Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
1.
J Psychiatr Res ; 178: 305-312, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39182445

ABSTRACT

Postpartum depression and depressive symptoms have a major impact on maternal and infant health and well-being, yet to date their aetiology remains unclear. One hypothesis suggests a link between these symptoms and variations in prenatal cortisol levels, but existing evidence is limited and inconclusive. This study aims to provide additional evidence to disentangle the relationship between prenatal cortisol concentrations and subsequent occurrence of postpartum depressive symptoms. Cortisol for all three trimesters of pregnancy was extracted from the hair of 775 women participating in the French ELFE cohort. Depressive symptomatology at two months postpartum was assessed through the Edinburgh Postpartum Depression Scale (EPDS). Associations between prenatal cortisol levels and EPDS scores were tested using propensity-score weighted logistic regression models to control for confounders. An increase in mean cortisol concentrations was observed from the first to the third trimester of pregnancy. No significant differences in hair cortisol concentrations were found during the first and second trimesters between women who experienced postpartum depressive symptoms and those who did not. However, an association was observed between third trimester hair cortisol concentrations and depressive symptoms at two months postpartum. Women whose cortisol concentrations fell within the second quartile had a higher risk of subsequent PPDS (aOR = 2.67, 95%CI [1.01, 7.08]). Using a large sample from the general population, we observed an association between hair cortisol levels during the third trimester of pregnancy and postpartum depressive symptoms. Nevertheless, our results suggest that future studies could benefit from investigating other biomarkers of the reactivity of the corticotropic axis.


Subject(s)
Depression, Postpartum , Hair , Hydrocortisone , Humans , Female , Hydrocortisone/metabolism , Hydrocortisone/analysis , Hair/chemistry , Pregnancy , Depression, Postpartum/metabolism , Adult , Cohort Studies , Young Adult , France/epidemiology , Pregnancy Trimesters , Pregnancy Complications
2.
Int J Mol Sci ; 25(16)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39201521

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.


Subject(s)
Biomarkers , Depression, Postpartum , Humans , Depression, Postpartum/metabolism , Female , Animals , Pituitary-Adrenal System/metabolism , Hypothalamo-Hypophyseal System/metabolism , Brain/metabolism , Brain/diagnostic imaging , Stress, Psychological/metabolism
3.
Cell ; 187(16): 4176-4192.e17, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38959890

ABSTRACT

Hypothalamic neural circuits regulate instinctive behaviors such as food seeking, the fight/flight response, socialization, and maternal care. Here, we identified microdeletions on chromosome Xq23 disrupting the brain-expressed transient receptor potential (TRP) channel 5 (TRPC5). This family of channels detects sensory stimuli and converts them into electrical signals interpretable by the brain. Male TRPC5 deletion carriers exhibited food seeking, obesity, anxiety, and autism, which were recapitulated in knockin male mice harboring a human loss-of-function TRPC5 mutation. Women carrying TRPC5 deletions had severe postpartum depression. As mothers, female knockin mice exhibited anhedonia and depression-like behavior with impaired care of offspring. Deletion of Trpc5 from oxytocin neurons in the hypothalamic paraventricular nucleus caused obesity in both sexes and postpartum depressive behavior in females, while Trpc5 overexpression in oxytocin neurons in knock-in mice reversed these phenotypes. We demonstrate that TRPC5 plays a pivotal role in mediating innate human behaviors fundamental to survival, including food seeking and maternal care.


Subject(s)
Depression, Postpartum , Neurons , Obesity , TRPC Cation Channels , Animals , Female , Mice , Obesity/metabolism , Obesity/genetics , Male , Humans , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Depression, Postpartum/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Mice, Inbred C57BL , Oxytocin/metabolism , Maternal Behavior
4.
Neurochem Res ; 49(9): 2615-2635, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38904910

ABSTRACT

Despite the increase in the prevalence of postpartum depression among maternal disorder, its treatment outcomes remain suboptimal. Studies have shown that exercise can reduce postpartum depressive episodes in the mother, but the effects of exercise during pregnancy on maternal behavior and the potential mechanisms involved remain poorly understood. From the second day of pregnancy to the day of birth, dams exercised for 1 h a day by running on a controlled wheel. The maternal behaviors of the dams were assessed on postpartum day 2 to postpartum day 8. Chronic restraint stress was applied from postpartum day 2 to day 12. Blood was collected on postpartum days 3 and 8, then subjected to ELISA to determine the serum concentration of prolactin. The weight of each dam and the food intake were recorded. Anxiety- and depression-like behavioral tests were conducted, and hippocampal neuroinflammation and prolactin receptor levels were measured. The dams exhibited elevated levels of anxiety and depression, decreased serum prolactin levels, decreased prolactin receptor expression, and activation of NLRP3-mediated neuroinflammation in the hippocampus following the induction of postpartum chronic restraint stress, which were reversed with controlled wheel running during pregnancy. Overall, the findings of this study revealed that the preventive effects of exercise during pregnancy on postpartum anxiety-and depression-like behaviors were accompanied by increased serum prolactin levels, hippocampal prolactin receptor expression and hippocampal NLRP3-mediated neuroinflammation.


