1
|
Neitzke EV, Dos Santos FG, Zanini BM, Cavalcante MB, Mason JB, Masternak MM, de Souza ICC, Schneider A. The influence of ovarian activity and menopause on mental health: Evidence from animal models and women. Physiol Behav 2025; 294:114886. [PMID: 40118132 DOI: 10.1016/j.physbeh.2025.114886] [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: 01/09/2025] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Hormonal variations occurring throughout the female reproductive cycle have a significant impact on physical and mental health, particularly due to the influence of estradiol (E2) and progesterone (P4). These changes are directly related to alterations in neurological systems, being associated with conditions such as premenstrual syndrome (PMS), premenstrual dysphoric disorder (PMDD), and mood disorders during hormonal transition phases, such as perimenopause and menopause. Studies conducted in humans and animal models indicate that these fluctuations affect neurotransmitters, neural plasticity, and patterns of brain activity, ultimately influencing quality of life and mental health. Despite extensive research on the topic, the interactions between sex hormones, mental health, and reproductive aging still require further investigation, emphasizing approaches that simultaneously address experimental and behavioral aspects. Thus, this review aims to sumarize findings about the influence of hormonal fluctuations throughout the female reproductive lifespan, including transitions such as perimenopause and menopause, on mental health. A comparative analysis of data from studies in animal models and humans was conducted, highlighting neuroendocrine, behavioral, and emotional mechanisms associated with hormonal changes and their impacts on female mental health.
Collapse
Affiliation(s)
- Ediana V Neitzke
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | | | - Bianka M Zanini
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | - Marcelo B Cavalcante
- Postgraduate Program in Medical Sciences, University of Fortaleza (UNIFOR), Fortaleza, CE, Brazil
| | - Jeffrey B Mason
- College of Veterinary Medicine, Department of Veterinary Clinical and Life Sciences, Center for Integrated BioSystems, Utah State University, Logan, UT, USA
| | - Michal M Masternak
- University of Central Florida, College of Medicine, Burnett School of Biomedical Sciences, Orlando, Florida, USA; Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Izabel C C de Souza
- Instituto de Biologia, Departamento de Morfologia, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas RS, Brazil.
| |
Collapse
|
2
|
Zhang K, He L, Li Z, Ding R, Han X, Chen B, Cao G, Ye JH, Li T, Fu R. Bridging Neurobiological Insights and Clinical Biomarkers in Postpartum Depression: A Narrative Review. Int J Mol Sci 2024; 25:8835. [PMID: 39201521 PMCID: PMC11354679 DOI: 10.3390/ijms25168835] [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: 07/27/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Postpartum depression (PPD) affects 174 million women worldwide and is characterized by profound sadness, anxiety, irritability, and debilitating fatigue, which disrupt maternal caregiving and the mother-infant relationship. Limited pharmacological interventions are currently available. Our understanding of the neurobiological pathophysiology of PPD remains incomplete, potentially hindering the development of novel treatment strategies. Recent hypotheses suggest that PPD is driven by a complex interplay of hormonal changes, neurotransmitter imbalances, inflammation, genetic factors, psychosocial stressors, and hypothalamic-pituitary-adrenal (HPA) axis dysregulation. This narrative review examines recent clinical studies on PPD within the past 15 years, emphasizing advancements in neuroimaging findings and blood biomarker detection. Additionally, we summarize recent laboratory work using animal models to mimic PPD, focusing on hormone withdrawal, HPA axis dysfunction, and perinatal stress theories. We also revisit neurobiological results from several brain regions associated with negative emotions, such as the amygdala, prefrontal cortex, hippocampus, and striatum. These insights aim to improve our understanding of PPD's neurobiological mechanisms, guiding future research for better early detection, prevention, and personalized treatment strategies for women affected by PPD and their families.
