1
|
Wei LC, Chiu HJ. Sex Steroid Control of Serotonergic System: Clinical Implications for Psychiatric Disorders and Addiction Treatment. Clin Exp Pharmacol Physiol 2025; 52:e70044. [PMID: 40264347 DOI: 10.1111/1440-1681.70044] [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: 01/19/2025] [Revised: 02/27/2025] [Accepted: 04/07/2025] [Indexed: 04/24/2025]
Abstract
The seminal work by Fink et al. on sex steroid regulation of mood, cognition, and memory has profound implications for understanding sex differences in psychiatric disorders and addiction treatment. Their findings that estradiol upregulates serotonin transporter (SERT) and 5-HT2A receptor expression, along with testosterone's reliance on aromatisation for its serotonergic effects, highlight key neurobiological mechanisms underlying psychiatric conditions. These insights are particularly relevant to addiction medicine, given serotonin's modulatory role in reward pathways and substance use disorders. Recent research by Gu et al. has demonstrated that estradiol reduces serotonin reuptake by downregulating the plasma membrane monoamine transporter (PMAT) through oestrogen receptor beta (ERβ) and MAPK/ERK signalling pathways, further elucidating the neurochemical underpinnings of mood disorders. Additionally, testosterone's effects on serotonergic regulation are dependent on its conversion to estradiol via aromatase, which influences the expression of SERT and 5-HT2A receptors in critical brain regions. This process may explain sex differences in psychiatric disorders and treatment responses, particularly in mood disorders and substance use disorders. From a clinical perspective, understanding aromatase activity's role in modulating serotonergic pathways may aid in predicting treatment responses, particularly for male patients undergoing testosterone replacement therapy. Furthermore, targeting ERβ as a potential treatment strategy could provide novel therapeutic avenues for managing depression and substance use disorders in women experiencing hormonal fluctuations. These findings underscore the importance of sex-specific considerations in psychiatric and addiction treatment paradigms.
Collapse
Affiliation(s)
- Lien-Chung Wei
- Department of Addiction Psychiatry, Taoyuan Psychiatric Center, Ministry of Health and Welfare, Taoyuan, Taiwan, ROC
- Department of Counseling and Industrial/Organizational Psychology, Ming Chuan University, Taoyuan, Taiwan, ROC
| | - Hsien-Jane Chiu
- Department of General Psychiatry, Taoyuan Psychiatric Center, Ministry of Health and Welfare, Taoyuan, Taiwan, ROC
- Institute of Hospital and Health Care Administration, National Yang Ming Chiao Tung University, Hsinchu, Taiwan, ROC
| |
Collapse
|
2
|
Zeng H, Le L, Zhou W, Chen Y, Xie W, Xiong B, Chen Y, Fu B, Qiu R. Ni/Pd Dual-Catalysis Strategy for C(sp 2)-Sb Cross-Coupling of Halostibines with Aryl Triflates and Applications of Products as Coupling Reagents, Ligands, and Anticancer Compounds. J Org Chem 2025; 90:7043-7048. [PMID: 40388973 DOI: 10.1021/acs.joc.5c00613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
A novel and efficient dual-catalysis strategy using nickel and palladium has been developed for the cross-coupling of halostibines with aryl triflates to form C(sp2)-Sb bonds. This approach shows a wide substrate scope and high functional group tolerance and could be conducted on a gram scale. The synthesized arylstibines also could be arylation reagents reacting with alkyl and phenyl alkenes to form olefins and ligands to regulate the hydrogenation of diphenylacetylene. In addition, synthesized arylstibine 3q also shows satisfactory anticancer activity against cancerous MDA-MB-231 cells.
