1
|
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
|
2
|
Batallas D, Gallego JJ, Casanova-Ferrer F, Fiorillo A, Rivas-Diaz P, López-Gramaje A, Arenas YM, Aparicio L, Escudero-García D, Durbán L, Rios MP, Benlloch S, Urios A, Hidalgo V, Montoliu C, Salvador A. Blood ammonia and eye-hand coordination negatively affect health-related quality of life in women with minimal hepatic encephalopathy. Qual Life Res 2025:10.1007/s11136-025-03920-3. [PMID: 40167844 DOI: 10.1007/s11136-025-03920-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE Minimal hepatic encephalopathy (MHE) is common in cirrhosis, leading to cognitive impairment and eye-hand coordination (EHC) alterations. Hyperammonemia plays a key role in MHE, contributing to motor and cognitive deficits. Elevated blood ammonia levels and impaired EHC correlate with neuropsychiatric dysfunction, yet their direct impact on health-related quality of life (HRQoL) is complex. This study examines the associations between blood ammonia, EHC, and HRQoL, and the moderating influence of sex on these associations. METHODS Eighty-seven cirrhotic patients (67 male) and 23 healthy volunteers (11 male), aged 44-80 years, performed the Psychometric Hepatic Encephalopathy Score (PHES) for MHE diagnosis, the Vienna Test System, bimanual and visuomotor tests, and completed the SF-36 questionnaire to measure HRQoL. Blood samples were taken to test ammonia levels. RESULTS Results indicated a significant association between elevated blood ammonia and impaired EHC among cirrhotic patients. However, increased blood ammonia and EHC did not directly predict HRQoL. Moderated moderation analysis revealed that women with MHE showed greater sensitivity to hyperammonemia and EHC deficits in tasks requiring fine motor control and stability skills (aiming, tapping, and bimanual coordination), which were linked to lower HRQoL in both physical and mental domains. In women without MHE, alterations in linear tracking were linked to worse HRQoL. These effects were not observed in men. CONCLUSIONS The findings underscore the sex-specific impacts of MHE, with women disproportionately affected by ammonia-related motor impairments and their subsequent influence on HRQoL. These results could contribute to developing targeted strategies to improve outcomes in this vulnerable population.
Collapse
Affiliation(s)
- Daniela Batallas
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
| | - Juan José Gallego
- INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
- Department of Pathology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain
| | | | | | - Pablo Rivas-Diaz
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
| | - Adrià López-Gramaje
- INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
- Department of Pathology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain
| | - Yaiza M Arenas
- INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
- Department of Pathology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain
| | - Luis Aparicio
- Anatomy and Embryology Department, University of Valencia, 46010, Valencia, Spain
| | - Desamparados Escudero-García
- Servicio de Medicina Digestiva, Departamento de Medicina, Hospital Clínico Universitario de Valencia, Universidad de Valencia, 46010, Valencia, Spain
| | - Lucía Durbán
- Servicio de Medicina Digestiva, Hospital Arnau de Vilanova, 46015, Valencia, Spain
| | - María-Pilar Rios
- Servicio de Medicina Digestiva, Hospital Arnau de Vilanova, 46015, Valencia, Spain
| | - Salvador Benlloch
- Servicio de Medicina Digestiva, Hospital Arnau de Vilanova, 46015, Valencia, Spain
- Universidad Cardenal Herrera-CEU Universities, 46115, Valencia, Spain
- CIBERehd, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Amparo Urios
- INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| | - Vanesa Hidalgo
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
- Department of Psychology and Sociology, Area of Psychobiology, University of Zaragoza, Teruel, Spain
| | - Carmina Montoliu
- INCLIVA Biomedical Research Institute, 46010, Valencia, Spain.
- Department of Pathology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain.
| | - Alicia Salvador
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, 28029, Madrid, Spain
| |
Collapse
|
3
|
Fleck L, Buss C, Bauer M, Stein M, Mekle R, Kock L, Klawitter H, Godara M, Ramler J, Entringer S, Endres M, Heim C. Early-Life Adversity Predicts Markers of Aging-Related Neuroinflammation, Neurodegeneration, and Cognitive Impairment in Women. Ann Neurol 2025; 97:642-656. [PMID: 39786167 PMCID: PMC11889533 DOI: 10.1002/ana.27161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/24/2024] [Accepted: 11/29/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVE Despite the overwhelming evidence for profound and longstanding effects of early-life stress (ELS) on inflammation, brain structure, and molecular aging, its impact on human brain aging and risk for neurodegenerative disease is poorly understood. We examined the impact of ELS severity in interaction with age on blood-based markers of neuroinflammation and neurodegeneration, brain volumes, and cognitive function in middle-aged women. METHODS We recruited 179 women (aged 30-60 years) with and without ELS exposure before the onset of puberty. Using Simoa technology, we assessed blood-based markers of neuroinflammation and neurodegeneration, including serum concentrations of glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL). We further obtained T1-weighted and T2-weighted magnetic resonance images to assess brain volumes and we assessed cognitive performance sensitive to early impairments associated with the development of dementia, using the Cambridge Neuropsychological Automated Test Battery. We used generalized additive models to examine nonlinear interaction effects of ELS severity and age on these outcomes. RESULTS Analyses revealed significant nonlinear interaction effects of ELS severity and age on NfL and GFAP serum concentrations, total and subcortical gray matter volume loss, increased third ventricular volume, and cognitive impairment. INTERPRETATION These findings suggest that ELS profoundly exacerbates peripheral, neurostructural, and cognitive markers of brain aging. Our results are critical for the development of novel early prevention strategies that target the impact of developmental stress on the brain to mitigate aging-related neurological diseases. ANN NEUROL 2025;97:642-656.
Collapse
Affiliation(s)
- Lara Fleck
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Claudia Buss
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
- Development, Health, and Disease Research ProgramUniversity of California, IrvineOrangeCA
- German Center for Mental HealthBerlinGermany
- German Center for Child and Adolescent HealthBerlinGermany
| | - Martin Bauer
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinExperimental and Clinical Research CenterBerlinGermany
| | - Maike Stein
- Department of Neurology with Experimental NeurologyCharité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Department of Neurology, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA
- Berlin Institute of Health at Charité—Universitätsmedizin BerlinDigital Health CenterBerlinGermany
- Center for Stroke Research BerlinCharité—Universitätsmedizin BerlinBerlinGermany
| | - Ralf Mekle
- Center for Stroke Research BerlinCharité—Universitätsmedizin BerlinBerlinGermany
| | - Lena Kock
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Heiko Klawitter
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Malvika Godara
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Judith Ramler
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Sonja Entringer
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
- Development, Health, and Disease Research ProgramUniversity of California, IrvineOrangeCA
- German Center for Mental HealthBerlinGermany
- German Center for Child and Adolescent HealthBerlinGermany
| | - Matthias Endres
- German Center for Mental HealthBerlinGermany
- Department of Neurology with Experimental NeurologyCharité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Center for Stroke Research BerlinCharité—Universitätsmedizin BerlinBerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, NeuroCure Cluster of ExcellenceBerlinGermany
- German Center for Neurodegenerative DiseasesBerlinGermany
- German Centre for Cardiovascular ResearchBerlinGermany
| | - Christine Heim
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
- German Center for Mental HealthBerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, NeuroCure Cluster of ExcellenceBerlinGermany
| |
Collapse
|
4
|
Xu L, Wang Z, Li Q. Global trends and projections of Parkinson's disease incidence: a 30-year analysis using GBD 2021 data. J Neurol 2025; 272:286. [PMID: 40131471 DOI: 10.1007/s00415-025-13030-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND AND OBJECTIVES Parkinson's disease (PD) is a neurodegenerative disorder marked by the progressive loss of dopaminergic neurons, leading to motor dysfunction and non-motor symptoms like cognitive decline and depression. With the aging global population, PD incidence is anticipated to rise, especially in regions with rapidly growing elderly populations. This study leverages Global Burden of Disease (GBD) 2021 data to analyze the burden of PD by region, sex, and age group, examining trends from 1992 to 2021 and projecting the future burden to 2030. METHODS Data from the GBD 2021 database for the years 1992-2021 were analyzed to assess age-standardized incidence rates (ASIR) and mortality of PD across socio-demographic index (SDI) regions, sex, and age groups. The Age-Period-Cohort (APC) model was used to explore temporal trends, while the Bayesian Age-Period-Cohort (BAPC) model projected future PD burden from 2022 to 2030. RESULTS From 1992 to 2021, global PD cases increased from 450,000 to 1.34 million, with crude incidence rates rising from 8.19 to 16.92 per 100,000 and ASIR from 11.54 to 15.63 per 100,000, indicating an annual net drift of 1.11% (95% CI 1.06%-1.17%), reflecting a growing burden driven by an aging population. All SDI regions saw a growth in PD burden, with the highest increases in middle- and high-middle-SDI regions, where male incidence was notably higher than female. Incidence rates escalated sharply in individuals aged 60 and older, peaking in those aged 85 and above. Projections suggest that by 2030, global PD cases will reach 1.93 million, with an ASIR of 27 per 100,000. DISCUSSION The findings highlight a sustained global increase in PD burden, particularly in middle- and high-income regions and among men. In low-SDI areas, PD burden may be underestimated due to limited healthcare access and diagnostic challenges. These results stress the urgent need for health policies focused on elderly populations, especially men, and call for effective prevention and intervention strategies to mitigate the future impact of PD.
Collapse
Affiliation(s)
- Libo Xu
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Zhenhao Wang
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Qingsong Li
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
5
|
Smith BL, Brooks-Patton B, Bollinger JL, Guzman TA, Brendle AH, Woodburn SC, Makela AG, Wohleb ES, Reyes TM. Prefrontal cortical microglial transcriptome relates to mouse offspring executive function deficits after perinatal opioid exposure in a sex-dependent manner. Brain Behav Immun 2025; 127:112-125. [PMID: 40068790 DOI: 10.1016/j.bbi.2025.03.016] [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: 10/22/2024] [Revised: 02/24/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025] Open
Abstract
Opioid use during pregnancy affects over 7% of pregnancies in the United States. While efforts have been directed at mitigating effects of prenatal opioid exposure acutely in the neonatal period, long-term neurodevelopmental studies in humans remain challenging. Using a preclinical model, we previously found that perinatal morphine (MO) exposure induces sex-dependent executive function deficits in adult offspring, and sexually divergent shifts in microglia phenotype. Therefore, this study used transcriptional profiling to test whether perinatal MO exposure would cause sex-specific transcriptional changes in microglia that would relate to offspring executive function outcomes in BXD F1 mice. Female C57BL/6 mice were given MO via the drinking water or saccharin only (SCH) one week prior to mating with DBA males, throughout gestation, and lactation until offspring were weaned. Offspring executive function was assessed in adulthood using the 5-choice serial reaction time task (5CSRTT), and microglia from the PFC were isolated and characterized via RNA-seq. In the 5CSRTT, male MO-exposed offspring had reduced accuracy and female MO-exposed offspring had increased inattentive behavior. There were a similar number of genes altered in female vs. male microglia, but only 3 differentially expressed genes were evident in both sexes. Further, hierarchical clustering analysis and WGCNA identified genes that related to behavioral deficits. Together, our data identify individual genes and pathways in microglia within each sex that may relate to executive function deficits observed after perinatal opioid exposure, even though the transcriptional profiles are highly divergent between the sexes.
