1
|
Szczerbowska-Boruchowska M, Chenczke A, Ruszczycki B, Wrobel P, Tokarczyk W, Stec P, Sowa KM, Ziomber-Lisiak A. Increased rubidium levels in brain regions involved in food intake in obese rats. Brain Struct Funct 2025; 230:66. [PMID: 40372485 DOI: 10.1007/s00429-025-02930-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
The hypothalamus, particularly its ventromedial and lateral regions, plays a pivotal role in homeostatic appetite regulation and is therefore a significant brain structure in the development of obesity. Additionally, the development of obesity can be caused by improper hedonic regulation, which involves neural circuits and systems associated with pleasure and reward. Several studies indicate a possible link between rubidium (Rb) and obesity, despite this element is not being typically considered influential in vital life processes. The present study, therefore, aims to investigate whether excessive body fat in obese animals alters rubidium levels in brain regions directly or indirectly involved in appetite regulation. The research was conducted on high-calorie diet (HCD)-induced obese rats (OB, n = 8) and their lean counterparts (L, n = 8). The determination of Rb levels in brain areas was performed using synchrotron radiation-based X-ray fluorescence microanalysis (SRXRF). The obtained results show a significantly higher level of Rb in all brain areas examined, although the increase in this element in obese individuals was not the same in all structures. The largest relative difference (over 70%) was observed for the orbitofrontal cortex, and the smallest (about 35%) for the amygdala. Principal component analysis with linear projections demonstrated a clear differentiation between the brain structures of obese and non-obese individuals based on the full elemental composition of tissues, while Rb was the only element that distinguished the obese group in each of the examined brain structures. The results obtained clearly confirm the increase in Rb levels in the brain structures responsible for regulating appetite in obesity.
Collapse
Affiliation(s)
| | - Aleksandra Chenczke
- Faculty of Physics and Applied Computer Science, AGH University of Krakow, Krakow, Poland
| | - Blazej Ruszczycki
- Faculty of Physics and Applied Computer Science, AGH University of Krakow, Krakow, Poland
| | - Pawel Wrobel
- Faculty of Physics and Applied Computer Science, AGH University of Krakow, Krakow, Poland
| | - Wiktoria Tokarczyk
- Faculty of Physics and Applied Computer Science, AGH University of Krakow, Krakow, Poland
| | - Patryk Stec
- Faculty of Physics and Applied Computer Science, AGH University of Krakow, Krakow, Poland
| | - Katarzyna M Sowa
- SOLARIS National Synchrotron Radiation Centre, Jagiellonian University, Krakow, Poland
| | | |
Collapse
|
2
|
Shipley J, Beadnall H, Butzkueven H, van der Walt A, Jokubaitis V. Impact of pregnancy on the maternal brain in health and multiple sclerosis. J Neurol Neurosurg Psychiatry 2025; 96:593-605. [PMID: 40132880 DOI: 10.1136/jnnp-2024-335319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
Multiple sclerosis (MS) is a chronic immune-mediated demyelinating disease of the central nervous system characterised by inflammatory lesions and neurodegeneration. Diagnosis often occurs in women of childbearing age, and therefore pregnancy is frequently encountered in women with MS. However, the effect of pregnancy on the MS brain is not well understood, including the impact on inflammatory lesion activity and rate of brain atrophy. Determining the effect of pregnancy on the MS brain is complex due to several confounding factors, including dynamic changes in brain volumes in healthy physiological (non-MS) states and the impact of withdrawing disease-modifying therapies for pregnancy on inflammatory lesion activity. This review first provides an in-depth overview of the profound structural neuroplasticity that occurs during pregnancy in healthy women without neurological disease and its association with maternal caregiving behaviours and maternal-infant attachment measures. These findings are integrated with results of MRI studies in pregnant women with MS to provide a perspective on the multifold influences on brain volume changes in this context. This review also explores the increase in inflammatory lesions observed on postpartum MRI in women with MS, which likely accrue in the postpartum phase mirroring clinical relapse dynamics. Key knowledge gaps are identified, and future research pathways are proposed to improve our understanding of how pregnancy impacts the brain in both healthy and MS states.
Collapse
Affiliation(s)
- Jessica Shipley
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Heidi Beadnall
- Brain & Mind Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Anneke van der Walt
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Yao J, Wu F, Xiu M. Interrelationships between glucose metabolism and gonadal hormones in female first-episode patients with schizophrenia. J Psychosom Res 2025; 192:112087. [PMID: 40068565 DOI: 10.1016/j.jpsychores.2025.112087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Schizophrenia (SCZ) is a complex mental disorder that typically emerges in late adolescence or early adulthood. The underlying molecular mechanisms of SCZ remain unclear. There is growing evidence supporting the involvement of abnormalities in metabolic and endocrine systems in the pathophysiology of SCZ. Our study was designed to explore the interrelationship between gonadal hormones and glucose metabolism in patients with SCZ. METHODS One hundred and twenty-four female first-episode patients diagnosed with SCZ according to DSM-IV were recruited. The glucose metabolism parameters, including fasting glucose, insulin, and insulin resistance index were measured. In addition, gonadal hormones, including follicle-stimulating hormone, testosterone, estradiol, progesterone, and luteinizing hormone were also determined in the same group of patients. RESULTS Abnormal glucose metabolism and sex hormones were observed in female DNFES patients, with a particularly high rate of insulin resistance (49.6 %). Correlation analysis revealed significant associations between glucose metabolic disturbance and sex hormones (all p < 0.05). Moreover, regression analysis adjusted for age, waist circumference, and BMI revealed that estradiol was negatively correlated with fasting insulin in female patients (β = -0.37, t = -4.0, p < 0.001). CONCLUSION These results provide evidence supporting the presence of disturbance in glucose metabolism and gonadal hormones at the onset of SCZ in female patients, suggesting that these dysregulations interact with each other to be involved in the disease's pathophysiological process.
Collapse
Affiliation(s)
- Jing Yao
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Fengchun Wu
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| | - Meihong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| |
Collapse
|
4
|
Xu HD, Wang TY, Wang C, Meng ZY, Chen JL, Zhou T, Wang LH, Zhao J. Study on the spectrum-effect correlation of phytoestrogenic activity of total flavonoids from Cynomorii Herba. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119646. [PMID: 40107476 DOI: 10.1016/j.jep.2025.119646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/16/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cynomorii Herba (CH), a traditional Chinese medicine, contains total flavonoids with potential phytoestrogenic activity, but the specific active components and mechanisms of action remain unclear. AIM OF THE STUDY To investigate the active components and mechanisms responsible for the phytoestrogenic activity of Cynomorii Herba Total Flavonoids (CHTF). MATERIALS AND METHODS A comprehensive fingerprint analysis of CHTF from 12 different regions was conducted using ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) technology. A spectrum-effect relationship model was established using grey relational analysis (GRA) and artificial neural network (ANN) methods, correlating the fingerprint peaks of CHTF with three pharmacological indicators: uterine index, MCF-7 cell proliferation rate, and estradiol (E2) content. Ultimately, molecular docking techniques were employed to verify the binding affinity of key flavonoid compounds to estrogen receptors (ER). RESULTS The sample from Alxa Left Banner (AQ), showed the strongest activity. A comprehensive analysis identified 16 key components closely related to phytoestrogenic activity, with (+)-catechin, procyanidin C1, and (-)-epicatechin being the main active constituents. Molecular docking studies showed that these compounds have good binding affinity to ERα and ERβ, confirming them as the main active substances responsible for the phytoestrogenic activity of CHTF. CONCLUSION CHTF exhibits significant phytoestrogenic activity by mediating ER, and CHTF from different regions shows various degrees of phytoestrogenic activity. There are 9 flavonoid and 7 non-flavonoid compounds in CHTF responsible for its phytoestrogenic activity.
Collapse
Affiliation(s)
- Hao-Dong Xu
- Department of Pharmacognosy, School of Pharmacy, Jiamusi University, Jiamusi, China
| | - Tian-Yu Wang
- Department of Pharmacognosy, School of Pharmacy, Jiamusi University, Jiamusi, China
| | - Chao Wang
- Department of Pharmacognosy, School of Pharmacy, Jiamusi University, Jiamusi, China
| | - Zhuo-Yi Meng
- Department of Pharmacognosy, School of Pharmacy, Jiamusi University, Jiamusi, China
| | - Jie-Lin Chen
- Department of Pharmacognosy, School of Pharmacy, Jiamusi University, Jiamusi, China
| | - Tong Zhou
- Department of Pharmacognosy, School of Pharmacy, Jiamusi University, Jiamusi, China
| | - Li-Hong Wang
- Department of Pharmacognosy, School of Pharmacy, Jiamusi University, Jiamusi, China.
| | - Ji Zhao
- Department of Pharmacognosy, School of Pharmacy, Jiamusi University, Jiamusi, China.
| |
Collapse
|
5
|
Gong W, Zhai Q, Wang Y, Shen A, Huang Y, Shi K, Huang Y, Song M, Yan R, Yao Z, Lu Q. Glymphatic function and choroid plexus volume is associated with systemic inflammation and oxidative stress in major depressive disorder. Brain Behav Immun 2025; 128:266-275. [PMID: 40220922 DOI: 10.1016/j.bbi.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/07/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Inflammatory processes were recognized as key factors in the pathophysiology of major depressive disorder (MDD). The choroid plexus (ChP) and glymphatic system played central roles in immune interactions between the brain and periphery. However, their specific roles in MDD and their relationship with systemic inflammation and oxidative stress remained unclear. METHODS This study finally included 665 MDD patients and 338 healthy controls. Clinical data and MRI scans were collected, and some patients also underwent blood routine and biochemical tests. ChP volume was manually segmented, and the diffusion tensor imaging along the perivascular space (DTI-ALPS) index, reflecting glymphatic function, was obtained through the FSL pipeline. The differences in these dices between groups were compared, and their associations with systemic inflammation and oxidative stress were analyzed. RESULTS MDD patients showed increased ChP volume (total: d = 0.316, p < 0.001; left: d = 0.317, p < 0.001; right: d = 0.268, p = 0.003) and decreased DTI-ALPS index (d = -0.144, p = 0.022), with a negative correlation between them (ρ = -0.135, p < 0.001). In MDD patients, lower DTI-ALPS index was correlated with higher LHR (ρ = -0.107, p = 0.025) and MHR (ρ = -0.126, p = 0.008). Larger right ChP volume was associated with higher MLR (ρ = 0.107, p = 0.009), SIRI (ρ = 0.086, p = 0.036), PIV (ρ = 0.086, p = 0.036), MHR (ρ = 0.136, p = 0.004), and PHR (ρ = 0.126, p = 0.008), while larger total ChP volume was correlated with higher MHR (ρ = 0.097, p = 0.042) and PHR (ρ = 0.114, p = 0.017). CONCLUSION MDD appeared to be accompanied by an increase in ChP volume and a decrease in glymphatic function, and these changes were related to systemic inflammation and oxidative stress.
Collapse
Affiliation(s)
- Wenyue Gong
- Nanjing Brain Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qinghua Zhai
- Nanjing Brain Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiwen Wang
- Nanjing Brain Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, China
| | - Azi Shen
- Nanjing Brain Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yinghong Huang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kaiyu Shi
- Nanjing Brain Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yingying Huang
- Nanjing Brain Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, China
| | - Moxuan Song
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Rui Yan
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhijian Yao
- Nanjing Brain Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, China; Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Qing Lu
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, China.
| |
Collapse
|
6
|
Haque R, Alam K, Gow J, Neville C, Keramat SA. Age and Gender Differences in the Relationship Between Chronic Pain and Dementia Among Older Australians. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2025; 28:562-570. [PMID: 39127253 DOI: 10.1016/j.jval.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/11/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVES Chronic pain is a highly debilitating condition that affects older adults and has the potential to increase their odds of experiencing cognitive impairment. The primary objective of this study was to examine the correlation between chronic pain and dementia. Additionally, this research endeavors to ascertain whether the association between chronic pain and dementia differs by age and gender. METHODS Cross-sectional data were derived from the Survey of Disability, Ageing, and Carers. A total of 20 671 and 20 081 participants aged 65 years and older in 2015 and 2018, respectively, were included in this study. The pooled association between chronic pain and dementia was assessed using a multivariable logistic regression model. Furthermore, the study also examined the multiplicative interaction effects between chronic pain and age, as well as chronic pain and gender, with dementia. RESULTS The pooled analysis demonstrated that chronic pain was associated with a heightened odds of dementia (adjusted odds ratio 1.95; 95% CI 1.85-2.05) among older Australians compared with their counterparts without chronic pain. The interaction effect indicated that individuals with chronic pain across all age groups exhibited increased odds of living with dementia. Additionally, women with chronic pain had higher odds of dementia compared with their counterparts without chronic pain and being male. CONCLUSIONS A continuous, coordinated, and tailored healthcare strategy is necessary to determine the pain management goals and explore early treatment options for chronic pain in older adults, particularly in groups with the greatest need.
Collapse
Affiliation(s)
- Rezwanul Haque
- School of Business, University of Southern Queensland, Toowoomba, Queensland, Australia.
| | - Khorshed Alam
- School of Business, University of Southern Queensland, Toowoomba, Queensland, Australia; Centre for Health Research, University of Southern Queensland, Toowoomba, Queensland, Australia
| | - Jeff Gow
- School of Business, University of Southern Queensland, Toowoomba, Queensland, Australia; Centre for Health Research, University of Southern Queensland, Toowoomba, Queensland, Australia; School of Accounting, Economics and Finance, University of KwaZulu-Natal, Durban, South Africa
| | - Christine Neville
- School of Nursing and Midwifery, University of Southern Queensland, Toowoomba, Queensland, Australia
| | - Syed Afroz Keramat
- Centre for Health Services Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
7
|
Wang X, Feng S, Deng Q, Wu C, Duan R, Yang L. The role of estrogen in Alzheimer's disease pathogenesis and therapeutic potential in women. Mol Cell Biochem 2025; 480:1983-1998. [PMID: 39088186 DOI: 10.1007/s11010-024-05071-4] [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: 04/11/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Estrogens are pivotal regulators of brain function throughout the lifespan, exerting profound effects from early embryonic development to aging. Extensive experimental evidence underscores the multifaceted protective roles of estrogens on neurons and neurotransmitter systems, particularly in the context of Alzheimer's disease (AD) pathogenesis. Studies have consistently revealed a greater risk of AD development in women compared to men, with postmenopausal women exhibiting heightened susceptibility. This connection between sex factors and long-term estrogen deprivation highlights the significance of estrogen signaling in AD progression. Estrogen's influence extends to key processes implicated in AD, including amyloid precursor protein (APP) processing and neuronal health maintenance mediated by brain-derived neurotrophic factor (BDNF). Reduced BDNF expression, often observed in AD, underscores estrogen's role in preserving neuronal integrity. Notably, hormone replacement therapy (HRT) has emerged as a sex-specific and time-dependent strategy for primary cardiovascular disease (CVD) prevention, offering an excellent risk profile against aging-related disorders like AD. Evidence suggests that HRT may mitigate AD onset and progression in postmenopausal women, further emphasizing the importance of estrogen signaling in AD pathophysiology. This review comprehensively examines the physiological and pathological changes associated with estrogen in AD, elucidating the therapeutic potential of estrogen-based interventions such as HRT. By synthesizing current knowledge, it aims to provide insights into the intricate interplay between estrogen signaling and AD pathogenesis, thereby informing future research directions and therapeutic strategies for this debilitating neurodegenerative disorder.
