
|
21
|
Higashida H, Furuhara K, Lopatina O, Gerasimenko M, Hori O, Hattori T, Hayashi Y, Cherepanov SM, Shabalova AA, Salmina AB, Minami K, Yuhi T, Tsuji C, Fu P, Liu Z, Luo S, Zhang A, Yokoyama S, Shuto S, Watanabe M, Fujiwara K, Munesue SI, Harashima A, Yamamoto Y. Oxytocin Dynamics in the Body and Brain Regulated by the Receptor for Advanced Glycation End-Products, CD38, CD157, and Nicotinamide Riboside. Front Neurosci 2022; 16:858070. [PMID: 35873827 PMCID: PMC9301327 DOI: 10.3389/fnins.2022.858070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/31/2022] [Indexed: 12/21/2022] Open
Abstract
Investigating the neurocircuit and synaptic sites of action of oxytocin (OT) in the brain is critical to the role of OT in social memory and behavior. To the same degree, it is important to understand how OT is transported to the brain from the peripheral circulation. To date, of these, many studies provide evidence that CD38, CD157, and receptor for advanced glycation end-products (RAGE) act as regulators of OT concentrations in the brain and blood. It has been shown that RAGE facilitates the uptake of OT in mother’s milk from the digestive tract to the cell surface of intestinal epithelial cells to the body fluid and subsequently into circulation in male mice. RAGE has been shown to recruit circulatory OT into the brain from blood at the endothelial cell surface of neurovascular units. Therefore, it can be said that extracellular OT concentrations in the brain (hypothalamus) could be determined by the transport of OT by RAGE from the circulation and release of OT from oxytocinergic neurons by CD38 and CD157 in mice. In addition, it has recently been found that gavage application of a precursor of nicotinamide adenine dinucleotide, nicotinamide riboside, for 12 days can increase brain OT in mice. Here, we review the evaluation of the new concept that RAGE is involved in the regulation of OT dynamics at the interface between the brain, blood, and intestine in the living body, mainly by summarizing our recent results due to the limited number of publications on related topics. And we also review other possible routes of OT recruitment to the brain.
Collapse
Affiliation(s)
- Haruhiro Higashida
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
- Laboratory of Social Brain Study, Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Professor V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
- *Correspondence: Haruhiro Higashida,
| | - Kazumi Furuhara
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Olga Lopatina
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
- Laboratory of Social Brain Study, Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Professor V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Maria Gerasimenko
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yasuhiko Hayashi
- Department of Neurosurgery, Kanazawa Medical University, Kanazawa, Japan
| | - Stanislav M. Cherepanov
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Anna A. Shabalova
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Alla B. Salmina
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
- Laboratory of Social Brain Study, Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Professor V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Kana Minami
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Teruko Yuhi
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Chiharu Tsuji
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - PinYue Fu
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Zhongyu Liu
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Shuxin Luo
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Anpei Zhang
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Shigeru Yokoyama
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Satoshi Shuto
- Faculty of Pharmaceutical Sciences, Center for Research and Education on Drug Discovery, Hokkaido University, Sapporo, Japan
| | - Mizuki Watanabe
- Faculty of Pharmaceutical Sciences, Center for Research and Education on Drug Discovery, Hokkaido University, Sapporo, Japan
| | - Koichi Fujiwara
- Faculty of Pharmaceutical Sciences, Center for Research and Education on Drug Discovery, Hokkaido University, Sapporo, Japan
| | - Sei-ichi Munesue
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Ai Harashima
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
22
|
Spinelli CP, Iwanaga J, Hur MS, Dumont AS, Tubbs RS. Discovery of a trans-sellar vascular supply for the pituitary gland. Anat Cell Biol 2022; 55:124-129. [PMID: 35599459 PMCID: PMC9256484 DOI: 10.5115/acb.21.255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/11/2022] [Accepted: 03/03/2022] [Indexed: 11/27/2022] Open
Abstract
The vasculature of the pituitary gland is discussed briefly and the details of an anatomical discovery of the vessels supplying the pituitary gland provided. Twenty latex injected cadaveric heads were dissected. Any vessels that were found to penetrate the sella turcica and travel to the pituitary gland were documented and measured. Additionally, 25 adult skulls were evaluated for the presence, size, and sites of bony foramina in the floor of sella turcica. Trans-sellar vessels were identified in 65% of specimens. There was a mean of 1.5 vessels per specimen consisting usually of a mixture of veins and arteries. The mean diameter of these vessels was 0.3 mm and the mean length from the sella turcica to the pituitary gland was 2.3 mm. These vessels were concentrated in the most concave part of the sella turcica. In bony specimens, the mean number of trans-sellar foramina was four. The diameter of these foramina ranged from 0.3 to 0.6 mm in size. The trans-sellar foramina were concentrated near the center part of the sella turcica and had no regular pattern. The pituitary gland receives at least some blood supply and drainage via vessels traveling along the septum of the sphenoidal sinuses and through the sella turcica. Knowledge of such vessels might lead to a better understanding of the vascular supply and drainage of the pituitary gland and would be useful during skull base approaches such as trans-nasal approaches to the pituitary gland.
Collapse
Affiliation(s)
- Casey P Spinelli
- Tulane University & Ochsner Clinic Neurosurgery Program, Tulane University School of Medicine, New Orleans, LA, USA
| | - Joe Iwanaga
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA.,Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Mi-Sun Hur
- Department of Anatomy, Catholic Kwandong University College of Medicine, Gangneung, Korea
| | - Aaron S Dumont
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - R Shane Tubbs
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA.,Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA.,Department of Neurosurgery and Ochsner Neuroscience Institute, Ochsner Health System, New Orleans, LA, USA.,Department of Anatomical Sciences, St. George's University, St. George's, Grenada.,Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA.,Department of Structural & Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA.,University of Queensland, Brisbane, Australia
| |
Collapse
|
23
|
Szafoni S, Piegza M. Progress in Personalized Psychiatric Therapy with the Example of Using Intranasal Oxytocin in PTSD Treatment. J Pers Med 2022; 12:1067. [PMID: 35887564 PMCID: PMC9317706 DOI: 10.3390/jpm12071067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/21/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a severe mental disorder that results in the frequent coexistence of other diseases, lowers patients' quality of life, and has a high annual cost of treatment. However, despite the variety of therapeutic approaches that exist, some patients still do not achieve the desired results. In addition, we may soon face an increase in the number of new PTSD cases because of the current global situation-both the COVID-19 pandemic and the ongoing armed conflicts. Hence, in recent years, many publications have sought a new, more personalized treatment approach. One such approach is the administration of intranasal oxytocin (INOXT), which, due to its pleiotropic effects, seems to be a promising therapeutic option. However, the current findings suggest that it might only be helpful for a limited, strictly selected group of patients.