Collapse
Affiliation(s)
- Brittany L Smith
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA.
| | - Brandon Brooks-Patton
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA
| | - Justin L Bollinger
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tess A Guzman
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alexander H Brendle
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Samuel C Woodburn
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Anna G Makela
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA
| | - Eric S Wohleb
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Teresa M Reyes
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
6
|
Castro CC, Vizuete A, Deniz BF, Wyse A, Netto CA. Sex-specific cognitive benefits and anti-inflammatory effects of coumestrol pretreatment in transient global cerebral ischemia. Mol Cell Neurosci 2025; 132:103991. [PMID: 39817970 DOI: 10.1016/j.mcn.2025.103991] [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: 08/19/2024] [Revised: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
Cerebral Global Ischemia (CGI) is a devastating neurological condition affecting millions globally each year, leading to significant inflammatory responses and long-term consequences, including delayed neuronal death and neurocognitive impairment. Following brain injury, resident microglial cells are activated, triggering pro-inflammatory cytokine expression and altering neuroimmune processes in a sex-dependent manner, particularly within the hippocampus. Coumestrol, a plant estrogen, is promoted as an alternative to post-menopausal hormone therapy due to its various mechanisms that enhance brain health, including its anti-inflammatory effects. This study aimed to investigate whether coumestrol pretreatment could attenuate the neuroinflammatory response following CGI by regulating pro-inflammatory pathways (GFAP, S100B, TNF-α, and IL-1β) and reversing CGI-induced memory loss. Male and female rats underwent CGI for 10 min or a sham surgery and received an ICV infusion of 20 μg of coumestrol or vehicle 1 h before CGI induction. Our findings revealed intriguing sex-specific effects of coumestrol pretreatment on gliosis following CGI and reperfusion, suggesting modulation of glial responses after ischemic insults. Coumestrol pre-administration significantly reduced levels of pro-inflammatory cytokines TNF-α and IL-1β during both reperfusion periods in both sexes, thereby mitigating CGI-induced neuroinflammation. Moreover, coumestrol pretreatment effectively reduced stroke-induced cognitive impairment, alleviating ischemia-induced memory deficits in both male and female rats. These results demonstrate the coumestrol's ability to attenuate cognitive deficits induced by CGI and highlight its potential sex-specific effects on inflammatory pathways. This study suggests that coumestrol modulates the glial and microglial inflammatory response, offering a promising approach to mitigate memory deficits associated with cerebral global ischemia.
Collapse
Affiliation(s)
- Cibele Canal Castro
- Department of Biochemistry, Institute for Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Adriana Vizuete
- Department of Biochemistry, Institute for Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bruna Ferrary Deniz
- Department of Physiology and Pharmacology, Universidade Federal de Pelotas, RS, Brazil
| | - Angela Wyse
- Department of Biochemistry, Institute for Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Department of Biochemistry, Institute for Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
7
|
Sakita M, Isobe W, Nonaka K, Murakami S, Miyachi R, Sakane K, Sugimoto S, Yamaguchi A, Yamamoto K. Age‑related changes in endoplasmic reticulum stress response‑associated protein expression in rat tibial nerves. Biomed Rep 2025; 22:50. [PMID: 39882333 PMCID: PMC11775640 DOI: 10.3892/br.2025.1928] [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: 10/17/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
In age-related peripheral neurodegeneration, changes in the promotion or inhibition of endoplasmic reticulum (ER) stress response related to the ubiquitin-proteasome degradation system (UPS), autophagy and apoptosis signaling factors during aging remain unclear. In the present study, the expression of ER stress response signaling-related protein factors was examined in tibial nerves during aging in rats. Tibial nerves were extracted from continuously housed rats at 20, 50, 70, 90 and 105 weeks of age. Expression of factors associated with ER stress-related degradation, including X-box binding protein 1 (XBP1s), eukaryotic translation initiation factor 2 subunit 1 (eIF2α), Beclin-1 (Becn1), and Caspase-3 (Casp3); ER stress-related repair, including binding immunoglobulin protein [also known as 78 kDa glucose-regulated protein (BiP/GRP78)], protein disulfide isomerase (PDI), brain-derived neurotrophic factor (BDNF) and the inflammatory cytokine IL6, was assessed by western blotting of tibial nerves from rats in each age group. Expression of XBP1s and Becn1, which promote UPS and autophagy, decreased significantly after 50 weeks of age. However, expression of eIF2α and Casp3, which inhibit new protein biosynthesis and promote apoptosis, increased significantly after 50 weeks. Expression of BiP/GRP78 and PDI, which are refolding factors for denatured proteins, showed a significant decrease after 50 (or 70) weeks of age. The expression of BDNF, a ligand protein for the repair cascade, showed a significant increase after 70 weeks of age, while that of IL6 increased significantly after 50 weeks of age. These results indicate that ER stress-related degradation (UPS and autophagy) and refolding repair functions are reduced in rat tibial nerves after 50 weeks, followed by enhanced apoptosis and inflammation. These findings shed light on the progression of age-related peripheral neurodegeneration in rats.
Collapse
Affiliation(s)
- Masahiro Sakita
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| | - Wataru Isobe
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
- Department of Rehabilitation, Mitsubishi Kyoto Hospital, Kyoto 615-8087, Japan
| | - Koji Nonaka
- Department of Rehabilitation, Faculty of Health Care Sciences, Naragakuen University, Nara 631-0003, Japan
| | - Shinichiro Murakami
- Department of Physical Therapy, Faculty of Health Care Sciences, Himeji-Dokkyo University, Himeji, Hyogo 670-0896, Japan
| | - Ryo Miyachi
- Department of Physical Therapy, Faculty of Health Care Sciences, Hokuriku University, Kanazawa, Ishikawa 920-1154, Japan
| | - Kento Sakane
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| | - Saki Sugimoto
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| | - Airi Yamaguchi
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| | - Koki Yamamoto
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| |
Collapse
|
8
|
Govil P, Kantrowitz JT. Negative Symptoms in Schizophrenia: An Update on Research Assessment and the Current and Upcoming Treatment Landscape. CNS Drugs 2025; 39:243-262. [PMID: 39799532 DOI: 10.1007/s40263-024-01151-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2024] [Indexed: 01/15/2025]
Abstract
The negative symptoms of schizophrenia include diminished emotional expression, avolition, alogia, anhedonia, and asociality, and due to their low responsiveness to available treatments, are a primary driver of functional disability in schizophrenia. This narrative review has the aim of providing a comprehensive overview of the current research developments in the treatment of negative symptoms in schizophrenia, and begins by introducing the concepts of primary, secondary, prominent, predominant, and broadly defined negative symptoms. We then compare and contrast commonly used research assessment scales for negative symptoms and review the evidence for the specific utility of widely available off-label and investigational treatments that have been studied for negative symptoms. Mechanism of action/putative treatments included are antipsychotics (D2R antagonists), N-methyl-D-aspartate receptor (NMDAR) and other glutamatergic modulators, serotonin receptor (5-HTR) modulators, anti-inflammatory agents, antidepressants, pro-dopaminergic modulators (non-D2R antagonists), acetylcholine modulators, oxytocin, and phosphodiesterase (PDE) inhibitors. With the caveat that no compounds are definitively proven as gold-standard treatments for broadly defined negative symptoms, the evidence base supports several potentially beneficial off-label and investigational medications for treating negative symptoms in schizophrenia, such as monotherapy with cariprazine, olanzapine, clozapine, and amisulpride, or adjunctive use of memantine, setrons such as ondansetron, minocycline, and antidepressants. These medications are widely available worldwide, generally tolerable and could be considered for an off-label, time-limited trial for a predesignated period of time, after which a decision to switch or stay can be made based on clinical response. Among investigational medications, NMDAR agonists, muscarinic agonists, and LB-102 remain under study. Suggestions for future research include reducing placebo effects by designing studies with a smaller number of high-quality study sites, potentially increasing the use of more precise rating scales for negative symptoms, and focused studies in people with predominant negative symptoms.
Collapse
Affiliation(s)
- Preetika Govil
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Joshua T Kantrowitz
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.
- College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
- Nathan Kline Institute, Orangeburg, NY, 10962, USA.
| |
Collapse
|
9
|
Lohkamp KJ, Timmer N, Solé Guardia G, Shenk J, Verweij V, Geenen B, Dederen PJ, Bakker L, Egitimci C, Yoldas R, Verhaeg M, Kothuis J, Nieuwenhuis D, Wiesmann M, Kiliaan AJ. Sex-Specific Adaptations in Alzheimer's Disease and Ischemic Stroke: A Longitudinal Study in Male and Female APP swe/PS1 dE9 Mice. Life (Basel) 2025; 15:333. [PMID: 40141679 PMCID: PMC11944048 DOI: 10.3390/life15030333] [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: 02/03/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
The long-term impact of stroke on Alzheimer's disease (AD) progression, particularly regarding sex-specific differences, remains unknown. Using a longitudinal study design, we investigated transient middle cerebral artery occlusion in 3.5-month-old APPswe/PS1dE9 (APP/PS1) and wild-type mice. In vivo, we assessed behavior, cerebral blood flow (CBF), and structural integrity by neuroimaging, as well as post-mortem myelin integrity (polarized light imaging, PLI), neuroinflammation, and amyloid beta (Aβ) deposition. APP/PS1 mice exhibited cognitive decline, white matter degeneration (reduced fractional anisotropy (FA) via diffusion tensor imaging (DTI)), and decreased myelin density via PLI. Despite early hypertension, APP/PS1 mice showed only sporadic hypoperfusion. Cortical thickening and hippocampal hypertrophy likely resulted from Aβ accumulation and neuroinflammation. Stroke-operated mice retained cognition despite cortical thinning and hippocampal atrophy due to cerebrovascular adaptation, including increased CBF in the hippocampus and thalamus. Stroke did not worsen AD pathology, nor did AD exacerbate stroke outcomes. Sex differences were found: female APP/PS1 mice had more severe Aβ deposition, hyperactivity, lower body weight, and reduced CBF but less neuroinflammation, suggesting potential neuroprotection. These findings highlight white matter degeneration and Aβ pathology as key drivers of cognitive decline in AD, with stroke-related deficits mitigated by (cerebro)vascular adaptation. Sex-specific therapies are crucial for AD and stroke.
Collapse
Affiliation(s)
- Klara J. Lohkamp
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Nienke Timmer
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Gemma Solé Guardia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Justin Shenk
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Vivienne Verweij
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Pieter J. Dederen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Lieke Bakker
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute (MHeNs), European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Cansu Egitimci
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Rengin Yoldas
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Minou Verhaeg
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Josine Kothuis
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Desirée Nieuwenhuis
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| |
Collapse
|
10
|
Bergendorf A, Park JH, Ball BK, Brubaker DK. Mouse-to-human modeling of microglia single-nuclei transcriptomics identifies immune signaling pathways and potential therapeutic candidates associated with Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637100. [PMID: 39975195 PMCID: PMC11839086 DOI: 10.1101/2025.02.07.637100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and behavior change. Studies have found that dysregulation of microglial cells is pivotal to AD pathology. These mechanisms have been studied in mouse models to uncover potential therapeutic biomarkers. Despite these findings, there are limitations to the translatable biological information from mice to humans due to differences in physiology, timeline of disease, and the heterogeneity of humans. To address the inter-species discrepancies, we developed a novel implementation of the Translatable Components Regression (TransComp-R) framework, which integrated microglia single-nuclei mouse and human transcriptomics data to identify biological pathways in mice predictive of human AD. We compared model variations with sparse and traditional principal component analysis. We found that both dimensionality reduction techniques encoded similar AD disease biology on mouse principal components with limited differences in technical performance. Several mouse sparse principal components explained high amounts of variance in humans and significantly differentiated human AD from control microglial cells. Additionally, we identified FDA-approved medications that induced gene expression profiles correlated with projections of healthy human microglia on mouse principal components. Such medications included cabergoline, selumetinib, and palbociclib. This computational framework may support uncovering cross-species disease insights and candidate pharmacological solutions from single-cell datasets.