Collapse
Affiliation(s)
- Huifan Zeng
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
- Department of Physiology, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, P. R. China
| | - Liyuan Le
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
- Department of Physiology, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, P. R. China
| | - Wenjun Zhou
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Youwen Chen
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Wuxing Xie
- Department of Physiology, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, P. R. China
| | - Biquan Xiong
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, P. R. China
| | - Yi Chen
- Department of Physiology, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, P. R. China
| | - Biao Fu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Renhua Qiu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
3
|
Wang W, Yang W, Wang F, Gao H, Liu K, Zhang J, Li Y, Zhang M, Zhou G, Hou Y, Bai G. Kunxinning granules alleviate perimenopausal syndrome by supplementing estrogen deficiency. Front Pharmacol 2025; 16:1554479. [PMID: 40206089 PMCID: PMC11979375 DOI: 10.3389/fphar.2025.1554479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction Ovarian function decline results in reduced estrogen levels, leading to endocrine disorders, oxidative stress damage, and excessive activation of inflammatory factors, all of which contribute to the development of premenstrual syndrome (PMS). Kunxinning Granules (KXN) has been clinically approved for PMS treatment, but its bioactive ingredients and mechanism of action remain unclear. This study aimed to investigate the active metabolites and molecular mechanism of KXN in treating PMS rats, laying a foundation for the clinical development of PMS treatment. Methods An ovariectomized (OVX) rat model was established to evaluate the efficacy of KXN in treating PMS. Molecular network (MN) analysis, combined with UPLC/Q-TOF-MS, identified prototype compounds in the samples and constructed a chemical classification map based on their structures. A network analysis and proteomics were conducted to predict potential pathways through which KXN regulates PMS. Quantitative metabolomics assays were used to confirm these potential pathways. Additionally, target prediction and binding enzyme activity detection elucidated the key active metabolites and mechanisms of action in KXN. Results KXN exhibited significant effectiveness in supplementing estrogen deficiency and uterine atrophy in the OVX model. We identified 16 absorbed metabolites as the potential pharmacological ingredients of KXN in vivo. The steroid hormone biosynthesis pathway, a crucial pathway of KXN in PMS, played a key role in KXN's effectiveness. KXN improved hormonal metabolic disorders by regulating this pathway. The main metabolites in KXN, including astragaloside IV, icariin and baohuoside I increased estradiol levels by enhancing the activity of CYP19A1, the representative enzyme in hormone biosynthesis pathway. Discussion This study shows that KXN could relieve anxiety, depression, and osteoporosis in PMS. This pharmacological effect is exerted through steroid hormone synthesis to address estrogen deficiency. The findings provide valuable insights into the underlying mechanisms and support its clinical application.
Collapse
Affiliation(s)
- Wenshuang Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Wen Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Fangwenting Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - He Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Kaixin Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Jinling Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yunjuan Li
- State Key Laboratory of Chinese Medicine Modernization, Tasly Pharmaceutical Group Co., Ltd., Tianjin, China
- Tianjin Key Laboratory of Component-Based Chinese Medicine, Tianjin, China
| | - Man Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Guirong Zhou
- State Key Laboratory of Chinese Medicine Modernization, Tasly Pharmaceutical Group Co., Ltd., Tianjin, China
- Tianjin Key Laboratory of Component-Based Chinese Medicine, Tianjin, China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| |
Collapse
|
4
|
Zhao H, Yu F, Wu W. The Mechanism by Which Estrogen Level Affects Knee Osteoarthritis Pain in Perimenopause and Non-Pharmacological Measures. Int J Mol Sci 2025; 26:2391. [PMID: 40141035 PMCID: PMC11942494 DOI: 10.3390/ijms26062391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Perimenopausal women have fluctuating estrogen levels, which often trigger a range of symptoms of perimenopausal syndromes as estrogen levels decrease. Changes in perimenopausal estrogen levels are closely related to pain in knee osteoarthritis (KOA), which has long been a research area of great interest in perimenopausal women. In recent years, it has been found that perimenopausal estrogen levels have an important role in KOA pain, namely, that estrogen can affect KOA pain through the regulation of inflammatory responses, inhibition of cellular senescence and apoptosis, and modulation of neurotransmitters, which may provide new ideas for KOA treatment. This study aims to describe the mechanism of estrogen level on knee osteoarthritis pain in perimenopause and related non-pharmacological measures, such as physical therapy, physical factor therapy, traditional Chinese medicine, and diet, which can provide a reference for the study and treatment of pain in perimenopausal women with KOA.