Subject(s)
Anxiety , Hippocampus , NLR Family, Pyrin Domain-Containing 3 Protein , Postpartum Period , Prolactin , Receptors, Prolactin , Animals , Female , Prolactin/blood , Prolactin/metabolism , Hippocampus/metabolism , Pregnancy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Anxiety/metabolism , Receptors, Prolactin/metabolism , Mice , Postpartum Period/metabolism , Physical Conditioning, Animal/physiology , Depression, Postpartum/metabolism , Depression, Postpartum/prevention & control , Depression/metabolism , Stress, Psychological/metabolism , Stress, Psychological/psychology , Running/physiology , Running/psychology
5.
Psychoneuroendocrinology ; 166: 107081, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38759520

ABSTRACT

BACKGROUND: Allopregnanolone (ALLO) is a metabolite of progesterone and a neuroactive steroid hormone. As a positive allosteric modulator of gamma-aminobutyric acid (GABA) receptors, ALLO seems to have antidepressant and anxiolytic effects, and was therefore approved as a specific medication for the treatment of postpartum depression in 2019. Despite the growing number of publications investigating ALLO levels, results on the biological and psychological correlates in the peripartum period remain inconsistent, possibly due to methodological challenges regarding measurement. To date, however, there is no systematic review examining the correlates, concentrations, and challenges in measuring ALLO in peripartum women. METHOD: A systematic literature search of PubMed and PsycINFO was conducted in August 2023. Original research articles that measured ALLO concentrations in peripartum women were included. Reports were excluded if they were not original research, included non-human subjects, did not include peripartum women, did not include ALLO measurement as an outcome, included (pharmacological) interventions, constituted method validations, or used the same cohort as another study. RESULTS: The literature search yielded 234 articles, and two articles were identified from other sources. After full-text screening, 19 articles (N = 1401) met the inclusion criteria, of which seven focused on biological correlates of ALLO and 12 on mood correlates. Of the latter, six found no association between ALLO and mood, four found a negative association, and two found a positive association. Overall, the results show an increase in ALLO levels during pregnancy and a decrease after birth, with levels then remaining low until six months postpartum. ALLO was most commonly measured in blood plasma and by gas chromatography-mass spectrometry (GC-MS). A significant matrix effect was found for blood serum and a significant method effect for radioimmunoassays (RIAs). A significant effect of time of measurement was found. CONCLUSION: ALLO measurement shows method and matrix effects. ALLO levels are higher when measured in serum compared to in plasma, and when measured using RIA compared to other methods. Time of measurement, study design, and standardization of measurement also influence the reliability of measurement and the interpretation of results.


Subject(s)
Depression, Postpartum , Peripartum Period , Pregnanolone , Humans , Pregnanolone/blood , Pregnanolone/analysis , Female , Pregnancy , Depression, Postpartum/blood , Depression, Postpartum/drug therapy , Depression, Postpartum/metabolism , Adult
6.
J Affect Disord ; 359: 241-252, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38768820

ABSTRACT

BACKGROUND: Postpartum depression (PPD) is a serious psychiatric disorder that has significantly adverse impacts on maternal health. Metabolic abnormalities in the brain are associated with numerous neurological disorders, yet the specific metabolic signaling pathways and brain regions involved in PPD remain unelucidated. METHODS: We performed behavioral test in the virgin and postpartum mice. We used mass spectrometry imaging (MSI) and targeted metabolomics analyses to investigate the metabolic alternation in the brain of GABAAR Delta-subunit-deficient (Gabrd-/-) postpartum mice, a specific preclinical animal model of PPD. Next, we performed mechanism studies including qPCR, Western blot, immunofluorescence staining, electron microscopy and primary astrocyte culture. In the specific knockdown and rescue experiments, we injected the adeno-associated virus into the central amygdala (CeA) of female mice. RESULTS: We identified that prostaglandin D2 (PGD2) downregulation in the CeA was the most outstanding alternation in PPD, and then validated that lipocalin-type prostaglandin D synthase (L-PGDS)/PGD2 downregulation plays a causal role in depressive behaviors derived from PPD in both wild-type and Gabrd-/- mice. Furthermore, we verified that L-PGDS/PGD2 signaling dysfunction-induced astrocytes atrophy is mediated by Src phosphorylation both in vitro and in vivo. LIMITATIONS: L-PGDS/PGD2 signaling dysfunction may be only responsible for the depressive behavior rather than maternal behaviors in the PPD, and it remains to be seen whether this mechanism is applicable to all depression types. CONCLUSION: Our study identified abnormalities in the L-PGDS/PGD2 signaling in the CeA, which inhibited Src phosphorylation and induced astrocyte atrophy, ultimately resulting in the development of PPD in mice.