Collapse
Affiliation(s)
- Alexander Bergendorf
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jee Hyun Park
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Brendan K. Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Douglas K. Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, OH 44106, USA
| |
Collapse
|
11
|
Guo R, Yan Z, Wang R, Guo T, Li H, Kong M, Guo W. Advances in Pharmacological Research on Icaritin: A Comprehensive Review. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:179-203. [PMID: 39880661 DOI: 10.1142/s0192415x25500089] [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: 01/31/2025]
Abstract
Epimedium has been widely used in traditional Chinese medicine for several thousands of years. This plant is known for tonifying kidney Yang, strengthening muscles and bones, and dispelling wind and dampness. It is worth noting that icaritin, a prenylated flavonoid isolated from Epimedium, has received increasing attention in recent years due to its wide range of pharmacological activities. Icaritin exhibits significant therapeutic potential against various diseases, such as osteoporosis, tumors (hepatocellular carcinoma, stomach cancer, breast cancer, and glioblastoma), cerebral ischemia skin injury, thrombocytopenia, and systemic lupus erythematosus. We review the pharmacological activities of icaritin and its potential molecular mechanisms for the treatment of related diseases. The data suggest that icaritin can have the pharmacological effects of mediating Wnt/[Formula: see text]-catenin, IL-6/JAK2/STAT3, AMPK/mTOR, PTEN/AKT, MAPK, NF-[Formula: see text]B, and other signaling pathways. This paper also discusses the progress of clinical trials of icaritin. Icaritin was approved by the State Food and Drug Administration in January 2022 for the treatment of advanced HCC, and has various clinical drug prospects. Although it has some disadvantages, including poor solubility, and low bioavailability, icaritin is still a prospective candidate for the development of naturally derived drugs, especially in the treatment of tumors and inflammatory diseases. This review aims to update and deepen the understanding of icaritin, and provide a theoretical basis for its further study.
Collapse
Affiliation(s)
- Ran Guo
- Henan Key Laboratory of Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Zhiping Yan
- Henan Key Laboratory of Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Rui Wang
- Henan Key Laboratory of Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Tongxuan Guo
- Henan Key Laboratory of Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Hao Li
- Henan Key Laboratory of Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Minyu Kong
- Henan Key Laboratory of Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Wenzhi Guo
- Henan Key Laboratory of Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
| |
Collapse
|
12
|
Jia CJ, Chen MN, Huang DD, Wu SF, Zeng CM, Liu ZQ, Wang MX, Huang YF, Yuan QJ, Zhang X. Edaravone promotes motoneuron survival and functional recovery after brachial plexus root avulsion and reimplantation in rats: Involvement of SIRT1/TFEB pathway. Int Immunopharmacol 2025; 145:113686. [PMID: 39642559 DOI: 10.1016/j.intimp.2024.113686] [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: 06/09/2024] [Revised: 10/31/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Brachial plexu root avulsion (BPRA) commonly causes extensive motoneuron death, motor axon degeneration and denervation of biceps, leading to devastating motor dysfunction in the upper limb. Edaravone (Eda) has been proven to exert anti-oxidative and neuroprotective effects on various neurological disorders. Herein, we aimed to investigate the efficacy profile and potential mechanisms of Eda on BPRA in vitro and in vivo models. METHODS Rats following BPRA and reimplantation surgery were intraperitoneally injected with Eda once daily. The motor function recovery of the affected forelimb was assessed by Terzis grooming test. Histological staining and transmission electron microscopy were performed to evaluate the morphological appearance of the spinal cord, musculocutaneous nerve, and biceps. Further in-depth studies to explore the underlying mechanisms of Eda were conducted using Western blotting, biochemical assays, and immunofluorescence in H2O2-induced NSC-34 cells. RESULTS Eda significantly accelerated motor function recovery, enhanced motoneuron survival, prevented motor axon descent, preserved myelin sheath integrity and attenuated muscle atrophy. Additionally, Eda treatment markedly suppressed oxidative stress-related indicators, downregulated apoptosis-related proteins, mitigated glial reactivity, and activated SIRT1 and TFEB. Notably, the neuroprotective effect of Eda was diminished by the SIRT1 inhibitor EX527 in H2O2-treated NSC-34 cells, suggesting that Eda regulated oxidative stress and apoptosis through SIRT1/TFEB-induced autophagy flux. CONCLUSIONS Eda enhanced motoneuron survival and axonal regeneration that promotes motor functional restoration by inhibiting oxidative stress and apoptosis via the SIRT1/TFEB-autophagy pathway. Thus, it may serve as a promising strategy in reimplantation surgery for the treatment of BPRA.
Collapse
Affiliation(s)
- Cai-Ju Jia
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Man-Ni Chen
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Dou-Dou Huang
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Shao-Feng Wu
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Chun-Ming Zeng
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Zhe-Qi Liu
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Meng-Xia Wang
- ShunDe Polytechnic College, School of Medicine & Health Care, FoShan, Guangdong, 528000, China.
| | - Yan-Feng Huang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| | - Qiu-Ju Yuan
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Shatin, N.T., Hong Kong SAR, China.
| | - Xie Zhang
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, 523808, China.
| |
Collapse
|
13
|
Sun Y, Wang W, Li Y, Wang H, Liang L, Wang X, Wang K, Bai W, Luan L, Qin L. Unveiling proteomic targets in the hypothalamus of ovariectomized and estradiol-treated rats: Insights into menopausal syndrome mechanisms. Ann Anat 2025; 257:152341. [PMID: 39326767 DOI: 10.1016/j.aanat.2024.152341] [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: 07/04/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Menopausal syndrome profoundly affects the physical and mental health of many women, drawing increasing attention from the medical community. However, its pathogenesis remains unclear. These symptoms are primarily driven by hormonal fluctuation. The hypothalamus, a key regulator of hormonal balance, potentially playing a critical role in the manifestation of menopausal syndrome. METHODS We simulated the low-estrogen menopausal state using ovariectomized rats, confirmed the success of ovariectomy via histological analysis of the uterus and vagina, followed by estrogen treatment. TMT-labeled quantitative proteomics, RTqPCR, targeted proteomics and Western blotting were used to identify differentially expressed proteins and their functions in the hypothalamus under low-estrogen conditions. RESULTS One-way ANOVA (p < 0.05) identified 295 differentially expressed proteins across the sham, ovariectomized and estrogen-treated groups. Post-ovariectomy, 103 differentially expressed proteins were upregulated and 93 were downregulated. Among these, 50 proteins were involved in hormones and neurotransmitters, immunity, metabolism and cardiovascular function. Notably, four proteins-Prkcg, Hsp90ab1, Ywhae, and Gad2-were identified as crucial regulators. CONCLUSIONS This study elucidates the central molecular mechanism of menopausal syndrome through bioinformatics analysis of differentially expressed proteins in the hypothalamus under low-estrogen conditions, providing novel targets for the treatment of related symptoms.
Collapse
Affiliation(s)
- Yanrong Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wenjuan Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yao Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266071, China
| | - Hanfei Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Lining Liang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xiangqiu Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ke Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wenpei Bai
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China.
| | - Liju Luan
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Lihua Qin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
14
|
K M M, Ghosh P, Nagappan K, Palaniswamy DS, Begum R, Islam MR, Tagde P, Shaikh NK, Farahim F, Mondal TK. From Gut Microbiomes to Infectious Pathogens: Neurological Disease Game Changers. Mol Neurobiol 2025; 62:1184-1204. [PMID: 38967904 DOI: 10.1007/s12035-024-04323-0] [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: 04/02/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024]
Abstract
Gut microbiota and infectious diseases affect neurological disorders, brain development, and function. Compounds generated in the gastrointestinal system by gut microbiota and infectious pathogens may mediate gut-brain interactions, which may circulate throughout the body and spread to numerous organs, including the brain. Studies shown that gut bacteria and disease-causing organisms may pass molecular signals to the brain, affecting neurological function, neurodevelopment, and neurodegenerative diseases. This article discusses microorganism-producing metabolites with neuromodulator activity, signaling routes from microbial flora to the brain, and the potential direct effects of gut bacteria and infectious pathogens on brain cells. The review also considered the neurological aspects of infectious diseases. The infectious diseases affecting neurological functions and the disease modifications have been discussed thoroughly. Recent discoveries and unique insights in this perspective need further validation. Research on the complex molecular interactions between gut bacteria, infectious pathogens, and the CNS provides valuable insights into the pathogenesis of neurodegenerative, behavioral, and psychiatric illnesses. This study may provide insights into advanced drug discovery processes for neurological disorders by considering the influence of microbial communities inside the human body.
Collapse
Affiliation(s)
- Muhasina K M
- Department of Pharmacognosy, JSS College of Pharmacy, Ooty, Tamil Nadu, 643001, India.
| | - Puja Ghosh
- Department of Pharmacognosy, JSS College of Pharmacy, Ooty, Tamil Nadu, 643001, India
| | - Krishnaveni Nagappan
- Department of Pharmaceutical Analysis, JSS College of Pharmacy, Ooty, Tamil Nadu, 643001, India
| | | | - Rahima Begum
- Department of Microbiology, Gono Bishwabidyalay, Dhaka, Bangladesh
| | - Md Rabiul Islam
- Tennessee State University Chemistry department 3500 John A Merritt Blvd, Nashville, TN, 37209, USA
| | - Priti Tagde
- PRISAL(Pharmaceutical Royal International Society), Branch Office Bhopal, Bhopal, Madhya Pradesh, 462042, India
| | - Nusrat K Shaikh
- Department of Quality Assurance, Smt. N. M, Padalia Pharmacy College, Navapura, Ahmedabad, 382 210, Gujarat, India
| | - Farha Farahim
- Department of Nursing, King Khalid University, Abha, 61413, Kingdom of Saudi Arabia
| | | |
Collapse
|
15
|
Nesbitt C, Van Der Walt A, Butzkueven H, Cheung AS, Jokubaitis VG. Exploring the role of sex hormones and gender diversity in multiple sclerosis. Nat Rev Neurol 2025; 21:48-62. [PMID: 39658653 DOI: 10.1038/s41582-024-01042-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 12/12/2024]
Abstract
Sex and sex hormones are thought to influence multiple sclerosis (MS) through effects on inflammation, myelination and neurodegeneration, and exogenous hormones have been explored for their therapeutic potential. However, our understanding of how sex hormones influence MS disease processes and outcomes remains incomplete. Furthermore, our current knowledge is derived primarily from studies that focus exclusively on cisgender populations with exclusion of gender-diverse people. Gender-affirming hormone therapy comprising exogenous sex hormones or sex hormone blocking agents are commonly used by transgender and gender-diverse individuals, and it could influence MS risk and outcomes at various stages of disease. A better understanding of the impact and potential therapeutic effects of both endogenous and exogenous sex hormones in MS is needed to improve care and outcomes for cisgender individuals and, moreover, for gender-diverse populations wherein an evidence base does not exist. In this Perspective, we discuss the effects of endogenous and exogenous sex hormones in MS, including their potential therapeutic benefits, and examine both established sex-based dimorphisms and the potential for gender-diverse dimorphisms. We advocate for future research that includes gender-diverse people to enhance our knowledge of the interplay of sex and sex hormones in MS, leading to the development of more effective and inclusive treatment strategies and improvement of care for all individuals with MS.