Collapse
Affiliation(s)
- Huiying Zhao
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (H.Z.); (F.Y.)
| | - Fan Yu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (H.Z.); (F.Y.)
| | - Wei Wu
- School of Athletic Performance, Shanghai University of Sports, Shanghai 200438, China
| |
Collapse
|
5
|
Chen M, Yuan L, Chen B, Chang H, Luo J, Zhang H, Chen Z, Kong J, Yi Y, Bai M, Dong M, Zhou H, Jiang H. SLC29A1 and SLC29A2 are human nicotinamide cell membrane transporters. Nat Commun 2025; 16:1181. [PMID: 39885119 PMCID: PMC11782521 DOI: 10.1038/s41467-025-56402-y] [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: 11/23/2023] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Nicotinamide (NAM), a main precursor of NAD+, is essential for cellular fuel respiration, energy production, and other cellular processes. Transporters for other precursors of NAD+ such as nicotinic acid and nicotinamide mononucleotide (NMN) have been identified, but the cellular transporter of nicotinamide has not been elucidated. Here, we demonstrate that equilibrative nucleoside transporter 1 and 2 (ENT1 and 2, encoded by SLC29A1 and 2) drive cellular nicotinamide uptake and establish nicotinamide metabolism homeostasis. In addition, ENT1/2 exhibits a strong capacity to change the cellular metabolite composition and the transcript, especially those related to nicotinamide. We further observe that ENT1/2 regulates cellular respiration and senescence, contributing by altering the NAD+ pool level and mitochondrial status. Changes to cellular respiration, mitochondrial status and senescence by ENT1/2 knockdown are reversed by NMN supplementation. Together, ENT1 and ENT2 act as both cellular nicotinamide-level keepers and nicotinamide biological regulators through their NAM transport functions.
Collapse
Affiliation(s)
- Mingyang Chen
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Luexiang Yuan
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Binxin Chen
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou, China
| | - Hui Chang
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Luo
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou, China
| | - Hengbin Zhang
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Zhongjian Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, China
| | - Jiao Kong
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Yaodong Yi
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Mengru Bai
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou, China
| | - Minlei Dong
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Huidi Jiang
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| |
Collapse
|
6
|
Yang X, Wang Z, Chen Y, Ding H, Fang Y, Ma X, Liu H, Guo J, Zhao J, Wang J, Lu W. ALKBH5 Reduces BMP15 mRNA Stability and Regulates Bovine Puberty Initiation Through an m6A-Dependent Pathway. Int J Mol Sci 2024; 25:11605. [PMID: 39519156 PMCID: PMC11546126 DOI: 10.3390/ijms252111605] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
The timing of puberty significantly influences subsequent reproductive performance in cattle. N6-methyladenosine (m6A) is a key epigenetic modification involved in the regulation of pubertal onset. However, limited research has investigated alterations in m6A methylation within the hypothalamic-pituitary-ovarian (HPO) axis during the onset of puberty. In this study, combined analysis of methylated RNA immunoprecipitation sequencing (MeRIP-Seq) and RNA sequencing (RNA-seq) is used to describe the overall modification pattern of m6A in the HPO axis, while GSEA, KEGG, and GO analyses are used to describe the enrichment pathways of differentially expressed genes and differentially methylated genes. The m6A modifications of the differential genes KL, IGSF10, PAPPA2, and BMP15 and the pathways of cell adhesion molecules (CAMs), TGF-β, cell cycle, and steroid hormone synthesis may play roles in regulating the function of the HPO axis tissue during pubertal transition. Notably, BMP15's m6A modification depends on the action of the demethylase ALKBH5, which is recognized by the reader protein YTHDF2, promoting bovine granulosa cell proliferation, steroid production, and estrogen secretion. This study reveals for the first time the modification mechanism of BMP15 m6A during the initiation of bovine puberty, which will provide useful information for improving the reproductive efficiency of Chinese beef cattle.
Collapse
Affiliation(s)
- Xiaorui Yang
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Ziming Wang
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Yue Chen
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - He Ding
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Yi Fang
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Xin Ma
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Hongyu Liu
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Jing Guo
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Jing Zhao
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Jun Wang
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Wenfa Lu
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; (X.Y.); (Z.W.); (Y.C.); (H.D.); (Y.F.); (X.M.); (H.L.); (J.G.); (J.Z.)
- Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| |
Collapse
|
7
|
Chen L, Xu T, Lou J, Zhang T, Wu S, Xie R, Xu J. The beneficial roles and mechanisms of estrogens in immune health and infection disease. Steroids 2024; 207:109426. [PMID: 38685461 DOI: 10.1016/j.steroids.2024.109426] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/28/2024] [Accepted: 04/21/2024] [Indexed: 05/02/2024]
Abstract
Multiple epidemiologic studies have revealed that gender is considered one of the important factors in the frequency and severity of certain infectious diseases, in which estrogens may play a vital role. There is growing evidence that estrogens as female sex hormone can modulate multiple biological functions outside of the reproductive system, such as in brain and cardiovascular system. However, it is largely unknown about the roles and mechanisms of estrogens/estrogen receptors in immune health and infection disease. Thence, by reading a lot of literature, we summarized the regulatory mechanisms of estrogens/estrogen receptors in immune cells and their roles in certain infectious diseases with gender differences. Therefore, estrogens may have therapeutic potentials to prevent and treat these infectious diseases, which needs further clinical investigation.
Collapse
Affiliation(s)
- Lan Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ting Xu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun Lou
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ting Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Sheng Wu
- Department of Gastroenterology, Liupanshui People's Hospital, Liupanshui City 553000, Guizhou Province, China
| | - Rui Xie
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Jingyu Xu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
8
|
Honan LE, Fraser-Spears R, Daws LC. Organic cation transporters in psychiatric and substance use disorders. Pharmacol Ther 2024; 253:108574. [PMID: 38072333 PMCID: PMC11052553 DOI: 10.1016/j.pharmthera.2023.108574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/01/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Psychiatric and substance use disorders inflict major public health burdens worldwide. Their widespread burden is compounded by a dearth of effective treatments, underscoring a dire need to uncover novel therapeutic targets. In this review, we summarize the literature implicating organic cation transporters (OCTs), including three subtypes of OCTs (OCT1, OCT2, and OCT3) and the plasma membrane monoamine transporter (PMAT), in the neurobiology of psychiatric and substance use disorders with an emphasis on mood and anxiety disorders, alcohol use disorder, and psychostimulant use disorder. OCTs transport monoamines with a low affinity but high capacity, situating them to play a central role in regulating monoamine homeostasis. Preclinical evidence discussed here suggests that OCTs may serve as promising targets for treatment of psychiatric and substance use disorders and encourage future research into their therapeutic potential.
Collapse
Affiliation(s)
- Lauren E Honan
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA
| | - Rheaclare Fraser-Spears
- University of the Incarnate Word, Feik School of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Lynette C Daws
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA; The University of Texas Health Science Center at San Antonio, Department of Pharmacology, USA.
| |
Collapse
|
9
|
Ahmed F, Vranic M, Hetty S, Mathioudaki A, Patsoukaki V, Fanni G, Pereira MJ, Eriksson JW. Increased OCT3 Expression in Adipose Tissue With Aging: Implications for Catecholamine and Lipid Turnover and Insulin Resistance in Women. Endocrinology 2023; 165:bqad172. [PMID: 37972266 PMCID: PMC10690730 DOI: 10.1210/endocr/bqad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Catecholamine-stimulated lipolysis is reduced with aging, which may promote adiposity and insulin resistance. Organic cation transporter 3 (OCT3), which is inhibited by estradiol (E2), mediates catecholamine transport into adipocytes for degradation, thus decreasing lipolysis. In this study, we investigated the association of OCT3 mRNA levels in subcutaneous adipose tissue (SAT) with aging and markers of insulin resistance in women. METHODS SAT biopsies were obtained from 66 women with (19) or without (47) type 2 diabetes (age 22-76 years, 20.0-40.1 kg/m2). OCT3 mRNA and protein levels were measured for group comparisons and correlation analysis. SAT was incubated with E2 and OCT3 mRNA levels were measured. Associations between OCT3 single nucleotide polymorphisms (SNPs) and diabetes-associated traits were assessed. RESULTS OCT3 mRNA and protein levels in SAT increased with aging. SAT from postmenopausal women had higher levels of OCT3 than premenopausal women, and there was a dose-dependent reduction in OCT3 mRNA levels in SAT treated with E2. OCT3 mRNA levels were negatively associated with markers of insulin resistance, and ex vivo lipolysis. OCT3 SNPs were associated with BMI, waist to hip ratio, and circulating lipids (eg, triglycerides). CONCLUSION OCT3 mRNA and protein levels in SAT increased with aging, and mRNA levels were negatively associated with markers of insulin resistance. E2 incubation downregulated OCT3 mRNA levels, which may explain lower OCT3 mRNA in premenopausal vs postmenopausal women. High OCT3 protein levels in adipose tissue may result in increased catecholamine degradation, and this can contribute to the reduction in lipolysis observed in women with aging.
Collapse
Affiliation(s)
- Fozia Ahmed
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden
| | - Milica Vranic
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden
| | - Susanne Hetty
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden
| | - Argyri Mathioudaki
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden
| | - Vagia Patsoukaki
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden
| | - Giovanni Fanni
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
10
|
Weber BL, Nicodemus MM, Hite AK, Spalding IR, Beaver JN, Scrimshaw LR, Kassis SK, Reichert JM, Ford MT, Russell CN, Hallal EM, Gilman TL. Heterotypic Stressors Unmask Behavioral Influences of PMAT Deficiency in Mice. Int J Mol Sci 2023; 24:16494. [PMID: 38003684 PMCID: PMC10671398 DOI: 10.3390/ijms242216494] [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: 09/08/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Certain life stressors having enduring physiological and behavioral consequences, in part by eliciting dramatic signaling shifts in monoamine neurotransmitters. High monoamine levels can overwhelm selective transporters like the serotonin transporter. This is when polyspecific transporters like plasma membrane monoamine transporter (PMAT, Slc29a4) are hypothesized to contribute most to monoaminergic signaling regulation. Here, we employed two distinct counterbalanced stressors-fear conditioning and swim stress-in mice to systematically determine how reductions in PMAT function affect heterotypic stressor responsivity. We hypothesized that male heterozygotes would exhibit augmented stressor responses relative to female heterozygotes. Decreased PMAT function enhanced context fear expression, an effect unexpectedly obscured by a sham stress condition. Impaired cued fear extinction retention and enhanced context fear expression in males were conversely unmasked by a sham swim condition. Abrogated corticosterone levels in male heterozygotes that underwent swim stress after context fear conditioning did not map onto any measured behaviors. In sum, male heterozygous mouse fear behaviors proved malleable in response to preceding stressor or sham stress exposure. Combined, these data indicate that reduced male PMAT function elicits a form of stress-responsive plasticity. Future studies should assess how PMAT is differentially affected across sexes and identify downstream consequences of the stress-shifted corticosterone dynamics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - T. Lee Gilman
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH 44240, USA
| |
Collapse
|
11
|
Kang WC, Park K, Kong CH, Kim DY, Lee YS, Jeon M, Kim MS, Jung SY, Hong J, Choi JH, Ryu JH. D-Pinitol attenuates postmenopausal symptoms in ovariectomized mice. Life Sci 2023; 333:122147. [PMID: 37802198 DOI: 10.1016/j.lfs.2023.122147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023]
Abstract
AIMS Menopause is a natural process in women that can lead to post-menopausal syndrome with symptoms such as hot flushes, weight gain, anxiety, cognitive decline, and depression. Hormonal replacement therapy is commonly prescribed. However, it has serious adverse effects. Herbal medicinal products and isoflavones are used as alternatives. D-Pinitol found in Pinaceae and Fabaceae families has anti-inflammatory and antioxidant effects. However, it has not received as much attention as isoflavones. In this study, we investigated whether D-pinitol could alleviate post-menopausal symptoms using an ovariectomized (OVX) mouse model. MAIN METHODS Female ICR mice were divided into six groups: sham (vehicle), OVX (vehicle), OVX + D-pinitol (10, 30, 100 mg/kg, p.o.), and OVX + estradiol (0.5 mg/kg, s.c.). Treatment with vehicle, D-pinitol, and estradiol began at seven weeks post ovariectomy. We employed several behavioral tests, hot-flush test, and Western blot analysis. KEY FINDINGS We found that D-pinitol treatment (30, 100 mg/kg, p.o.) reversed cognitive dysfunction in OVX mice (novel object recognition and Y-maze test). Additionally, D-pinitol alleviated anxiety-like behaviors (elevated plus-maze) and reversed depressive-like behaviors (splash test, tail suspension test). It also normalized increased basal tail skin temperature in OVX mice. Moreover, D-pinitol administration reversed decreased expression of ERβ and synaptophysin and phosphorylation of ERK and PI3K-Akt-GSK-3β induced by OVX in the hippocampus and prefrontal cortex. SIGNIFICANCE These findings indicate that D-pinitol might be a promising candidate for treating post-menopausal symptoms by increasing ERβ and synaptophysin expression levels and activation of ERK or PI3K-Akt-GSK-3β signaling pathway, at least in part.
Collapse
Affiliation(s)
- Woo Chang Kang
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Keontae Park
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Chang Hyeon Kong
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Do Yeon Kim
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yong Seung Lee
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Mijin Jeon
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Min Seo Kim
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seo Yun Jung
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jung Hye Choi
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Oriental Pharmaceutical Science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jong Hoon Ryu
- Department of Biomedical and Pharmaceutical science, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Oriental Pharmaceutical Science, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
12
|
Weber BL, Nicodemus MM, Hite AK, Spalding IR, Beaver JN, Scrimshaw LR, Kassis SK, Reichert JM, Ford MT, Russell CN, Hallal EM, Gilman TL. Heterotypic stressors unmask behavioral influences of PMAT deficiency in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555632. [PMID: 37693400 PMCID: PMC10491137 DOI: 10.1101/2023.08.30.555632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Certain life stressors having enduring physiological and behavioral consequences, in part by eliciting dramatic signaling shifts in monoamine neurotransmitters. High monoamine levels can overwhelm selective transporters like the serotonin transporter. This is when polyspecific transporters like plasma membrane monoamine transporter (PMAT, Slc29a4) are hypothesized to contribute most to monoaminergic signaling regulation. Here, we employed two distinct counterbalanced stressors - fear conditioning, and swim stress - in mice to systematically determine how reductions in PMAT function affect heterotypic stressor responsivity. We hypothesized male heterozygotes would exhibit augmented stressor responses relative to female heterozygotes. Decreased PMAT function enhanced context fear expression, an effect unexpectedly obscured by a sham stress condition. Impaired cued fear extinction retention and enhanced context fear expression in males were conversely unmasked by a sham swim condition. Abrogated corticosterone levels in male heterozygotes that underwent swim stress after context fear conditioning did not map on to any measured behaviors. In sum, male heterozygous mouse fear behaviors proved malleable in response to preceding stressor or sham stress exposure. Combined, these data indicate reduced male PMAT function elicits a form of stress-responsive plasticity. Future studies should assess how PMAT is differentially affected across sexes and identify downstream consequences of the stress-shifted corticosterone dynamics.