Subject(s)
Astrocytes , Atrophy , Depression, Postpartum , Disease Models, Animal , Prostaglandin D2 , Signal Transduction , Animals , Astrocytes/pathology , Astrocytes/metabolism , Female , Depression, Postpartum/pathology , Depression, Postpartum/metabolism , Mice , Signal Transduction/physiology , Prostaglandin D2/metabolism , Central Amygdaloid Nucleus/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipocalins/genetics , Lipocalins/metabolism , src-Family Kinases/metabolism , Mice, Knockout
7.
Brain Behav Immun ; 119: 220-235, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38599497

ABSTRACT

Postpartum depression (PPD) is a severe mental disorder that affects approximately 10---20% of women after childbirth. The precise mechanism underlying PPD pathogenesis remains elusive, thus limiting the development of therapeutics. Gut microbiota dysbiosis is considered to contribute to major depressive disorder. However, the associations between gut microbiota and PPD remain unanswered. Here, we established a mouse PPD model by sudden ovarian steroid withdrawal after hormone-simulated pseudopregnancy-human (HSP-H) in ovariectomy (OVX) mouse. Ovarian hormone withdrawal induced depression-like and anxiety-like behaviors and an altered gut microbiota composition. Fecal microbiota transplantation (FMT) from PPD mice to antibiotic cocktail-treated mice induced depression-like and anxiety-like behaviors and neuropathological changes in the hippocampus of the recipient mice. FMT from healthy mice to PPD mice attenuated the depression-like and anxiety-like behaviors as well as the inflammation mediated by the NOD-like receptor protein (NLRP)-3/caspase-1 signaling pathway both in the gut and the hippocampus, increased fecal short-chain fatty acids (SCFAs) levels and alleviated gut dysbiosis with increased SCFA-producing bacteria and reduced Akkermansia in the PPD mice. Also, downregulation of NLRP3 in the hippocampus mitigated depression-like behaviors in PPD mice and overexpression of NLRP3 in the hippocampal dentate gyrus induced depression-like behaviors in naïve female mice. Intriguingly, FMT from healthy mice failed to alleviate depression-like behaviors in PPD mice with NLRP3 overexpression in the hippocampus. Our results highlighted the NLRP3 inflammasome as a key component within the microbiota-gut-brain axis, suggesting that targeting the gut microbiota may be a therapeutic strategy for PPD.


Subject(s)
Depression, Postpartum , Disease Models, Animal , Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Hippocampus , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Female , Dysbiosis/metabolism , Hippocampus/metabolism , Mice , Gastrointestinal Microbiome/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Fecal Microbiota Transplantation/methods , Depression, Postpartum/metabolism , Mice, Inbred C57BL , Depression/metabolism , Neuroinflammatory Diseases/metabolism , Behavior, Animal/physiology , Anxiety/metabolism , Brain-Gut Axis/physiology , Inflammation/metabolism , Ovariectomy
8.
Article in English | MEDLINE | ID: mdl-38484929

ABSTRACT

Postpartum depression (PPD) is a serious mental health problem that can negatively affect future generations. BDNF/AKT/mTOR signaling in the frontal lobe and hippocampus in mice is associated with depression, but its role in mice with PPD and their offspring is unknown. This study was aimed at investigating the effects of esketamine (ESK), a drug approved for treatment of refractory depression, on the BDNF/AKT/mTOR pathway in mice with PPD and their offspring. A model of chronic unpredictable mild stress with pregnancy was used. ESK was injected into postpartum mice, and behavioral tests were conducted to predict the severity of symptoms at the end of lactation and in the offspring after adulthood. Both mice with PPD and their offspring showed significant anxiety- and depression-like behaviors that were ameliorated with the ESK intervention. ESK enhanced exploratory behavior in unfamiliar environments, increased the preference for sucrose, and ameliorated the impaired BDNF/AKT/mTOR signaling in the frontal and hippocampal regions in mice. Thus, ESK may have great potential in treating PPD and decreasing the incidence of depression in offspring.