Collapse
Affiliation(s)
- Cassie Nesbitt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia.
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| | - Anneke Van Der Walt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Ada S Cheung
- Trans Health Research Group, Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia.
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| |
Collapse
|
16
|
Bahramiazar P, Abdollahzade N, Tartibian B, Ahmadiasl N, Yaghoob Nezhad F. The Role of Estrogen in Brain MicroRNAs Regulation. Adv Pharm Bull 2024; 14:819-835. [PMID: 40190672 PMCID: PMC11970499 DOI: 10.34172/apb.39216] [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: 08/28/2022] [Revised: 08/12/2024] [Accepted: 09/08/2024] [Indexed: 04/09/2025] Open
Abstract
Purpose This review aims to elucidate the role of estrogen-sensitive microRNAs (miRNAs) in modulating brain functions and disorders, highlighting the protective effects of estrogen on the central nervous system. Methods A comprehensive literature review was conducted, examining the relationship between estrogen, miRNAs, and cognitive health. The study focused on experimental data comparing cognitive impairments between genders and the mechanisms of estrogen's effects on brain function. Results Cognitive impairments are less prevalent in women of reproductive age compared to men, indicating estrogen's neuroprotective role. Estrogen modulates gene expression through specific receptors, while miRNAs regulate approximately 30% of protein-coding genes in mammals. These miRNAs play critical roles in synaptic plasticity and neuronal survival. The review identifies several estrogen-sensitive miRNAs and their potential involvement in brain disorders. Conclusion The interplay between estrogen and miRNAs offers valuable insights into the molecular mechanisms underlying cognitive health and disease. Understanding these relationships may lead to novel therapeutic strategies for addressing various brain disorders, particularly those associated with hormonal changes and aging.
Collapse
Affiliation(s)
- Peyvand Bahramiazar
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naseh Abdollahzade
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Bakhtyar Tartibian
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Allameh Tabataba’i University, Tehran, Iran
| | - Naser Ahmadiasl
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
17
|
Gao Z, Ding C, Huang X, Liu Y, Fan W, Song S. Estrogen receptor α aggravates intestinal inflammation via promoting the activation of NLRP3 inflammasome. Int Immunopharmacol 2024; 143:113425. [PMID: 39426237 DOI: 10.1016/j.intimp.2024.113425] [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: 08/26/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Activation of the NLRP3 inflammasome and estrogen receptor α (ERα) has been shown to increase the risk of inflammatory bowel diseases (IBD) or promote disease recurrence. In previous work, we demonstrated that ERα regulated the transcription of NLRP3. However, the precise mechanism by which ERα modulates NLRP3 in IBD models remains unclear. In this study, we induced IBD in wild-type mice using DSS or TNBS, followed by treatment with the ERα-specific agonist PPT. The results showed that IBD symptoms and intestinal inflammation responses were significantly exacerbated after PPT treatment. Furthermore, the activation of ERα by PPT led to a marked increase in the expression of NLRP3 and pro-inflammatory cytokines, including IL-1β and IL-18, suggesting that ERα activation exacerbated intestinal inflammation and impaired mucosal healing during the recovery phase of inflammation. In contrast, ERα-knockout mice exhibited only mild symptoms when exposed to DSS or TNBS, with a concurrent reduction in NLRP3 expression, indicating that ERα plays a role in inflammation susceptibility. Similar findings were observed in NCM-460 cells, where the inflammation response was attenuated in ERα-knockdown cells. Importantly, we demonstrated that ERα interacted with the NLRP3 inflammasome and promoted its assembly. Collectively, we propose an underlying pathogenesis of IBD, that is, ERα can interact with the NLRP3 inflammasome and promote its expression and assembly, thereby exacerbating intestinal inflammation in IBD models. Therefore, ERα could serve as a potential therapeutic target for NLRP3 inflammasome-associated intestinal inflammation.
Collapse
Affiliation(s)
- Zhangshan Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Chenchen Ding
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Xi Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Yapei Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Wentao Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Suquan Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China.
| |
Collapse
|
18
|
Lee SH, Shin HS, So YH, Lee DH, An BS, Lee GS, Jung EM. Maternal exposure to 4-tert-octylphenol causes alterations in the morphology and function of microglia in the offspring mouse brain. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136258. [PMID: 39471617 DOI: 10.1016/j.jhazmat.2024.136258] [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: 06/20/2024] [Revised: 10/01/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
4-tert-Octylphenol (OP), an endocrine disrupting chemical is widely used in the production of industrial products. Prenatal exposure to endocrine-disrupting chemicals negatively affects the brain. However, the influence of OP exposure during neurodevelopment in adult offspring remains unclear. Thus, in the present study, we investigated the effects of maternal OP exposure on brain development in adult offspring by analyzing primary glial cell cultures and mice. Our findings revealed that OP exposure led to a specific increase in the mRNA expression of the ionized calcium-binding adapter molecule 1 (Iba-1) and the proportion of amoeboid microglia in the primary glial cell culture and adult offspring mice. Exposure to OP increased the transcriptional activation of Iba-1 and estrogen response element, which were counteracted by estrogen receptor antagonists ICI 182,780. Moreover, OP exposure increased the nuclear localization of the estrogen receptor. Remarkably, OP exposure decreased the mRNA expression levels of proinflammatory cytokines and genes associated with immune response in the brains of the offspring. OP exposure upregulated actin filament-related genes and altered cytoskeletal gene expression, as demonstrated by microarray analysis. The morphological changes in microglia did not result in an inflammatory response following lipopolysaccharide treatment. Taken together, the effects of OP exposure during neurodevelopment persist into adulthood, resulting in microglial dysfunction mediated by estrogen receptor signaling pathways in the brains of adult offspring mice.
Collapse
Affiliation(s)
- Seung Hyun Lee
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea; Institute for Future Earth, Pusan National University, Busan, Republic of Korea
| | - Hyun Seung Shin
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea; Institute for Future Earth, Pusan National University, Busan, Republic of Korea
| | - Yun Hee So
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea; Institute for Future Earth, Pusan National University, Busan, Republic of Korea
| | - Dong Hun Lee
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea; Institute for Future Earth, Pusan National University, Busan, Republic of Korea
| | - Beum-Soo An
- Department of Biomaterials Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Republic of Korea
| | - Geun-Shik Lee
- Department of Veterinary Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Eui-Man Jung
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea; Institute for Future Earth, Pusan National University, Busan, Republic of Korea.
| |
Collapse
|
19
|
Tan X, Cao M, Zhao Y, Yi L, Li Y, He C, Li QX, Dong Y. Neuroprotection of isoorientin against microglia activation induced by lipopolysaccharide via regulating GSK3β, NF-κb and Nrf2/HO-1 pathways. Immunopharmacol Immunotoxicol 2024; 46:741-750. [PMID: 39245870 DOI: 10.1080/08923973.2024.2399249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
Background: Isoorientin (ISO), a flavone C-glycoside, is a glycogen synthase kinase 3β (GSK3β) substrate-competitive inhibitor. ISO has potential in treatment of Alzheimer's disease (AD). An excessive activation of GSK3β can lead to neuroinflammation causing neuronal damage. Microglia cells, as resident immune cells of the central nervous system, mediate neuroinflammation. Here, we studied the effects of ISO on microglial activation to alleviate neuroinflammation. Methods: Effects of ISO were observed upon the stimulation of mouse microglia BV2 or SIM-A9 cells by lipopolysaccharide (LPS). Lithium chloride (LiCl) was the positive control as a GSK3β inhibitor. The release of TNF-α and NO were analyzed by ELISA and Griess assays, while expressions of COX-2, Iba-1, BDNF, GSK3β, NF-κB p65, IκB, Nrf2 and HO-1 were detected by Western blotting. In the co-culture model of SIM-A9 cells and differentiated SH-SY5Y human neuroblastoma cells, effects of ISO on microglia-mediated neuronal damage were evaluated with the MTS assay. Results: ISO significantly inhibited the production of TNF-α (p < 0.01), NO (p < 0.001) and the expression of COX-2 (p < 0.01) and Iba-1 (p < 0.05) induced by LPS, and increased BDNF. The cell viability of SH-SY5Y was inhibited by LPS in the co-culture, which was prevented by ISO pretreatment. ISO increased the expression of p-GSK3β (Ser9), IκB and HO-1 in the cytoplasm, decreased NF-κB p65 and increased Nrf2 in the nucleus compared with the LPS group. Conclusion: ISO attenuated the activation of microglia through regulating the GSK3β, NF-κB and Nrf2/HO-1 signaling pathways to exert neuroprotection.
Collapse
Affiliation(s)
- Xiaoqin Tan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawaii, USA
- Medical Department, Wuhan City Collge, Wuhan, China
| | - Mindie Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Yijing Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Lang Yi
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Yingui Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Changhong He
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Qing X Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Yan Dong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| |
Collapse
|
20
|
Alsegiani AS, Shah ZA. Age-dependent sex differences in cofilin1 pathway (LIMK1/SSH1) and its association with AD biomarkers after chronic systemic inflammation in mice. Neurobiol Aging 2024; 144:43-55. [PMID: 39265451 DOI: 10.1016/j.neurobiolaging.2024.09.003] [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: 04/23/2024] [Revised: 08/15/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024]
Abstract
Chronic systemic inflammation (CSI) results in neuroinflammation and neurodegeneration. Cofilin1 is a stress protein that activates microglia and induces neuroinflammation, but its role in CSI at different aging stages remains unidentified. Therefore, the study aims to identify cofilin1 and its upstream regulators LIMK1 and SSH1 after CSI in young-, middle-, and advanced-aged mice. CSI was induced by injecting the male and female mice with a sub-lethal dose of Lipopolysaccharide weekly for six weeks. The results showed that normal male mice did not show cofilin pathway dysregulation, but a significant dysregulation was observed in CSI advanced-aged mice. In females, cofilin1 dysregulation was observed in healthy and CSI advanced-aged mice, while significant cofilin1 dysregulation was observed in middle-aged mice during CSI. Furthermore, cofilin1 pathway dysregulations correlated with Alzheimer's disease (AD) biomarkers in the brain and saliva, astrocyte activation, synaptic degeneration, neurobehavioral impairments, gut-microbiota abnormalities, and circulatory inflammation. These results provide new insights into cofilin1 sex and age-dependent mechanistic differences that might help identify targets for modulating neuroinflammation and early onset of AD.
Collapse
Affiliation(s)
- Amsha S Alsegiani
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA.
| |
Collapse
|
21
|
Rymbai E, Roy D, Jupudi S, Srinivasadesikan V. The identification of c-Abl inhibitors as potential agents for Parkinson's disease: a preliminary in silico approach. Mol Divers 2024; 28:4051-4065. [PMID: 38273156 DOI: 10.1007/s11030-023-10796-3] [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: 08/15/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024]
Abstract
Parkinson's disease (PD) is the most common movement disorder worldwide. PD is primarily associated with the mutation, overexpression, and phosphorylation of α-synuclein. At the molecular level, the upstream protein c-Abl, a tyrosine kinase, has been shown to regulate α-synuclein activation and expression patterns. This study aimed to identify potential c-Abl inhibitors through in silico approaches. Molecular docking was performed using PyRx software, followed by Prime MM-GBSA studies. BBB permeability and toxicity were predicted using CBligand and ProTox-II, respectively. ADME was assessed using QikProp. Molecular dynamics were carried out using Desmond (Academic version). DFT calculations were performed using the Gaussian 16 suite program. The binding scores of the top hits, norimatinib, DB07326, and entinostat were - 11.8 kcal/mol, - 11.8 kcal/mol, and - 10.8 kcal/mol, respectively. These hits displayed drug-likeness with acceptable ADME properties, except for the standard, nilotinib, which violated Lipinski's rule of five. Similarly, the molecular dynamics showed that the top hits remained stable during the 100 ns simulation. DFT results indicate DB04739 as a potent reactive hit. While based on toxicity prediction, entinostat may be a potential candidate for preclinical and clinical testing in PD. Further studies are warranted to confirm the activity and efficacy of these ligands for PD.