Collapse
Affiliation(s)
- Sandra Szafoni
- Department of Psychiatry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 42-612 Tarnowskie Góry, Poland;
| | | |
Collapse
|
24
|
Takayama K. Peptide Tool-Driven Functional Elucidation of Biomolecules Related to Endocrine System and Metabolism. Chem Pharm Bull (Tokyo) 2022; 70:413-419. [PMID: 35650039 DOI: 10.1248/cpb.c22-00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The enhancement of basic research based on biomolecule-derived peptides has the potential to elucidate their biological function and lead to the development of new drugs. In this review, two biomolecules, namely "neuromedin U (NMU)" and "myostatin," are discussed. NMU, a neuropeptide first isolated from the porcine spinal cord, non-selectively activates two types of receptors (NMUR1 and NMUR2) and displays a variety of physiological actions, including appetite suppression. The development of receptor-selective regulators helps elucidate each receptor's detailed biological roles. A structure-activity relationship (SAR) study was conducted to achieve this purpose using the amidated C-terminal core structure of NMU for receptor activation. Through obtaining receptor-selective hexapeptide agonists, molecular functions of the core structure were clarified. Myostatin is a negative regulator of skeletal muscle growth and has attracted attention as a target for treating atrophic muscle disorders. Although the protein inhibitors, such as antibodies and receptor-decoys have been developed, the inhibition by smaller molecules, including peptides, is less advanced. Focusing on the inactivation mechanism by prodomain proteins derived from myostatin-precursor, a first mid-sized α-helical myostatin-inhibitory peptide (23-mer) was identified from the mouse sequence. The detailed SAR study based on this peptide afforded the structural requirements for effective inhibition. The subsequent computer simulation proposed the docking mode at the activin type I receptor binding site of myostatin. The resulting development of potent inhibitors suggested the existence of a more appropriate binding mode linked to their β-sheet forming properties, suggesting that further investigations might be needed.
Collapse
Affiliation(s)
- Kentaro Takayama
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University
| |
Collapse
|
25
|
Ye C, Cheng M, Ma L, Zhang T, Sun Z, Yu C, Wang J, Dou Y. Oxytocin Nanogels Inhibit Innate Inflammatory Response for Early Intervention in Alzheimer's Disease. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21822-21835. [PMID: 35510352 DOI: 10.1021/acsami.2c00007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Prevention of Alzheimer's disease (AD) is a global imperative, but reliable early interventions are currently lacking. Microglia-mediated chronic neuroinflammation is thought to occur in the early stage of AD and plays a critical role in AD pathogenesis. Here, oxytocin (OT)-loaded angiopep-2-modified chitosan nanogels (AOC NGs) were designed for early treatment of AD via inhibiting innate inflammatory response. Through the effective transcytosis of angiopep-2, AOC NGs were driven intravenously to cross the blood-brain barrier, enter the brain, and enrich in brain areas affected by AD. A large amount of OT was then released and specifically bound to the pathological upregulated OT receptor, thus effectively inhibiting microglial activation and reducing inflammatory cytokine levels through blocking the ERK/p38 MAPK and COX-2/iNOS NF-κB signaling pathways. Consecutive weekly intravenous administration of AOC NGs into 12-week-old young APP/PS1 mice, representing the early stage of AD, remarkably slowed the progression of Aβ deposition and neuronal apoptosis in the APP/PS1 mice as they aged and ultimately prevented cognitive impairment and delayed hippocampal atrophy. Together, the findings suggest that AOC NGs, which show good biosafety, can serve as a promising therapeutic candidate to combat neuroinflammation for early prevention of AD.
Collapse
Affiliation(s)
- Caihua Ye
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Meng Cheng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Lin Ma
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Tianzhu Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Junping Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| |
Collapse
|
26
|
Neural circuit control of innate behaviors. SCIENCE CHINA. LIFE SCIENCES 2022; 65:466-499. [PMID: 34985643 DOI: 10.1007/s11427-021-2043-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022]
Abstract
All animals possess a plethora of innate behaviors that do not require extensive learning and are fundamental for their survival and propagation. With the advent of newly-developed techniques such as viral tracing and optogenetic and chemogenetic tools, recent studies are gradually unraveling neural circuits underlying different innate behaviors. Here, we summarize current development in our understanding of the neural circuits controlling predation, feeding, male-typical mating, and urination, highlighting the role of genetically defined neurons and their connections in sensory triggering, sensory to motor/motivation transformation, motor/motivation encoding during these different behaviors. Along the way, we discuss possible mechanisms underlying binge-eating disorder and the pro-social effects of the neuropeptide oxytocin, elucidating the clinical relevance of studying neural circuits underlying essential innate functions. Finally, we discuss some exciting brain structures recurrently appearing in the regulation of different behaviors, which suggests both divergence and convergence in the neural encoding of specific innate behaviors. Going forward, we emphasize the importance of multi-angle and cross-species dissections in delineating neural circuits that control innate behaviors.
Collapse
|
27
|
Liu X, Zhao Y, Dou J, Hou Q, Cheng J, Jiang X. Bioeffects of Inhaled Nanoplastics on Neurons and Alteration of Animal Behaviors through Deposition in the Brain. NANO LETTERS 2022; 22:1091-1099. [PMID: 35089039 DOI: 10.1021/acs.nanolett.1c04184] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The potential toxicity of nanoplastics on plants has previously been illustrated, but whether nanoplastics could cause neurotoxicity, especially to higher animals, remains unclear. We now demonstrate that nanoplastics can be deposited in the brain via nasal inhalation, triggering neuron toxicity and altering the animal behavior. Polystyrene nanoparticles (PS-NPs) of PS-COOH and PS-NH2 are used as models for nanoplastics. We designed a microfluidic chip to evaluate the PS-NPs with different concentrations, surface ligands, and sizes to interact with neurons. Smaller PS-NPs can induce more cellular uptake than larger PS-NPs. PS-NPs with a size of 80 nm can reach and deposit in the brain of mice via aerosol inhalation. Mice inhaling PS-NPs exhibit fewer activities in comparison to those inhaling water droplets. An obvious neurotoxicity of the nanoplastics could be observed from the results of the inhibition of AChE activities. Our results show the potential significance of the physiochemical properties of organic nanoplastics on depositing in mammalian brains by nasal inhalation.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, People's Republic of China
- CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, University of Chinese Academy of Sciences, No. 11 Zhongguancun Beiyitiao, Beijing 100190, People's Republic of China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, People's Republic of China
| | - Yingcan Zhao
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, People's Republic of China
| | - Jiabin Dou
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, People's Republic of China
- CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, University of Chinese Academy of Sciences, No. 11 Zhongguancun Beiyitiao, Beijing 100190, People's Republic of China
| | - Qinghong Hou
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, People's Republic of China
| | - Jinxiong Cheng
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, People's Republic of China
| | - Xingyu Jiang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, People's Republic of China
- CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, University of Chinese Academy of Sciences, No. 11 Zhongguancun Beiyitiao, Beijing 100190, People's Republic of China
| |
Collapse
|
28
|
Yao S, Kendrick KM. Effects of Intranasal Administration of Oxytocin and Vasopressin on Social Cognition and Potential Routes and Mechanisms of Action. Pharmaceutics 2022; 14:323. [PMID: 35214056 PMCID: PMC8874551 DOI: 10.3390/pharmaceutics14020323] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 12/03/2022] Open
Abstract
Acute and chronic administration of intranasal oxytocin and vasopressin have been extensively utilized in both animal models and human preclinical and clinical studies over the last few decades to modulate various aspects of social cognition and their underlying neural mechanisms, although effects are not always consistent. The use of an intranasal route of administration is largely driven by evidence that it permits neuropeptides to penetrate directly into the brain by circumventing the blood-brain barrier, which has been considered relatively impermeable to them. However, this interpretation has been the subject of considerable debate. In this review, we will focus on research in both animal models and humans, which investigates the different potential routes via which these intranasally administered neuropeptides may be producing their various effects on social cognition. We will also consider the contribution of different methods of intranasal application and additionally the importance of dose magnitude and frequency for influencing G protein-coupled receptor signaling and subsequent functional outcomes. Overall, we conclude that while some functional effects of intranasal oxytocin and vasopressin in the domain of social cognition may result from direct penetration into the brain following intranasal administration, others may be contributed by the neuropeptides either entering the peripheral circulation and crossing the blood-brain barrier and/or producing vagal stimulation via peripheral receptors. Furthermore, to complicate matters, functional effects via these routes may differ, and both dose magnitude and frequency can produce very different functional outcomes and therefore need to be optimized to produce desired effects.
Collapse
Affiliation(s)
- Shuxia Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Keith Maurice Kendrick
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| |
Collapse
|
29
|
Ghazy AA, Soliman OA, Elbahnasi AI, Alawy AY, Mansour AM, Gowayed MA. Role of Oxytocin in Different Neuropsychiatric, Neurodegenerative, and Neurodevelopmental Disorders. Rev Physiol Biochem Pharmacol 2022; 186:95-134. [PMID: 36416982 DOI: 10.1007/112_2022_72] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Oxytocin has recently gained significant attention because of its role in the pathophysiology and management of dominant neuropsychiatric disorders. Oxytocin, a peptide hormone synthesized in the hypothalamus, is released into different brain regions, acting as a neurotransmitter. Receptors for oxytocin are present in many areas of the brain, including the hypothalamus, amygdala, and nucleus accumbens, which have been involved in the pathophysiology of depression, anxiety, schizophrenia, autism, Alzheimer's disease, Parkinson's disease, and attention deficit hyperactivity disorder. Animal studies have spotlighted the role of oxytocin in social, behavioral, pair bonding, and mother-infant bonding. Furthermore, oxytocin protects fetal neurons against injury during childbirth and affects various behaviors, assuming its possible neuroprotective characteristics. In this review, we discuss some of the concepts and mechanisms related to the role of oxytocin in the pathophysiology and management of some neuropsychiatric, neurodegenerative, and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Aya A Ghazy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Omar A Soliman
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Aya I Elbahnasi
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Aya Y Alawy
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Amira Ma Mansour
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Mennatallah A Gowayed
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt.
| |
Collapse
|
30
|
Current knowledge, challenges, new perspectives of the study, and treatments of Autism Spectrum Disorder. Reprod Toxicol 2021; 106:82-93. [PMID: 34695561 DOI: 10.1016/j.reprotox.2021.10.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 01/12/2023]
Abstract
Over the past 70 years, the understanding of Autism Spectrum Disorder (ASD) improved greatly and is characterized as a heterogeneous neuropsychiatric syndrome. ASD is characterized by difficulties in social communication, restricted and repetitive behavior, interests, or activities. And it is often described as a combination of genetic predisposition and environmental factors. There are many treatments and approaches to ASD, including pharmacological therapies with antipsychotics, antidepressants, mood regulators, stimulants, and behavioral ones. However, no treatment is capable of reverting ASD. This review provides an overview of animal models of autism. We summarized genetic and environmental models and then valproic acid treatment as a useful model for ASD. As well as the main therapies and approaches used in the treatment, relating them to the neurochemical pathways altered in ASD, emphasizing the pharmacological potential of peptides and bioinspired compounds found in animal venoms as a possible future treatment for ASD.
Collapse
|
31
|
Hatakawa Y, Tanaka A, Furubayashi T, Nakamura R, Konishi M, Akizawa T, Sakane T. Direct Delivery of ANA-TA9, a Peptide Capable of Aβ Hydrolysis, to the Brain by Intranasal Administration. Pharmaceutics 2021; 13:1673. [PMID: 34683967 PMCID: PMC8538057 DOI: 10.3390/pharmaceutics13101673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 11/17/2022] Open
Abstract
We have recently reported Catalytides (Catalytic peptides) JAL-TA9 (YKGSGFRMI) and ANA-TA9 (SKGQAYRMI), which are the first Catalytides found to cleave Aβ42. Although the Catalytides must be delivered to the brain parenchyma to treat Alzheimer's disease, the blood-brain barrier (BBB) limits their entry into the brain from the systemic circulation. To avoid the BBB, the direct route from the nasal cavity to the brain was used in this study. The animal studies using rats and mice clarified that the plasma clearance of ANA-TA9 was more rapid than in vitro degradation in the plasma, whole blood, and the cerebrospinal fluid (CSF). The brain concentrations of ANA-TA9 were higher after nasal administration than those after intraperitoneal administration, despite a much lower plasma concentration after nasal administration, suggesting the direct delivery of ANA-TA9 to the brain from the nasal cavity. Similar findings were observed for its transport to CSF after nasal and intravenous administration. The concentration of ANA-TA9 in the olfactory bulb reached the peak at 5 min, whereas those in the frontal and occipital brains was 30 min, suggesting the sequential backward translocation of ANA-TA9 in the brain. In conclusion, ANA-TA9 was efficiently delivered to the brain by nasal application, as compared to other routes.
Collapse
Affiliation(s)
- Yusuke Hatakawa
- Laboratory of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba, Sendai 980-8578, Japan;
| | - Akiko Tanaka
- Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-Machi 4-19-1 Higashinada, Kobe, Hyogo 658-8558, Japan; (A.T.); (T.F.)
| | - Tomoyuki Furubayashi
- Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-Machi 4-19-1 Higashinada, Kobe, Hyogo 658-8558, Japan; (A.T.); (T.F.)
| | - Rina Nakamura
- O-Force Co., Ltd., 3454 Irino Kuroshio-Cho, Hata-Gun, Kochi 789-1931, Japan; (R.N.); (T.A.)
- Laboratory of Pharmacology, School of Medicine, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Motomi Konishi
- Department of Integrative Pharmaceutical Science, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka 573-0101, Japan;
| | - Toshifumi Akizawa
- O-Force Co., Ltd., 3454 Irino Kuroshio-Cho, Hata-Gun, Kochi 789-1931, Japan; (R.N.); (T.A.)
- Laboratory of Pharmacology, School of Medicine, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Toshiyasu Sakane
- Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-Machi 4-19-1 Higashinada, Kobe, Hyogo 658-8558, Japan; (A.T.); (T.F.)
| |
Collapse
|
32
|
King CE, Griffin WC, Lopez MF, Becker HC. Activation of hypothalamic oxytocin neurons reduces binge-like alcohol drinking through signaling at central oxytocin receptors. Neuropsychopharmacology 2021; 46:1950-1957. [PMID: 34127796 PMCID: PMC8429589 DOI: 10.1038/s41386-021-01046-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/04/2021] [Accepted: 05/18/2021] [Indexed: 02/05/2023]
Abstract
Preclinical and clinical evidence suggests that exogenous administration of oxytocin (OT) may hold promise as a therapeutic strategy for reducing heavy alcohol drinking. However, it remains unknown whether these effects are mediated by stimulation of endogenous sources of OT and signaling at oxytocin receptors (OTR) in brain or in the periphery. To address this question, we employed a targeted chemogenetic approach to examine whether selective activation of OT-containing neurons in the paraventricular nucleus of the hypothalamus (PVN) alters alcohol consumption in a binge-like drinking ("Drinking-in-the-Dark"; DID) model. Adult male Oxt-IRES-Cre mice received bilateral infusion of a Cre-dependent virus containing an excitatory DREADD (AAV8-hSyn-DIO-hM3Dq-mCherry) or control virus (AAV8-hSyn-DIO-mCherry) into the PVN. Chemogenetic activation of PVNOT+ neurons following clozapine-N-oxide injection reduced binge-like alcohol drinking in a similar manner as systemic administration of the neuropeptide. Pretreatment with a brain-penetrant OTR antagonist (L-368,899) reversed this effect while systemic administration of a peripherally restricted OTR antagonist (Atosiban) did not alter reduced alcohol drinking following chemogenetic activation of PVNOT+ neurons. Altogether, these data are the first to demonstrate that targeted activation of hypothalamic (endogenous) OT reduces alcohol consumption, providing further evidence that this neuropeptide plays a role in regulation of alcohol self-administration behavior. Further, results indicate that the ability OT to reduce alcohol drinking is mediated by signaling at OTR in the brain.
Collapse
Affiliation(s)
- Courtney E King
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- RHJ Department of Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|
33
|
Prenatal exposure to valproic acid and treatment with intranasal oxytocin have sex-specific effects on behavior in Long Evans rats. Behav Pharmacol 2021; 32:561-570. [PMID: 34494987 DOI: 10.1097/fbp.0000000000000650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social behaviors and communication. In rodents and humans, prenatal exposure to antiepileptic valproic acid is associated with an increased risk for autistic-like characteristics. One potential treatment is oxytocin, a prosocial neuropeptide that can be delivered intranasally. However, the sex-specific effects of valproic acid exposure and intranasal oxytocin treatment on behavior have not been fully explored. Pregnant Long Evans rats were administered valproic acid (500 mg/kg) or saline midday on gestational day 12, and after weaning, male and female pups were assigned to control (saline-saline), valproic acid-saline, or valproic acid-oxytocin groups. Oxytocin (0.8 IU/kg) or saline was delivered intranasally 30-60 min before tests for anxiety-like behaviors (elevated plus maze), social interactions (sociability) and sociosexual behaviors (partner preference, 50 kHz vocalizations and scent marking). Prenatal exposure to valproic acid resulted in sex-specific differences in behavior. When compared to controls, valproic acid males showed enhanced anxiety-like behaviors in adolescence and fewer scent marks in adulthood, while valproic acid females showed reduced sexual (partner) preference as adults. Intranasal oxytocin was anxiolytic for valproic acid males, but moderately anxiogenic for valproic acid females, and in both sexes it surprisingly impaired social interactions in the sociability test. Furthermore, intranasal oxytocin failed to improve sociosexual deficits in valproic acid rats. These findings highlight the importance of conducting preclinical studies in both sexes, and suggest that oxytocin may be an effective treatment in animal models with heightened anxiety-like behaviors.
Collapse
|
34
|
Intranasal Administration for Pain: Oxytocin and Other Polypeptides. Pharmaceutics 2021; 13:pharmaceutics13071088. [PMID: 34371778 PMCID: PMC8309171 DOI: 10.3390/pharmaceutics13071088] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 11/16/2022] Open
Abstract
Pain, particularly chronic pain, remains one of the most debilitating and difficult-to-treat conditions in medicine. Chronic pain is difficult to treat, in part because it is associated with plastic changes in the peripheral and central nervous systems. Polypeptides are linear organic polymers that are highly selective molecules for neurotransmitter and other nervous system receptors sites, including those associated with pain and analgesia, and so have tremendous potential in pain therapeutics. However, delivery of polypeptides to the nervous system is largely limited due to rapid degradation within the peripheral circulation as well as the blood–brain barrier. One strategy that has been shown to be successful in nervous system deposition of polypeptides is intranasal (IN) delivery. In this narrative review, we discuss the delivery of polypeptides to the peripheral and central nervous systems following IN administration. We briefly discuss the mechanism of delivery via the nasal–cerebral pathway. We review recent studies that demonstrate that polypeptides such as oxytocin, delivered IN, not only reach key pain-modulating regions in the nervous system but, in doing so, evoke significant analgesic effects. IN administration of polypeptides has tremendous potential to provide a non-invasive, rapid and effective method of delivery to the nervous system for chronic pain treatment and management.
Collapse
|
35
|
Hatakawa Y, Nakamura R, Konishi M, Sakane T, Tanaka A, Matsuda A, Saito M, Akizawa T. Amyloid beta cleavage by ANA-TA9, a synthetic peptide from the ANA/BTG3 Box A region. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12146. [PMID: 33816760 PMCID: PMC8012241 DOI: 10.1002/trc2.12146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/12/2020] [Accepted: 01/06/2021] [Indexed: 11/08/2022]
Abstract
INTRODUCTION We recently discovered a short synthetic peptide derived from the ANA/BTG3 protein Box A region called ANA-TA9 (SKGQAYRMI), which possesses catalytic activity. Herein we demonstrated the proteolytic activity of ANA-TA9 against amyloid beta 42 (Aβ42). METHODS The proteolytic activity of ANA-TA9 against both the authentic soluble form Aβ42 (a-Aβ42) and the solid insoluble form Aβ42 (s-Aβ42) was analyzed by high-performance liquid chromatography and mass spectrometry. Plasma clearance, brain uptake, and cell viability were examined. RESULTS ANA-TA9 cleaved not only a-Aβ42 but also s-Aβ42. Proteolytic activity was partially inhibited by 4-(2-aminoethyl) benzenesulfonyl fluoride hydrochloride, a serine protease inhibitor. Plasma clearance was very rapid, and the brain concentration indicated efficient brain delivery of ANA-TA9 via nasal application. Cell viability analysis indicated that ANA-TA9 did not display toxicity. DISCUSSION ANA-TA9 is an attractive potential candidate for the development of novel peptide drugs in Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Yusuke Hatakawa
- Pharmaceutical TechnologyKobe Pharmaceutical UniversityHigashinadaKobeJapan
| | - Rina Nakamura
- O‐Force Co., LtdHata‐gunKochiJapan
- Laboratory of PharmacologySchool of MedicineKohasuOko‐choKochi UniversityNankokuKochiJapan
| | - Motomi Konishi
- Department of Integrative Pharmaceutical ScienceFaculty of Pharmaceutical SciencesSetsunan UniversityHirakataOsakaJapan
| | - Toshiyasu Sakane
- Pharmaceutical TechnologyKobe Pharmaceutical UniversityHigashinadaKobeJapan
| | - Akiko Tanaka
- Pharmaceutical TechnologyKobe Pharmaceutical UniversityHigashinadaKobeJapan
| | - Akira Matsuda
- Laboratory of Medicinal and Biochemical AnalysisFaculty of Pharmaceutical SciencesHiroshima International UniversityKureHiroshimaJapan
| | - Motoaki Saito
- Laboratory of PharmacologySchool of MedicineKohasuOko‐choKochi UniversityNankokuKochiJapan
| | - Toshifumi Akizawa
- O‐Force Co., LtdHata‐gunKochiJapan
- Laboratory of PharmacologySchool of MedicineKohasuOko‐choKochi UniversityNankokuKochiJapan
| |
Collapse
|
36
|
Munesue SI, Liang M, Harashima A, Zhong J, Furuhara K, Boitsova EB, Cherepanov SM, Gerasimenko M, Yuhi T, Yamamoto Y, Higashida H. Transport of oxytocin to the brain after peripheral administration by membrane-bound or soluble forms of receptors for advanced glycation end-products. J Neuroendocrinol 2021; 33:e12963. [PMID: 33733541 DOI: 10.1111/jne.12963] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/22/2022]
Abstract
Oxytocin (OT) is a neuropeptide hormone. Single and repetitive administration of OT increases social interaction and maternal behaviour in humans and mammals. Recently, it was found that the receptor for advanced glycation end-products (RAGE) is an OT-binding protein and plays a critical role in the uptake of OT to the brain after peripheral OT administration. Here, we address some unanswered questions on RAGE-dependent OT transport. First, we found that, after intranasal OT administration, the OT concentration increased in the extracellular space of the medial prefrontal cortex (mPFC) of wild-type male mice, as measured by push-pull microperfusion. No increase of OT in the mPFC was observed in RAGE knockout male mice. Second, in a reconstituted in vitro blood-brain barrier system, inclusion of the soluble form of RAGE (endogenous secretory RAGE [esRAGE]), an alternative splicing variant, in the luminal (blood) side had no effect on the transport of OT to the abluminal (brain) chamber. Third, OT concentrations in the cerebrospinal fluid after i.p. OT injection were slightly higher in male mice overexpressing esRAGE (esRAGE transgenic) compared to those in wild-type male mice, although this did not reach statistical significance. Although more extensive confirmation is necessary because of the small number of experiments in the present study, the reported data support the hypothesis that RAGE may be involved in the transport of OT to the mPFC from the circulation. These results suggest that the soluble form of RAGE in the plasma does not function as a decoy in vitro.
Collapse
Affiliation(s)
- Sei-Ichi Munesue
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - MingKun Liang
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Ai Harashima
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Jing Zhong
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Kazumi Furuhara
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Elizabeta B Boitsova
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
- Laboratory for Social Brain Studies, Research Institute of Molecular Medicine and Pathobiochemistry, Department of Biochemistry, Krasnoyarsk State Medical University named after Prof. V. F. Voino-Yasentsky, Krasnoyarsk, Russia
| | - Stanislav M Cherepanov
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Maria Gerasimenko
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Teruko Yuhi
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Haruhiro Higashida
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University, Kanazawa, Japan
- Laboratory for Social Brain Studies, Research Institute of Molecular Medicine and Pathobiochemistry, Department of Biochemistry, Krasnoyarsk State Medical University named after Prof. V. F. Voino-Yasentsky, Krasnoyarsk, Russia
| |
Collapse
|
37
|
Dalvi AV, Ravi PR, Uppuluri CT, Mahajan RR, Katke SV, Deshpande VS. Thermosensitive nasal in situ gelling systems of rufinamide formulated using modified tamarind seed xyloglucan for direct nose-to-brain delivery: design, physical characterization, and in vivo evaluation. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021. [DOI: 10.1007/s40005-020-00505-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
38
|
Fukuda M, Kanazawa T, Iioka S, Oguma T, Iwasa R, Masuoka S, Suzuki N, Kosuge Y, Suzuki T. Quantitative analysis of inulin distribution in the brain focused on nose-to-brain route via olfactory epithelium by reverse esophageal cannulation. J Control Release 2021; 332:493-501. [PMID: 33647429 DOI: 10.1016/j.jconrel.2021.02.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/15/2022]
Abstract
This study aimed to determine the effect of intranasal dosing speed and administrating volume of nose-to-brain delivery on candidates for peptide drugs (molecular weight ca. 1-10 kDa). Using inulin as the model molecule of a peptide drug, intranasal administration by cannulation from the airway side through the esophagus was tested in mice. This was done to determine the quantitative distribution levels of the drug in the brain and cerebral spinal fluid (CSF). Distribution levels were increased with slower and constant speed (5 μL/min), with higher dosing volume equivalent to nasal volume per body weight in mice (25 μL), and were recorded 0.27% injected dose per gram of tissue (ID/g) in the brain, and 0.24% injected dose per milliliter (ID/mL) in the CSF at 60 min. Then, brain distribution resulting from reverse cannulation was two times more than that of the typical intranasal administration method using a micropipette. In addition, the percentage of inulin estimated to reach the brain via direct transport (%DTP) during reverse cannulation was estimated to be 93%, suggesting that ~95% of the total dose was transferred directly to the brain via the olfactory mucosa. These results show that distribution of the peptide drug in the brain was increased through constant administration at a slow and constant speed.
Collapse
Affiliation(s)
- Mitsuyoshi Fukuda
- Laboratory of Pharmaceutics, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Takanori Kanazawa
- Laboratory of Pharmaceutics, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan; Department of Pharmaceutical Engineering and Drug Delivery Sciences, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Shingo Iioka
- Laboratory of Pharmaceutics, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Takayuki Oguma
- Laboratory of Pharmaceutics, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Ryohei Iwasa
- Laboratory of Pharmaceutics, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Saki Masuoka
- Laboratory of Pharmaceutics, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Naoto Suzuki
- Laboratory of Pharmaceutics, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Yasuhiro Kosuge
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Toyofumi Suzuki
- Laboratory of Pharmaceutics, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan.
| |
Collapse
|
39
|
Che X, Cai J, Liu Y, Xu T, Yang J, Wu C. Oxytocin signaling in the treatment of drug addiction: Therapeutic opportunities and challenges. Pharmacol Ther 2021; 223:107820. [PMID: 33600854 DOI: 10.1016/j.pharmthera.2021.107820] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 12/13/2022]
Abstract
Drug addiction is one of the leading causes of mortality worldwide. Despite great advances were achieved in understanding the neurobiology of drug addiction, the therapeutic options are severely limited, with poor effectiveness and serious side effects. The neuropeptide oxytocin (OXT) is well known for its effects on uterine contraction, sexual/maternal behaviors, social affiliation, stress and learning/memory by interacting with the OXT receptor and other neuromodulators. Emerging evidence suggests that the acute or chronic exposure to drugs can affect the OXT system. Additionally, OXT administration can ameliorate a wide range of abused drug-induced neurobehavioral changes. Overall, OXT not only suppresses drug reward in the binge stage of drug addiction, but also reduces stress responses and social impairments during the withdrawal stage and, finally, prevents drug/cue/stress-induced reinstatement. More importantly, clinical studies have also shown that OXT can exert beneficial effects on reducing substance use disorders of a series of drugs, such as heroin, cocaine, alcohol, cannabis and nicotine. Thus, the present review focuses on the role of OXT in treating drug addiction, including the preclinical and clinical therapeutic potential of OXT and its analogs on the neurobiological perspectives of drugs, to provide a better insight of the efficacy of OXT as a clinical addiction therapeutic agent.
Collapse
Affiliation(s)
- Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Tianyu Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China.
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China.
| |
Collapse
|
40
|
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
|
41
|
Quintana DS, Lischke A, Grace S, Scheele D, Ma Y, Becker B. Advances in the field of intranasal oxytocin research: lessons learned and future directions for clinical research. Mol Psychiatry 2021; 26:80-91. [PMID: 32807845 PMCID: PMC7815514 DOI: 10.1038/s41380-020-00864-7] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 07/16/2020] [Accepted: 08/05/2020] [Indexed: 01/01/2023]
Abstract
Reports on the modulatory role of the neuropeptide oxytocin on social cognition and behavior have steadily increased over the last two decades, stimulating considerable interest in its psychiatric application. Basic and clinical research in humans primarily employs intranasal application protocols. This approach assumes that intranasal administration increases oxytocin levels in the central nervous system via a direct nose-to-brain route, which in turn acts upon centrally-located oxytocin receptors to exert its behavioral effects. However, debates have emerged on whether intranasally administered oxytocin enters the brain via the nose-to-brain route and whether this route leads to functionally relevant increases in central oxytocin levels. In this review we outline recent advances from human and animal research that provide converging evidence for functionally relevant effects of the intranasal oxytocin administration route, suggesting that direct nose-to-brain delivery underlies the behavioral effects of oxytocin on social cognition and behavior. Moreover, advances in previously debated methodological issues, such as pre-registration, reproducibility, statistical power, interpretation of non-significant results, dosage, and sex differences are discussed and integrated with suggestions for the next steps in translating intranasal oxytocin into psychiatric applications.
Collapse
Affiliation(s)
- Daniel S Quintana
- Norwegian Centre for Mental Disorders Research (NORMENT), University of Oslo and Oslo University Hospital, Oslo, Norway.
| | - Alexander Lischke
- Department of Psychology, University of Greifswald, Greifswald, Germany
| | - Sally Grace
- School of Psychology, Australian Catholic University, Melbourne, Australia
| | - Dirk Scheele
- Division of Medical Psychology, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
- Department of Psychiatry, School of Medicine & Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Yina Ma
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
| | - Benjamin Becker
- The Clinical Hospital of the Chengdu Brain Science Institute, Key Laboratory for NeuroInformation, School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
42
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
43
|
Brain and Nasal Cavity Anatomy of the Cynomolgus Monkey: Species Differences from the Viewpoint of Direct Delivery from the Nose to the Brain. Pharmaceutics 2020; 12:pharmaceutics12121227. [PMID: 33352847 PMCID: PMC7766477 DOI: 10.3390/pharmaceutics12121227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 11/17/2022] Open
Abstract
Based on structural data on the nasal cavity and brain of the cynomolgus monkey, species differences in the olfactory bulb and cribriform plate were discussed from the viewpoint of direct delivery from the nose to the brain. Structural 3D data on the cynomolgus monkey skull were obtained using X-ray computed tomography. The dimensions of the nasal cavity of the cynomolgus monkey were 5 mm width × 20 mm height × 60 mm depth. The nasal cavity was very narrow and the olfactory region was far from the nostrils, similar to rats and humans. The weight and size of the monkey brain were 70 g and 55 mm width × 40 mm height × 70 mm depth. The olfactory bulb of monkeys is plate-like, while that of humans and rats is bulbar, suggesting that the olfactory area connected with the brain of monkeys is narrow. Although the structure of the monkey nasal cavity is similar to that of humans, the size and shape of the olfactory bulb are different, which is likely to result in low estimation of direct delivery from the nose to the brain in monkeys.
Collapse
|
44
|
Chen Y, Li Q, Zhang Q, Kou J, Zhang Y, Cui H, Wernicke J, Montag C, Becker B, Kendrick KM, Yao S. The Effects of Intranasal Oxytocin on Neural and Behavioral Responses to Social Touch in the Form of Massage. Front Neurosci 2020; 14:589878. [PMID: 33343285 PMCID: PMC7746800 DOI: 10.3389/fnins.2020.589878] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Manually-administered massage can potently increase endogenous oxytocin concentrations and neural activity in social cognition and reward regions and intranasal oxytocin can increase the pleasantness of social touch. In the present study, we investigated whether intranasal oxytocin modulates behavioral and neural responses to foot massage applied manually or by machine using a randomized placebo-controlled within-subject pharmaco-fMRI design. 46 male participants underwent blocks of massage of each type where they both received and imagined receiving the massage. Intranasal oxytocin significantly increased subjective pleasantness ratings of the manual but not the machine massage and neural responses in key regions involved in reward (orbitofrontal cortex, dorsal striatum and ventral tegmental area), social cognition (superior temporal sulcus and inferior parietal lobule), emotion and salience (amygdala and anterior cingulate and insula) and default mode networks (medial prefrontal cortex, parahippocampal gyrus, posterior cingulate, and precuneus) as well as a number of sensory and motor processing regions. Both neural and behavioral effects of oxytocin occurred independent of whether subjects thought the massage was applied by a male or female masseur. These findings support the importance of oxytocin for enhancing positive behavioral and neural responses to social touch in the form of manually administered massage and that a combination of intranasal oxytocin and massage may have therapeutic potential in autism. CLINICAL TRIALS REGISTRATION The Effects of Oxytocin on Social Touch; registration ID: NCT03278860; URL: https://clinicaltrials.gov/ct2/show/NCT03278860.
Collapse
Affiliation(s)
- Yuanshu Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qin Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qianqian Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Juan Kou
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yingying Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Han Cui
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jennifer Wernicke
- Department of Molecular Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany
| | - Christian Montag
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Molecular Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany
| | - Benjamin Becker
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Keith M. Kendrick
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shuxia Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
45
|
Inoue D, Furubayashi T, Tanaka A, Sakane T, Sugano K. Effect of Cerebrospinal Fluid Circulation on Nose-to-Brain Direct Delivery and Distribution of Caffeine in Rats. Mol Pharm 2020; 17:4067-4076. [PMID: 32955898 DOI: 10.1021/acs.molpharmaceut.0c00495] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Direct drug delivery from nose to brain has drawn much attention as an effective strategy for the treatment of central nervous system diseases. After intranasal administration, drug molecules can be directly delivered from the nose to the brain. However, the detailed mechanism for this direct delivery to the brain has not been elucidated. In the present study, the effect of the activation of the cerebral fluid circulation (the glymphatic system) on the efficacy of direct delivery from nose to brain was investigated. Because the glymphatic system is activated by some anesthetic regimens, the differences in brain delivery and the pharmacokinetics under anesthetic and conscious conditions were compared in rats. Under urethane anesthesia, direct delivery from the nose to the brain was facilitated, whereas the brain uptake from the systemic circulation via the blood-brain barrier was decreased. In addition, both the brain uptake of caffeine injected into the subarachnoid cerebrospinal fluid (CSF) and the extracerebral clearance of caffeine after intrastriatal injection were enhanced under anesthesia. For intranasal administration, caffeine was transported directly from the nose to the CSF and then delivered into the brain parenchyma by the CSF circulation. The results obtained in the present study clarified that the direct delivery from nose to brain could be facilitated by anesthesia. These findings suggest that fluid circulation in the brain can contribute to a wider cerebral distribution of the drug after direct delivery from nose to brain.
Collapse
Affiliation(s)
- Daisuke Inoue
- Molecular Pharmaceutics Laboratory, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan.,Department of Pharmaceutics, School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama 703-8516, Japan
| | - Tomoyuki Furubayashi
- Department of Pharmaceutics, School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama 703-8516, Japan.,Laboratory of Pharmaceutical Technology, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada, Kobe 658-8558, Japan
| | - Akiko Tanaka
- Laboratory of Pharmaceutical Technology, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada, Kobe 658-8558, Japan
| | - Toshiyasu Sakane
- Laboratory of Pharmaceutical Technology, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada, Kobe 658-8558, Japan
| | - Kiyohiko Sugano
- Molecular Pharmaceutics Laboratory, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| |
Collapse
|
46
|
Oxytocin Reduces Brain Injury and Maintains Blood–Brain Barrier Integrity After Ischemic Stroke in Mice. Neuromolecular Med 2020; 22:557-571. [DOI: 10.1007/s12017-020-08613-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/29/2020] [Indexed: 12/16/2022]
|
47
|
Maejima Y, Horita S, Otsuka A, Hidema S, Nishimori K, Shimomura K. Oral oxytocin delivery with proton pump inhibitor pretreatment decreases food intake. Peptides 2020; 128:170312. [PMID: 32298773 DOI: 10.1016/j.peptides.2020.170312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/25/2022]
Abstract
Oxytocin (Oxt) is considered as a potential agent to treat multiple neuropsychiatric disorders, obesity and metabolic syndrome. Although the mechanisms underlying these effects remain unclear, nasal administration is considered to be a potential way to deliver Oxt into blood vessels. The development of an easier, more stable and efficient way is expected. A recent study demonstrated that orally administered Oxt can be transmitted into blood if it is prevented from degradation in stomach and reaches the intestinal tract. In this study, we pretreated mice with a proton pump inhibitor (PPI), omeprazole (20 mg/kg), and administered capsulized Oxt (0.25 mg), so that the Oxt can be prevented from degradation by pepsin due to the low pH in stomach and reach the intestinal tract. Functionally, these mice showed a similar decrease in food intake to those who underwent intraperitoneal administration. We also confirmed that this method dramatically increased plasma Oxt levels and the expression of neural activation marker c-Fos protein in the paraventricular and suprachiasmatic nucleus. Our study showed that by pretreating mice with PPI, Oxt in a gelatin-coated capsule can prevent Oxt from degradation by pepsin in stomach, and reach the bloodstream in an effective concentration. These results indicate that our method is a promising oral delivery of Oxt and should be investigated further for other peptide agents based on peripheral injection or nasal administration.
Collapse
Affiliation(s)
- Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 960-1295, Fukushima, Japan.
| | - Shoichiro Horita
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 960-1295, Fukushima, Japan
| | - Ayano Otsuka
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University Sendai-shi, 981-8555, Miyagi, Japan
| | - Shizu Hidema
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University Sendai-shi, 981-8555, Miyagi, Japan
| | - Katsuhiko Nishimori
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University Sendai-shi, 981-8555, Miyagi, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 960-1295, Fukushima, Japan
| |
Collapse
|
48
|
Lefter R, Ciobica A, Antioch I, Ababei DC, Hritcu L, Luca AC. Oxytocin Differentiated Effects According to the Administration Route in a Prenatal Valproic Acid-Induced Rat Model of Autism. ACTA ACUST UNITED AC 2020; 56:medicina56060267. [PMID: 32485966 PMCID: PMC7353871 DOI: 10.3390/medicina56060267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/14/2020] [Accepted: 05/25/2020] [Indexed: 12/31/2022]
Abstract
Background and objectives: The hormone oxytocin (OXT) has already been reported in both human and animal studies for its promising therapeutic potential in autism spectrum disorder (ASD), but the comparative effectiveness of various administration routes, whether central or peripheral has been insufficiently studied. In the present study, we examined the effects of intranasal (IN) vs. intraperitoneal (IP) oxytocin in a valproic-acid (VPA) autistic rat model, focusing on cognitive and mood behavioral disturbances, gastrointestinal transit and central oxidative stress status. Materials and Methods: VPA prenatally-exposed rats (500 mg/kg; age 90 days) in small groups of 5 (n = 20 total) were given OXT by IP injection (10 mg/kg) for 8 days consecutively or by an adapted IN pipetting protocol (12 IU/kg, 20 μL/day) for 4 consecutive days. Behavioral tests were performed during the last three days of OXT treatment, and OXT was administrated 20 minutes before each behavioral testing for each rat. Biochemical determination of oxidative stress markers in the temporal area included superoxide dismutase (SOD), glutathione peroxidase (GPx) and malondialdehyde (MDA). A brief quantitative assessment of fecal discharge over a period of 24 hours was performed at the end of the OXT treatment to determine differences in intestinal transit. Results: OXT improved behavioral and oxidative stress status in both routes of administration, but IN treatment had significantly better outcome in improving short-term memory, alleviating depressive manifestations and mitigating lipid peroxidation in the temporal lobes. Significant correlations were also found between behavioral parameters and oxidative stress status in rats after OXT administration. The quantitative evaluation of the gastrointestinal (GI) transit indicated lower fecal pellet counts in the VPA group and homogenous average values for the control and both OXT treated groups. Conclusions: The data from the present study suggest OXT IN administration to be more efficient than IP injections in alleviating autistic cognitive and mood dysfunctions in a VPA-induced rat model. OXT effects on the cognitive and mood behavior of autistic rats may be associated with its effects on oxidative stress. Additionally, present results provide preliminary evidence that OXT may have a balancing effect on gastrointestinal motility.
Collapse
Affiliation(s)
- Radu Lefter
- Center of Biomedical Research, Romanian Academy, B dul Carol I, No 8, 700505 Iasi, Romania;
| | - Alin Ciobica
- Department of Research, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, No 11, 700506 Iasi, Romania;
- Correspondence: (A.C.); (L.H.)
| | - Iulia Antioch
- Department of Research, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, No 11, 700506 Iasi, Romania;
| | - Daniela Carmen Ababei
- “Grigore T.Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.C.A.); (A.-C.L.)
| | - Luminita Hritcu
- Faculty of Veterinary Medicine, University of Agricultural Sciencies and Veterinary Medicine “Ion Ionescu de la Brad” of Iasi, 3rd Mihail Sadoveanu Alley, 700490 Iasi, Romania
- Correspondence: (A.C.); (L.H.)
| | - Alina-Costina Luca
- “Grigore T.Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.C.A.); (A.-C.L.)
| |
Collapse
|
49
|
Islam SU, Shehzad A, Ahmed MB, Lee YS. Intranasal Delivery of Nanoformulations: A Potential Way of Treatment for Neurological Disorders. Molecules 2020; 25:molecules25081929. [PMID: 32326318 PMCID: PMC7221820 DOI: 10.3390/molecules25081929] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/17/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Although the global prevalence of neurological disorders such as Parkinson’s disease, Alzheimer’s disease, glioblastoma, epilepsy, and multiple sclerosis is steadily increasing, effective delivery of drug molecules in therapeutic quantities to the central nervous system (CNS) is still lacking. The blood brain barrier (BBB) is the major obstacle for the entry of drugs into the brain, as it comprises a tight layer of endothelial cells surrounded by astrocyte foot processes that limit drugs’ entry. In recent times, intranasal drug delivery has emerged as a reliable method to bypass the BBB and treat neurological diseases. The intranasal route for drug delivery to the brain with both solution and particulate formulations has been demonstrated repeatedly in preclinical models, including in human trials. The key features determining the efficacy of drug delivery via the intranasal route include delivery to the olfactory area of the nares, a longer retention time at the nasal mucosal surface, enhanced penetration of the drugs through the nasal epithelia, and reduced drug metabolism in the nasal cavity. This review describes important neurological disorders, challenges in drug delivery to the disordered CNS, and new nasal delivery techniques designed to overcome these challenges and facilitate more efficient and targeted drug delivery. The potential for treatment possibilities with intranasal transfer of drugs will increase with the development of more effective formulations and delivery devices.
Collapse
Affiliation(s)
- Salman Ul Islam
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (S.U.I.); (M.B.A.)
| | - Adeeb Shehzad
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Muhammad Bilal Ahmed
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (S.U.I.); (M.B.A.)
| | - Young Sup Lee
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (S.U.I.); (M.B.A.)
- Correspondence: ; Tel.: +82-53-950-6353; Fax: +82-53-943-2762
| |
Collapse
|
50
|
Romano A, Friuli M, Cifani C, Gaetani S. Oxytocin in the neural control of eating: At the crossroad between homeostatic and non-homeostatic signals. Neuropharmacology 2020; 171:108082. [PMID: 32259527 DOI: 10.1016/j.neuropharm.2020.108082] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 12/21/2022]
Abstract
The understanding of the biological substrates regulating feeding behavior is relevant to address the health problems related to food overconsumption. Several studies have expanded the conventional view of the homeostatic regulation of body weight mainly orchestrated by the hypothalamus, to include also the non-homeostatic control of appetite. Such processes include food reward and are mainly coordinated by the activation of the central mesolimbic dopaminergic pathway. The identification of endogenous systems acting as a bridge between homoeostatic and non-homeostatic pathways might represent a significant step toward the development of drugs for the treatment of aberrant eating patterns. Oxytocin is a hypothalamic hormone that is directly secreted into the brain and reaches the blood circulation through the neurohypophysis. Oxytocin regulates a variety of physiologic functions, including eating and metabolism. In the last years both preclinical and clinical studies well characterized oxytocin for its effects in reducing food intake and body weight. In the present review we summarize the role played by oxytocin in the control of both homeostatic and non-homeostatic eating, within cognitive, metabolic and reward mechanisms, to mostly highlight its potential therapeutic effects as a new pharmacological approach for the development of drugs for eating disorders. We conclude that the central oxytocinergic system is possibly one of the mechanisms that coordinate energy balance at the crossroads between homeostatic and non-homeostatic mechanisms. This concept should foster studies aimed at exploring the possible exploitation of oxytocin in the treatment of aberrant eating patterns. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Marzia Friuli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 9, 62032, Camerino, MC, Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|