Subject(s)
Depression, Postpartum , Ketamine , Animals , Female , Mice , Pregnancy , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Antidepressive Agents/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Depression/psychology , Depression, Postpartum/drug therapy , Depression, Postpartum/metabolism , Disease Models, Animal , Hippocampus/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stress, Psychological/drug therapy , TOR Serine-Threonine Kinases/metabolism
9.
Arch Womens Ment Health ; 27(3): 435-445, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38214755

ABSTRACT

PURPOSE: Women are at high risk of stress, anxiety, and depression during the postpartum but the ways in which these different types of psychological distress are related to cortisol regulation is not clear. We examined the distinct association of each type of distress with women's average cortisol level, cortisol awakening response (CAR), cortisol decline across the day (diurnal slope), and overall amount of cortisol secretion across the day (AUCG). METHODS: At 6 months postpartum, a diverse group of 58 women completed measures of depression, anxiety, perceived stress, and life stressors. Each woman provided 4 salivary samples for cortisol assay from waking to bedtime on each of 2 consecutive days. Linear regressions were used to examine associations of stress, anxiety and depression to each of the 4 cortisol measures, controlling for number of stressful life events. RESULTS: Depressive symptoms were associated with less of a rise in the CAR (ß = -.46, p = 0.01), steeper diurnal slope (ß = .51, p = 0.006), and higher average cortisol level (ß = .42, p = .01). Women who met the clinical cutoff for an anxiety disorder had lower overall cortisol output (ß = -.29, p = 0.03). Stress was not related to any cortisol metric. CONCLUSIONS: Findings suggest that stress is less associated with cortisol alterations in the postpartum than are more severe types of psychological distress. Anxiety and depression may have distinct and opposite profiles of cortisol dysregulation. Results indicate that mental health assessment is critical even in the later postpartum so that interventions can be initiated to reduce emotional suffering and the risk of impaired cortisol regulation.


Subject(s)
Anxiety , Depression , Hydrocortisone , Postpartum Period , Saliva , Stress, Psychological , Humans , Female , Hydrocortisone/analysis , Hydrocortisone/metabolism , Adult , Stress, Psychological/metabolism , Stress, Psychological/psychology , Saliva/chemistry , Postpartum Period/psychology , Anxiety/psychology , Anxiety/metabolism , Depression/psychology , Depression/metabolism , Circadian Rhythm/physiology , Depression, Postpartum/psychology , Depression, Postpartum/metabolism , Young Adult
10.
Mol Psychiatry ; 29(4): 962-973, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38212371

ABSTRACT

The absence of non-invasive tests that can monitor the status of the brain is a major obstacle for psychiatric care. In order to address this need, we assessed the feasibility of using tissue-specific gene expression to determine the origin of extracellular vesicle (EV) mRNAs in peripheral blood. Using the placenta as a model, we discovered that 26 messenger RNAs that are specifically expressed in the placenta are present in EVs circulating in maternal blood. Twenty-three of these transcripts were either exclusively or highly expressed in maternal blood during pregnancy only and not in the postpartum period, verifying the feasibility of using tissue-specific gene expression to infer the tissue of origin for EV mRNAs. Using the same bioinformatic approach, which provides better specificity than isolating L1 cell-adhesion molecule containing EVs, we discovered that 181 mRNAs that are specifically expressed in the female brain are also present in EVs circulating in maternal blood. Gene set enrichment analysis revealed that these transcripts, which are involved in synaptic functions and myelination, are enriched for genes implicated in mood disorders, schizophrenia, and substance use disorders. The EV mRNA levels of 13 of these female brain-specific transcripts are associated with postpartum depression (adjusted p-vals = 3 × 10-5 to 0.08), raising the possibility that they can be used to infer the state of the brain. In order to determine the extent to which EV mRNAs reflect transcription in the brain, we compared mRNAs isolated from cells and EVs in an iPSC-derived brain microphysiological system differentiated for 3 and 9 weeks. We discovered that, although cellular and extracellular mRNA levels are not identical, they do correlate, and it is possible to extrapolate cellular RNA expression changes in the brain via EV mRNA levels. Our findings bring EV mRNAs to the forefront of peripheral biomarker development efforts in psychiatric diseases by demonstrating the feasibility of inferring transcriptional changes in the brain via blood EV mRNA levels.