Collapse
Affiliation(s)
- Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India.
| | - Dhritiman Roy
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
| | - Srikanth Jupudi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Venkatesan Srinivasadesikan
- Department of Sciences and Humanities, Vignan's Foundation for Science, Technology and Research (Deemed to be University), Vadlamudi, Guntur, Andhra Pradesh, India
| |
Collapse
|
22
|
Munteanu C, Onose G, Rotariu M, Poștaru M, Turnea M, Galaction AI. Role of Microbiota-Derived Hydrogen Sulfide (H 2S) in Modulating the Gut-Brain Axis: Implications for Alzheimer's and Parkinson's Disease Pathogenesis. Biomedicines 2024; 12:2670. [PMID: 39767577 PMCID: PMC11727295 DOI: 10.3390/biomedicines12122670] [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: 10/27/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025] Open
Abstract
Microbiota-derived hydrogen sulfide (H2S) plays a crucial role in modulating the gut-brain axis, with significant implications for neurodegenerative diseases such as Alzheimer's and Parkinson's. H2S is produced by sulfate-reducing bacteria in the gut and acts as a critical signaling molecule influencing brain health via various pathways, including regulating inflammation, oxidative stress, and immune responses. H2S maintains gut barrier integrity at physiological levels and prevents systemic inflammation, which could impact neuroinflammation. However, as H2S has a dual role or a Janus face, excessive H2S production, often resulting from gut dysbiosis, can compromise the intestinal barrier and exacerbate neurodegenerative processes by promoting neuroinflammation and glial cell dysfunction. This imbalance is linked to the early pathogenesis of Alzheimer's and Parkinson's diseases, where the overproduction of H2S exacerbates beta-amyloid deposition, tau hyperphosphorylation, and alpha-synuclein aggregation, driving neuroinflammatory responses and neuronal damage. Targeting gut microbiota to restore H2S homeostasis through dietary interventions, probiotics, prebiotics, and fecal microbiota transplantation presents a promising therapeutic approach. By rebalancing the microbiota-derived H2S, these strategies may mitigate neurodegeneration and offer novel treatments for Alzheimer's and Parkinson's diseases, underscoring the critical role of the gut-brain axis in maintaining central nervous system health.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (M.P.); (M.T.); (A.I.G.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (M.P.); (M.T.); (A.I.G.)
| | - Mădălina Poștaru
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (M.P.); (M.T.); (A.I.G.)
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (M.P.); (M.T.); (A.I.G.)
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (M.P.); (M.T.); (A.I.G.)
| |
Collapse
|
23
|
Dow LF, Pathirage R, Erickson HE, Amani E, Ronning DR, Trippier PC. Synthesis and biological characterization of a 17β hydroxysteroid dehydrogenase type 10 (17β-HSD10) inhibitor. RSC Med Chem 2024:d4md00733f. [PMID: 39618963 PMCID: PMC11605429 DOI: 10.1039/d4md00733f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Alzheimer's disease (AD) is estimated to affect over 55 million people across the world. Small molecule treatment options are limited to symptom management with no impact on disease progression. The need for new protein targets and small molecule hit compounds is unmet and urgent. Hydroxysteroid 17-β dehydrogenase type 10 (17β-HSD10) is a mitochondrial enzyme known to bind amyloid beta, a hallmark of AD, and potentiate its toxicity to neurons. Identification of small molecules capable of interacting with 17β-HSD10 may drive drug discovery efforts for AD. The screening compound BCC0100281 (1), was previously identified as an inhibitor of 17β-HSD10. Herein we report the first synthetic access to the hit compound following a convergent pathway starting from simple heterocyclic building blocks. The compound was found to be toxic to 'neuron-like' cells, specifically those of neuroblastoma origin, providing a potential hit compound for cancer drug discovery, wherein the protein is known to be overexpressed. However, assay of synthetic intermediates identified novel scaffolds with effect to rescue amyloid beta-induced cytotoxicity, showcasing the power of organic synthesis and medicinal chemistry to optimize hit compounds.
Collapse
Affiliation(s)
- Louise F Dow
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha Nebraska 68198 USA
| | - Rasangi Pathirage
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha Nebraska 68198 USA
| | - Helen E Erickson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha Nebraska 68198 USA
| | - Edrees Amani
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha Nebraska 68198 USA
| | - Donald R Ronning
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha Nebraska 68198 USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center Omaha Nebraska 68198 USA
- UNMC Center for Drug Design and Innovation, University of Nebraska Medical Center Omaha Nebraska 68198 USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha Nebraska 68198 USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center Omaha Nebraska 68198 USA
- UNMC Center for Drug Design and Innovation, University of Nebraska Medical Center Omaha Nebraska 68198 USA
| |
Collapse
|
24
|
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
|
25
|
Yang B, Zhu Y, Li K, Wang F, Liu B, Zhou Q, Tai Y, Liu Z, Yang L, Ba R, Lei C, Ren H, Xu Z, Pang A, Yang X. Machine learning model base on metabolomics and proteomics to predict cognitive impairment in Parkinson's disease. NPJ Parkinsons Dis 2024; 10:187. [PMID: 39394257 PMCID: PMC11470017 DOI: 10.1038/s41531-024-00795-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/05/2024] [Indexed: 10/13/2024] Open
Abstract
There is an urgent need to identify predictive biomarkers of Parkinson's disease (PD) with cognitive impairment (PDCI) in order to individualize patient management, ensure timely intervention, and improve prognosis. The aim of this study was to screen for these biomarkers by comparing the plasma proteome and metabolome of PD patients with or without cognitive impairment. Proteomics and metabolomics analyses were performed on a discover cohort. A machine learning model was used to identify candidate protein and metabolite biomarkers of PDCI, which were validated in an independent cohort. The predictive ability of these biomarkers for PDCI was evaluated by plotting receiver operating characteristic curves and calculating the area under the curve (AUC). Moreover, we assessed the predictive ability of these proteins in combination with neuroimaging. In the discover cohort (n = 100), we identified 25 protein features with best results in the machine learning model, including top-ranked PSAP and H3C15. The two-proteins were used for model construction, achieving an Area under the curve (AUC) of 0.951 in the train set and AUC of 0.981 in the test set. Similarly, the model gives a rank list of endogenous metabolite features, Glycocholic Acid and 6-Methylnicotinamide were two top features. Combining these two markers further got the AUC of 0.969 in train set and 0.867 in the test set. To validate the performance of the protein biomarkers, we performed targeted analysis of selected proteins (H3C15 and PSAP) and proteins likely associated with PDCI (NCAM2 and LAMB2) using parallel reaction monitoring in validation cohort (n = 116). The AUC of the classifier built with H3C15 and PSAP is 0.813. Moreover, when combining H3C15, PSAP, NCAM2, and LAMB2, the model achieved AUC of 0.983 in the train set, AUC of 0.981 in the test set, and AUC of 0.839 in the validation set. Furthermore, we verified that these protein markers we discovered can improve the predictive effect of neuroimaging on PDCI: the classifier built with neuroimaging features had AUC of 0.833, which improved to 0.905 when combined with H3C15. Taken together, our integrated proteomics and metabolomics analysis successfully identified potential biomarkers for PDCI. Additionally, H3C15 showed promise in enhancing the predictive performance of neuroimaging for cognitive impairment.
Collapse
Affiliation(s)
- Baiyuan Yang
- Department of Neurology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan Province, China
| | - Yongyun Zhu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Kelu Li
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fang Wang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Bin Liu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Qian Zhou
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuchao Tai
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhaochao Liu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Lin Yang
- Department of Neurology, The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Ruiqiong Ba
- Department of Neurology, Qujing City First People's Hospital, Qujing, Yunnan Province, China
| | - Chunyan Lei
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hui Ren
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhong Xu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Ailan Pang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Xinglong Yang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
26
|
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
|
27
|
Jiang X, Yu X, Hu S, Dai H, Zhang H, Hang Y, Xie X, Yang Y, Wu F. Effects of E2 on the IDO1-mediated metabolic KYN pathway in OVX female mice. J Cell Mol Med 2024; 28:e70179. [PMID: 39467780 PMCID: PMC11518696 DOI: 10.1111/jcmm.70179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/12/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
The aim of this study was to investigate the role of 17β-estradiol (E2)-mediated oestrogen receptor (ER) in modulating the depressive-like behaviours of ovariectomy (OVX) mice and the associated mechanisms. E2 was administrated in OVX mice. The behaviour and physiological changes of OVX mice including immobility time in tail suspension test (TST) and forced swimming test (FST), levels of serum E2, inflammatory mediators, oxidative stress factors, indoleamine2,3-dioxygenase 1 (IDO1) and the neurotransmitters mediated by IDO1 activation were then recorded. Cell injury models established by lipopolysaccharide (LPS) or H2O2 stimulation in HT22 and BV2 cells were employed to further explore the mechanisms of E2's function. E2 treatment improved OVX-induced increase of immobility time in FST and TST. Meanwhile, E2 ameliorated the changes of inflammatory factors (NF-κB, TNF-α and IL-6), IDO1, IDO1-mediated TRP/KYN pathway and oxidative stress factors (iNOS, MDA, GSH and SOD) in the hippocampus of OVX mice. Interestingly, ERβ inhibitor abolished E2's inhibitory effects on the inflammation and IDO1-mediated TRP/KYN pathway; ERβ inhibitor also abolished E2's anti-oxidative stress effect. In cell experiments, ERβ small interfering RNA (siRNA) pretreatment reversed E2's anti-inflammatory effect on LPS-treated HT22 and BV2 cells and E2's inhibitory effect on IDO1 expression in LPS-treated BV2 cells. ERβ siRNA pretreatment also reversed E2's anti-oxidation effect on H2O2-treated HT22 cells. E2 exert the antidepressant function in OVX mice via ERβ-modulated suppression of NF-κB-mediated inflammatory pathway, oxidative stress factors and IDO1-mediated TRP/KYN pathway in the hippocampus.
Collapse
Affiliation(s)
- Xi Jiang
- Department of PharmacyZhejiang University Mingzhou HospitalNingboChina
| | - Xuefeng Yu
- Department of PharmacyZhejiang Pharmaceutical UniversityNingboChina
| | - Shuran Hu
- Department of NeurosurgerySouth Yunnan Central Hospital of Yunnan Province (The First People's Hospital of Honghe Prefecture)MengziChina
| | - Huidan Dai
- Department of PharmacyZhejiang Pharmaceutical UniversityNingboChina
| | - Hanqin Zhang
- Department of PharmacyZhejiang Pharmaceutical UniversityNingboChina
| | - Yuyang Hang
- Department of PharmacyZhejiang Pharmaceutical UniversityNingboChina
| | - Xupei Xie
- Department of PharmacyShaoxing Second HospitalShaoxingChina
| | - Yubo Yang
- Department of PharmacyZhejiang University Mingzhou HospitalNingboChina
| | - Fan Wu
- Department of PharmacyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| |
Collapse
|
28
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
29
|
Hagarty-Waite KA, Emmons HA, Fordahl SC, Erikson KM. The Influence of Strain and Sex on High Fat Diet-Associated Alterations of Dopamine Neurochemistry in Mice. Nutrients 2024; 16:3301. [PMID: 39408267 PMCID: PMC11479034 DOI: 10.3390/nu16193301] [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: 08/19/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Objective: The objective of this study was to determine the influence of sex and strain on striatal and nucleus accumbens dopamine neurochemistry and dopamine-related behavior due to a high-saturated-fat diet (HFD). Methods: Male and female C57B6/J (B6J) and Balb/cJ (Balb/c) mice were randomly assigned to a control-fat diet (CFD) containing 10% kcal fat/g or a mineral-matched HFD containing 60% kcal fat/g for 12 weeks. Results: Intraperitoneal glucose tolerance testing (IPGTT) and elevated plus maze experiments (EPM) confirmed that an HFD produced marked blunting of glucose clearance and increased anxiety-like behavior, respectively, in male and female B6J mice. Electrically evoked dopamine release in the striatum and reuptake in the nucleus accumbens (NAc), as measured by ex vivo fast scan cyclic voltammetry, was reduced for HFD-fed B6J females. Impairment in glucose metabolism explained HFD-induced changes in dopamine neurochemistry for B6J males and, to a lesser extent, Balb/c males. The relative expressions of protein markers associated with the activation of microglia, ionized calcium binding adaptor molecule (Iba1) and cluster of differentiation molecule 11b (CD11b) in the striatum were increased due to an HFD for B6J males but were unchanged or decreased amongst HFD-fed Balb/c mice. Conclusions: Our findings demonstrate that strain and sex influence the insulin- and microglia-dependent mechanisms of alterations to dopamine neurochemistry and associated behavior due to an HFD.
Collapse
Affiliation(s)
| | | | | | - Keith M. Erikson
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (K.A.H.-W.); (H.A.E.); (S.C.F.)
| |
Collapse
|
30
|
Wan M, Liu Y, Li D, Snyder R, Elkin L, Day C, Rodriguez J, Grunseich C, Mahley R, Watts J, Cheung V. The enhancer RNA, AANCR, regulates APOE expression in astrocytes and microglia. Nucleic Acids Res 2024; 52:10235-10254. [PMID: 39162226 PMCID: PMC11417409 DOI: 10.1093/nar/gkae696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
Enhancers, critical regulatory elements within the human genome, are often transcribed into enhancer RNAs. The dysregulation of enhancers leads to diseases collectively termed enhanceropathies. While it is known that enhancers play a role in diseases by regulating gene expression, the specific mechanisms by which individual enhancers cause diseases are not well understood. Studies of individual enhancers are needed to fill this gap. This study delves into the role of APOE-activating noncoding RNA, AANCR, in the central nervous system, elucidating its function as a genetic modifier in Alzheimer's Disease. We employed RNA interference, RNaseH-mediated degradation, and single-molecule RNA fluorescence in situ hybridization to demonstrate that mere transcription of AANCR is insufficient; rather, its transcripts are crucial for promoting APOE expression. Our findings revealed that AANCR is induced by ATM-mediated ERK phosphorylation and subsequent AP-1 transcription factor activation. Once activated, AANCR enhances APOE expression, which in turn imparts an inflammatory phenotype to astrocytes. These findings demonstrate that AANCR is a key enhancer RNA in some cell types within the nervous system, pivotal for regulating APOE expression and influencing inflammatory responses, underscoring its potential as a therapeutic target in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ma Wan
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Yaojuan Liu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dongjun Li
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryan J Snyder
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Lillian B Elkin
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Christopher R Day
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Joseph Rodriguez
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Christopher Grunseich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert W Mahley
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Pathology and Medicine, University of California, San Francisco, CA, USA
| | - Jason A Watts
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Vivian G Cheung
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
31
|
Mading A, Chotritthirong Y, Chulikhit Y, Daodee S, Boonyarat C, Khamphukdee C, Sukketsiri W, Kwankhao P, Pitiporn S, Monthakantirat O. Effectiveness of Tri-Kaysorn-Mas Extract in Ameliorating Cognitive-like Behavior Deficits in Ovariectomized Mice via Activation of Multiple Mechanisms. Pharmaceuticals (Basel) 2024; 17:1182. [PMID: 39338344 PMCID: PMC11435318 DOI: 10.3390/ph17091182] [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: 06/26/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Postmenopausal women have a higher probability of experiencing cognitive alterations compared to men, suggesting that the decline in female hormones may contribute to cognitive deterioration. Thailand traditionally uses Tri-Kaysorn-Mas (TKM), a blend of three medicinal herbs, as a tonic to stimulate appetite and relieve dyspepsia. Due to its antioxidant and anti-acetylcholinesterase activities, we investigated the effects of TKM (50 and 100 mg/kg/day, p.o., for 8 weeks) on cognitive deficits and their underlying causes in an ovariectomized (OVX) mouse model of menopause. OVX mice showed cognitive impairment in the Y-maze, novel object recognition task (NORT), and Morris water maze (MWM) behavioral tests, along with atrophic changes to the uterus, altered levels of serum 17β-estradiol, and down-regulated expression of estrogen receptors (ERα and ERβ). These behavioral effects were reversed by TKM. TKM decreased malondialdehyde (MDA) levels and mitigated oxidative stress in the brain by enhancing the activity of superoxide dismutase (SOD) and catalase (CAT) and by up-regulating the antioxidant-related gene Nrf2 while down-regulating Keap1. TKM also counteracted OVX-induced neurodegeneration by enhancing the expression of the neurogenesis-related genes BDNF and CREB. The results indicate that TKM extract alleviates oxidative brain damage and neurodegeneration while enhancing cognitive behavior in OVX mice, significantly improving cognitive deficiencies related to menopause/ovariectomy through multiple targets.
Collapse
Affiliation(s)
- Abdulwaris Mading
- Graduate School of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (A.M.); (Y.C.)
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (S.D.); (C.B.)
| | - Yutthana Chotritthirong
- Graduate School of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (A.M.); (Y.C.)
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (S.D.); (C.B.)
| | - Yaowared Chulikhit
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (S.D.); (C.B.)
| | - Supawadee Daodee
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (S.D.); (C.B.)
| | - Chantana Boonyarat
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (S.D.); (C.B.)
| | - Charinya Khamphukdee
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Sciences, Kaen University, Khon Kaen 40002, Thailand;
| | - Wanida Sukketsiri
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand;
| | - Pakakrong Kwankhao
- Department of Pharmacy, Chao Phya Abhaibhubejhr Hospital, Ministry of Public Health, Prachinburi 25000, Thailand; (P.K.); (S.P.)
| | - Supaporn Pitiporn
- Department of Pharmacy, Chao Phya Abhaibhubejhr Hospital, Ministry of Public Health, Prachinburi 25000, Thailand; (P.K.); (S.P.)
| | - Orawan Monthakantirat
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (S.D.); (C.B.)
| |
Collapse
|
32
|
Chao YW, Tung YT, Yang SC, Shirakawa H, Su LH, Loe PY, Chiu WC. The Effects of Rice Bran on Neuroinflammation and Gut Microbiota in Ovariectomized Mice Fed a Drink with Fructose. Nutrients 2024; 16:2980. [PMID: 39275295 PMCID: PMC11397027 DOI: 10.3390/nu16172980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024] Open
Abstract
Rice bran, which is abundant in dietary fiber and phytochemicals, provides multiple health benefits. Nonetheless, its effects on neuroinflammation and gut microbiota in postmenopausal conditions are still not well understood. This study investigated the effects of rice bran and/or tea seed oil supplementation in d-galactose-injected ovariectomized (OVX) old mice fed a fructose drink. The combination of d-galactose injection, ovariectomy, and fructose drink administration creates a comprehensive model that simulates aging in females under multiple metabolic stressors, including oxidative stress, estrogen deficiency, and high-sugar diets, and allows the study of their combined impact on metabolic disorders and related diseases. Eight-week-old and 6-8-month-old female C57BL/6 mice were used. The mice were divided into six groups: a sham + young mice, a sham + old mice, an OVX + soybean oil, an OVX + soybean oil with rice bran, an OVX + tea seed oil (TO), and an OVX + TO with rice bran diet group. The OVX groups were subcutaneously injected with d-galactose (100 mg/kg/day) and received a 15% (v/v) fructose drink. The rice bran and tea seed oil supplementation formed 10% of the diet (w/w). The results showed that the rice bran with TO diet increased the number of short-chain fatty acid (SCFA)-producing Clostridia and reduced the number of endotoxin-producing Tannerellaceae, which mitigated imbalances in the gut-liver-brain axis. Rice bran supplementation reduced the relative weight of the liver, levels of hepatic triglycerides and total cholesterol; aspartate transaminase and alanine aminotransferase activity; brain levels of proinflammatory cytokines, including interleukin-1β and tumor necrosis factor-α; and plasma 8-hydroxy-2-deoxyguanosine. This study concludes that rice bran inhibits hepatic fat accumulation, which mitigates peripheral metaflammation and oxidative damage and reduces neuroinflammation in the brain.
Collapse
Affiliation(s)
- Yu-Wen Chao
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Tang Tung
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
- Advanced Plant and Food Crop Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Suh-Ching Yang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Hitoshi Shirakawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Li-Han Su
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Pei-Yu Loe
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Wan-Chun Chiu
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Department of Nutrition, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| |
Collapse
|
33
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [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: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
34
|
Briceno Silva G, Arvelaez Pascucci J, Karim H, Kaur G, Olivas Lerma R, Mann AK, Gnanasekaran S, Thomas Garcia KD. Influence of the Onset of Menopause on the Risk of Developing Alzheimer's Disease. Cureus 2024; 16:e69124. [PMID: 39262936 PMCID: PMC11387275 DOI: 10.7759/cureus.69124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 09/13/2024] Open
Abstract
Menopause is a natural phase marked by the permanent cessation of menstrual cycles, occurring when the production of reproductive hormones from the ovaries stops for at least 12 consecutive months. Studies have suggested a potential connection between menopause and a heightened risk of developing Alzheimer's disease (AD), underscoring the significant role of reduced estrogen levels in the development of AD. Estrogen plays a crucial role in brain metabolism, influencing energy metabolism, synaptic plasticity, and cognitive functions. The cognitive benefits associated with hormone replacement therapy (HRT) are believed to be linked to estrogen's neuroprotective effects, either through direct action on the brain or indirectly by improving cardiovascular health. Extensive literature supports the positive impact of estrogen on brain cells. While the physiological effects of estrogen on the brain have not been consistently replicated in clinical trials, further research is crucial to provide more definitive recommendations to menopausal patients regarding the influence of HRT on AD. This review aims to comprehensively explore the interplay between menopause and AD, as well as the potential of HRT to mitigate cognitive decline in post-menopausal individuals.
Collapse
Affiliation(s)
| | | | - Hajira Karim
- Internal Medicine, Istanbul Medipol University, Istanbul, TUR
| | - Gurpreet Kaur
- Neurosurgery, Institute of Human Behaviour and Allied Sciences, New Delhi, IND
| | | | | | - Sulochana Gnanasekaran
- Internal Medicine, New York Medical College, St. Mary's and St. Clare's Hospital, Passaic, USA
| | | |
Collapse
|
35
|
Lu S, Zhao Q, Guan Y, Sun Z, Li W, Guo S, Zhang A. The communication mechanism of the gut-brain axis and its effect on central nervous system diseases: A systematic review. Biomed Pharmacother 2024; 178:117207. [PMID: 39067168 DOI: 10.1016/j.biopha.2024.117207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
Gut microbiota is involved in intricate and active metabolic processes the host's brain function, especially its role in immune responses, secondary metabolism, and symbiotic connections with the host. Gut microbiota can promote the production of essential metabolites, neurotransmitters, and other neuroactive chemicals that affect the development and treatment of central nervous system diseases. This article introduces the relevant pathways and manners of the communication between the brain and gut, summarizes a comprehensive overview of the current research status of key gut microbiota metabolites that affect the functions of the nervous system, revealing those adverse factors that affect typical communication between the brain-gut axis, and outlining the efforts made by researchers to alleviate these neurological diseases through targeted microbial interventions. The relevant pathways and manners of communication between the brain and gut contribute to the experimental design of new treatment plans and drug development. The factors that may cause changes in gut microbiota and affect metabolites, as well as current intervention methods are summarized, which helps improve gut microbiota brain dialogue, prevent adverse triggering factors from interfering with the gut microbiota system, and minimize neuropathological changes.
Collapse
Affiliation(s)
- Shengwen Lu
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Qiqi Zhao
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Yu Guan
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Zhiwen Sun
- Department of Gastroenterology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Wenhao Li
- School of Basic Medical Science of Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Sifan Guo
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China
| | - Aihua Zhang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China; INTI International University, Nilai 71800, Malaysia.
| |
Collapse
|
36
|
Walker KA, Rhodes ST, Liberman DA, Gore AC, Bell MR. Microglial responses to inflammatory challenge in adult rats altered by developmental exposure to polychlorinated biphenyls in a sex-specific manner. Neurotoxicology 2024; 104:95-115. [PMID: 39038526 PMCID: PMC11548868 DOI: 10.1016/j.neuro.2024.07.009] [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: 02/01/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Polychlorinated biphenyls are ubiquitous environmental contaminants linkedc with peripheral immune and neural dysfunction. Neuroimmune signaling is critical to brain development and later health; however, effects of PCBs on neuroimmune processes are largely undescribed. This study extends our previous work in neonatal or adolescent rats by investigating longer-term effects of perinatal PCB exposure on later neuroimmune responses to an inflammatory challenge in adulthood. Male and female Sprague-Dawley rats were exposed to a low-dose, environmentally relevant, mixture of PCBs (Aroclors 1242, 1248, and 1254, 1:1:1, 20 μg / kg dam BW per gestational day) or oil control during gestation and via lactation. Upon reaching adulthood, rats were given a mild inflammatory challenge with lipopolysaccharide (LPS, 50 μg / kg BW, ip) or saline control and then euthanized 3 hours later for gene expression analysis or 24 hours later for immunohistochemical labeling of Iba1+ microglia. PCB exposure did not alter gene expression or microglial morphology independently, but instead interacted with the LPS challenge in brain region- and sex-specific ways. In the female hypothalamus, PCB exposure blunted LPS responses of neuroimmune and neuromodulatory genes without changing microglial morphology. In the female prefrontal cortex, PCBs shifted Iba1+ cells from reactive to hyperramified morphology in response to LPS. Conversely, in the male hypothalamus, PCBs shifted cell phenotypes from hyperramified to reactive morphologies in response to LPS. The results highlight the potential for long-lasting effects of environmental contaminants that are differentially revealed over a lifetime, sometimes only after a secondary challenge. These neuroimmune endpoints are possible mechanisms for PCB effects on a range of neural dysfunction in adulthood, including mental health and neurodegenerative disorders. The findings suggest possible interactions with other environmental challenges that also influence neuroimmune systems.
Collapse
Affiliation(s)
- Katherine A Walker
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Simone T Rhodes
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Deborah A Liberman
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy and Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA.
| | - Margaret R Bell
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA; Division of Pharmacology and Toxicology, College of Pharmacy and Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
37
|
Yamamoto EA, Koike S, Wong C, Dennis LE, Luther MN, Scatena A, Khambadkone S, Iliff JJ, Lim MM, Levendovszky SR, Elliott JE, Barisano G, Müller-Oehring EM, Morales AM, Baker FC, Nagel BJ, Piantino J. Biological sex and BMI influence the longitudinal evolution of adolescent and young adult MRI-visible perivascular spaces. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.17.608337. [PMID: 39229241 PMCID: PMC11370374 DOI: 10.1101/2024.08.17.608337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background and Purpose An association recently emerged between magnetic resonance imaging (MRI)-visible perivascular spaces (MV-PVS) with intracerebral solute clearance and neuroinflammation, in adults. However, it is unknown how MV-PVS change throughout adolescence and what factors influence MV-PVS volume and morphology. This study assesses the temporal evolution of MV-PVS volume in adolescents and young adults, and secondarily evaluates the relationship between MV-PVS, age, sex, and body mass index (BMI). Materials and Methods This analysis included a 783 participant cohort from the longitudinal multicenter National Consortium on Alcohol and Neurodevelopment in Adolescence study that involved up to 6 imaging visits spanning 5 years. Healthy adolescents aged 12-21 years at study entry with at least two MRI scans were included. The primary outcome was mean MV-PVS volume (mm 3 /white matter cm 3 ). Results On average, males had greater MV-PVS volume at all ages compared to females. A linear mixed-effect model for MV-PVS volume was performed. Mean BMI and increases in a person's BMI were associated with increases in MV-PVS volume over time. In females only, changes in BMI correlated with MV-PVS volume. One unit increase in BMI above a person's average BMI was associated with a 0.021 mm 3 /cm 3 increase in MV-PVS volume (p<0.001). Conclusion This longitudinal study showed sex differences in MV-PVS features during adolescence and young adulthood. Importantly, we report that increases in BMI from a person's mean BMI are associated with increases in MV-PVS volume in females only. These findings suggest a potential link between MV-PVS, sex, and BMI that warrants future study.
Collapse
|
38
|
Nyarko-Danquah I, Pajarillo E, Kim S, Digman A, Multani HK, Ajayi I, Son DS, Aschner M, Lee E. Microglial Sp1 induced LRRK2 upregulation in response to manganese exposure, and 17β-estradiol afforded protection against this manganese toxicity. Neurotoxicology 2024; 103:105-114. [PMID: 38857675 PMCID: PMC11972059 DOI: 10.1016/j.neuro.2024.05.007] [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: 02/12/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024]
Abstract
Chronic exposure to elevated levels of manganese (Mn) causes a neurological disorder referred to as manganism, presenting symptoms similar to those of Parkinson's disease (PD), yet the mechanisms by which Mn induces its neurotoxicity are not completely understood. 17β-estradiol (E2) affords neuroprotection against Mn toxicity in various neural cell types including microglia. Our previous studies have shown that leucine-rich repeat kinase 2 (LRRK2) mediates Mn-induced inflammatory toxicity in microglia. The LRRK2 promoter sequences contain three putative binding sites of the transcription factor (TF), specificity protein 1 (Sp1), which increases LRRK2 promoter activity. In the present study, we tested if the Sp1-LRRK2 pathway plays a role in both Mn toxicity and the protection afforded by E2 against Mn toxicity in BV2 microglial cells. The results showed that Mn induced cytotoxicity, oxidative stress, and tumor necrosis factor-α production, which were attenuated by an LRRK2 inhibitor, GSK2578215A. The overexpression of Sp1 increased LRRK2 promoter activity, mRNA and protein levels, while inhibition of Sp1 with its pharmacological inhibitor, mithramycin A, attenuated the Mn-induced increases in LRRK2 expression. Furthermore, E2 attenuated the Mn-induced Sp1 expression by decreasing the expression of Sp1 via the promotion of the ubiquitin-dependent degradation pathway, which was accompanied by increased protein levels of RING finger protein 4, the E3-ligase of Sp1, Sp1 ubiquitination, and SUMOylation. Taken together, our novel findings suggest that Sp1 serves as a critical TF in Mn-induced LRRK2 expression as well as in the protection afforded by E2 against Mn toxicity through reduction of LRRK2 expression in microglia.
Collapse
Affiliation(s)
- Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Harpreet Kaur Multani
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Itunu Ajayi
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA.
| |
Collapse
|
39
|
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
|
40
|
Siddiqui N, Sharma A, Kesharwani A, Anurag, Parihar VK. Exploring role of natural compounds in molecular alterations associated with brain ageing: A perspective towards nutrition for ageing brain. Ageing Res Rev 2024; 97:102282. [PMID: 38548242 DOI: 10.1016/j.arr.2024.102282] [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: 08/30/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/12/2024]
Abstract
Aging refers to complete deterioration of physiological integrity and function. By midcentury, adults over 60 years of age and children under 15 years will begin to outnumber people in working age. This shift will bring multiple global challenges for economy, health, and society. Eventually, aging is a natural process playing a vital function in growth and development during pediatric stage, maturation during adult stage, and functional depletion. Tissues experience negative consequences with enhanced genomic instability, deregulated nutrient sensing, mitochondrial dysfunction, and decline in performance on cognitive tasks. As brain ages, its volume decreases, neurons & glia get inflamed, vasculature becomes less developed, blood pressure increases with a risk of stroke, ischemia, and cognitive deficits. Diminished cellular functions leads to progressive reduction in functional and emotional capacity with higher possibility of disease and finally death. This review overviews cellular as well as molecular aspects of aging, biological pathway related to accelerated brain aging, and strategies minimizing cognitive aging. Age-related changes include altered bioenergetics, decreased neuroplasticity and flexibility, aberrant neural activity, deregulated Ca2+ homeostasis in neurons, buildup of reactive oxygen species, and neuro-inflammation. Unprecedented progress has been achieved in recent studies, particularly in terms of how herbal or natural substances affect genetic pathways and biological functions that have been preserved through evolution. Herein, the present work provides an overview of ageing and age-related disorders and explore the molecular mechanisms that underlie therapeutic effects of herbal and natural chemicals on neuropathological signs of brain aging.