Collapse
Affiliation(s)
- Keyi Zhang
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Lingxuan He
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Zhuoen Li
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Ruxuan Ding
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Xiaojiao Han
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Bingqing Chen
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Guoxin Cao
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ 07103, USA;
| | - Tian Li
- Department of Gynecology and Obstetrics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Rao Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| |
Collapse
|
3
|
Acosta MC, Hussein M, Saltzman W. Effects of acute inhibition of dopamine β-hydroxylase on neural responses to pups in adult virgin male California mice (Peromyscus californicus). Behav Brain Res 2024; 471:115116. [PMID: 38897419 DOI: 10.1016/j.bbr.2024.115116] [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: 03/22/2024] [Revised: 06/09/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
The neural mechanisms underlying paternal care in biparental mammals are not well understood. The California mouse (Peromyscus californicus) is a biparental rodent in which virtually all fathers are attracted to pups, while virgin males vary widely in their behavior toward unrelated infants, ranging from attacking to avoiding to huddling and grooming pups. We previously showed that pharmacologically inhibiting the synthesis of the neurotransmitter norepinephrine (NE) with the dopamine β-hydroxylase inhibitor nepicastat reduced the propensity of virgin male and female California mice to interact with pups. The current study tested the hypothesis that nepicastat would reduce pup-induced c-Fos immunoreactivity, a cellular marker of neural activity, in the medial preoptic area (MPOA), medial amygdala (MeA), basolateral amygdala (BLA), and bed nucleus of the stria terminalis (BNST), brain regions implicated in the control of parental behavior and/or anxiety. Virgin males were injected with nepicastat (75 mg/kg, i.p.) or vehicle 2 hours prior to exposure to either an unrelated pup or novel object for 60 minutes (n = 4-6 mice per group). Immediately following the 60-minute stimulus exposure, mice were euthanized and their brains were collected for c-Fos immunohistochemistry. Nepicastat reduced c-Fos expression in the MeA and MPOA of pup-exposed virgin males compared to vehicle-injected controls. In contrast, nepicastat did not alter c-Fos expression in any of the above brain regions following exposure to a novel object. Overall, these results suggest that the noradrenergic system might influence MeA and MPOA function to promote behavioral interactions with pups in virgin males.
Collapse
Affiliation(s)
- Melina C Acosta
- Graduate Program in Neuroscience, University of California, Riverside, CA 92521, USA; Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, CA 92521, USA
| | - Manal Hussein
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, CA 92521, USA
| | - Wendy Saltzman
- Graduate Program in Neuroscience, University of California, Riverside, CA 92521, USA; Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
4
|
Xie H, Xie Z, Luan F, Zeng J, Zhang X, Chen L, Zeng N, Liu R. Potential therapeutic effects of Chinese herbal medicine in postpartum depression: Mechanisms and future directions. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117785. [PMID: 38262525 DOI: 10.1016/j.jep.2024.117785] [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: 07/02/2023] [Revised: 11/15/2023] [Accepted: 01/15/2024] [Indexed: 01/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Postpartum depression (PPD) is a common psychiatric disorder in women after childbirth. Per data from epidemiologic studies, PPD affects about 5%-26.32% of postpartum mothers worldwide. Biological factors underlying this condition are multiple and complex and have received extensive inquiries for the roles they play in PPD. Chinese herbal medicine (CHM), which is widely used as a complementary and alternative therapy for neurological disorders, possesses multi-component, multi-target, multi-access, and low side effect therapeutic characteristics. CHM has already shown efficacy in the treatment of PPD, and a lot more research exploring the mechanisms of its potential therapeutic effects is being conducted. AIM OF THE REVIEW This review provides an in-depth and comprehensive overview of the underlying mechanisms of PPD, as well as samples the progress made in researching the potential role of CHM in treating the disorder. MATERIALS AND METHODS Literature was searched comprehensively in scholarly electronic databases, including PubMed, Web of Science, Scopus, CNKI and WanFang DATA, using the search terms "postpartum depression", "genetic", "hormone", "immune", "neuroinflammation", "inflammation", "neurotransmitter", "neurogenesis", "brain-gut axis", "traditional Chinese medicine", "Chinese herbal medicine", "herb", and an assorted combination of these terms. RESULTS PPD is closely associated with genetics, as well as with the hormones, immune inflammatory, and neurotransmitter systems, neurogenesis, and gut microbes, and these biological factors often interact and work together to cause PPD. For example, inflammatory factors could suppress the production of the neurotransmitter serotonin by inducing the regulation of tryptophan-kynurenine in the direction of neurotoxicity. Many CHM constituents improve anxiety- and depression-like behaviors by interfering with the above-mentioned mechanisms and have shown decent efficacy clinically against PPD. For example, Shen-Qi-Jie-Yu-Fang invigorates the neuroendocrine system by boosting the hormone levels of hypothalamic pituitary adrenal (HPA) and hypothalamic pituitary gonadal (HPG) axes, regulating the imbalance of Treg/T-helper cells (Th) 17 and Th1/Th2, and modulating neurotransmitter system to play antidepressant roles. The Shenguiren Mixture interferes with the extracellular signal-regulated kinase (ERK) pathway to enhance the number, morphology and apoptosis of neurons in the hippocampus of PPD rats. Other herbal extracts and active ingredients of CHM, such as Paeoniflorin, hypericin, timosaponin B-III and more, also manage depression by remedying the neuroendocrine system and reducing neuroinflammation. CONCLUSIONS The pathogenesis of PPD is complex and diverse, with the main pathogenesis not clear. Still, CHM constituents, like Shen-Qi-Jie-Yu-Fang, the Shenguiren Mixture, Paeoniflorin, hypericin and other Chinese Medicinal Formulae, active monomers and Crude extracts, treats PPD through multifaceted interventions. Therefore, developing more CHM components for the treatment of PPD is an essential step forward.