Subject(s)
Biomarkers , Brain , Extracellular Vesicles , RNA, Messenger , Female , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Humans , RNA, Messenger/metabolism , Brain/metabolism , Biomarkers/blood , Biomarkers/metabolism , Pregnancy , Placenta/metabolism , Gene Expression/genetics , Adult , Depression, Postpartum/genetics , Depression, Postpartum/metabolism
11.
J Affect Disord ; 348: 283-296, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38159656

ABSTRACT

AIMS: To assess the effect of the translocator protein 18 kDa (TSPO) on postpartum depression and explore its mechanism. METHODS: Postpartum depression (PPD) mouse model was established, and flow cytometry, immunofluorescence, Western blot analysis, real-time quantitative PCR, adeno-associated virus (AAV), co-immunoprecipitation-mass spectrometry and immunofluorescence co-staining were used to detect the effect of TSPO ligand ZBD-2 on PPD mice. RESULTS: ZBD-2 inhibits the overactivation of microglia in the hippocampus and amygdala of PPD model mice. ZBD-2 not only inhibited the inflammation but also repressed the burst of reactive oxygen species (ROS) and mitochondrial ROS (mtROS). Meanwhile, ZBD-2 protects mitochondria from LPS-induced damages through inhibiting the influx of calcium. ZBD-2 modulated the calcium influx by increasing the level of translocase of the outer mitochondrial membrane 40 (TOM40) and reducing the interaction of TSPO and TOM40. In addition, the effect of ZBD-2 was partially dependent on anti-oxidative process. Knockdown of TOM40 by adeno-associated virus (AAV) in the hippocampus or amygdala dramatically reduced the effect of ZBD-2 on PPD, indicating that TOM40 mediates the effect of ZBD-2 on PPD. CONCLUSIONS: TOM40 is required for the effect of ZBD-2 on treating anxiety and depression in PPD mice. This study reveals the role of microglia TSPO in PPD development and provides the new therapeutic strategy for PPD.


Subject(s)
Depression, Postpartum , Microglia , Animals , Female , Mice , Calcium/metabolism , Carrier Proteins , Depression, Postpartum/drug therapy , Depression, Postpartum/metabolism , Homeostasis , Microglia/metabolism , Mitochondrial Membranes/metabolism , Reactive Oxygen Species/metabolism , Receptors, GABA/metabolism
12.
Genes (Basel) ; 14(6)2023 06 08.
Article in English | MEDLINE | ID: mdl-37372414

ABSTRACT

Brexanolone, a formulation of the neurosteroid allopregnanolone (ALLO), is approved for treating postpartum depression (PPD) and is being investigated for therapeutic efficacy across numerous neuropsychiatric disorders. Given ALLO's beneficial effects on mood in women with PPD compared to healthy control women, we sought to characterize and compare the cellular response to ALLO in women with (n = 9) or without (n = 10, i.e., Controls) past PPD, utilizing our previously established patient-derived lymphoblastoid cell lines (LCLs). To mimic in vivo PPD ALLO-treatment, LCLs were exposed to ALLO or DMSO vehicle for 60 h and RNA-sequenced to detect differentially expressed genes (DEGs, pnominal < 0.05). Between ALLO-treated Control and PPD LCLs, 269 DEGs were identified, including Glutamate Decarboxylase 1 (GAD1), which was decreased 2-fold in PPD. Network analysis of PPD:ALLO DEGs revealed enriched terms related to synaptic activity and cholesterol biosynthesis. Within-diagnosis analyses (i.e., DMSO vs. ALLO) detected 265 ALLO-induced DEGs in Control LCLs compared to only 98 within PPD LCLs, with just 11 DEGs overlapping. Likewise, the gene ontologies underlying ALLO-induced DEGs in PPD and Control LCLs were divergent. These data suggest that ALLO may activate unique and opposing molecular pathways in women with PPD, which may be tied to its antidepressant mechanism.


Subject(s)
Depression, Postpartum , Pregnanolone , Humans , Female , Pregnanolone/pharmacology , Pregnanolone/metabolism , Pregnanolone/therapeutic use , Depression, Postpartum/drug therapy , Depression, Postpartum/genetics , Depression, Postpartum/metabolism , Transcriptome/genetics , Dimethyl Sulfoxide , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use
13.
Mol Psychiatry ; 28(7): 3023-3032, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36782063