Collapse
Affiliation(s)
- Nazia Siddiqui
- Department of Pharmaceutical Technology, MIET, Meerut 250005, India
| | - Alok Sharma
- Department of Pharmaceutical Technology, MIET, Meerut 250005, India.
| | - Anuradha Kesharwani
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur 844102, India
| | - Anurag
- Department of Pharmaceutical Technology, MIET, Meerut 250005, India
| | - Vipan Kumar Parihar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur 844102, India.
| |
Collapse
|
41
|
Han T, Xu Y, Sun L, Hashimoto M, Wei J. Microglial response to aging and neuroinflammation in the development of neurodegenerative diseases. Neural Regen Res 2024; 19:1241-1248. [PMID: 37905870 PMCID: PMC11467914 DOI: 10.4103/1673-5374.385845] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/30/2023] [Accepted: 07/17/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cellular senescence and chronic inflammation in response to aging are considered to be indicators of brain aging; they have a great impact on the aging process and are the main risk factors for neurodegeneration. Reviewing the microglial response to aging and neuroinflammation in neurodegenerative diseases will help understand the importance of microglia in neurodegenerative diseases. This review describes the origin and function of microglia and focuses on the role of different states of the microglial response to aging and chronic inflammation on the occurrence and development of neurodegenerative diseases, including Alzheimer's disease, Huntington's chorea, and Parkinson's disease. This review also describes the potential benefits of treating neurodegenerative diseases by modulating changes in microglial states. Therefore, inducing a shift from the neurotoxic to neuroprotective microglial state in neurodegenerative diseases induced by aging and chronic inflammation holds promise for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Tingting Han
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yuxiang Xu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lin Sun
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng, Henan Province, China
| | - Makoto Hashimoto
- Department of Basic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| |
Collapse
|
42
|
Li S, Liu W, Huang Z, Lin H, Ning Y, Li Z. Sex-specific associations between sex hormones and clinical symptoms in late-life schizophrenia. Schizophr Res 2024; 267:1-7. [PMID: 38492529 DOI: 10.1016/j.schres.2024.03.002] [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: 07/25/2023] [Revised: 02/14/2024] [Accepted: 03/08/2024] [Indexed: 03/18/2024]
Abstract
The prevalence of late-life schizophrenia is increasing with high burden. It is well-documented that schizophrenia affects men and women differently in terms of symptoms. Sex hormones, which play a role in the pathology and symptoms of schizophrenia, are greatly affected by aging. To the best of our knowledge, this is a study to examine the sex differences in psychiatric symptoms and their correlation with sex hormones in participants with late-life schizophrenia. Positive and Negative Syndrome Scale (PANSS) factors were evaluated. Testosterone, follicle-stimulating hormone (FSH), luteinizing hormone (LH), estradiol, progesterone, and prolactin were measured. Male participants with late-life schizophrenia had more severe negative symptoms than female participants (z = -2.56, P = 0.010), while female participants had more severe anxiety/depression compared to male participants (z = 2.64, P = 0.008). Testosterone levels in male participants were positively associated with negative symptoms (β = 0.23, t = 2.27, P = 0.025), while there was no significant association between sex hormones and symptoms in female participants. In conclusion, higher testosterone levels were associated with more severe negative symptoms in male participants with late-life schizophrenia, suggesting that attention should be paid to the sex differences in late-life schizophrenia in clinical practice.
Collapse
Affiliation(s)
- Shuyun Li
- The Affiliated Brain Hospital of 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, Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Weijian Liu
- Peking University Sixth Hospital, Peking University, Beijing, China
| | - Zebin Huang
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hong Lin
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuping Ning
- The Affiliated Brain Hospital of 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, Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| | - Zezhi Li
- The Affiliated Brain Hospital of 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, Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
43
|
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: 0] [Impact Index Per Article: 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
|
44
|
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
|
45
|
Ma’arif B, Anwar MF, Hidayatullah H, Muslikh FA, Suryadinata A, Sugihantoro H, Mirza DM, Maulina N, Taek MM. Effect of polar fractions of Marsilea crenata C. Presl. leaves in zebrafish locomotor activity. J Adv Pharm Technol Res 2024; 15:125-129. [PMID: 38903546 PMCID: PMC11186547 DOI: 10.4103/japtr.japtr_241_23] [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: 04/30/2023] [Revised: 11/01/2023] [Accepted: 02/02/2024] [Indexed: 06/22/2024] Open
Abstract
Neurodegenerative diseases (NDs) are pathological conditions initiated by the loss of neuronal cell structure and the progressive decline in function caused by prolonged neuroinflammation. Postmenopausal women are at a high risk of experiencing NDs due to estrogen deficiency in their bodies, necessitating the administration of phytoestrogens as a replacement for estrogen in the body. One alternative therapy is administering phytoestrogens, estrogen-like substances from plants, which can be obtained from Marsilea crenata C. Presl. leaves. The purpose of this study was to determine whether administration of the n-butanol fraction (BF) and water fraction (WF) of M. crenata leaves could increase locomotor activity in rotenone-induced zebrafish. Treatment was given to each group of zebrafish with BF and WF at doses of 2.5; 5; 10; and 20 ppm to determine the locomotor activity. Then an analysis was carried out by looking at each movement of the zebrafish swimming for 1 min at the time of observation on days 0, 7, 14, 21, and 28. The result showed that BF and WF significantly increased the locomotor activity of zebrafish at the optimum dose of 20 ppm for BF and 5 ppm for WF compared to the negative control. This concludes that the polar fraction of M. crenata leaves is proven to have the potential to prevent ND progressivity.
Collapse
Affiliation(s)
- Burhan Ma’arif
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Mohamad Fajrul Anwar
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Hidayatullah Hidayatullah
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Faisal Akhmal Muslikh
- Department of Pharmacy, Faculty of Pharmacy, Bhakti Wiyata Institute of Health Sciences, Kediri, East Java, Indonesia
| | - Arief Suryadinata
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Hajar Sugihantoro
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Denis Mery Mirza
- Department of Pharmacy, Faculty of Medicine, Islamic University of Malang, Malang, Indonesia
| | - Novia Maulina
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Maximus M. Taek
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Widya Mandira Catholic University, Kupang, Indonesia
| |
Collapse
|
46
|
Mengelkoch S, Slavich GM. Sex Differences in Stress Susceptibility as a Key Mechanism Underlying Depression Risk. Curr Psychiatry Rep 2024; 26:157-165. [PMID: 38470558 PMCID: PMC10978685 DOI: 10.1007/s11920-024-01490-8] [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] [Accepted: 01/26/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE OF REVIEW Although females are at relatively greater risk for a variety of disorders, including depression, the biological mechanisms underlying this striking health disparity remain unclear. To address this issue, we highlight sex differences in stress susceptibility as a key mechanism potentially driving this effect and describe the interacting inflammatory, hormonal, epigenomic, and social-environmental mechanisms involved. RECENT FINDINGS Using the Social Signal Transduction Theory of Depression as a theoretical framework, women's elevated risk for depression may stem from a tight link between life stress, inflammation, and depression in women. Further, research finds hormonal contraceptive use alters cortisol and inflammatory reactivity to acute stress in ways that may increase depression risk in females. Finally, beyond established epigenetic mechanisms, mothers may transfer risk for depression to their female offspring through stressful family environments, which influence stress generation and stress-related gene expression. Together, these findings provide initial, biologically plausible clues that may help explain the relatively greater risk for depression in females vs. males. Looking forward, much more research is needed to address the longstanding underrepresentation of females in biomedical research on the biology of stress and depression.
Collapse
Affiliation(s)
- Summer Mengelkoch
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA.
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| |
Collapse
|
47
|
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
|
48
|
Laaksonen S, Saraste M, Nylund M, Hinz R, Snellman A, Rinne J, Matilainen M, Airas L. Sex-driven variability in TSPO-expressing microglia in MS patients and healthy individuals. Front Neurol 2024; 15:1352116. [PMID: 38445263 PMCID: PMC10913932 DOI: 10.3389/fneur.2024.1352116] [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: 12/07/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Background Males with multiple sclerosis (MS) have a higher risk for disability progression than females, but the reasons for this are unclear. Objective We hypothesized that potential differences in TSPO-expressing microglia between female and male MS patients could contribute to sex differences in clinical disease progression. Methods The study cohort consisted of 102 MS patients (mean (SD) age 45.3 (9.7) years, median (IQR) disease duration 12.1 (7.0-17.2) years, 72% females, 74% relapsing-remitting MS) and 76 age- and sex-matched healthy controls. TSPO-expressing microglia were measured using the TSPO-binding radioligand [11C](R)-PK11195 and brain positron emission tomography (PET). TSPO-binding was quantified as distribution volume ratio (DVR) in normal-appearing white matter (NAWM), thalamus, whole brain and cortical gray matter (cGM). Results Male MS patients had higher DVRs compared to female patients in the whole brain [1.22 (0.04) vs. 1.20 (0.02), p = 0.002], NAWM [1.24 (0.06) vs. 1.21 (0.05), p = 0.006], thalamus [1.37 (0.08) vs. 1.32 (0.02), p = 0.008] and cGM [1.25 (0.04) vs. 1.23 (0.04), p = 0.028]. Similarly, healthy men had higher DVRs compared to healthy women except for cGM. Of the studied subgroups, secondary progressive male MS patients had the highest DVRs in all regions, while female controls had the lowest DVRs. Conclusion We observed higher TSPO-binding in males compared to females among people with MS and in healthy individuals. This sex-driven inherent variability in TSPO-expressing microglia may predispose male MS patients to greater likelihood of disease progression.
Collapse
Affiliation(s)
- Sini Laaksonen
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Maija Saraste
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Marjo Nylund
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, United Kingdom
| | - Anniina Snellman
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Juha Rinne
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Markus Matilainen
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Laura Airas
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| |
Collapse
|
49
|
Karnik SJ, Margetts TJ, Wang HS, Movila A, Oblak AL, Fehrenbacher JC, Kacena MA, Plotkin LI. Mind the Gap: Unraveling the Intricate Dance Between Alzheimer's Disease and Related Dementias and Bone Health. Curr Osteoporos Rep 2024; 22:165-176. [PMID: 38285083 PMCID: PMC10912190 DOI: 10.1007/s11914-023-00847-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/30/2024]
Abstract
PURPOSE OF REVIEW This review examines the linked pathophysiology of Alzheimer's disease/related dementia (AD/ADRD) and bone disorders like osteoporosis. The emphasis is on "inflammaging"-a low-level inflammation common to both, and its implications in an aging population. RECENT FINDINGS Aging intensifies both ADRD and bone deterioration. Notably, ADRD patients have a heightened fracture risk, impacting morbidity and mortality, though it is uncertain if fractures worsen ADRD. Therapeutically, agents targeting inflammation pathways, especially Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) and TNF-α, appear beneficial for both conditions. Additionally, treatments like Sirtuin 1 (SIRT-1), known for anti-inflammatory and neuroprotective properties, are gaining attention. The interconnectedness of AD/ADRD and bone health necessitates a unified treatment approach. By addressing shared mechanisms, we can potentially transform therapeutic strategies, enriching our understanding and refining care in our aging society. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Sonali J Karnik
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tyler J Margetts
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hannah S Wang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alexandru Movila
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adrian L Oblak
- Department of Radiology & Imaging Sciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jill C Fehrenbacher
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
| | - Lilian I Plotkin
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
50
|
Dadwal S, Heneka MT. Microglia heterogeneity in health and disease. FEBS Open Bio 2024; 14:217-229. [PMID: 37945346 PMCID: PMC10839410 DOI: 10.1002/2211-5463.13735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/12/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), have received significant attention due to their critical roles in maintaining brain homeostasis and mediating cerebral immune responses. Understanding the origin of microglia has been a subject of great interest, and emerging evidence suggests that microglia consist of multiple subpopulations with unique molecular and functional characteristics. These subpopulations of microglia may exhibit specialized roles in response to different environmental cues as in disease conditions. The newfound understanding of microglial heterogeneity has significant implications for elucidating their roles in both physiological and pathological conditions. In the context of disease, microglia have been studied rigorously as they play a very important role in neuroinflammation. Dysregulated microglial activation and function contribute to chronic inflammation. Further exploration of microglial heterogeneity and their interactions with other cell types in the CNS will undoubtedly pave the way to novel therapeutic strategies targeting microglia-mediated pathologies. In this review, we discuss the latest advances in the field of microglia research, focusing specifically on the origin and subpopulations of microglia, the populations of microglia types in the brains of patients with neurodegenerative diseases, and how microglia are regulated in the healthy CNS.
Collapse
Affiliation(s)
- Shilauni Dadwal
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgBelvalLuxembourg
| | - Michael T. Heneka
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgBelvalLuxembourg
- Division of Infectious Diseases and ImmunologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| |
Collapse
|