Collapse
Affiliation(s)
- Hongxiao Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China.
| | - Zhiqiang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China.
| | - Fei Luan
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, PR China.
| | - Jiuseng Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China.
| | - Xiumeng Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China.
| | - Li Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China; Department of Pharmacy, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, PR China.
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China.
| | - Rong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China.
| |
Collapse
|
5
|
Chen K, Yang J, Li F, Chen J, Chen M, Shao H, He C, Cai D, Zhang X, Wang L, Luo Y, Cheng B, Wang J. Molecular basis underlying default mode network functional abnormalities in postpartum depression with and without anxiety. Hum Brain Mapp 2024; 45:e26657. [PMID: 38544486 PMCID: PMC10973776 DOI: 10.1002/hbm.26657] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/04/2024] [Accepted: 02/27/2024] [Indexed: 11/12/2024] Open
Abstract
Although Postpartum depression (PPD) and PPD with anxiety (PPD-A) have been well characterized as functional disruptions within or between multiple brain systems, however, how to quantitatively delineate brain functional system irregularity and the molecular basis of functional abnormalities in PPD and PPD-A remains unclear. Here, brain sample entropy (SampEn), resting-state functional connectivity (RSFC), transcriptomic and neurotransmitter density data were used to investigate brain functional system irregularity, functional connectivity abnormalities and associated molecular basis for PPD and PPD-A. PPD-A exhibited higher SampEn in medial prefrontal cortex (MPFC) and posterior cingulate cortex (PPC) than healthy postnatal women (HPW) and PPD while PPD showed lower SampEn in PPC compared to HPW and PPD-A. The functional connectivity analysis with MPFC and PPC as seed areas revealed decreased functional couplings between PCC and paracentral lobule and between MPFC and angular gyrus in PPD compared to both PPD-A and HPW. Moreover, abnormal SampEn and functional connectivity were associated with estrogenic level and clinical symptoms load. Importantly, spatial association analyses between functional changes and transcriptome and neurotransmitter density maps revealed that these functional changes were primarily associated with synaptic signaling, neuron projection, neurotransmitter level regulation, amino acid metabolism, cyclic adenosine monophosphate (cAMP) signaling pathways, and neurotransmitters of 5-hydroxytryptamine (5-HT), norepinephrine, glutamate, dopamine and so on. These results reveal abnormal brain entropy and functional connectivities primarily in default mode network (DMN) and link these changes to transcriptome and neurotransmitters to establish the molecular basis for PPD and PPD-A for the first time. Our findings highlight the important role of DMN in neuropathology of PPD and PPD-A.
Collapse
Affiliation(s)
- Kexuan Chen
- Faculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
- Medical SchoolKunming University of Science and TechnologyKunmingChina
| | - Jia Yang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Fang Li
- Medical SchoolKunming University of Science and TechnologyKunmingChina
| | - Jin Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Meiling Chen
- Department of Clinical Psychology, the First People's Hospital of Yunnan ProvinceThe Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Heng Shao
- Department of Geriatrics, the First People's Hospital of Yunnan ProvinceThe Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Chongjun He
- People's Hospital of Lijiangthe Affiliated Hospital of Kunming University of Science and TechnologyLijiangChina
| | - Defang Cai
- The Second People's Hospital of Yuxithe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Xing Zhang
- The Second People's Hospital of Yuxithe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Libo Wang
- The Second People's Hospital of Yuxithe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Yuejia Luo
- Medical SchoolKunming University of Science and TechnologyKunmingChina
- Center for Brain Disorders and Cognitive Sciences, School of PsychologyShenzhen UniversityShenzhenChina
- The State Key Lab of Cognitive and Learning, Faculty of PsychologyBeijing Normal UniversityBeijingChina
| | - Bochao Cheng
- Department of RadiologyWest China Second University Hospital of Sichuan UniversityChengduChina
| | - Jiaojian Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| |
Collapse
|
6
|
Zubkov E, Riabova O, Zorkina Y, Egorova A, Ushakova V, Lepioshkin A, Novoselova E, Abramova O, Morozova A, Chekhonin V, Makarov V. Antidepressant-like Effect of the Eburnamine-Type Molecule Vindeburnol in Rat and Mouse Models of Ultrasound-Induced Depression. ACS Chem Neurosci 2024; 15:560-571. [PMID: 38216514 DOI: 10.1021/acschemneuro.3c00590] [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] [Indexed: 01/14/2024] Open
Abstract
Vindeburnol (VIND, RU24722, BC19), a synthetic molecule derived from the eburnamine-vincamine alkaloid group, has many neuropsychopharmacological effects, but its antidepressant-like effects are poorly understood and have only been described in a few patents. To reliably estimate this effect, vindeburnol was studied in a model of long-term variable-frequency ultrasound (US) exposure at 20-45 kHz in male Wistar rats and BALB/c mice. Vindeburnol was administered chronically for 21 days against a background of simultaneous ultrasound exposure at a dose of 20 mg/kg intraperitoneally (IP). Using four behavioral tests, the sucrose preference test (SPT), the social interaction test (SIT), the open field test (OFT), and the forced swimming test (FST), we found that the treatment with the compound diminished depression-like symptoms in mice and rats. The compound restored the ultrasound-related reduced sucrose consumption to control levels and increased social interaction time in mice and rats compared with those in ultrasound-exposed animals. Vindeburnol showed contraversive results of horizontal and vertical activity in both species and generally did not increase locomotor activity. At the same time, the compound showed a specific effect in the FST, significantly reducing the immobility time. Moreover, we found an increase in norepinephrine, dopamine, and its metabolite levels in the brainstem, as well as an increase in dopamine, 3-methoxytyramine, and 3,4-dihydroxyphenylacetic acid levels in the striatum. We also observed a statistically significant increase in tyrosine hydroxylase (TH) levels in the region containing the locus coeruleus (LC). We suggest that using its distinct chemical structure and pharmacological activity as a starting point could boost antidepressant drug discovery.
Collapse
Affiliation(s)
- Eugene Zubkov
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 23 Kropotkinsky Pereulok, 119034 Moscow, Russia
| | - Olga Riabova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Yana Zorkina
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 23 Kropotkinsky Pereulok, 119034 Moscow, Russia
| | - Anna Egorova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Valeriya Ushakova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 23 Kropotkinsky Pereulok, 119034 Moscow, Russia
| | - Alexander Lepioshkin
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Elena Novoselova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Olga Abramova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 23 Kropotkinsky Pereulok, 119034 Moscow, Russia
| | - Anna Morozova
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 23 Kropotkinsky Pereulok, 119034 Moscow, Russia
| | - Vladimir Chekhonin
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 23 Kropotkinsky Pereulok, 119034 Moscow, Russia
| | - Vadim Makarov
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| |
Collapse
|
7
|
Zhao J, He Y, Duan Y, Ma Y, Dong H, Zhang X, Fang R, Zhang Y, Yu M, Huang F. HDAC6 Deficiency Has Moderate Effects on Behaviors and Parkinson's Disease Pathology in Mice. Int J Mol Sci 2023; 24:9975. [PMID: 37373121 DOI: 10.3390/ijms24129975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Histone deacetylase 6 (HDAC6) is involved in the regulation of protein aggregation and neuroinflammation, but its role in Parkinson's disease (PD) remains controversial. In this study, Hdac6-/- mice were generated by CRISPR-Cas9 technology for exploring the effect of HDAC6 on the pathological progression of PD. We found that male Hdac6-/- mice exhibit hyperactivity and certain anxiety. In the acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice, though motor injury was slightly alleviated by HDAC6 deficiency, dopamine (DA) depletion in the striatum, the decrease in the number of DA neurons in the substantia nigra (SN) and the reduction in DA neuronal terminals were not affected. In addition, activation of glial cells and the expression of α-synuclein, as well as the levels of apoptosis-related proteins in the nigrostriatal pathway, were not changed in MPTP-injected wild-type and Hdac6-/- mice. Therefore, HDAC6 deficiency leads to moderate alterations of behaviors and Parkinson's disease pathology in mice.
Collapse
Affiliation(s)
- Jiayin Zhao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yongtao He
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yufei Duan
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yuanyuan Ma
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Hongtian Dong
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Xiaoshuang Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Rong Fang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yunhe Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Fang Huang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|
8
|
Moreira LKS, Moreira CVL, Custódio CHX, Dias MLP, Rosa DA, Ferreira-Neto ML, Colombari E, Costa EA, Fajemiroye JO, Pedrino GR. Post-partum depression: From clinical understanding to preclinical assessments. Front Psychiatry 2023; 14:1173635. [PMID: 37143780 PMCID: PMC10151489 DOI: 10.3389/fpsyt.2023.1173635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023] Open
Abstract
Post-partum depression (PPD) with varying clinical manifestations affecting new parents remains underdiagnosed and poorly treated. This minireview revisits the pharmacotherapy, and relevant etiological basis, capable of advancing preclinical research frameworks. Maternal tasks accompanied by numerous behavioral readouts demand modeling different paradigms that reflect the complex and heterogenous nature of PPD. Hence, effective PPD-like characterization in animals towards the discovery of pharmacological intervention demands research that deepens our understanding of the roles of hormonal and non-hormonal components and mediators of this psychiatric disorder.