ABSTRACT

Postpartum depression (PPD) is a leading cause of morbidity and mortality among women. Clinically, the administration and withdrawal of supraphysiologic estradiol and progesterone (E2 + P) can cause affective symptom reoccurrence in women with a history of PPD, but not matched controls. To investigate the cellular basis underlying this differential affective response, lymphoblastoid cell lines (LCLs) were derived from women with and without past PPD and compared transcriptomically in hormone conditions mimicking pregnancy and parturition: supraphysiologic E2 + P-addback; supraphysiologic E2 + P-withdrawal; and no added E2 + P (Baseline). RNA-sequencing identified unique differentially expressed genes (DEGs) in all hormone conditions, but the majority tended to be downregulated in PPD and observed in E2 + P-addback. Two of these DEGs were evolutionarily conserved cellular stress regulators: IMPACT, an integrative response protein maintaining translational homeostasis, and WWTR1, a transcriptional coactivator in the 'Hippo' pathway mediating cell proliferation and survival. Correspondingly, significant gene network modules were linked to cell cycle progression, estrogen response, and immune dysregulation, suggesting innate differences in intracellular signaling in PPD. In certain hormone conditions, PPD LCLs displayed increased GATA3 expression (an upstream regulator of IMPACT and WWTR1) and differentially phosphorylated eiF2α (the ultimate downstream target of IMPACT). Taken together, these transcriptomic data primarily implicate innately dysregulated cellular responses as potentially influencing mood and/or escalating PPD risk. Furthermore, the intrinsic downregulation of IMPACT's translation and WWTR1's transcription networks may suggest a novel link between PPD and a compromised ability to maintain homeostasis in the context of cellular stress occurring during pregnancy and parturition.


Subject(s)
Depression, Postpartum , Pregnancy , Female , Humans , Depression, Postpartum/genetics , Depression, Postpartum/metabolism , Gene Regulatory Networks/genetics , Estradiol , Progesterone , Estrogens
14.
Behav Brain Res ; 438: 114208, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36356720

ABSTRACT

BACKGROUND: Postpartum depression (PPD) causes maternal mortality, and has a high disability rate. In recent years, studies have suggested the Sirt1 gene to be involved in the pathogenesis of depression. Resveratrol (RSV), an activator of Sirt1, has been investigated in depressive behavior. However, its effect on PPD remains to be thoroughly elucidated. METHODS: We employed a mice model with bilateral oophorectomy combined with hormone-simulated pregnancy to assess postpartum depression-like behavior. The behavioral tests were performed 2 days after the withdrawal of estradiol benzoate. RSV was administered subcutaneously to the PPD model mice. Several behavioral tests were executed, including the open field test, forced swimming test, and tail suspension test. Western blot analyses and immunofluorescence staining were used to evaluate protein expression levels of SIRT1, autophagy markers, and the AKT/mTOR. RESULTS: Postpartum depressive-like behavior was triggered following the withdrawal of estradiol benzoate after hormone-stimulated-pregnancy. RSV improved postpartum depressive-like behavior of mice via its upregulation of the SIRT1 and autophagy markers, such as Beclin1, ATG5 and LC3B. Also, the downregulation of the p62 protein expression was observed. More importantly, we also detected the inhibition of phosphorylated AKT and mTOR in the hippocampus of postpartum depressive-like mice. CONCLUSION: RSV could alleviate postpartum depression-like behavior in mice by stimulating the SIRT1, induce autophagy and inhibit the AKT/ mTOR signaling pathway.


Subject(s)
Depression, Postpartum , Sirtuin 1 , Animals , Female , Mice , Pregnancy , Autophagy , Depression, Postpartum/drug therapy , Depression, Postpartum/metabolism , Hormones , Proto-Oncogene Proteins c-akt/metabolism , Resveratrol/pharmacology , Sirtuin 1/metabolism , TOR Serine-Threonine Kinases/metabolism
15.
Acta Neurobiol Exp (Wars) ; 82(3): 347-357, 2022.
Article in English | MEDLINE | ID: mdl-36214717

ABSTRACT

Postpartum depression (PPD) is the most common type of puerperal mental syndrome and affects maternal physical and mental health and even the growth and development of infants. Paeoniflorin exerts a potential antidepressive effect; however, the functional roles and potential mechanisms of paeoniflorin in PPD are still largely unknown. PPD rat models were prepared by withdrawing hormone­simulated pregnancy (HSP), and subjects were treated with paeoniflorin and fluoxetine or plasmids. The sucrose preference test (SPT), forced swimming test (FST) and tail suspension test (TST) were used to monitor depression­like behavior in rats. A radioimmunoassay was utilized for estradiol (E2) and progesterone (P) measurements. ELISA was performed to detect serum corticosterone (Cor), hippocampal allopregnanolone (Allo), IL­1ß and TNF­α levels. Expression of the E2 receptors ERα and ERß was detected by qPCR. Western blotting was used to detect TSPO, BDNF and mTOR phosphorylation. Paeoniflorin drastically increased the sucrose preference of rats while decreasing the immobility time in the FST and TST in PPD models. Moreover, paeoniflorin intervention upregulated serum E2, hippocampal Allo, ERα, and ERß levels but degraded P, serum Cor, IL­1ß, TNF­α and ERα/ERß levels. Mechanistically, paeoniflorin promoted TSPO and BDNF­mTOR pathway activation in PPD rats. Furthermore, suppression of TSPO or the BDNF­mTOR pathway partially reversed the effects of paeoniflorin on depression­like behaviors, hormone levels, and inflammatory cytokine release. Paeoniflorin may improve symptoms of PPD by regulating the TSPO and BDNF­mTOR pathways, indicating that paeoniflorin may be an effective anti­PPD and antidepressant drug, providing evidence for the future treatment of PPD.