Collapse
Affiliation(s)
- Xinyi Wang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
8
|
Elbeltagy M, Khraisat B, AlZoubi L, Hmoud L, AlJeady A, Yousef M, Salman A. The neuroprotective effects of Piracetam on cisplatin-induced cognitive decline. Int J Neurosci 2025; 135:337-344. [PMID: 38153438 DOI: 10.1080/00207454.2023.2300733] [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: 08/03/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 12/29/2023]
Abstract
AIM This work explores the effect of Cisplatin-a chemotherapeutic agent known to cause deterioration in cognitive function in cancer patients, and spatial memory in mice. It also investigates the potential neuroprotective effects of Piracetam, which is a nootropic drug recognized for improving cognitive ability. MATERIALS AND METHODS The study incorporates four groups of mice receiving varied medication regimens, with memory tested using the Novel Location Recognition (NLR) method. RESULTS The findings from our study revealed that memory decline and a suppression of cellular proliferation were observed in adult male mice subjected to Cisplatin treatment; furthermore, a decline in antioxidant efficacy within the hippocampal dentate gyrus was evident. Moreover, analysis of treatment effects on the animals' weight revealed that the Cisplatin and Piracetam group exhibited the most significant weight loss during drug administration. Despite the significant weight loss, the simultaneous use of Cisplatin and Piracetam demonstrated a notable improvement in memory and an augmentation of hippocampal proliferation and antioxidant effect. LIMITATIONS It is important to note that our study was hampered by budget limits, a lack of additional animals, and mice's low tolerance for protracted treatment. CONCLUSIONS Should the outcomes of Piracetam observed in this investigation be applicable to patients, it might offer a relatively straightforward approach to mitigate the cognitive impacts endured by cancer survivors following exposure to chemotherapy. Future research will be needed to study Piracetam's effect on mice with brain cancer after Cisplatin treatment in order to extrapolate the results onto cancer patients.
Collapse
Affiliation(s)
- Maha Elbeltagy
- Faculty of Medicine, The University of Jordan, Amman, Jordan
- Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| | - Bann Khraisat
- Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Lujain AlZoubi
- Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Leen Hmoud
- Faculty of Dentistry, The University of Jordan, Amman, Jordan
| | | | - Mohammed Yousef
- Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Ahmed Salman
- Faculty of Medicine, The University of Jordan, Amman, Jordan
- Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| |
Collapse
|
9
|
Liu S, Yin N, Zhao Y, Yan B, Li S, Gao S. A highly sensitive electrochemical aptasensor for detecting broad-spectrum estrogen molecules in clinical samples. Talanta 2025; 283:127071. [PMID: 39447399 DOI: 10.1016/j.talanta.2024.127071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
The lack of sensitive and accurate monitoring methods for in vivo estrogen levels presents challenges for better prevention of estrogen-induced diseases. We have developed a label-free electrochemical aptasensor that demonstrates high sensitivity and selectivity for the broad-spectrum detection of estrogen molecules. This biosensor uses gold nanoparticles for electrochemical signal amplification and aptamers for broad-spectrum target recognition, enabling precise detection of trace amounts of estrogen in serum samples. The aptasensor demonstrates high sensitivity in estrogen detection, with a linear detection range of 0.01-1 nM and a minimum detection limit of 3 pM. It also exhibits excellent selectivity and interference resistance, with a detection error of less than 19 %, even in the presence of high concentrations of other biological substances. Additionally, molecular dynamics simulations were performed to provide insights into the molecular mechanism of aptamer broad-spectrum recognition and construction principles underlying the sensor. We anticipate that this aptasensor will serve as a robust, convenient, and cost-effective detection method, offering a valuable solution for the prevention of estrogen-induced diseases.
Collapse
Affiliation(s)
- Siyao Liu
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ning Yin
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
| | - Ya Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China.
| | - Shengjie Li
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China.
| | - Shunxiang Gao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China.
| |
Collapse
|
10
|
Molina Calistro L, Arancibia Y, Olivera MA, Domke S, Torres RF. Interaction of GPER-1 with the endocrine signaling axis in breast cancer. Front Endocrinol (Lausanne) 2025; 16:1494411. [PMID: 39936103 PMCID: PMC11811623 DOI: 10.3389/fendo.2025.1494411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
G Protein-Coupled Estrogen Receptor 1 (GPER-1) is a membrane estrogen receptor that has emerged as a key player in breast cancer development and progression. In addition to its direct influence on estrogen signaling, a crucial interaction between GPER-1 and the hypothalamic-pituitary-gonadal (HPG) axis has been evidenced. The novel and complex relationship between GPER-1 and HPG implies a hormonal regulation with important homeostatic effects on general organ development and reproductive tissues, but also on the pathophysiology of cancer, especially breast cancer. Recent research points to a great versatility of GPER-1, interacting with classical estrogen receptors and with signaling pathways related to inflammation. Importantly, through its activation by environmental and synthetic estrogens, GPER-1 is associated with hormone therapy resistance in breast cancer. These findings open new perspectives in the understanding of breast tumor development and raise the possibility of future applications in the design of more personalized and effective therapeutic approaches.
Collapse
Affiliation(s)
| | - Yennyfer Arancibia
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | | | - Sigrid Domke
- Facultad de Ciencias para el cuidado de la salud, Universidad San Sebastián, Puerto Montt, Chile
| | | |
Collapse
|
11
|
Liang Y, Fu W, Tang Y, Ye H, Wang Y, Sun C, Xiang Y, Xiong W, Cui M, Chen Y, Wang T, Deng Y. Selective Activation of G Protein-Coupled Estrogen Receptor 1 (GPER1) Reduces ER Stress and Pyroptosis via AMPK Signaling Pathway in Experimental Subarachnoid Hemorrhage. Mol Neurobiol 2025; 62:871-884. [PMID: 38935231 DOI: 10.1007/s12035-024-04312-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
Neuroinflammation is a critical pathogenic event following hemorrhagic stroke. Endoplasmic reticulum (ER) stress-induced apoptosis and nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing protein 3(NLRP3)-associated pyroptosis can contribute to the escalation of neuroinflammatory responses, leading to increased brain damage. G protein-coupled estrogen receptor 1(GPER1), as the most extensively characterized brain-derived estrogen, was reported to trigger neuroprotective effects. However, the anti-apoptotic and anti-pyroptotic effect of GPER1 activation and the underlying mechanism has not been fully elucidated. We established the experimental SAH model by intravascular perforation. The GPER1 selective agonist G1 was intravenously administered 1 h following SAH. For mechanistic exploration, the selective inhibitor of adenosine monophosphate-activated protein kinase (AMPK), dorsomorphin, was administered via intracerebroventricular injection 30 min prior to SAH induction. Post-SAH assessments included SAH grade, the short-term and long-term neurological outcomes, brain edema, cerebral blood flow, transmission electron microscopy (TEM), western blot (WB), ELISA, TUNEL staining, Fluoro-Jade C staining (FJC), and immunofluorescence staining. The expression of GPER1 was observed to elevate at 6 h and peaked at 24 h subsequent to SAH, predominantly co-localized with neurons. Post-treatment with G1 markedly ameliorated both the short-term and long-term neurological deficits of SAH mouse, as well as inhibiting the expression of neuronal ER stress-associated apoptotic proteins (i.e., CHOP, GRP78, Caspase-12, Cleaved Caspase-3, Bax, Bcl2) and pyroptosis-associated proteins (i.e., NLRP3, ASC, Cleaved Caspase-1). Additionally, our research revealed that inhibition of AMPK with dorsomorphin attenuated the neuroprotective effects of G1. This was accompanied by modifications in the molecular pathways associated with ER stress-induced apoptosis and pyroptosis. These data herein elucidated that GPER1 exerted neuroprotective effects by mitigating neuroinflammation in an AMPK-dependent manner, which modulates neuronal ER stress-associated apoptosis and pyroptosis. Boosting the anti-apoptotic and anti-pyroptotic effect by activating GPER1 may be an efficient treatment strategy for SAH patients.
Collapse
Affiliation(s)
- Yidan Liang
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400016, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400016, China
| | - Wenqiao Fu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yin Tang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hongjiang Ye
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yanglingxi Wang
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400016, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400016, China
| | - Chao Sun
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400016, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400016, China
| | - Yi Xiang
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400016, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400016, China
| | - Weiming Xiong
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400016, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400016, China
| | - Min Cui
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400016, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400016, China
| | - Yuanlin Chen
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ting Wang
- Department of Psychology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Yongbing Deng
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400016, China.
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400016, China.
| |
Collapse
|
12
|
Taylor S, Adhikari R. The Effect of Statin Treatment on Synaptogenesis in the Hippocampus. Biol Res Nurs 2025; 27:71-80. [PMID: 39165164 DOI: 10.1177/10998004241270079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Deranged lipid homeostasis has been implicated in neurodegenerative diseases. Cholesterol reducing compounds such as statins have received special attention for the possibility that they may be able to ameliorate or prevent cognitive loss associated with neurodegeneration. However, there is much dissension concerning the actual effect of statins on cognitive function. The aim of this study is to investigate the effects of pitavastatin on hippocampal synaptogenesis because the hippocampus is crucial for memory formation. We also evaluated the effects of pitavastatin on local hippocampal estrogen synthesized in the hippocampus itself and its effect on Brain-Derived Neurotrophic Factor (BDNF). Using a hippocampal cell line, H19-7, we found that hippocampal neurons exposed to pitavastatin demonstrate a significant reduction in the synaptic marker postsynaptic density protein 95 (psd-95). The pitavastatin treated neurons also exhibited decreased production of local estrogen and their expression of BDNF mRNA was decreased. These results suggest that statins reduce the ability of hippocampal neurons to form synapses by restricting the production of local estrogen. Because neural connections in the hippocampus are crucial for memory formation, our findings implicate statins as medications that may compromise cognitive function.
Collapse
Affiliation(s)
- Sara Taylor
- UNC-CH Division of Clinical Laboratory Science, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
13
|
Vassal M, Martins F, Monteiro B, Tambaro S, Martinez-Murillo R, Rebelo S. Emerging Pro-neurogenic Therapeutic Strategies for Neurodegenerative Diseases: A Review of Pre-clinical and Clinical Research. Mol Neurobiol 2025; 62:46-76. [PMID: 38816676 PMCID: PMC11711580 DOI: 10.1007/s12035-024-04246-w] [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: 01/03/2024] [Accepted: 05/14/2024] [Indexed: 06/01/2024]
Abstract
The neuroscience community has largely accepted the notion that functional neurons can be generated from neural stem cells in the adult brain, especially in two brain regions: the subventricular zone of the lateral ventricles and the subgranular zone in the dentate gyrus of the hippocampus. However, impaired neurogenesis has been observed in some neurodegenerative diseases, particularly in Alzheimer's, Parkinson's, and Huntington's diseases, and also in Lewy Body dementia. Therefore, restoration of neurogenic function in neurodegenerative diseases emerges as a potential therapeutic strategy to counteract, or at least delay, disease progression. Considering this, the present study summarizes the different neuronal niches, provides a collection of the therapeutic potential of different pro-neurogenic strategies in pre-clinical and clinical research, providing details about their possible modes of action, to guide future research and clinical practice.
Collapse
Affiliation(s)
- Mariana Vassal
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Filipa Martins
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Bruno Monteiro
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Huddinge, Sweden
| | - Ricardo Martinez-Murillo
- Neurovascular Research Group, Department of Translational Neurobiology, Cajal Institute (CSIC), Madrid, Spain
| | - Sandra Rebelo
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
14
|
Qi Z, Zhai Y, Han Y, Li K, Wang T, Li P, Li J, Zhou X, Zhao X, Song W. Genetic Evidence for Estrogenic Effects of Benzophenone-2 on Zebrafish Neurodevelopment and Its Signaling Mechanism. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:21433-21449. [PMID: 39579127 DOI: 10.1021/acs.est.4c06892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Estrogens play a crucial role in regulating various biological responses during the early stages of neurodevelopment. Benzophenone-2 (BP2), a widely used organic ultraviolet (UV) filter, has been proven as an estrogenic compound, whereas the estrogenic effects of BP2 on neurodevelopment remain largely unknown. Here, we investigated the neurodevelopmental toxicity of BP2 by exposing zebrafish embryos from 2 to 120 h postfertilization (hpf) at environmentally relevant concentrations. We demonstrated that early life exposure to BP2 induced multiple concentration-dependent impairments in the nervous system, including hypoactivity, abnormal brain morphology, impaired neurocyte proliferation, shortened axon, and increased neurocyte apoptosis. Moreover, metabolomic profiling revealed a decrease in dopamine (DA) and its metabolites in BP2-treated larvae. Using E2 treatment and morpholino knockdown assays, we provided strong genetic evidence that the BP2-induced behavioral disorders were associated with estrogen-dependent signaling, especially estrogen receptors 2a and 2b (esr2). Subsequently, transcriptomic profiling indicated that the activation of esr2 further inhibited the expression of LIM homeobox transcription factor 1 β a (lmx1ba), which is vital for normal neurodevelopment. Consistently, the overexpression of lmx1ba and inhibition of esr2 obviously alleviated BP2-caused neurotoxicity, uncovering a seminal role of esr2 and lmx1ba in BP2-induced neurodevelopmental toxicity. Our findings provide the first evidence in fish that BP2 can induce neurodevelopmental deficits and brain dysfunction and offer novel insights into the mechanisms of toxicity of BP2 as well as other emerging benzophenones.