Collapse
Affiliation(s)
| | | | | | - Matheus L. P. Dias
- Institute of Biological Sciences, Federal University of Goiás, Goiania, GO, Brazil
| | - Daniel A. Rosa
- Institute of Biological Sciences, Federal University of Goiás, Goiania, GO, Brazil
| | - Marcos L. Ferreira-Neto
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Elson A. Costa
- Institute of Biological Sciences, Federal University of Goiás, Goiania, GO, Brazil
| | - James O. Fajemiroye
- Institute of Biological Sciences, Federal University of Goiás, Goiania, GO, Brazil
- Graduate Program in Pharmaceutical Sciences, Campus Arthur Wesley Archibald, Evangelical University of Goiás, Anápolis, Brazil
- *Correspondence: James O. Fajemiroye,
| | - Gustavo R. Pedrino
- Institute of Biological Sciences, Federal University of Goiás, Goiania, GO, Brazil
| |
Collapse
|
9
|
Rincón-Cortés M, Grace AA. Dopamine downregulation in novel rodent models useful for the study of postpartum depression. Front Behav Neurosci 2022; 16:1065558. [PMID: 36620861 PMCID: PMC9812956 DOI: 10.3389/fnbeh.2022.1065558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Postpartum depression (PPD) is the most common psychiatric disorder following childbirth and is characterized by maternal mood disturbances, impaired maternal responses, and disrupted caregiving- all of which negatively impact offspring development. Since PPD has detrimental consequences for both mother and child, clinical and preclinical research has focused on identifying brain changes associated with this disorder. In humans, PPD is linked to dysregulated mesolimbic dopamine (DA) system function and altered neural responses (i.e., decreased reward-related activity) to infant-related cues, which are considered hallmark features of PPD. In accordance, rodent models employing translational risk factors useful for the study of PPD have demonstrated alterations in mesolimbic DA system structure and function, and these changes are reviewed here. We also present two novel rodent models based on postpartum adversity exposure (i.e., pup removal, scarcity-adversity) which result in PPD-relevant behavioral changes (e.g., disrupted mother-infant interactions, deficits in maternal behavior, depressive-like phenotypes) and attenuated ventral tegmental area (VTA) DA neuron activity consistent with a hypodopaminergic state. Furthermore, we highlight open questions and future directions for these rodent models. In sum, human and rodent studies converge in showing blunted mesolimbic DA function (i.e., DA downregulation) in PPD. We propose that reduced activity of VTA DA neurons, resulting in downregulation of the mesolimbic DA system, interferes with reward-related processes necessary for maternal motivation and responsiveness. Thus, the mesolimbic DA system may constitute a therapeutic target for ameliorating reward-related deficits in PPD.
Collapse
Affiliation(s)
- Millie Rincón-Cortés
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States,*Correspondence: Millie Rincón-Cortés
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
10
|
Labaka A, Gómez-Lazaro E, Goñi-Balentziaga O, Pérez-Tejada J, Vegas O, Garmendia L. Venlafaxine reduces the striatal il6/il10 ratio and increases hippocampal GR expression in female mice subjected to chronic social instability stress. Stress 2021; 24:561-571. [PMID: 33769212 DOI: 10.1080/10253890.2021.1895111] [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] [Indexed: 10/21/2022] Open
Abstract
Women are twice as likely as men to develop depression and antidepressant treatment is more frequent in females. Moreover, neuroinflammatory changes related to affective disorders differ in accordance with sex. Despite this evidence, female populations have been largely omitted from preclinical experiments studying antidepressants. The aim of this work is to analyze the potential restorative effect of venlafaxine on an animal model of depression. Female CD1 mice were subjected to chronic social instability (CSI) stress for 7 weeks, and were administered venlafaxine during the last 3 weeks of the stress period. Behavioral and physiological parameters were then analyzed. Stressed mice showed a decreased sucrose preference and increased whisking behavior, and had a lower body weight, higher plasma corticosterone levels and increased hypothalamic GR expression. They also had lower levels of 5-HT, 5-HIAA and NA and a higher KYN/TRYP ratio in the hippocampus. Moreover, CSI increased striatal IL-6 mRNA expression levels. Venlafaxine treatment reduced the striatal IL-6/IL-10 ratio and increased hippocampal GR expression, although it did not reverse stress-induced behavioral changes. In conclusion, seven weeks of exposure to CSI produced depressive-like alterations in female mice. The venlafaxine treatment regimen was found to have a modest anti-inflammatory effect in the striatum and increased hippocampal GR mRNA, although it failed to redress stress-induced behavioral disturbances.
Collapse
Affiliation(s)
- Ainitze Labaka
- Department of Basic Psychological Processes and their Development, University of the Basque Country UPV/EHU, San Sebastian, Spain
- Department of Nursing II, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, San Sebastian, Spain
| | - Eneritz Gómez-Lazaro
- Department of Basic Psychological Processes and their Development, University of the Basque Country UPV/EHU, San Sebastian, Spain
| | - Olatz Goñi-Balentziaga
- Department of Basic Psychological Processes and their Development, University of the Basque Country UPV/EHU, San Sebastian, Spain
| | - Joana Pérez-Tejada
- Department of Basic Psychological Processes and their Development, University of the Basque Country UPV/EHU, San Sebastian, Spain
- Oncologic Center (Onkologikoa), San Sebastian, Spain
| | - Oscar Vegas
- Department of Basic Psychological Processes and their Development, University of the Basque Country UPV/EHU, San Sebastian, Spain
- Biodonostia Institute, San Sebastian, Spain
| | - Larraitz Garmendia
- Department of Basic Psychological Processes and their Development, University of the Basque Country UPV/EHU, San Sebastian, Spain
| |
Collapse
|
11
|
Choi Y, Yoon M. The Effects of Androstenone on the Plasma Serotonin, β-Endorphin, and Cortisol Concentrations in Thoroughbred Horses. Animals (Basel) 2021; 11:ani11061694. [PMID: 34200209 PMCID: PMC8226760 DOI: 10.3390/ani11061694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary The development of horse managing tools is needed to prevent accidents and to improve welfare for domestic horses because the safety hazards for people who are exposed to horses are well documented. Androstenone, a pheromone secreted from boars, changes the behavior of dogs to become less excited. In horses, a previous study showed that a specific receptor for androstenone was expressed in the vomeronasal organ and nasal cavity. Horses treated with androstenone also showed more compliant behaviors. Thus, this study was conducted to investigate the mechanism of androstenone for changing horse behaviors. The change in the plasma concentrations of serotonin, β-endorphin, and cortisol in response to the treatment of androstenone was evaluated using an immunoassay. The results of this study demonstrated that androstenone may control the neuroendocrine system of horses, resulting in behavioral changes. This is the first work that studies the mechanism of pheromone treatment in horses and can be applied for further study about the effect of pheromone therapy on horses. Abstract Androstenone influences the changing behaviors of animals. Previous studies discovered that an androstenone receptor was expressed in horses and treatment with androstenone induced horses to be more compliant. As changes in the level of neuroendocrine factors result in animal behavioral changes, the objective of the study was to monitor the changes in the concentrations of 5-HT, β-endorphin, and cortisol in response to androstenone. Eight thoroughbred horses (five mares and three geldings) were treated with androstenone diluted in jojoba oil (10 µg/mL) and only oil for a control cross-overly. A handler applied the treatments to the horses′ nostril and rubbed for 5 s. Blood samples were collected before, 15, 30, and 60 min after each treatment. The concentrations of each neurotransmitter were analyzed by enzyme-linked immunosorbent assay. The concentrations of each neurotransmitter after the treatment were compared to its baseline concentration. The concentration of 5-HT of the androstenone-treated horses remained consistent throughout the experiment, while the concentration of the control group significantly decreased over time. The plasma concentration of β-endorphin in the androstenone-treated group also remained constant, whereas the concentration increased in the control group. Cortisol levels did not change in either the treated or untreated groups. An androstenone treatment triggers changes in the secretion of 5-HT and β-endorphin in horses.
Collapse
|
12
|
Hagiwara A, Sugiyama N, Ohtsuka T. Impaired experience-dependent maternal care in presynaptic active zone protein CAST-deficient dams. Sci Rep 2020; 10:5238. [PMID: 32251313 PMCID: PMC7090055 DOI: 10.1038/s41598-020-62072-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/09/2020] [Indexed: 11/24/2022] Open
Abstract
Although sociological studies affirm the importance of parental care in the survival of offspring, maltreatment—including child neglect—remains prevalent in many countries. While child neglect is well known to affect child development, the causes of maternal neglect are poorly understood. Here, we found that female mice with a deletion mutation of CAST (a presynaptic release-machinery protein) showed significantly reduced weaning rate when primiparous and a recovered rate when multiparous. Indeed, when nurturing, primiparous and nulliparous CAST knock out (KO) mice exhibited less crouching time than control mice and moved greater distances. Contrary to expectations, plasma oxytocin (OXT) was not significantly reduced in CAST KO mice even though terminals of magnocellular neurons in the posterior pituitary expressed CAST. We further found that compared with control mice, CAST KO mice drank significantly less water when nurturing and had a greater preference for sucrose during pregnancy. We suggest that deficiency in presynaptic release-machinery protein impairs the facilitation of some maternal behaviours, which can be compensated for by experience and learning.
Collapse
Affiliation(s)
- Akari Hagiwara
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Naoko Sugiyama
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan.
| |
Collapse
|
13
|
Arévalo L, Campbell P. Placental effects on the maternal brain revealed by disrupted placental gene expression in mouse hybrids. Proc Biol Sci 2020; 287:20192563. [PMID: 31937228 PMCID: PMC7003458 DOI: 10.1098/rspb.2019.2563] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022] Open
Abstract
The mammalian placenta is both the physical interface between mother and fetus, and the source of endocrine signals that target the maternal hypothalamus, priming females for parturition, lactation and motherhood. Despite the importance of this connection, the effects of altered placental signalling on the maternal brain are insufficiently studied. Here, we show that placental dysfunction alters gene expression in the maternal brain, with the potential to affect maternal behaviour. Using a cross between the house mouse and the Algerian mouse, in which hybrid placental development is abnormal, we sequenced late-gestation placental and maternal medial preoptic area transcriptomes and quantified differential expression and placenta-maternal brain co-expression between normal and hybrid pregnancies. The expression of Fmn1 and Drd3 was significantly altered in the brains of females exposed to hybrid placentas. Most strikingly, expression patterns of placenta-specific gene families and Drd3 in the brains of house mouse females carrying hybrid litters matched those of female Algerian mice, the paternal species in the cross. Our results indicate that the paternally derived placental genome can influence the expression of maternal-fetal communication genes, including placental hormones, suggesting an effect of the offspring's father on the mother's brain.
Collapse
|
14
|
Winokur SB, Lopes KL, Moparthi Y, Pereira M. Depression-related disturbances in rat maternal behaviour are associated with altered monoamine levels within mesocorticolimbic structures. J Neuroendocrinol 2019; 31:e12766. [PMID: 31265182 DOI: 10.1111/jne.12766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/27/2019] [Accepted: 06/27/2019] [Indexed: 12/25/2022]
Abstract
The ability of mothers to sensitively attune their maternal responses to the needs of their developing young is fundamental to a healthy mother-young relationship. The biological mechanisms that govern how mothers adjust caregiving to the dynamic changes in the demands of the young remain an open question. In the present study, we examined whether changes in monoamine levels, within discrete mesocorticolimbic structures involved in cognitive and motivational processes key to parenting, modulate this flexibility in caregiving across the postpartum period. The present study used a Wistar-Kyoto (WKY) animal model of depression and control Sprague-Dawley (SD) rats, which differ dramatically in their cognitive, motivational, and parenting performance. Levels of the monoamine neurotransmitters, dopamine, noradrenaline and serotonin, as well as their major metabolites, were measured within the medial prefrontal cortex, striatum, nucleus accumbens and medial preoptic area of SD and WKY mothers at early (postpartum day [PPD]7-8), late (PPD15-16) and weaning (PPD25) postpartum stages using high-performance liquid chromatography with electrochemical detection. Consistent with our prior work, we find that caregiving of SD mothers declined as the postpartum period progressed. Relative to nulliparous females, early postpartum mothers had lower intracellular concentrations of monoamines, as well as lower noradrenaline turnover, and an elevated serotonin turnover within most structures. Postpartum behavioural trajectories subsequently corresponded to a progressive increase in all three monoamine levels within multiple structures. Compared to SD mothers, WKY mothers were inconsistent and disorganised in caring for their offspring and exhibit profound deficits in maternal behaviour. Additionally, WKY mothers had generally lower levels of all three monoamines, as well as different patterns of change across the postpartum period, compared to SD mothers, suggesting dysfunctional central monoamine pathways in WKY mothers as they transition and experience motherhood. Taken together, the results of the present study suggest a role for monoamines at multiple mesocorticolimbic structures with repect to modulating caregiving behaviours attuned to the changing needs of the young.
Collapse
Affiliation(s)
- Sarah B Winokur
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, USA
| | - Keianna L Lopes
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, USA
| | - Yashaswani Moparthi
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, USA
| | - Mariana Pereira
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
15
|
Post C, Leuner B. The maternal reward system in postpartum depression. Arch Womens Ment Health 2019; 22:417-429. [PMID: 30554286 PMCID: PMC6784840 DOI: 10.1007/s00737-018-0926-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/07/2018] [Indexed: 12/12/2022]
Abstract
The experience of motherhood is most often emotionally positive and rewarding, but for many new mothers suffering from postpartum depression (PPD), this is not the case. Preclinical and clinical research has sought to uncover brain changes underlying PPD in order to gain a better understanding of how this disorder develops. This review focuses on the mesolimbic dopamine system, particularly the ventral tegmental area-nucleus accumbens pathway which has been implicated in the regulation of critical functions disrupted in PPD including mood, motivation, and mothering. Specifically, we discuss normative changes in the mesolimbic system during motherhood in both rodents and humans and how these are impacted in PPD. We also consider modulation of mesolimbic dopamine by the hypothalamic neuropeptide oxytocin and how oxytocin-dopamine interactions regulate mood and mothering during the postpartum period. In addition to providing an overview of reward mechanisms in PPD, our goal is to highlight open questions which warrant further research.
Collapse
Affiliation(s)
- Caitlin Post
- Department of Psychology, The Ohio State University, 1835 Neil Avenue, Columbus, OH, USA
| | - Benedetta Leuner
- Department of Psychology, The Ohio State University, 1835 Neil Avenue, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
16
|
Hu Z, Du X, Yang Y, Botchway BOA, Fang M. Progesterone and fluoxetine treatments of postpartum depressive-like behavior in rat model. Cell Biol Int 2019; 43:539-552. [PMID: 30811083 DOI: 10.1002/cbin.11123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/23/2019] [Indexed: 12/19/2022]
Abstract
Research studies have indicated that alterations in plasma progesterone levels might be associated with the hippocampal synaptic plasticity of postpartum depressive-like behavior. Herein, we assess both progesterone and fluoxetine effects in adult female Sprague-Dawley rats with postpartum depressive-like behavior. Depressive-like behavior of postpartum rats was established using chronic ultra-mild stress (CUMS) method for 1 week from gestation day 15. Postpartum rats that showed depressive-like behavior were treated with either progesterone (subcutaneously, 0.5 mg/kg) from gestation day 17 to gestation day 22 or fluoxetine (by gavage, 10 mg/kg/day) for 4 weeks after birth. Open field and sucrose preference tests were conducted at the start, week 2 and week 4 postpartum. Golgi staining, immunofluorescence and Western blot analyses of rats' hippocampi were conducted on week 4 postpartum. Results showed CUMS increases depressive-like behavior, however, treatment with progesterone and fluoxetine improves this behavior. Both progesterone and fluoxetine treatments increase the numbers of dendritic spines pyramidal neurons in the CA3 region of the hippocampus as well as protein expression levels of microtubule-associated protein 2 (MAP-2) and synaptophysin (SYP). CUMS-induced decrement of MAP-2 and SYP protein expressions can be prevented by treatment with progesterone in advanced pregnant stage and fluoxetine in the postpartum period.
Collapse
Affiliation(s)
- Zhiying Hu
- Hangzhou Red Cross Hospital, Hangzhou, China
| | - Xiaoxue Du
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yang Yang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
17
|
Zhang L, Cao LL, Yang DD, Ding JH, Guo XD, Xue TF, Zhao XJ, Sun XL. Establishment and evaluation of a novel mouse model of peri/postmenopausal depression. Heliyon 2019; 5:e01195. [PMID: 30839939 PMCID: PMC6365542 DOI: 10.1016/j.heliyon.2019.e01195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/20/2018] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
Women are believed to be more vulnerable to develop depressive symptoms during the perimenopause compared to postmenopause. The traditional bilateral ovariectomy and chronic mild stress (CMS) stimulation animal model produces a postmenopausal depressive-like state but the transition from perimenopausal period to postmenopausal period was ignored. Thus we establish a novel animal model in which the mice were stimulated by CMS for three months and removed the ovaries by two-step operation, and then evaluate whether this novel model could be much better for preclinical study used as a peri/postmenopause depressive model. The present study systemically evaluated the changes induced by two-step ovariectomy plus CMS in the mice. The depression-like behaviors, the levels of corticosterone, estrogen, pro-inflammatory factors, neurotransmitters, as well as brain-derived neurotrophic factor were determined; the changes of estrogen receptors, serotonin receptors, uterine weight and bone microarchitecture were also observed. The results show that the behaviors and biochemical indexes of mice changed gradually over time. Our study suggests that this two-step ovariectomy operation plus CMS successfully establishes a more reasonable peri/postmenopausal depression animal model which effectively simulates the clinical symptoms of peri/postmenopausal depressive women.
Collapse
Affiliation(s)
- Ling Zhang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Lu-Lu Cao
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Dan-Dan Yang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jian-Hua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xu-Dong Guo
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Teng-Fei Xue
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiao-Jie Zhao
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiu-Lan Sun
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| |
Collapse
|