Subject(s)
Brain-Derived Neurotrophic Factor , Depression, Postpartum , Glucosides , Monoterpenes , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , Carrier Proteins/metabolism , Corticosterone/metabolism , Depression/drug therapy , Depression, Postpartum/drug therapy , Depression, Postpartum/metabolism , Disease Models, Animal , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Fluoxetine/therapeutic use , Glucosides/pharmacology , Hippocampus/metabolism , Humans , Monoterpenes/pharmacology , Pregnancy , Pregnanolone/metabolism , Progesterone/metabolism , Rats , Receptors, GABA-A/metabolism , Stress, Psychological , Sucrose , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
Mol Psychiatry ; 27(6): 2858-2867, 2022 06.
Article in English | MEDLINE | ID: mdl-35365803

ABSTRACT

Postpartum depression (PPD) affects 1 in 7 women and has negative mental health consequences for both mother and child. However, the precise biological mechanisms behind the disorder are unknown. Therefore, we performed the largest transcriptome-wide association study (TWAS) for PPD (482 cases, 859 controls) to date using RNA-sequencing in whole blood and deconvoluted cell types. No transcriptional changes were observed in whole blood. B-cells showed a majority of transcriptome-wide significant results (891 transcripts representing 789 genes) with pathway analyses implicating altered B-cell activation and insulin resistance. Integration of other data types revealed cell type-specific DNA methylation loci and disease-associated eQTLs (deQTLs), but not hormones/neuropeptides (estradiol, progesterone, oxytocin, BDNF), serve as regulators for part of the transcriptional differences between cases and controls. Further, deQTLs were enriched for several brain region-specific eQTLs, but no overlap with MDD risk loci was observed. Altogether, our results constitute a convergence of evidence for pathways most affected in PPD with data across different biological mechanisms.


Subject(s)
Depression, Postpartum , Genome-Wide Association Study , Insulin Resistance , Depression, Postpartum/genetics , Depression, Postpartum/metabolism , Female , Genome-Wide Association Study/methods , Humans , Insulin Resistance/genetics , Transcriptome/genetics
17.
Curr Neuropharmacol ; 20(10): 1988-2000, 2022.
Article in English | MEDLINE | ID: mdl-35236264

ABSTRACT

BACKGROUND: The substantial female hormone fluctuations associated with pregnancy and postpartum have been linked to a greater risk of developing depressive symptoms, particularly in high-risk women (HRW), i.e. those with histories of mood sensitivity to female hormone fluctuations. We have shown that glutamate (Glu) levels in the medial prefrontal cortex (MPFC) decrease during perimenopause, a period of increased risk of developing a major depressive episode. Our team has also demonstrated that percentage gray matter (%GM), another neural correlate of maternal brain health, decreases in the MPFC during pregnancy. OBJECTIVE: To investigate MPFC Glu levels and %GM from late pregnancy up to 7 weeks postpartum in HRW and healthy pregnant women (HPW). METHODS: Single-voxel spectra were acquired from the MPFC of 41 HPW and 22 HRW using 3- Tesla in vivo proton magnetic resonance spectroscopy at five different time points. RESULTS: We observed a statistically significant interaction between time and group for the metabolite Glu, with Glu levels being lower for HRW during pregnancy and early postpartum (p<0.05). MPFC %GM was initially lower during pregnancy and then significantly increased over time in both groups (p<0.01). CONCLUSION: This investigation suggests that the vulnerability towards PPD is associated with unique fluctuations of MPFC Glu levels during pregnancy and early postpartum period. Our results also suggest that the decline in MPFC %GM associated with pregnancy seems to progressively recover over time. Further investigations are needed to determine the specific role that female hormones play on the physiological changes in %GM during pregnancy and postpartum.