Collapse
Affiliation(s)
- Zhipeng Qi
- Key Laboratory of Human Genetics and Environmental Medicine, Key Laboratory of Environment and Health, Center for Medical Statistics and Data Analysis, Xuzhou Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Yue Zhai
- School of Nursing, Jilin University, Changchun 130021, China
| | - Yi Han
- Key Laboratory of Human Genetics and Environmental Medicine, Key Laboratory of Environment and Health, Center for Medical Statistics and Data Analysis, Xuzhou Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Keying Li
- Key Laboratory of Human Genetics and Environmental Medicine, Key Laboratory of Environment and Health, Center for Medical Statistics and Data Analysis, Xuzhou Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Tianchen Wang
- Key Laboratory of Human Genetics and Environmental Medicine, Key Laboratory of Environment and Health, Center for Medical Statistics and Data Analysis, Xuzhou Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Peng Li
- Key Laboratory of Human Genetics and Environmental Medicine, Key Laboratory of Environment and Health, Center for Medical Statistics and Data Analysis, Xuzhou Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Jianan Li
- Key Laboratory of Human Genetics and Environmental Medicine, Key Laboratory of Environment and Health, Center for Medical Statistics and Data Analysis, Xuzhou Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiaomai Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Xinying Zhao
- Key Laboratory of Human Genetics and Environmental Medicine, Key Laboratory of Environment and Health, Center for Medical Statistics and Data Analysis, Xuzhou Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Weiyi Song
- Key Laboratory of Human Genetics and Environmental Medicine, Key Laboratory of Environment and Health, Center for Medical Statistics and Data Analysis, Xuzhou Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
15
|
Lewitus VJ, Kim J, Blackwell KT. Sex and estradiol effects in the rodent dorsal striatum. Eur J Neurosci 2024; 60:6962-6986. [PMID: 39573926 DOI: 10.1111/ejn.16607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
17β-Estradiol (E2) is a sex hormone that acts on many brain regions to produce changes in neuronal activity and learning. A key brain region sensitive to E2 is the dorsal striatum (also called caudate-putamen), which controls motor behaviour, goal-directed learning and habit learning. In adult rodents, oestrogen receptors (ERs) in the dorsal striatum are localized to the plasma membrane and include ERα, ERβ and G protein-coupled ER (GPER). E2, either naturally produced or exogenously applied, may influence neuronal excitability, basal synaptic transmission and long-term synaptic potentiation. These effects may be due to direct action on signalling pathways or may be due to changes in dopamine availability. In particular, estradiol influences dopamine release, dopamine receptor expression and dopamine transporter expression. We review the cellular effects that E2 has in the dorsal striatum, distinguishing between exogenously applied E2 and the oestrous cycle, as well as its influence on dorsal striatal-dependent motor and learning behaviour.
Collapse
|
16
|
Hu Q, Wang J, Liang J, Xiu M, Zhang S, Wu F. Gonadal hormone abnormalities in young patients with first-episode schizophrenia. Int J Neuropsychopharmacol 2024; 27:pyae063. [PMID: 39657134 DOI: 10.1093/ijnp/pyae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Gonadal hormones have been reported to be involved in the molecular mechanisms of schizophrenia (SCH). However, only a few studies have examined the gonadal hormone dysfunctions in first-episode schizophrenia (FES) patients, in particular in young patients with SCH. This study was designed to investigate the sex differences in gonadal hormones in young and antipsychotic-naïve FES patients. METHODS One hundred and sixty-two patients with SCH and 74 healthy controls were recruited, and blood gonadal hormones, including estradiol (E2), follicular-stimulating hormone (FSH), progesterone (PROG), luteinizing hormone (LH), and testosterone (TESTO), were measured in young FES patients and controls. RESULTS We found that both male and female young FES patients showed gonadal hormone disturbances at the onset of psychosis. Male patients exhibited a significantly higher rate of abnormal E2 (25.6% vs 3.9%), while female patients had higher rates of abnormal FSH (0% vs 5.3%), PROG (0% vs 21.1%), LH (3.5% vs 17.1%), and TESTO (3.5% vs 13.2%) (all P < .05). Multivariate logistic regression analysis further identified that specific gonadal hormone indices, including E2, LH, and TESTO, were factors associated with sex differences in young FES patients, after controlling for age, smoking status, and body mass index. CONCLUSIONS Our study reveals an overall gonadal hormone imbalance in young antipsychotic-naïve FES patients, highlighting sex differences at the onset of psychosis. Our study provides a foundation for further research into the role of gonadal hormones in the pathophysiology of SCH and the potential for personalized medicine approaches based on hormonal balance. Future studies were warranted to explore these differences and their implications for clinical practice to improve the treatment outcomes for individuals suffering from SCH.
Collapse
Affiliation(s)
- Qiang Hu
- Department of Psychiatry, Zhenjiang Mental Health Center, Zhenjiang, China
| | | | - Jing Liang
- Qingdao Mental Health Center, Qingdao, China
| | - Meihong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Shuangli Zhang
- Department of Psychiatry, The Third Hospital of Quzhou, Quzhou, China
| | - Fengchun Wu
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
17
|
Stein TP. Does Bisphenol A (BPA) Exposure Cause Human Diseases? Biomedicines 2024; 12:2678. [PMID: 39767585 PMCID: PMC11727305 DOI: 10.3390/biomedicines12122678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Autism spectrum disorders (ASDs), attention-deficit disorder (ADHD), Parkinson's disease (PD), polycystic ovary disease (PCOS), and Alzheimer's disease (AD) have all been linked to exposure to bisphenol A (BPA). METHODS This paper is a review and discussion of the published literature. RESULTS Animal studies have shown BPA to be a broad-spectrum endocrine disruptor. BPA is metabolized via the glucuronidation pathway, which involves the addition of glucose to the target molecule, and is catalyzed by uridine 5'-diphospho-glucuronosyltransferases (UGTs). Evidence of compromised glucuronidation has been found for ASD, DHD, PD, and PCOS. Genetic polymorphisms that alter the catalytic activity of the UGTs and efflux transporters involved are common. There are two ways to interpret the findings of associations between BPA glucuronidation efficiency and disease, a 'direct' pathway and an 'indirect' pathway. With the 'direct' pathway, free BPA is the actual causative agent. Compromised BPA detoxification leads to higher concentrations of free BPA in vulnerable tissues. Decreased BPA detoxification leads to increased exposure of vulnerable tissues to free BPA, where it can function as an endocrine disruptor. With the 'indirect' pathway, BPA is not the causative agent. BPA serves as a marker for the decreased glucuronidation efficiency of another unknown compound of endogenous origin detoxified by a similar combination of UGTs and efflux transporters as BPA. It is this compound(s), acting as an endocrine disruptor, that leads to a metabolic environment that favors disease development over an extended time period. CONCLUSION A review of the existing literature supports the indirect 'marker' hypothesis over the 'direct' hypothesis.
Collapse
Affiliation(s)
- T Peter Stein
- Rowan-Virtua School of Translational Biomedical Engineering and Sciences and School of Osteopathic Medicine, 2 Medical Center Drive, Stratford, NJ 08084, USA
| |
Collapse
|
18
|
Lasisi-Sholola AS, Hammed SO, Ajike RA, Akhigbe RE, Afolabi OA. Estrogen replacement therapy reverses spatial memory loss and pyramidal cell neurodegeneration in the prefrontal cortex of lead-exposed ovariectomized Wistar rats. Curr Res Toxicol 2024; 7:100200. [PMID: 39583742 PMCID: PMC11582547 DOI: 10.1016/j.crtox.2024.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/16/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Although menopause is a component of chronological aging, it may be induced by exposure to heavy metals like lead. Interestingly, lead exposure, just like the postmenopausal state, has been associated with spatial memory loss and neurodegeneration; however, the impact of hormone replacement therapy (HRT) on menopause and lead-induced spatial memory loss and neurodegeneration is yet to be reported. AIM The present study investigated the effect and associated mechanism of HRT on ovariectomized-driven menopausal state and lead exposure-induced spatial memory loss and neurodegeneration. MATERIALS AND METHODS Thirty adult female Wistar rats were randomized into 6 groups (n = 5 rats/group); the sham-operated vehicle-treated, ovariectomized (OVX), OVX + HRT, lead-exposed, OVX + lead, and OVX + Lead + HRT groups. Treatment was daily via gavage and lasted for 28 days. RESULTS Ovariectomy and lead exposure impaired spatial memory deficit evidenced by a significant reduction in novel arm entry, time spent in the novel arm, alternation, time exploring novel and familiar objects, and discrimination index. These findings were accompanied by a marked distortion in the histology of the prefrontal cortex, and a decline in serum dopamine level and pyramidal neurons. In addition, ovariectomy and lead exposure induced metabolic disruption (as depicted by a marked rise in lactate level and lactate dehydrogenase and creatinine kinase activities), oxidative stress (evidenced by a significant increase in MDA level, and decrease in GSH level, and SOD and catalase activities), inflammation (as shown by significant upregulation of myeloperoxidase activity, and TNF-α and IL-1β), and apoptosis (evidenced by a rise in caspase 3 activity) of the prefrontal cortex. The observed biochemical and histological perturbations were attenuated by HRT. CONCLUSIONS This study revealed that HRT attenuated ovariectomy and lead-exposure-induced spatial memory deficit and pyramidal neurodegeneration by suppressing oxidative stress, inflammation, and apoptosis of the prefrontal cortex.
Collapse
Affiliation(s)
- Abiodun Shukrat Lasisi-Sholola
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - Sodiq Opeyemi Hammed
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Richard Adedamola Ajike
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Roland Eghoghosoa Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - Oladele Ayobami Afolabi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
19
|
Cao J, Chen H, Zhang Y, Kang Y, Zhou S, Liao Z, Gao L, Yin J, Jing Y. Androgen deprivation exacerbates AD pathology by promoting the loss of microglia in an age-dependent manner. Life Sci 2024; 355:122973. [PMID: 39142510 DOI: 10.1016/j.lfs.2024.122973] [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: 04/30/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
AIMS Microglial cells are integral to the pathogenesis of Alzheimer's disease (AD). The observed sex disparity in AD prevalence, with a notable predominance in women, implies a potential influence of sex hormones, such as androgens, on disease mechanisms. Despite this, the specific effects of androgens on microglia remain unclear. This study is designed to delineate the interplay between androgens and the survival and inflammatory profile of microglial cells, as well as to explore their contribution to the progression of AD. METHODS AND KEY FINDINGS To create a chronic androgen deficiency model, 3-month-old wild-type (WT) mice and APP/PS1 mice underwent bilateral orchiectomy (ORX), with age-matched sham-operated controls. Cognitive and memory were evaluated at 5 and 12 months, paralleled by assessments of amyloid-beta (Aβ) and microglial morphology in hippocampal and cortical areas. The ORX treatment in mice resulted in diminished microglial populations and morphological alterations, alongside an increase in Aβ plaques and a concomitant decline in cognitive performance that exacerbated over time. In vitro, dihydrotestosterone (DHT) was found to stimulate microglial proliferation and ameliorate Aβ1-42-induced apoptosis. SIGNIFICANCE These findings suggested that androgens may exert a protective role, maintaining the normal proliferation and functionality of microglial cells. This preservation could potentially slow the progression of AD. As a result, our study provided a conceptual framework for the development of novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Jiaxin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Haichao Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yishu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yiting Kang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Siwei Zhou
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Zirui Liao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Liping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jie Yin
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yuhong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China; Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, People's Republic of China.
| |
Collapse
|
20
|
Abdelmissih S, Hosny SA, Elwi HM, Sayed WM, Eshra MA, Shaker OG, Samir NF. Chronic Caffeine Consumption, Alone or Combined with Agomelatine or Quetiapine, Reduces the Maximum EEG Peak, As Linked to Cortical Neurodegeneration, Ovarian Estrogen Receptor Alpha, and Melatonin Receptor 2. Psychopharmacology (Berl) 2024; 241:2073-2101. [PMID: 38842700 PMCID: PMC11442587 DOI: 10.1007/s00213-024-06619-4] [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: 07/31/2023] [Accepted: 05/16/2024] [Indexed: 06/07/2024]
Abstract
RATIONALE Evidence of the effects of chronic caffeine (CAFF)-containing beverages, alone or in combination with agomelatine (AGO) or quetiapine (QUET), on electroencephalography (EEG), which is relevant to cognition, epileptogenesis, and ovarian function, remains lacking. Estrogenic, adenosinergic, and melatonergic signaling is possibly linked to the dynamics of these substances. OBJECTIVES The brain and ovarian effects of CAFF were compared with those of AGO + CAFF and QUET + CAFF. The implications of estrogenic, adenosinergic, and melatonergic signaling and the brain-ovarian crosstalk were investigated. METHODS Adult female rats were administered AGO (10 mg/kg), QUET (10 mg/kg), CAFF, AGO + CAFF, or QUET + CAFF, once daily for 8 weeks. EEG, estrous cycle progression, and microstructure of the brain and ovaries were examined. Brain and ovarian 17β-estradiol (E2), antimullerian hormone (AMH), estrogen receptor alpha (E2Rα), adenosine receptor 2A (A2AR), and melatonin receptor 2 (MT2R) were assessed. RESULTS CAFF, alone or combined with AGO or QUET, reduced the maximum EEG peak, which was positively linked to ovarian E2Rα, negatively correlated to cortical neurodegeneration and ovarian MT2R, and associated with cystic ovaries. A large corpus luteum emerged with AGO + CAFF and QUET + CAFF, antagonizing the CAFF-mediated increased ovarian A2AR and reduced cortical E2Rα. AGO + CAFF provoked TTP delay and increased ovarian AMH, while QUET + CAFF slowed source EEG frequency to δ range and increased brain E2. CONCLUSIONS CAFF treatment triggered brain and ovarian derangements partially antagonized with concurrent AGO or QUET administration but with no overt affection of estrus cycle progression. Estrogenic, adenosinergic, and melatonergic signaling and brain-ovarian crosstalk may explain these effects.
Collapse
Affiliation(s)
- Sherine Abdelmissih
- Department of Medical Pharmacology, Faculty of Medicine Kasr Al-Ainy, Cairo University, Cairo, Egypt.
| | - Sara Adel Hosny
- Department of Medical Histology, Faculty of Medicine Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Heba M Elwi
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Walaa Mohamed Sayed
- Department of Anatomy and Embryology, Faculty of Medicine Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Mohamed Ali Eshra
- Department of Medical Physiology, Faculty of Medicine Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Olfat Gamil Shaker
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Nancy F Samir
- Department of Medical Physiology, Faculty of Medicine Kasr Al-Ainy, Cairo University, Cairo, Egypt
| |
Collapse
|
21
|
Bondy E. Considering the role of estradiol in the psychoneuroimmunology of perimenopausal depression. Brain Behav Immun Health 2024; 40:100830. [PMID: 39161877 PMCID: PMC11331712 DOI: 10.1016/j.bbih.2024.100830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/24/2024] [Accepted: 07/20/2024] [Indexed: 08/21/2024] Open
Abstract
In recent years, a burgeoning field of research has focused on women's mental health and psychiatric conditions associated with perinatal and postpartum periods. An emerging trend points to the link between hormone fluctuations during pregnancy and postpartum that have immunologic consequences in cases of perinatal depression and postpartum psychosis. The transition to menopause (or "perimenopause") has garnered comparatively less attention, but existing studies point to the influential interaction of hormonal and immune pathways. Moreover, the role of this cross talk in perturbing neural networks has been implicated in risk for cognitive decline, but relatively less work has focused on the depressed brain during perimenopause. This brief review brings a psychoneuroimmunology lens to depression during the perimenopausal period by providing an overview of existing knowledge and suggestions for future research to intertwine these bodies of work.
Collapse
Affiliation(s)
- Erin Bondy
- Department of Psychiatry, University of North Carolina School of Medicine, USA
| |
Collapse
|
22
|
Zhang J, Liu ZH, Zhao KM, Dang Z, Liu Y, Liu Y. Transformation of 17β-estradiol to estrone by unknown wild-type enzyme in commercial arylsulfatase from Helix pomatia. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1246:124293. [PMID: 39236428 DOI: 10.1016/j.jchromb.2024.124293] [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: 04/22/2024] [Revised: 08/02/2024] [Accepted: 08/25/2024] [Indexed: 09/07/2024]
Abstract
This work for the first time reported the complete transformation of 17β-estradiol (E2) to estrone (E1) by unknown wild-type enzyme present in the widely used commercial arylsulfatase derived from Helix pomatia. It was found that acetate could effectively inhibit the unknown enzyme with a half inhibitory concentration (IC50) of 140.9 μM, while phosphate and citrate showed no inhibition. Since the buffer solutions with phosphate and citrate have been used in the enzymatic hydrolysis of natural estrogen conjugates for decades, the transformation of E2 to E1 likely occurred during such procedure, inevitably leading to overestimated E1, but underestimated E2. It was further suggested that acetate should be used to prevent this undesirable transformation during the enzymatic hydrolysis of natural estrogen conjugates.
Collapse
Affiliation(s)
- Jun Zhang
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Ze-Hua Liu
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, Guangdong, China; Key Lab Pollution Control & Ecosystem Restoration in Industry Cluster, Ministry of Education, Guangzhou 510006, Guangdong, China; Guangdong Provincial Key Laboratory of Solid Wastes Pollution Control and Recycling, Guangzhou 510006, Guangdong, China; Guangdong Provincial Engineering and Technology Research Center for Environment Risk Prevention and Emergency Disposal, South China University of Technology, Guangzhou 510006, Guangdong, China.
| | - Ke-Meng Zhao
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Zhi Dang
- Key Lab Pollution Control & Ecosystem Restoration in Industry Cluster, Ministry of Education, Guangzhou 510006, Guangdong, China
| | - Yun Liu
- South China Institute of Environmental Sciences, Ministry of Ecology and Environment of the People's Republic of China, Guangzhou 510655, China
| | - Yu Liu
- Engineering Laboratory of Low-Carbon Unconventional Water Resources Utilization and Water Quality Assurance, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| |
Collapse
|
23
|
Ihalainen JK, Mikkonen RS, Ackerman KE, Heikura IA, Mjøsund K, Valtonen M, Hackney AC. Beyond Menstrual Dysfunction: Does Altered Endocrine Function Caused by Problematic Low Energy Availability Impair Health and Sports Performance in Female Athletes? Sports Med 2024; 54:2267-2289. [PMID: 38995599 PMCID: PMC11393114 DOI: 10.1007/s40279-024-02065-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/13/2024]
Abstract
Low energy availability, particularly when problematic (i.e., prolonged and/or severe), has numerous negative consequences for health and sports performance as characterized in relative energy deficiency in sport. These consequences may be driven by disturbances in endocrine function, although scientific evidence clearly linking endocrine dysfunction to decreased sports performance and blunted or diminished training adaptations is limited. We describe how low energy availability-induced changes in sex hormones manifest as menstrual dysfunction and accompanying hormonal dysfunction in other endocrine axes that lead to adverse health outcomes, including negative bone health, impaired metabolic activity, undesired outcomes for body composition, altered immune response, problematic cardiovascular outcomes, iron deficiency, as well as impaired endurance performance and force production, all of which ultimately may influence athlete health and performance. Where identifiable menstrual dysfunction indicates hypothalamic-pituitary-ovarian axis dysfunction, concomitant disturbances in other hormonal axes and their impact on the athlete's health and sports performance must be recognized as well. Given that the margin between podium positions and "losing" in competitive sports can be very small, several important questions regarding low energy availability, endocrinology, and the mechanisms behind impaired training adaptations and sports performance have yet to be explored.
Collapse
Affiliation(s)
- Johanna K Ihalainen
- Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, PO Box 35, 40014, Jyväskylä, Finland.
- Finnish Institute of High Performance Sport KIHU, Jyväskylä, Finland.
| | - Ritva S Mikkonen
- Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, PO Box 35, 40014, Jyväskylä, Finland
- Sports Technology Unit, Faculty of Sport and Health Sciences, University of Jyväskylä, Vuokatti, Finland
| | - Kathryn E Ackerman
- Wu Tsai Female Athlete Program, Division of Sports Medicine, Boston Children's Hospital, Boston, MA, USA
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ida A Heikura
- Canadian Sport Institute-Pacific, Victoria, BC, Canada
- Exercise Science, Physical and Health Education, University of Victoria, Victoria, BC, Canada
| | - Katja Mjøsund
- Paavo Nurmi Centre and Unit for Health and Physical Activity, University of Turku, Turku, Finland
- National Olympic Training Centre Helsinki, Helsinki, Finland
| | - Maarit Valtonen
- Finnish Institute of High Performance Sport KIHU, Jyväskylä, Finland
| | - Anthony C Hackney
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
24
|
Sadek KM, Khalifa NE, Alshial EE, Abdelnour SA, Mohamed AAR, Noreldin AE. Potential hazards of bisphenol A on the male reproductive system: Induction of programmed cell death in testicular cells. J Biochem Mol Toxicol 2024; 38:e23844. [PMID: 39252451 DOI: 10.1002/jbt.23844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/10/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024]
Abstract
A common industrial chemical known as bisphenol A (BPA) has been linked to endocrine disruption and can interfere with hormonal signaling pathways in humans and animals. This comprehensive review aims to explore the detrimental consequences of BPA on reproductive organ performance and apoptosis induction, shedding light on the emerging body of evidence from laboratory animal studies. Historically, most studies investigating the connection between BPA and reproductive tissue function have mainly leaned on laboratory animal models. These studies have provided crucial insights into the harmful effects of BPA on several facets of reproduction. This review consolidates an increasing literature that correlates exposure to BPA in the environment with a negative impact on human health. It also integrates findings from laboratory studies conducted on diverse species, collectively bolstering the mounting evidence that environmental BPA exposure can be detrimental to both humans and animals, particularly to reproductive health. Furthermore, this article explores the fundamental processes by which BPA triggers cell death and apoptosis in testicular cells. By elucidating these mechanisms, this review aids a deeper understanding of the complex interactions between BPA and reproductive tissues.
Collapse
Affiliation(s)
- Kadry M Sadek
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Norhan E Khalifa
- Department of Physiology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Eman E Alshial
- Department of Biochemistry, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Sameh A Abdelnour
- Department of Animal Production, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Amany A-R Mohamed
- Departmentof Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed E Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
25
|
Barrón-González M, Rivera-Antonio AM, Jarillo-Luna RA, Santiago-Quintana JM, Levaro-Loquio D, Pérez-Capistran T, Guerra-Araiza CH, Soriano-Ursúa MA, Farfán-García ED. Borolatonin limits cognitive deficit and neuron loss while increasing proBDNF in ovariectomised rats. Fundam Clin Pharmacol 2024; 38:730-741. [PMID: 38423984 DOI: 10.1111/fcp.12997] [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: 11/21/2023] [Revised: 01/31/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Borolatonin is a potential therapeutic agent for some neuronal diseases such as Alzheimer's disease (AD). Its administration exerts ameliorative effects such as those induced by the equimolar administration of melatonin in behavioral tests on male rats and in neuronal immunohistochemistry assays. OBJECTIVE In this study, motivated by sex differences in neurobiology and the incidence of AD, the ability of borolatonin to induce changes in female rats was assessed. METHODS Effects of borolatonin were measured by the evaluation of both behavioral and immunohistopathologic approaches; additionally, its ability to limit amyloid toxicity was determined in vitro. RESULTS Surprisingly, behavioral changes were similar to those reported in male rats, but not those evaluated by immunoassays regarding neuronal survival; while pro-brain-derived neurotrophic factor (BDNF) immunoreactivity and the limitation of toxicity by amyloid in vitro were observed for the first time. CONCLUSION Borolatonin administration induced changes in female rats. Differences induced by the administration of borolatonin or melatonin could be related to the differences in the production of steroid hormones in sex dependence. Further studies are required to clarify the possible mechanism and origin of differences in disturbed memory caused by the gonadectomy procedure between male and female rats.
Collapse
Affiliation(s)
- Mónica Barrón-González
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Astrid M Rivera-Antonio
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, ESM-IPN, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Rosa A Jarillo-Luna
- Laboratorio de Morfología, Sección de Estudios de Posgrado e Investigación, ESM-IPN, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - José M Santiago-Quintana
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - David Levaro-Loquio
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Teresa Pérez-Capistran
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Christian H Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - Marvin A Soriano-Ursúa
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Eunice D Farfán-García
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| |
Collapse
|
26
|
Kulkarni J, Gurvich C, Mu E, Molloy G, Lovell S, Mansberg G, Horton S, Morton E, Uppal T, Cashell C, de Castella A, Reisel D, Dear L, Weatherburn-Reeves N, Harris K, Pietrobon K, Teagle K, Kim BY, Newson L, Szoeke C. Menopause depression: Under recognised and poorly treated. Aust N Z J Psychiatry 2024; 58:636-640. [PMID: 38761367 PMCID: PMC11308326 DOI: 10.1177/00048674241253944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Menopause is a biological process experienced by all people assigned female at birth. A significant number of women experience mental ill health related to the major brain gonadal hormone shifts that occur in their midlife. There is poor understanding and management of the complex mental ill health issues, with the biological brain hormone changes receiving little formal attention. The current treatment advice is to manage this special type of mental ill health in the same way that all mental ill health is managed. This leads to poor outcomes for women and their families. Many women leave the workforce earlier than expected due to menopause-related depression and anxiety, with subsequent loss of salary and superannuation. Others describe being unable to adequately parent or maintain meaningful relationships - all ending in a poor quality of life. We are a large and diverse group of national and international clinicians, lived experience and social community advocates, all working together to innovate the current approaches available for women with menopausal mental ill health. Above all, true innovation is only possible when the woman with lived experience of menopause is front and centre of this debate.
Collapse
Affiliation(s)
- Jayashri Kulkarni
- HER Centre Australia, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Caroline Gurvich
- HER Centre Australia, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Eveline Mu
- HER Centre Australia, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Grace Molloy
- Menopause Friendly Australia, Surry Hills, NSW, Australia
| | - Sonya Lovell
- Dear Menopause Australia, Sydney, NSW, Australia
| | | | | | - Erin Morton
- Health Data and Clinical Trials, Flinders University, Adelaide, SA, Australia
| | - Talat Uppal
- Women’s Health Road, Frenchs Forest, NSW, Australia
| | - Ceri Cashell
- Avalon Family Medical Practice, Avalon Beach, NSW, Australia
| | - Anthony de Castella
- HER Centre Australia, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | | | | | | | | | - Kerry Pietrobon
- Lived Experience & Menopause Advocate , Brunswick, VIC, Australia
| | | | - Bo Youn Kim
- WA Country Health Service, Perth, WA, Australia
| | | | - Cassandra Szoeke
- Healthy Ageing Program, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
27
|
Żabińska M, Wiśniewska K, Węgrzyn G, Pierzynowska K. Exploring the physiological role of the G protein-coupled estrogen receptor (GPER) and its associations with human diseases. Psychoneuroendocrinology 2024; 166:107070. [PMID: 38733757 DOI: 10.1016/j.psyneuen.2024.107070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/15/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Estrogen is a group of hormones that collaborate with the nervous system to impact the overall well-being of all genders. It influences many processes, including those occurring in the central nervous system, affecting learning and memory, and playing roles in neurodegenerative diseases and mental disorders. The hormone's action is mediated by specific receptors. Significant roles of classical estrogen receptors, ERα and ERβ, in various diseases were known since many years, but after identifying a structurally and locationally distinct receptor, the G protein-coupled estrogen receptor (GPER), its role in human physiology and pathophysiology was investigated. This review compiles GPER-related information, highlighting its impact on homeostasis and diseases, while putting special attention on functions and dysfunctions of this receptor in neurobiology and biobehavioral processes. Understanding the receptor modulation possibilities is essential for therapy, as disruptions in receptors can lead to diseases or disorders, irrespective of correct estrogen levels. We conclude that studies on the GPER receptor have the potential to develop therapies that regulate estrogen and positively impact human health.
Collapse
Affiliation(s)
- Magdalena Żabińska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland
| | - Karolina Wiśniewska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland
| | - Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland.
| |
Collapse
|
28
|
Yu J, Zhang Y, Yao H, Zhang Z, Yang X, Zhu W, Xu J. ERβ activation improves nonylphenol-induced depression and neurotransmitter secretion disruption via the TPH2/5-HT pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116521. [PMID: 38850708 DOI: 10.1016/j.ecoenv.2024.116521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
The aim of this study is to investigate the role of estrogen receptor β (ERβ) in nonylphenol (NP) - induced depression - like behavior in rats and its impact on the regulation of the TPH2/5-HT pathway. In the in vitro experiment, rat basophilic leukaemia cells (RBL-2H3) cells were divided into the four groups: blank group, NP group (20 μM), ERβ agonist group (0.01 μM), and NP+ERβ agonist group (20 μM+0.01 μM). For the in vivo experiment, 72 adult male Sprague-Dawley rats were randomly divided into following six groups: the Control, NP (40 mg/kg) group, ERβ agonist (2 mg/kg, Diarylpropionitrile (DPN)) group, ERβ inhibitor (0.1 mg/kg, 4-(2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl) phenol (PHTPP)) group, NP+ERβ agonist (40 mg/kg NP + 2 mg/kg DPN) group, and NP+ERβ inhibitor (40 mg/kg NP + 0.1 mg/kg PHTPP) group, with 12 rats in each group. Each rat in drug group were given NP by gavage and/or received a single intraperitoneal injection of DPN 2 mg/kg or PHTPP 0.1 mg/kg. Both in vivo and in vitro, NP group showed a decrease in the expression levels of ERβ, tryptophan hydroxylase (TPH1), and tryptophan hydroxylase-2 (TPH2) genes and proteins, and reduced levels of DA, NE, and 5-hydroxytryptophan (5-HT) neurotransmitters. RBL-2H3 cells showed signs of cell shrinkage, with rounded cells, increased suspension and more loosely arranged cells. The effectiveness of the ERβ agonist stimulation exhibited an increase exceeding 60% in RBL-2H3 cells. The application of ERβ agonist resulted in an alleviation the aforementioned alterations. ERβ agonist activated the TPH2/5-HT signaling pathways. Compared to the control group, the NP content in the brain tissue of the NP group was significantly increased. The latency to eat for the rats was longer and the amount of food consumed was lower, and the rats had prolonged immobility time in the behavioral experiment of rats. The expression levels of ERβ, TPH1, TPH2, 5-HT and 5-HITT proteins were decreased in the NP group, suggesting NP-induced depression-like behaviours as well as disturbances in the secretion of serum hormones and monoamine neurotransmitters. In the NP group, the midline raphe nucleus showed an elongated nucleus with a dark purplish-blue colour, nuclear atrophy, displacement and pale cytoplasm. ERβ might ameliorate NP-induced depression-like behaviors, and secretion disorders of serum hormones and monoamine neurotransmitters via activating TPH2/5-HT signaling pathways.
Collapse
Affiliation(s)
- Jie Yu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Yujie Zhang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Hao Yao
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Ziping Zhang
- Department of Clinical Laboratory, Zunyi Medical and Pharmaceutical College, Zunyi 563006, PR China
| | - Xiao Yang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Wei Zhu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Jie Xu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China.
| |
Collapse
|
29
|
Chen L, Zhang Y, Wang Z, Zhang Z, Wang J, Zhu G, Yang S. Activation of GPER1 by G1 prevents PTSD-like behaviors in mice: Illustrating the mechanisms from BDNF/TrkB to mitochondria and synaptic connection. CNS Neurosci Ther 2024; 30:e14855. [PMID: 38992889 PMCID: PMC11239537 DOI: 10.1111/cns.14855] [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: 04/12/2024] [Revised: 06/11/2024] [Accepted: 06/29/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND G1 is a specific agonist of G protein-coupled estrogen receptor 1 (GPER1), which binds and activates GPER1 to exert various neurological functions. However, the preventive effect of G1 on post-traumatic stress disorder (PTSD) and its mechanisms are unclear. OBJECTIVE To evaluate the protective effect of G1 against synaptic and mitochondrial impairments and to investigate the mechanism of G1 to improve PTSD from brain-derived neurotrophic factor (BDNF)/tyrosine kinase receptor B (TrkB) signaling. METHODS This study initially detected GPER1 expression in the hippocampus of single prolonged stress (SPS) mice, utilizing both Western blot and immunofluorescence staining. Subsequently, the effects of G1 on PTSD-like behaviors, synaptic, and mitochondrial functions in SPS mice were investigated. Additionally, the involvement of BDNF/TrkB signaling involved in the protection was further confirmed using GPER1 antagonist and TrkB inhibitor, respectively. RESULTS The expression of GPER1 was reduced in the hippocampus of SPS mice, and G1 treatment given for 14 consecutive days significantly improved PTSD-like behaviors in SPS mice compared with model group. Electrophysiological local field potential (LFP) results showed that G1 administration for 14 consecutive days could reverse the abnormal changes in the gamma oscillation in the CA1 region of SPS mice. Meanwhile, G1 administration for 14 consecutive days could significantly improve the abnormal expression of synaptic proteins, increase the expression of mitochondria-related proteins, increase the number of synapses in the hippocampus, and ameliorate the damage of hippocampal mitochondrial structure in SPS mice. In addition, G15 (GPER1 inhibitor) and ANA-12 (TrkB inhibitor) blocked the ameliorative effects of G1 on PTSD-like behaviors and aberrant expression of hippocampal synaptic and mitochondrial proteins in SPS mice and inhibited the reparative effects of G1 on structural damage to hippocampal mitochondria, respectively. CONCLUSION G1 improved PTSD-like behaviors in SPS mice, possibly by increasing hippocampal GPER1 expression and promoting BDNF/TrkB signaling to repair synaptic and mitochondrial functional impairments. This study would provide critical mechanism for the prevention and treatment of PTSD.
Collapse
Affiliation(s)
- Lixia Chen
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Yang Zhang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Zisheng Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Zhengrong Zhang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Jingji Wang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Shaojie Yang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
30
|
Burmistrov DE, Gudkov SV, Franceschi C, Vedunova MV. Sex as a Determinant of Age-Related Changes in the Brain. Int J Mol Sci 2024; 25:7122. [PMID: 39000227 PMCID: PMC11241365 DOI: 10.3390/ijms25137122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The notion of notable anatomical, biochemical, and behavioral distinctions within male and female brains has been a contentious topic of interest within the scientific community over several decades. Advancements in neuroimaging and molecular biological techniques have increasingly elucidated common mechanisms characterizing brain aging while also revealing disparities between sexes in these processes. Variations in cognitive functions; susceptibility to and progression of neurodegenerative conditions, notably Alzheimer's and Parkinson's diseases; and notable disparities in life expectancy between sexes, underscore the significance of evaluating aging within the framework of gender differences. This comprehensive review surveys contemporary literature on the restructuring of brain structures and fundamental processes unfolding in the aging brain at cellular and molecular levels, with a focus on gender distinctions. Additionally, the review delves into age-related cognitive alterations, exploring factors influencing the acceleration or deceleration of aging, with particular attention to estrogen's hormonal support of the central nervous system.
Collapse
Affiliation(s)
- Dmitriy E. Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
| | - Sergey V. Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| |
Collapse
|
31
|
Al-Tameemi ZKA, Khanam R, Shetty PJ. Bisphenol-A Leaching from Polycarbonate 5-Gallon Water Bottles in the UAE: A Comprehensive Study. Nepal J Epidemiol 2024; 14:1302-1309. [PMID: 39280642 PMCID: PMC11396560 DOI: 10.3126/nje.v14i1.59934] [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: 03/09/2024] [Revised: 03/24/2024] [Accepted: 03/30/2024] [Indexed: 09/18/2024] Open
Abstract
Bisphenol A (BPA) is widely used around the world in the production of Polycarbonate (PC) plastics. Notably, the ubiquitous 5-gallon water bottles in the UAE are primarily made of PC plastic, making them a significant concern as bottled water is the region's main supply of drinking water. These bottles undergo temperature variations during storage and transportation, potentially leading to harmful BPA (Bisphenol A) leaching. This study analyzed 40 PC 5-gallon water bottles from two local brands A and B, with 20 bottles per brand, under two conditions: room temperature and outdoor sunlight exposure for a month. BPA levels were assessed at 0, 15, and 30 days, following ethical approval. Liquid-liquid extraction and ELISA assays were conducted, with comprehensive kit validation. The results revealed a significant increase in BPA concentration over time, particularly in bottles exposed to elevated temperatures (Day 30 outdoor-stored samples exhibited the highest concentration at 9.05 ± 2.30 μg/L). Brand B consistently exhibited higher BPA concentrations across different samples and environments. This study emphasizes the link between BPA content and storage time, highlighting the need for preventive measures to reduce BPA exposure. Individuals should be aware of potential health risks associated with prolonged storage in plastic containers and consider safer alternatives.
Collapse
Affiliation(s)
| | - Razia Khanam
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, UAE
| | - Preetha J Shetty
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, UAE
| |
Collapse
|
32
|
Balan I, Boero G, Chéry SL, McFarland MH, Lopez AG, Morrow AL. Neuroactive Steroids, Toll-like Receptors, and Neuroimmune Regulation: Insights into Their Impact on Neuropsychiatric Disorders. Life (Basel) 2024; 14:582. [PMID: 38792602 PMCID: PMC11122352 DOI: 10.3390/life14050582] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/18/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Pregnane neuroactive steroids, notably allopregnanolone and pregnenolone, exhibit efficacy in mitigating inflammatory signals triggered by toll-like receptor (TLR) activation, thus attenuating the production of inflammatory factors. Clinical studies highlight their therapeutic potential, particularly in conditions like postpartum depression (PPD), where the FDA-approved compound brexanolone, an intravenous formulation of allopregnanolone, effectively suppresses TLR-mediated inflammatory pathways, predicting symptom improvement. Additionally, pregnane neurosteroids exhibit trophic and anti-inflammatory properties, stimulating the production of vital trophic proteins and anti-inflammatory factors. Androstane neuroactive steroids, including estrogens and androgens, along with dehydroepiandrosterone (DHEA), display diverse effects on TLR expression and activation. Notably, androstenediol (ADIOL), an androstane neurosteroid, emerges as a potent anti-inflammatory agent, promising for therapeutic interventions. The dysregulation of immune responses via TLR signaling alongside reduced levels of endogenous neurosteroids significantly contributes to symptom severity across various neuropsychiatric disorders. Neuroactive steroids, such as allopregnanolone, demonstrate efficacy in alleviating symptoms of various neuropsychiatric disorders and modulating neuroimmune responses, offering potential intervention avenues. This review emphasizes the significant therapeutic potential of neuroactive steroids in modulating TLR signaling pathways, particularly in addressing inflammatory processes associated with neuropsychiatric disorders. It advances our understanding of the complex interplay between neuroactive steroids and immune responses, paving the way for personalized treatment strategies tailored to individual needs and providing insights for future research aimed at unraveling the intricacies of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Irina Balan
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Giorgia Boero
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA;
| | - Samantha Lucenell Chéry
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Minna H. McFarland
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alejandro G. Lopez
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
33
|
Balu D, Valencia-Olvera AC, Deshpande A, Narayanam S, Konasani S, Pattisapu S, York JM, Thatcher GRJ, LaDu MJ, Tai LM. Estradiol improves behavior in FAD transgenic mice that express APOE3 but not APOE4 after ovariectomy. Front Endocrinol (Lausanne) 2024; 15:1374825. [PMID: 38742194 PMCID: PMC11089251 DOI: 10.3389/fendo.2024.1374825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
Increasing evidence suggests that female individuals have a higher Alzheimer's disease (AD) risk associated with post-menopausal loss of circulating estradiol (E2). However, clinical data are conflicting on whether E2 lowers AD risk. One potential contributing factor is APOE. The greatest genetic risk factor for AD is APOE4, a factor that is pronounced in female individuals post-menopause. Clinical data suggests that APOE impacts the response of AD patients to E2 replacement therapy. However, whether APOE4 prevents, is neutral, or promotes any positive effects of E2 is unclear. Therefore, our goal was to determine whether APOE modulates the impact of E2 on behavior and AD pathology in vivo. To that end, mice that express human APOE3 (E3FAD) or APOE4 (E4FAD) and overproduce Aβ42 were ovariectomized at either 4 months (early) or 8 months (late) and treated with vehicle or E2 for 4 months. In E3FAD mice, we found that E2 mitigated the detrimental effect of ovariectomy on memory, with no effect on Aβ in the early paradigm and only improved learning in the late paradigm. Although E2 lowered Aβ in E4FAD mice in the early paradigm, there was no impact on learning or memory, possibly due to higher Aβ pathology compared to E3FAD mice. In the late paradigm, there was no effect on learning/memory and Aβ pathology in E4FAD mice. Collectively, these data support the idea that, in the presence of Aβ pathology, APOE impacts the response to E2 supplementation post-menopause.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ana C. Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ashwini Deshpande
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Saharsh Narayanam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Sravya Konasani
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Shreya Pattisapu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Jason M. York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
34
|
Creinin MD, Cagnacci A, Spaczyński RZ, Stute P, Chabbert-Buffet N, Korver T, Simoncini T. Experts' view on the role of oestrogens in combined oral contraceptives: emphasis on oestetrol (E4). Front Glob Womens Health 2024; 5:1395863. [PMID: 38655395 PMCID: PMC11035732 DOI: 10.3389/fgwh.2024.1395863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction The evolution of contraception has been crucial for public health and reproductive well-being. Over the past 60 years, combined oral contraceptives (COCs) have remained an important part of the contraceptive landscape worldwide; continued development has worked toward maintaining efficacy and improving safety. Methods Seven global experts convened to discuss the clinical relevance of the oestrogen in COCs, focusing on the impact of the new oestrogen, oestetrol (E4). Participants then commented through an online forum on the summary content and other participants' feedback. We prepared this report to describe the experts' views, their follow-up from the open forum and the evidence supporting their views. Results Ethinylestradiol (EE) and oestradiol (E2) affect receptors similarly whereas E4 has differential effects, especially in the liver and breast. Adequate oestrogen doses in COCs ensure regular bleeding and user acceptability. EE and E4 have longer half-lives than E2; accordingly, COCs with EE and E4 offer more predictable bleeding than those with E2. Oestrogen type and progestin influence VTE risk; E2 poses a lower risk than EE; although promising, E4/DRSP VTE risk is lacking population-based data. COCs alleviate menstrual symptoms, impact mental health, cognition, libido, skin, and bone health. Conclusion Oestrogens play an important role in the contraceptive efficacy, bleeding patterns, and overall tolerability/safety of COCs. Recent studies exploring E4 combined with DRSP show promising results compared to traditional formulations, but more definitive conclusions await further research.
Collapse
Affiliation(s)
- M. D. Creinin
- Department of Obstetrics and Gynecology, University of California, Davis, Sacramento, CA, United States
| | - A. Cagnacci
- Academic Unit of Obstetrics and Gynecology, DINOGMI, IRCCS-Azienda Ospedaliera Universitaria San Martino di Genova, Genova, Italy
| | - R. Z. Spaczyński
- Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| | - P. Stute
- Department of Obstetrics and Gynecology, Bern University Hospital, Bern, Switzerland
| | - N. Chabbert-Buffet
- Gynécologie—Obstétrique et Médecine de la Reproduction—Maternité, Hospital Tenon, Paris, France
| | - T. Korver
- Reprovision Clinical Consultancy, Oss, Netherlands
| | - T. Simoncini
- Division of Obstetrics and Gynecology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| |
Collapse
|
35
|
Oltra J, Segura B, Strafella AP, van Eimeren T, Ibarretxe-Bilbao N, Diez-Cirarda M, Eggers C, Lucas-Jiménez O, Monté-Rubio GC, Ojeda N, Peña J, Ruppert MC, Sala-Llonch R, Theis H, Uribe C, Junque C. A multi-site study on sex differences in cortical thickness in non-demented Parkinson's disease. NPJ Parkinsons Dis 2024; 10:69. [PMID: 38521776 PMCID: PMC10960793 DOI: 10.1038/s41531-024-00686-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
Clinical, cognitive, and atrophy characteristics depending on sex have been previously reported in Parkinson's disease (PD). However, though sex differences in cortical gray matter measures in early drug naïve patients have been described, little is known about differences in cortical thickness (CTh) as the disease advances. Our multi-site sample comprised 211 non-demented PD patients (64.45% males; mean age 65.58 ± 8.44 years old; mean disease duration 6.42 ± 5.11 years) and 86 healthy controls (50% males; mean age 65.49 ± 9.33 years old) with available T1-weighted 3 T MRI data from four international research centers. Sex differences in regional mean CTh estimations were analyzed using generalized linear models. The relation of CTh in regions showing sex differences with age, disease duration, and age of onset was examined through multiple linear regression. PD males showed thinner cortex than PD females in six frontal (bilateral caudal middle frontal, bilateral superior frontal, left precentral and right pars orbitalis), three parietal (bilateral inferior parietal and left supramarginal), and one limbic region (right posterior cingulate). In PD males, lower CTh values in nine out of ten regions were associated with longer disease duration and older age, whereas in PD females, lower CTh was associated with older age but with longer disease duration only in one region. Overall, male patients show a more widespread pattern of reduced CTh compared with female patients. Disease duration seems more relevant to explain reduced CTh in male patients, suggesting worse prognostic over time. Further studies should explore sex-specific cortical atrophy trajectories using large longitudinal multi-site data.
Collapse
Affiliation(s)
- Javier Oltra
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Faculty of Medicine, Clínic Campus, Carrer de Casanova, 143, Ala Nord, 5th floor, 08036, Barcelona, Catalonia, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Carrer del Rosselló, 149, 08036, Barcelona, Catalonia, Spain
| | - Barbara Segura
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Faculty of Medicine, Clínic Campus, Carrer de Casanova, 143, Ala Nord, 5th floor, 08036, Barcelona, Catalonia, Spain.
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Carrer del Rosselló, 149, 08036, Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Hospital Clínic Barcelona, Carrer de Villarroel, 170, 08036, Barcelona, Catalonia, Spain.
| | - Antonio P Strafella
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., M5T 1R8, Toronto, ON, Canada
- Edmond J. Safra Parkinson Disease Program, Neurology Division, Toronto Western Hospital & Krembil Brain Institute, University Health Network, University of Toronto, 399 Bathurst Street, M5T 2S8, Toronto, ON, Canada
| | - Thilo van Eimeren
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, University Medical Center Cologne, Kerpener Straße, 62, 50937, Cologne, Germany
- Department of Neurology, University Medical Center Cologne, Kerpener Straße, 62, 50937, Cologne, Germany
| | - Naroa Ibarretxe-Bilbao
- Department of Psychology, Faculty of Health Sciences, University of Deusto, Avenida de las Universidades, 24, 48007, Bilbao, Basque Country, Spain
| | - Maria Diez-Cirarda
- Department of Neurology, Hospital Clínico San Carlos, Health Research Institute 'San Carlos' (IdISCC), Complutense University of Madrid, Calle del Profesor Martín Lagos, s/n, 28040, Madrid, Spain
| | - Carsten Eggers
- Department of Neurology, University Medical Center Cologne, Kerpener Straße, 62, 50937, Cologne, Germany
- Department of Neurology, University Hospital of Giessen and Marburg, Center for Mind, Brain and Behavior, University of Marburg and Giessen Universiy, Hans-Meerwein-Straße, 6, 35043, Marburg, Germany
| | - Olaia Lucas-Jiménez
- Department of Psychology, Faculty of Health Sciences, University of Deusto, Avenida de las Universidades, 24, 48007, Bilbao, Basque Country, Spain
| | - Gemma C Monté-Rubio
- Centre for Comparative Medicine and Bioimaging (CMCiB), Gemans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916, Badalona, Catalonia, Spain
| | - Natalia Ojeda
- Department of Psychology, Faculty of Health Sciences, University of Deusto, Avenida de las Universidades, 24, 48007, Bilbao, Basque Country, Spain
| | - Javier Peña
- Department of Psychology, Faculty of Health Sciences, University of Deusto, Avenida de las Universidades, 24, 48007, Bilbao, Basque Country, Spain
| | - Marina C Ruppert
- Department of Neurology, University Hospital of Giessen and Marburg, Center for Mind, Brain and Behavior, University of Marburg and Giessen Universiy, Hans-Meerwein-Straße, 6, 35043, Marburg, Germany
| | - Roser Sala-Llonch
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Carrer del Rosselló, 149, 08036, Barcelona, Catalonia, Spain
- Department of Biomedicine, Institute of Neurosciences, University of Barcelona, Faculty of Medicine, Clínic Campus, Carrer de Casanova, 143, Ala Nord, 5th floor, 08036, Barcelona, Catalonia, Spain
- Biomedical Imaging Group, Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN: CB06/01/1039-ISCIII), Carrer de Casanova, 143, 08036, Barcelona, Catalonia, Spain
| | - Hendrik Theis
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, University Medical Center Cologne, Kerpener Straße, 62, 50937, Cologne, Germany
- Department of Neurology, University Medical Center Cologne, Kerpener Straße, 62, 50937, Cologne, Germany
| | - Carme Uribe
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., M5T 1R8, Toronto, ON, Canada
| | - Carme Junque
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Faculty of Medicine, Clínic Campus, Carrer de Casanova, 143, Ala Nord, 5th floor, 08036, Barcelona, Catalonia, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Carrer del Rosselló, 149, 08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Hospital Clínic Barcelona, Carrer de Villarroel, 170, 08036, Barcelona, Catalonia, Spain
| |
Collapse
|
36
|
Baek DC, Kang JY, Lee JS, Lee EJ, Son CG. Linking alterations in estrogen receptor expression to memory deficits and depressive behavior in an ovariectomy mouse model. Sci Rep 2024; 14:6854. [PMID: 38514828 PMCID: PMC10958029 DOI: 10.1038/s41598-024-57611-z] [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: 11/17/2023] [Accepted: 03/20/2024] [Indexed: 03/23/2024] Open
Abstract
The high risk of neurological disorders in postmenopausal women is an emerging medical issue. Based on the hypothesis of altered estrogen receptors (ERα and β) after the decline of estrogen production, we investigated the changes in ERs expressions across brain regions and depressive/amnesic behaviors. C57BL/6J female mice were ovariectomized (OVX) to establish a menopausal condition. Along with behavior tests (anxiety, depression, and memory), the expression of ERs, microglial activity, and neuronal activity was measured in six brain regions (hippocampus, prefrontal cortex, striatum, raphe nucleus, amygdala, and hypothalamus) from 4 to 12 weeks after OVX. Mice exhibited anxiety- and depressive-like behaviors, as well as memory impairment. These behavioral alterations have been linked to a suppression in the expression of ERβ. The decreased ERβ expression coincided with microglial-derived neuroinflammation, as indicated by notable activations of Ionized calcium-binding adapter molecule 1 and Interleukin-1beta. Additionally, the activity of brain-derived neurotrophic factor (BDNF), particularly in the hippocampus, decreased in a time-dependent manner from 4 to 12 weeks post-OVX. Our study provides evidence shedding light on the susceptibility to memory impairment and depression in women after menopause. This susceptibility is associated with the suppression of ERβ and alteration of ERα in six brain regions.
Collapse
Affiliation(s)
- Dong-Cheol Baek
- Institute of Bioscience & Integrative Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea
| | - Ji-Yun Kang
- Institute of Bioscience & Integrative Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea
| | - Jin-Seok Lee
- Institute of Bioscience & Integrative Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea
| | - Eun-Jung Lee
- Department of Korean Rehabilitation Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea
| | - Chang-Gue Son
- Institute of Bioscience & Integrative Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea.
| |
Collapse
|
37
|
Piñon-Teal WL, Ogilvie JM. G protein-coupled estrogen receptor expression in postnatal developing mouse retina. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1331298. [PMID: 38984123 PMCID: PMC11182193 DOI: 10.3389/fopht.2024.1331298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/13/2024] [Indexed: 07/11/2024]
Abstract
Introduction Estrogen has emerged as a multifaceted signaling molecule in the retina, playing an important role in neural development and providing neuroprotection in adults. It interacts with two receptor types: classical estrogen receptors (ERs) alpha and beta, and G protein-coupled estrogen receptor (Gper). Gper differs from classical ERs in structure, localization, and signaling. Here we provide the first report of the temporal and spatial properties of Gper transcript and protein expression in the developing and mature mouse retina. Methods We applied qRT-PCR to determine Gper transcript expression in wild type mouse retina from P0-P21. Immunohistochemistry and Western blot were used to determine Gper protein expression and localization at the same time points. Results Gper expression showed a 6-fold increase during postnatal development, peaking at P14. Relative total Gper expression exhibited a significant decrease during retinal development, although variations emerged in the timing of changes among different forms of the protein. Gper immunoreactivity was seen in retinal ganglion cells (RGCs) throughout development and also in somas in the position of horizontal cells at early time points. Immunoreactivity was observed in the cytoplasm and Golgi at all time points, in the nucleus at early time points, and in RGC axons as the retina matured. Discussion In conclusion, our study illuminates the spatial and temporal expression patterns of Gper in the developing mouse retina and provides a vital foundation for further investigations into the role of Gper in retinal development and degeneration.
Collapse
Affiliation(s)
| | - Judith Mosinger Ogilvie
- Department of Biology, Saint Louis University, St. Louis, MO, United States
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
38
|
Oltra J, Habich A, Schwarz CG, Nedelska Z, Przybelski SA, Inguanzo A, Diaz‐Galvan P, Lowe VJ, Oppedal K, Gonzalez MC, Philippi N, Blanc F, Barkhof F, Lemstra AW, Hort J, Padovani A, Rektorova I, Bonanni L, Massa F, Kramberger MG, Taylor J, Snædal JG, Walker Z, Antonini A, Dierks T, Segura B, Junque C, Westman E, Boeve BF, Aarsland D, Kantarci K, Ferreira D. Sex differences in brain atrophy in dementia with Lewy bodies. Alzheimers Dement 2024; 20:1815-1826. [PMID: 38131463 PMCID: PMC10947875 DOI: 10.1002/alz.13571] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/13/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023]
Abstract
INTRODUCTION Sex influences neurodegeneration, but it has been poorly investigated in dementia with Lewy bodies (DLB). We investigated sex differences in brain atrophy in DLB using magnetic resonance imaging (MRI). METHODS We included 436 patients from the European-DLB consortium and the Mayo Clinic. Sex differences and sex-by-age interactions were assessed through visual atrophy rating scales (n = 327; 73 ± 8 years, 62% males) and automated estimations of regional gray matter volume and cortical thickness (n = 165; 69 ± 9 years, 72% males). RESULTS We found a higher likelihood of frontal atrophy and smaller volumes in six cortical regions in males and thinner olfactory cortices in females. There were significant sex-by-age interactions in volume (six regions) and cortical thickness (seven regions) across the entire cortex. DISCUSSION We demonstrate that males have more widespread cortical atrophy at younger ages, but differences tend to disappear with increasing age, with males and females converging around the age of 75. HIGHLIGHTS Male DLB patients had higher odds for frontal atrophy on radiological visual rating scales. Male DLB patients displayed a widespread pattern of cortical gray matter alterations on automated methods. Sex differences in gray matter measures in DLB tended to disappear with increasing age.
Collapse
Affiliation(s)
- Javier Oltra
- Medical Psychology UnitDepartment of MedicineInstitute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS)BarcelonaCataloniaSpain
- Division of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Annegret Habich
- Division of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
- University Hospital of Psychiatry and Psychotherapy Bern, University of BernBernSwitzerland
| | | | - Zuzana Nedelska
- Memory ClinicDepartment of NeurologyCharles University2nd Faculty of Medicine and Motol University HospitalPragueCzech Republic
| | | | - Anna Inguanzo
- Division of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | | | - Val J. Lowe
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | - Ketil Oppedal
- Center for Age‐Related MedicineStavanger University HospitalStavangerNorway
- Stavanger Medical Imaging Laboratory (SMIL)Department of RadiologyStavanger University HospitalStavangerNorway
- The Norwegian Centre for Movement DisordersStavanger University HospitalStavangerNorway
| | - Maria C. Gonzalez
- Center for Age‐Related MedicineStavanger University HospitalStavangerNorway
- Stavanger Medical Imaging Laboratory (SMIL)Department of RadiologyStavanger University HospitalStavangerNorway
- The Norwegian Centre for Movement DisordersStavanger University HospitalStavangerNorway
- Department of Quality and Health TechnologyFaculty of Health SciencesUniversity of StavangerStavangerNorway
| | - Nathalie Philippi
- Geriatrics and Neurology UnitsResearch and Resources Memory Center (CM2R)Hôpitaux Universitaires de StrasbourgStrasbourgFrance
- ICube Laboratory (CNRS, UMR 7357)StrasbourgFrance
| | - Frederic Blanc
- Geriatrics and Neurology UnitsResearch and Resources Memory Center (CM2R)Hôpitaux Universitaires de StrasbourgStrasbourgFrance
- ICube Laboratory (CNRS, UMR 7357)StrasbourgFrance
| | - Frederik Barkhof
- Department of Radiology & Nuclear Medicine (AMC)Amsterdam UMC, Vrije UniversiteitAmsterdamthe Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing (CMIC)University College LondonLondonUK
| | - Afina W. Lemstra
- Alzheimer Center AmsterdamNeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VumcAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VumcAmsterdamThe Netherlands
| | - Jakub Hort
- Memory ClinicDepartment of NeurologyCharles University2nd Faculty of Medicine and Motol University HospitalPragueCzech Republic
| | - Alessandro Padovani
- Neurology UnitDepartment of Clinical and Experimental Sciences (DSCS)University of BresciaBresciaItaly
| | - Irena Rektorova
- Brain and Mind ResearchCentral European Institute of Technology (CEITET)Masaryk UniversityBrnoCzech Republic
| | - Laura Bonanni
- Department of Medicine and Aging Sciences University G. d'Annunzio of Chieti‐Pescara ChietiChietiItaly
| | - Federico Massa
- Department of NeuroscienceRehabilitationOphthalmology, Genetics, Maternal and Child HealthUniversity of GenovaGenovaItaly
| | | | - John‐Paul Taylor
- Translational and Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | | | - Zuzana Walker
- Division of PsychiatryUniversity College LondonLondonUK
- St Margaret's HospitalEssex Partnership University NHS Foundation TrustEssexUK
| | - Angelo Antonini
- Parkinson and Movement Disorders UnitStudy Center on Neurodegeneration (CESNE)PadovaItaly
| | - Thomas Dierks
- University Hospital of Psychiatry and Psychotherapy Bern, University of BernBernSwitzerland
| | - Barbara Segura
- Medical Psychology UnitDepartment of MedicineInstitute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS)BarcelonaCataloniaSpain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaCataloniaSpain
| | - Carme Junque
- Medical Psychology UnitDepartment of MedicineInstitute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS)BarcelonaCataloniaSpain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaCataloniaSpain
| | - Eric Westman
- Division of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | | | - Dag Aarsland
- Center for Age‐Related MedicineStavanger University HospitalStavangerNorway
- Department of Old Age PsychiatryInstitute of PsychiatryPsychology & Neuroscience (IoPPN)King's College LondonLondonUK
| | | | - Daniel Ferreira
- Division of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
- Facultad de Ciencias de la SaludUniversidad Fernando Pessoa CanariasLas PalmasEspaña
| |
Collapse
|
39
|
Zhao J, He C, Fan X, Wang L, Zhao L, Liu H, Shen W, Jiang S, Pei K, Gao J, Qi Y, Liu Y, Zhao J, Zhang R, Lu C, Tong J, Huai J. Tripeptidyl peptidase II coordinates the homeostasis of calcium and lipids in the central nervous system and its depletion causes presenile dementia in female mice through calcium/lipid dyshomeostasis-induced autophagic degradation of CYP19A1. Theranostics 2024; 14:1390-1429. [PMID: 38389851 PMCID: PMC10879859 DOI: 10.7150/thno.92571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: Tripeptidyl peptidase II (TPP2) has been proven to be related to human immune and neurological diseases. It is generally considered as a cytosolic protein which forms the largest known protease complex in eukaryotic cells to operate mostly downstream of proteasomes for degradation of longer peptides. However, this canonical function of TPP2 cannot explain its role in a wide variety of biological and pathogenic processes. The mechanistic interrelationships and hierarchical order of these processes have yet to be clarified. Methods: Animals, cells, plasmids, and viruses established and/or used in this study include: TPP2 knockout mouse line, TPP2 conditional knockout mouse lines (different neural cell type oriented), TRE-TPP2 knockin mouse line on the C57BL/6 background; 293T cells with depletion of TPP2, ATF6, IRE1, PERK, SYVN1, UCHL1, ATG5, CEPT1, or CCTα, respectively; 293T cells stably expressing TPP2, TPP2 S449A, TPP2 S449T, or CCTα-KDEL proteins on the TPP2-depleted background; Plasmids for eukaryotic transient expression of rat CYP19A1-Flag, CYP19A1 S118A-Flag, CYP19A1 S118D-Flag, Sac I ML GFP Strand 11 Long, OMMGFP 1-10, G-CEPIA1er, GCAMP2, CEPIA3mt, ACC-GFP, or SERCA1-GFP; AAV2 carrying the expression cassette of mouse CYP19A1-3 X Flag-T2A-ZsGreen. Techniques used in this study include: Flow cytometry, Immunofluorescence (IF) staining, Immunohistochemical (IHC) staining, Luxol fast blue (LFB) staining, β-galactosidase staining, Lipid droplet (LD) staining, Calcium (Ca2+) staining, Stimulated emission depletion (STED) imaging, Transmission electron microscopic imaging, Two-photon imaging, Terminal deoxynucleotidyl transferase (TdT) dUTP nick-end Labeling (TUNEL) assay, Bromodeoxyuridine (BrdU) assay, Enzymatic activity assay, Proximity ligation assay (PLA), In vivo electrophysiological recording, Long-term potentiation (LTP) recording, Split-GFP-based mitochondria-associated membrane (MAM) detection, Immunoprecipitation (IP), Cellular fractionation, In situ hybridization, Semi-quantitative RT-PCR, Immunoblot, Mass spectrometry-based lipidomics, metabolomics, proteomics, Primary hippocampal neuron culture and Morris water maze (MWM) test. Results: We found that TPP2, independent of its enzymatic activity, plays a crucial role in maintaining the homeostasis of intracellular Ca2+ and phosphatidylcholine (PC) in the central nervous system (CNS) of mice. In consistence with the critical importance of Ca2+ and PC in the CNS, TPP2 gene ablation causes presenile dementia in female mice, which is closely associated with Ca2+/PC dysregulation-induced endoplasmic reticulum (ER) stress, abnormal autophagic degradation of CYP19A1 (aromatase), and estrogen depletion. This work therefore uncovers a new role of TPP2 in lipogenesis and neurosteroidogenesis which is tightly related to cognitive function of adult female mice. Conclusion: Our study reveals a crucial role of TPP2 in controlling homeostasis of Ca2+ and lipids in CNS, and its deficiency causes sexual dimorphism in dementia. Thus, this study is not only of great significance for elucidating the pathogenesis of dementia and its futural treatment, but also for interpreting the role of TPP2 in other systems and their related disorders.
Collapse
Affiliation(s)
- Jin Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Chengtong He
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Xueyu Fan
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Lin Wang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Liao Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Hui Liu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Wujun Shen
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Sanwei Jiang
- Henan International Key Laboratory for Noninvasive Neuromodulation, Department of Physiology & Pathology, Xinxiang Medical University, Xinxiang, PR China
| | - Kaixuan Pei
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Jingjing Gao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Yawei Qi
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Yang Liu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Junqiang Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
| | - Ruiling Zhang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
| | - Chengbiao Lu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
- Henan International Key Laboratory for Noninvasive Neuromodulation, Department of Physiology & Pathology, Xinxiang Medical University, Xinxiang, PR China
- Senior author for electrophysiological experiments and related analysis
| | - Jia Tong
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Jisen Huai
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| |
Collapse
|
40
|
Velikic G, Maric DM, Maric DL, Supic G, Puletic M, Dulic O, Vojvodic D. Harnessing the Stem Cell Niche in Regenerative Medicine: Innovative Avenue to Combat Neurodegenerative Diseases. Int J Mol Sci 2024; 25:993. [PMID: 38256066 PMCID: PMC10816024 DOI: 10.3390/ijms25020993] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Regenerative medicine harnesses the body's innate capacity for self-repair to restore malfunctioning tissues and organs. Stem cell therapies represent a key regenerative strategy, but to effectively harness their potential necessitates a nuanced understanding of the stem cell niche. This specialized microenvironment regulates critical stem cell behaviors including quiescence, activation, differentiation, and homing. Emerging research reveals that dysfunction within endogenous neural stem cell niches contributes to neurodegenerative pathologies and impedes regeneration. Strategies such as modifying signaling pathways, or epigenetic interventions to restore niche homeostasis and signaling, hold promise for revitalizing neurogenesis and neural repair in diseases like Alzheimer's and Parkinson's. Comparative studies of highly regenerative species provide evolutionary clues into niche-mediated renewal mechanisms. Leveraging endogenous bioelectric cues and crosstalk between gut, brain, and vascular niches further illuminates promising therapeutic opportunities. Emerging techniques like single-cell transcriptomics, organoids, microfluidics, artificial intelligence, in silico modeling, and transdifferentiation will continue to unravel niche complexity. By providing a comprehensive synthesis integrating diverse views on niche components, developmental transitions, and dynamics, this review unveils new layers of complexity integral to niche behavior and function, which unveil novel prospects to modulate niche function and provide revolutionary treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Gordana Velikic
- Department for Research and Development, Clinic Orto MD-Parks Dr. Dragi Hospital, 21000 Novi Sad, Serbia
- Hajim School of Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Dusan M. Maric
- Department for Research and Development, Clinic Orto MD-Parks Dr. Dragi Hospital, 21000 Novi Sad, Serbia
- Faculty of Stomatology Pancevo, University Business Academy, 26000 Pancevo, Serbia;
| | - Dusica L. Maric
- Department of Anatomy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Gordana Supic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Miljan Puletic
- Faculty of Stomatology Pancevo, University Business Academy, 26000 Pancevo, Serbia;
| | - Oliver Dulic
- Department of Surgery, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia;
| | - Danilo Vojvodic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| |
Collapse
|
41
|
Abramiuk M, Mertowska P, Frankowska K, Świechowska-Starek P, Satora M, Polak G, Dymanowska-Dyjak I, Grywalska E. How Can Selected Dietary Ingredients Influence the Development and Progression of Endometriosis? Nutrients 2024; 16:154. [PMID: 38201982 PMCID: PMC10781184 DOI: 10.3390/nu16010154] [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: 11/18/2023] [Revised: 12/30/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
Endometriosis is a chronic, hormone-dependent disease characterized by the presence of endometrial tissue in ectopic locations. Since the treatment options for this disease are still limited, and the cure rate is unsatisfactory, the search for ways to treat symptoms and modify the course of the disease is of key importance in improving the quality of life of patients with endometriosis. So far, the literature has shown that nutrition can influence endometriosis through hormonal modification and altering the inflammatory or oxidative response. Since the importance of nutrition in this disease is still a subject of scientific research, we aimed to summarize the current knowledge on the role of dietary modifications in endometriosis. Our review showed that nutrients with anti-inflammatory and antioxidant properties, including most vitamins and several trace elements, may influence the pathogenesis of endometriosis and can be considered as the nutrients preventing the development of endometriosis. However, despite the many discoveries described in this review, further interdisciplinary research on this topic seems to be extremely important, as in the future, it may result in the development of personalized therapies supporting the treatment of endometriosis.
Collapse
Affiliation(s)
- Monika Abramiuk
- Independent Laboratory of Minimally Invasive Gynaecology and Gynaecological Endocrinology, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland; (G.P.); (I.D.-D.)
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Karolina Frankowska
- 1st Chair and Department of Oncological Gynecology and Gynecology, Students’ Scientific Association, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland; (K.F.); (M.S.)
| | - Paulina Świechowska-Starek
- 1st Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland;
| | - Małgorzata Satora
- 1st Chair and Department of Oncological Gynecology and Gynecology, Students’ Scientific Association, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland; (K.F.); (M.S.)
| | - Grzegorz Polak
- Independent Laboratory of Minimally Invasive Gynaecology and Gynaecological Endocrinology, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland; (G.P.); (I.D.-D.)
| | - Izabela Dymanowska-Dyjak
- Independent Laboratory of Minimally Invasive Gynaecology and Gynaecological Endocrinology, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland; (G.P.); (I.D.-D.)
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland; (P.M.); (E.G.)
| |
Collapse
|
42
|
Kuck MJ, Begde A, Hawkins K, Hogervorst E. Alzheimer's Disease and (Phyto) Estrogen Treatment: Modification of Effects by Age, Type of Treatment, and Duration of Use. J Alzheimers Dis 2024; 101:S217-S234. [PMID: 39422947 DOI: 10.3233/jad-231415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Background There is a continued debate on whether menopausal hormone therapy (MHT) protects women against Alzheimer's disease (AD). It is also unclear whether phytoestrogen could be an alternative treatment for AD. Objective To investigate whether mixed study findings may be due to differences in age at initiation of MHT and duration of prescription of different types of MHT using meta-analyses. Methods After a systematic literature search, meta-analyses were carried out using Cochrane Revman 5.4.1.software including data from large nationwide studies of registered medically diagnosed AD and prescribed MHT. These analyses were stratified for duration and type of treatment, by age at start of prescription of therapy. Insufficient quality data were available for phytoestrogen treatment and AD meta-analyses. Results A total of 912,157 women were included from five registries, of whom 278,495 had developed AD during follow-up. Meta-analyses suggested a small increased AD risk after 5-10 years prescription of combination MHT regardless of age, and over 10 years only in women younger than 60 years of age. No association was seen for estrogen alone for women younger than 60 years of age, but AD risk did increase for women over 60 years of age for up to 5 years of MHT prescriptions. Conclusions Combination MHT should probably be prescribed for less than 5 years after menopause to reduce risk for AD, while estrogen alone should not be prescribed to women over 60. For phytoestrogen, small treatment trials suggested some benefit of tempeh (fermented soy), which should be investigated further.
Collapse
Affiliation(s)
- M J Kuck
- School of Sports Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Ahmet Begde
- School of Sports Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Katie Hawkins
- School of Sports Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Eef Hogervorst
- School of Sports Exercise and Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
43
|
Li Q, Wang L, Jia Y, Yang M, Zhang H, Hu J. Nontargeted Analysis Reveals a Broad Range of Bioactive Pollutants in Drinking Water by Estrogen Receptor Affinity-Mass Spectrometry. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:21327-21336. [PMID: 38059695 DOI: 10.1021/acs.est.3c05060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Exposure to environmental endocrine-disrupting chemicals (EDCs) can cause extensive health issues. However, specific EDCs remain elusive. This work aimed at performing nontargeted identification of estrogen receptor α (ERα)-active compounds using an ERα protein affinity assay combined with high-resolution mass spectrometry in the source and drinking water sampled from major rivers in China. Fifty-one potential ERα-active compounds across 13 categories were identified. For the first time, diisodecyl phenyl phosphate was found to have antiestrogenic activity, and three chemicals (galaxolidone, bensulfuron methyl, and UV234) were plausible ERα ligands. Among the 51 identified compounds, 12 were detected in the aquatic environment for the first time, and the concentration of N-phenyl-2-naphthylamine, a widely used antioxidant in rubber products, was up to 1469 and 1190 ng/L in source and drinking water, respectively. This study demonstrated the widespread presence of known and unknown ERα estrogenic and antiestrogenic pollutants in the major rivers that serve as key sources of drinking water in China and the low removal efficiency of these chemicals in drinking water treatment plants.
Collapse
Affiliation(s)
- Qiang Li
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Lei Wang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Yingting Jia
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Min Yang
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Haifeng Zhang
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jianying Hu
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
44
|
Zeidan RS, McElroy T, Rathor L, Martenson MS, Lin Y, Mankowski RT. Sex differences in frailty among older adults. Exp Gerontol 2023; 184:112333. [PMID: 37993077 DOI: 10.1016/j.exger.2023.112333] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/24/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023]
Abstract
By definition, aging is a natural, gradual and continuous process. On the other hand, frailty reflects the increase in vulnerability to stressors and shortens the time without disease (health span) while longevity refers to the length of life (lifespan). The average life expectancy has significantly increased during the last few decades. A longer lifespan has been accompanied by an increase in frailty and decreased independence in older adults, with major differences existing between men and women. For example, women tend to live longer than men but also experience higher rates of frailty and disability. Sex differences prevent optimization of lifestyle interventions and therapies to effectively prevent frailty. Sex differences in frailty and aging are rooted in a complex interplay between uncontrollable (genetic, epigenetic, physiological), and controllable factors (psychosocial and lifestyle factors). Thus, understanding the underlying causes of sex differences in frailty and aging is essential for developing personalized interventions to promote healthy aging and improve quality of life in older men and women. In this review, we have discussed the key contributors and knowledge gaps related to sex differences in aging and frailty.
Collapse
Affiliation(s)
- Rola S Zeidan
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America; Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Taylor McElroy
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America; Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Laxmi Rathor
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Matthew S Martenson
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Yi Lin
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Robert T Mankowski
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| |
Collapse
|
45
|
Kraemer RR, Kraemer BR. The effects of peripheral hormone responses to exercise on adult hippocampal neurogenesis. Front Endocrinol (Lausanne) 2023; 14:1202349. [PMID: 38084331 PMCID: PMC10710532 DOI: 10.3389/fendo.2023.1202349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Over the last decade, a considerable amount of new data have revealed the beneficial effects of exercise on hippocampal neurogenesis and the maintenance or improvement of cognitive function. Investigations with animal models, as well as human studies, have yielded novel understanding of the mechanisms through which endocrine signaling can stimulate neurogenesis, as well as the effects of exercise on acute and/or chronic levels of these circulating hormones. Considering the effects of aging on the decline of specific endocrine factors that affect brain health, insights in this area of research are particularly important. In this review, we discuss how different forms of exercise influence the peripheral production of specific endocrine factors, with particular emphasis on brain-derived neurotrophic factor, growth hormone, insulin-like growth factor-1, ghrelin, estrogen, testosterone, irisin, vascular endothelial growth factor, erythropoietin, and cortisol. We also describe mechanisms through which these endocrine responses to exercise induce cellular changes that increase hippocampal neurogenesis and improve cognitive function.
Collapse
Affiliation(s)
- Robert R. Kraemer
- Department of Kinesiology and Health Studies, Southeastern Louisiana University, Hammond, LA, United States
| | - Bradley R. Kraemer
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL, United States
| |
Collapse
|
46
|
Karaca E, Yarim M. Naringenin stimulates aromatase expression and alleviates the clinical and histopathological findings of experimental autoimmune encephalomyelitis in C57bl6 mice. Histochem Cell Biol 2023; 160:477-490. [PMID: 37378907 DOI: 10.1007/s00418-023-02217-1] [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] [Accepted: 06/02/2023] [Indexed: 06/29/2023]
Abstract
This study was conducted to demonstrate the possible protective and therapeutic effects of naringenin, an estrogenically effective flavonoid, in experimental autoimmune encephalomyelitis (EAE), which is the rodent model of multiple sclerosis. For this purpose, 50 12-week-old C57BL6 male mice were divided into five groups; control, naringenin, EAE, prophylactic naringenin + EAE, and EAE + therapeutic naringenin. The EAE model was induced with myelin oligodendrocyte glycoprotein(35-55), and naringenin (50 mg/kg) was administered by oral gavage. The prophylactic and therapeutic effects of naringenin were examined according to clinical, histopathological, immunohistochemical, electron microscopic, and RT-PCR (aromatase, 3βHSD, estrogen receptors, and progesterone receptor expression) parameters. The acute EAE model was successfully induced, along with its clinical and histopathological findings. RT-PCR showed that expression of aromatase, 3βHSD, estrogen receptor-β, and progesterone receptor gene decreased, while estrogen receptor-α increased after EAE induction. Electron microscopic analysis showed mitochondrial damage and degenerative changes in myelinated axons and neurons in EAE, which could be behind the downregulation in the expressions of neurosteroid enzymes. Aromatase immunopositivity rates also decreased in EAE, while estrogen receptor α and β, and progesterone receptor immunopositivity rates increased. Naringenin improved aromatase immunopositivity rates and gene expression in both prophylactic and therapeutic use. Clinical and histopathological findings revealed that EAE findings were alleviated in both prophylactic and therapeutic groups, along with significantly decreased inflammatory cell infiltrations in the white matter of the spinal cords. In conclusion, naringenin could provide long-term beneficial effects even in prophylactic use due to stimulating aromatase expression, but it could not prevent or eliminate the EAE model's lesions completely.
Collapse
Affiliation(s)
- Efe Karaca
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, 55200, Atakum, Samsun, Turkey.
| | - Murat Yarim
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, 55200, Atakum, Samsun, Turkey
| |
Collapse
|
47
|
Quan Y, Xu J, Xu Q, Guo Z, Ou R, Shang H, Wei Q. Association between the risk and severity of Parkinson's disease and plasma homocysteine, vitamin B12 and folate levels: a systematic review and meta-analysis. Front Aging Neurosci 2023; 15:1254824. [PMID: 37941998 PMCID: PMC10628521 DOI: 10.3389/fnagi.2023.1254824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/25/2023] [Indexed: 11/10/2023] Open
Abstract
Background Parkinson's disease (PD) is recognized as the second most prevalent progressive neurodegenerative disease among the elderly. However, the relationship between PD and plasma homocysteine (Hcy), vitamin B12, and folate has yielded inconsistent results in previous studies. Hence, in order to address this ambiguity, we conducted a meta-analysis to summarize the existing evidence. Methods Suitable studies published prior to May 2023 were identified by searching PubMed, EMBASE, Medline, Ovid, and Web of Science. The methodological quality of eligible studies was assessed using the Newcastle-Ottawa Quality Assessment Scale (NOS). Meta-analysis and publication bias were then performed using R version 4.3.1. Results The results of our meta-analysis, consisting of case-control and cross-sectional studies, showed that PD patients had lower folate and vitamin B12 levels (SMD [95%CI]: -0.30[-0.39, -0.22], p < 0.001 for Vitamin B12; SMD [95%CI]: -0.20 [-0.28, -0.13], p < 0.001 for folate), but a significant higher Hcy level (SMD [95%CI]: 0.86 [0.59, 1.14], p < 0.001) than healthy people. Meanwhile, PD was significantly related to hyperhomocysteinemia (SMD [95%]: 2.02 [1.26, 2.78], p < 0.001) rather than plasma Hcy below 15 μmol/L (SMD [95%]: -0.31 [-0.62, 0.00], p = 0.05). Subgroup analysis revealed associations between the Hcy level of PD patients and region (p = 0.03), age (p = 0.03), levodopa therapy (p = 0.03), Hoehn and Yahr stage (p < 0.001), and cognitive impairment (p < 0.001). However, gender (p = 0.38) and sample size (p = 0.49) were not associated. Conclusion Hcy, vitamin B12, and folic acid potentially predict the onset and development of PD. Additionally, multiple factors were linked to Hcy levels in PD patients. Further studies are needed to comprehend their roles in PD.
Collapse
Affiliation(s)
- Yuxin Quan
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jisen Xu
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qing Xu
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiqing Guo
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
48
|
Raposo M, Hübener-Schmid J, Ferreira AF, Vieira Melo AR, Vasconcelos J, Pires P, Kay T, Garcia-Moreno H, Giunti P, Santana MM, Pereira de Almeida L, Infante J, van de Warrenburg BP, de Vries JJ, Faber J, Klockgether T, Casadei N, Admard J, Schöls L, Riess O, Lima M. Blood transcriptome sequencing identifies biomarkers able to track disease stages in spinocerebellar ataxia type 3. Brain 2023; 146:4132-4143. [PMID: 37071051 DOI: 10.1093/brain/awad128] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/19/2022] [Accepted: 03/27/2023] [Indexed: 04/19/2023] Open
Abstract
Transcriptional dysregulation has been described in spinocerebellar ataxia type 3/Machado-Joseph disease (SCA3/MJD), an autosomal dominant ataxia caused by a polyglutamine expansion in the ataxin-3 protein. As ataxin-3 is ubiquitously expressed, transcriptional alterations in blood may reflect early changes that start before clinical onset and might serve as peripheral biomarkers in clinical and research settings. Our goal was to describe enriched pathways and report dysregulated genes, which can track disease onset, severity or progression in carriers of the ATXN3 mutation (pre-ataxic subjects and patients). Global dysregulation patterns were identified by RNA sequencing of blood samples from 40 carriers of ATXN3 mutation and 20 controls and further compared with transcriptomic data from post-mortem cerebellum samples of MJD patients and controls. Ten genes-ABCA1, CEP72, PTGDS, SAFB2, SFSWAP, CCDC88C, SH2B1, LTBP4, MEG3 and TSPOAP1-whose expression in blood was altered in the pre-ataxic stage and simultaneously, correlated with ataxia severity in the overt disease stage, were analysed by quantitative real-time PCR in blood samples from an independent set of 170 SCA3/MJD subjects and 57 controls. Pathway enrichment analysis indicated the Gαi signalling and the oestrogen receptor signalling to be similarly affected in blood and cerebellum. SAFB2, SFSWAP and LTBP4 were consistently dysregulated in pre-ataxic subjects compared to controls, displaying a combined discriminatory ability of 79%. In patients, ataxia severity was associated with higher levels of MEG3 and TSPOAP1. We propose expression levels of SAFB2, SFSWAP and LTBP4 as well as MEG3 and TSPOAP1 as stratification markers of SCA3/MJD progression, deserving further validation in longitudinal studies and in independent cohorts.
Collapse
Affiliation(s)
- Mafalda Raposo
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Faculdade de Ciências e Tecnologia, Universidade dos Açores, 9500-321 Ponta Delgada, Portugal
| | - Jeannette Hübener-Schmid
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, 72072 Tübingen, Germany
| | - Ana F Ferreira
- Faculdade de Ciências e Tecnologia, Universidade dos Açores, 9500-321 Ponta Delgada, Portugal
| | - Ana Rosa Vieira Melo
- Faculdade de Ciências e Tecnologia, Universidade dos Açores, 9500-321 Ponta Delgada, Portugal
| | - João Vasconcelos
- Serviço de Neurologia, Hospital do Divino Espírito Santo, 9500-370 Ponta Delgada, Portugal
| | - Paula Pires
- Serviço de Neurologia, Hospital do Santo Espírito da Ilha Terceira, 9700-049 Angra do Heroísmo, Portugal
| | - Teresa Kay
- Serviço de Genética Clínica, Hospital D. Estefânia, 1169-045 Lisboa, Portugal
| | - Hector Garcia-Moreno
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Department of Neurogenetics, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK
| | - Paola Giunti
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Department of Neurogenetics, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK
| | - Magda M Santana
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3000-075, Portugal
| | - Luis Pereira de Almeida
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3000-075, Portugal
| | - Jon Infante
- Neurology Service, University Hospital Marqués de Valdecilla-IDIVAL, Universidad de Cantabria, Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Bart P van de Warrenburg
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, 6525 EN Nijmegen, The Netherlands
| | - Jeroen J de Vries
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9700 AD Groningen, The Netherlands
| | - Jennifer Faber
- Department of Neurology, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Thomas Klockgether
- Department of Neurology, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
- NGS Competence Center Tübingen, 72016 Tübingen, Germany
| | - Jakob Admard
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
- NGS Competence Center Tübingen, 72016 Tübingen, Germany
| | - Ludger Schöls
- Department for Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center for Neurology, University of Tübingen, 72016 Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), 72016 Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, 72072 Tübingen, Germany
- NGS Competence Center Tübingen, 72016 Tübingen, Germany
| | - Manuela Lima
- Faculdade de Ciências e Tecnologia, Universidade dos Açores, 9500-321 Ponta Delgada, Portugal
| |
Collapse
|
49
|
Zhang C, Niu JG, Kong XR, Mi XJ, Liu Q, Chen FF, Rong WF, Liu J. G protein-coupled estrogen receptor 1 deficiency impairs adult hippocampal neurogenesis in mice with schizophrenia. J Chem Neuroanat 2023; 132:102319. [PMID: 37495162 DOI: 10.1016/j.jchemneu.2023.102319] [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: 04/18/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 07/28/2023]
Abstract
OBJECTIVE This study aimed to confirm that G protein-coupled estrogen receptor 1 (GPER1) deficiency affects cognitive function by reducing hippocampal neurogenesis via the PKA/ERK/IGF-I signaling pathway in mice with schizophrenia (SZ). METHODS Mice were divided into four groups, namely, KO Con, WT Con, KO Con, and WT SZ (n = 12 in each group). All mice were accustomed to the behavioral equipment overnight in the testing service room. The experimental conditions were consistent with those in the animal house. Forced swimming test and Y-maze test were conducted. Neuronal differentiation and maturation were detected using immunofluorescence and confocal imaging. The protein in the PKA/ERK/IGF-I signaling pathway was tested using Western blot analysis. RESULTS GPER1 KO aggravated depression during forced swimming test and decreased cognitive ability during Y-maze test in the mouse model of dizocilpine maleate (MK-801)-induced SZ. Immunofluorescence and confocal imaging results demonstrated that GPER1 knockout reduced adult hippocampal dentate gyrus neurogenesis. Furthermore, GPER1-KO aggravated the hippocampal damage induced by MK-801 in mice through the PKA/ERK/IGF-I signaling pathway. CONCLUSIONS GPER1 deficiency reduced adult hippocampal neurogenesis and neuron survival by regulating the PKA/ERK/IGF-I signaling pathway in the MK-801-induced mouse model of SZ.
Collapse
Affiliation(s)
- Chun Zhang
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan 750004, China
| | - Jian-Guo Niu
- Department of Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xue-Rui Kong
- Department of Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xiao-Juan Mi
- Department of Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Qiang Liu
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei-Fei Chen
- Department of Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Wei-Fang Rong
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan 750004, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Juan Liu
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| |
Collapse
|
50
|
Ramli NZ, Yahaya MF, Mohd Fahami NA, Abdul Manan H, Singh M, Damanhuri HA. Brain volumetric changes in menopausal women and its association with cognitive function: a structured review. Front Aging Neurosci 2023; 15:1158001. [PMID: 37818479 PMCID: PMC10561270 DOI: 10.3389/fnagi.2023.1158001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 09/04/2023] [Indexed: 10/12/2023] Open
Abstract
The menopausal transition has been proposed to put women at risk for undesirable neurological symptoms, including cognitive decline. Previous studies suggest that alterations in the hormonal milieu modulate brain structures associated with cognitive function. This structured review provides an overview of the relevant studies that have utilized MRI to report volumetric differences in the brain following menopause, and its correlations with the evaluated cognitive functions. We performed an electronic literature search using Medline (Ovid) and Scopus to identify studies that assessed the influence of menopause on brain structure with MRI. Fourteen studies met the inclusion criteria. Brain volumetric differences have been reported most frequently in the frontal and temporal cortices as well as the hippocampus. These regions are important for higher cognitive tasks and memory. Additionally, the deficit in verbal and visuospatial memory in postmenopausal women has been associated with smaller regional brain volumes. Nevertheless, the limited number of eligible studies and cross-sectional study designs warrant further research to draw more robust conclusions.
Collapse
Affiliation(s)
- Nur Zuliani Ramli
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nur Azlina Mohd Fahami
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Hanani Abdul Manan
- Functional Image Processing Laboratory, Department of Radiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Hanafi Ahmad Damanhuri
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|