Collapse
Affiliation(s)
- Brady L Weber
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Marissa M Nicodemus
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Allianna K Hite
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Isabella R Spalding
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Jasmin N Beaver
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Lauren R Scrimshaw
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Sarah K Kassis
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Julie M Reichert
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Matthew T Ford
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Cameron N Russell
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Elayna M Hallal
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - T Lee Gilman
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| |
Collapse
|
13
|
Weber BL, Beaver JN, Gilman TL. Summarizing studies using constitutive genetic deficiency to investigate behavioural influences of uptake 2 monoamine transporters. Basic Clin Pharmacol Toxicol 2023; 133:439-458. [PMID: 36316031 PMCID: PMC10657738 DOI: 10.1111/bcpt.13810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/27/2022]
Abstract
Burgeoning literature demonstrates that monoamine transporters with high transport capacity but lower substrate affinity (i.e., uptake 2) contribute meaningfully to regulation of monoamine neurotransmitter signalling. However, studying behavioural influences of uptake 2 is hindered by an absence of selective inhibitors largely free of off-target, confounding effects. This contrasts with study of monoamine transporters with low transport capacity but high substrate affinity (i.e., uptake 1), for which there are many reasonably selective inhibitors. To circumvent this dearth of pharmacological tools for studying uptake 2, researchers have instead employed mice with constitutive genetic deficiency in three separate transporters. By studying baseline behavioural shifts, plus behavioural responses to environmental and pharmacological manipulations-the latter primarily targeting uptake 1-investigators have been creatively characterizing the behavioural, and often sex-specific, influences of uptake 2. This non-systematic mini review summarizes current uptake 2 behaviour literature, highlighting emphases on stress responsivity in organic cation transporter 2 (OCT2) work, psychostimulant responsivity in OCT3 and plasma membrane monoamine transporter (PMAT) investigations, and antidepressant responsivity in all three. Collectively, this small but growing body of work reiterates the necessity for development of selective uptake 2-inhibiting drugs, with reviewed studies suggesting that these might advance personalized treatment approaches.
Collapse
Affiliation(s)
- Brady L Weber
- Department of Psychological Sciences & Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Jasmin N Beaver
- Department of Psychological Sciences & Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - T Lee Gilman
- Department of Psychological Sciences & Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| |
Collapse
|
14
|
Alcantara-Zapata DE, Lucero N, De Gregorio N, Astudillo Cornejo P, Ibarra Villanueva C, Baltodano-Calle MJ, Gonzales GF, Behn C. Women's mood at high altitude. sexual dimorphism in hypoxic stress modulation by the tryptophan-melatonin axis. Front Physiol 2023; 13:1099276. [PMID: 36733695 PMCID: PMC9887123 DOI: 10.3389/fphys.2022.1099276] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Sexual (and gender)-dimorphism in tolerance to hypobaric hypoxia increasingly matters for a differential surveillance of human activities at high altitude (HA). At low altitudes, the prevalence of anxiety and depression in women has already been found to double when compared with men; it could be expected to even increase on exposure to HA. In purposefully caring for the health of women at HA, the present work explores the potential involvement of the tryptophan (Trp)-melatonin axis in mood changes on exposure to hypobaric hypoxia. The present work highlights some already known anxiogenic effects of HA exposure. Hypoxia and insomnia reduce serotonin (5-HT) availability; the latter defect being expressed as failure of brown adipose tissue (BAT) activation and mood disorders. Rapid eye movement (REM) sleep organization and synapsis restoration that are additionally affected by hypoxia impair memory consolidation. Affective complaints may thus surge, evolving into anxiety and depression. Sex-related differences in neural network organization and hormonal changes during the menstrual cycle, and certainly also during the life cycle, underscore the possibility of 5-HT-related mood alterations, particularly in women on HA exposure. The mean brain rate of 5-HT synthesis at sea level is already 1.5-fold higher in males than in females. sexual dimorphism also evidences the overexpression effects of SERT, a 5-HT transporter protein. Gonadal and thyroid hormones, as influenced by HA exposure, further modulate 5-HT availability and its effects in women. Besides caring for adequate oxygenation and maintenance of one's body core temperature, special precautions concerning women sojourning at HA should include close observations of hormonal cycles and, perhaps, also trials with targeted antidepressants.
Collapse
Affiliation(s)
- D. E. Alcantara-Zapata
- Laboratorio de Endocrinología y Reproducción, Laboratorios de Investigación y Desarrollo (LID), Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - N. Lucero
- Occupational Health Program, School of Public Health, University of Chile, Santiago, Chile
| | - N. De Gregorio
- Laboratory of Extreme Environments, Department of Physiology and Biophysics, Biomedical Science Institute (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - P. Astudillo Cornejo
- Occupational Ergonomics Program, Department of Kinesiology, University of Atacama, Copiapó, Chile
| | - C. Ibarra Villanueva
- Occupational Ergonomics Program, Department of Kinesiology, University of Atacama, Copiapó, Chile
| | - M. J. Baltodano-Calle
- Laboratorio de Endocrinología y Reproducción, Laboratorios de Investigación y Desarrollo (LID), Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - G. F. Gonzales
- Laboratorio de Endocrinología y Reproducción, Laboratorios de Investigación y Desarrollo (LID), Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
- High Altitude Research Institute, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - C. Behn
- Laboratory of Extreme Environments, Department of Physiology and Biophysics, Biomedical Science Institute (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Faculty of Medicine, University of Atacama, Copiapó, Chile
| |
Collapse
|
15
|
Han Y, Gu S, Li Y, Qian X, Wang F, Huang JH. Neuroendocrine pathogenesis of perimenopausal depression. Front Psychiatry 2023; 14:1162501. [PMID: 37065890 PMCID: PMC10098367 DOI: 10.3389/fpsyt.2023.1162501] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/13/2023] [Indexed: 04/18/2023] Open
Abstract
With the development of social economics and the increase of working pressure, more and more women are suffering from long-term serious stress and showing symptoms of perimenopausal depression (PMD). The incidence rate of PMD is increasing, and the physical and mental health are seriously affected. However, due to the lack of accurate knowledge of pathophysiology, its diagnosis and treatment cannot be accurately executed. By consulting the relevant literature in recent years, this paper elaborates the neuroendocrine mechanism of perimenopausal depression from the aspects of epigenetic changes, monoamine neurotransmitter and receptor hypothesis, glial cell-induced neuroinflammation, estrogen receptor, interaction between HPA axis and HPG axis, and micro-organism-brain gut axis. The purpose is to probe into new ways of treatment of PMD by providing new knowledge about the neuroendocrine mechanism and treatment of PMD.
Collapse
Affiliation(s)
- Yuping Han
- Department of Psychology, Medical School, Jiangsu University, Zhenjiang, China
| | - Simeng Gu
- Department of Psychology, Medical School, Jiangsu University, Zhenjiang, China
- *Correspondence: Simeng Gu,
| | - Yumeng Li
- Department of Psychology, Medical School, Jiangsu University, Zhenjiang, China
| | - Xin Qian
- Department of Psychology, Medical School, Jiangsu University, Zhenjiang, China
| | - Fushun Wang
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu, Sichuan, China
| | - Jason H. Huang
- Department of Neurosurgery, Baylor Scott and White Health, Temple, TX, United States
- Department of Surgery, Texas A&M University, Temple, TX, United States
| |
Collapse
|
16
|
Jin LP, Zhang C, Xie Q, Xu J, Wang L, Yang LC, Huang EF, Wan DCC, Hu C. Design, synthesis and biological activity against estrogen receptor-dependent breast cancer of furo[1]benzofuran derivatives. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|