Subject(s)
Depression, Postpartum , Depressive Disorder, Major , Depression, Postpartum/metabolism , Depressive Disorder, Major/metabolism , Female , Glutamic Acid/metabolism , Gray Matter/diagnostic imaging , Gray Matter/metabolism , Hormones/metabolism , Humans , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/metabolism , Pregnancy , Prospective Studies
18.
Neurosci Lett ; 761: 136112, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34265417

ABSTRACT

Recent evidence has confirmed the association of glucocorticoid receptor (GR) gene variants with the "stress" endocrine axis in postpartum depression (PPD). Sirtuin 1(SIRT1) is an NAD+-dependent histone deacetylase and transcriptional enhancer of GR. However, to date, the function of the SIRT1 gene in the regulation of GR expression in PPD remains to be fully determined. A hormone-stimulated pregnancy (HSP) and subsequent "postpartum" withdrawal of estrogen was employed to mimic the fluctuations in estradiol associated with pregnancy and postpartum. We confirmed that estradiol benzoate withdrawal (EW)-rats displayed depression- and anxiety-like behaviors. These behavioral dysfunctions are associated with attenuated expression of SIRT1 and GR in the hippocampus. To assess the role of SIRT1, as well as its regulatory target directly, a selective SIRT1 activator (SRT2104) was infused into the hippocampus of EW-rats. We found that pharmacological activation of hippocampal SIRT1 blocks the development of depression-related, but not anxiety-related, phenotypes of PPD. In addition, the activation of SIRT1 leads to an increase in hippocampal GR expression in EW-rats. We further confirmed that SIRT1 physically interacts with GR in a glucocorticoid-dependent manner. Taken together, our results suggest that neuropathology in PPD is caused, at least in part, by the inhibition of the SIRT1-GR signaling pathway. Elevating SIRT1 levels, either pharmacologically or through other means, could represent a therapeutic strategy for PPD.


Subject(s)
Depression, Postpartum/metabolism , Receptors, Glucocorticoid/metabolism , Sirtuin 1/metabolism , Animals , Female , HEK293 Cells , Hippocampus/metabolism , Humans , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/genetics , Sirtuin 1/genetics , Up-Regulation
19.
Sci Rep ; 11(1): 8518, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33875712

ABSTRACT

Gestational stress can increase postpartum depression in women. To treat maternal depression, fluoxetine (FLX) is most commonly prescribed. While FLX may be effective for the mother, at high doses it may have adverse effects on the fetus. As environmental enrichment (EE) can reduce maternal stress effects, we hypothesized that a subthreshold dose of FLX increases the impact of EE to reduce anxiety and depression-like behavior in postpartum dams exposed to gestational stress. We evaluated this hypothesis in mice and to assess underlying mechanisms we additionally measured hypothalamic-pituitary-adrenal (HPA) axis function and brain levels of the hormone oxytocin, which are thought to be implicated in postpartum depression. Gestational stress increased anxiety- and depression-like behavior in postpartum dams. This was accompanied by an increase in HPA axis function and a decrease in whole-brain oxytocin levels in dams. A combination of FLX and EE remediated the behavioral, HPA axis and oxytocin changes induced by gestational stress. Central administration of an oxytocin receptor antagonist prevented the remediating effect of FLX + EE, indicating that brain oxytocin contributes to the effect of FLX + EE. These findings suggest that oxytocin is causally involved in FLX + EE mediated remediation of postpartum stress-related behaviors, and HPA axis function in postpartum dams.


Subject(s)
Depression, Postpartum/drug therapy , Fluoxetine/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Oxytocin/metabolism , Pituitary-Adrenal System/drug effects , Postpartum Period/drug effects , Stress, Psychological/drug therapy , Animals , Anxiety/drug therapy , Anxiety/metabolism , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Brain/drug effects , Brain/metabolism , Depression, Postpartum/metabolism , Disease Models, Animal , Female , Hypothalamo-Hypophyseal System/metabolism , Male , Maternal Behavior/drug effects , Mice , Pituitary-Adrenal System/metabolism , Postpartum Period/metabolism , Pregnancy , Receptors, Oxytocin/metabolism , Stress, Psychological/metabolism
20.
J Perinat Neonatal Nurs ; 35(1): 4-7, 2021.
Article in English | MEDLINE | ID: mdl-33528179

ABSTRACT

Depression onset during and after pregnancy is prevalent and associated with significant implications for maternal, child, and family health. Although environmental risk factors important to the expression of pregnancy-related depression are well known, knowledge of the genetic underpinning is limited. Given the joint contribution of environmental and genetic factors to depression risk liability, DNA methylation presents itself as an ideal biomarker to investigate basic mechanisms and opportunities for translational research to care for pregnancy-related depression health outcomes. This article is an introduction to DNA methylation and its potential to serve as a marker of depression risk during pregnancy and the postpartum. This commentary discusses current clinical uses of DNA methylation-based testing and how it may be applied to perinatal depression clinical care and management.


Subject(s)
DNA Methylation , Depression, Postpartum/metabolism , Protein Precursors/metabolism , Adult , Depression, Postpartum/genetics , Female , Humans , Maternal Behavior , Perinatal Care , Pregnancy , Receptors, Oxytocin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL