1
|
Gui Y, Zhou G, Cui S, Li H, Lu H, Zhao H. The left amygdala is genetically sexually-dimorphic: multi-omics analysis of structural MRI volumes. Transl Psychiatry 2025; 15:17. [PMID: 39843917 PMCID: PMC11754786 DOI: 10.1038/s41398-025-03223-8] [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] [Received: 03/19/2024] [Revised: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Brain anatomy plays a key role in complex behaviors and mental disorders that are sexually divergent. While our understanding of the sex differences in the brain anatomy remains relatively limited, particularly of the underlying genetic and molecular mechanisms that contribute to these differences. We performed the largest study of sex differences in brain volumes (N = 33,208) by examining sex differences both in the raw brain volumes and after controlling the whole brain volumes. Genetic correlation analysis revealed sex differences only in the left amygdala. We compared transcriptome differences between males and females using data from GTEx and characterized cell-type compositions using GTEx bulk amygdala RNA-seq data and LIBD amygdala single-cell reference profiles. We also constructed polygenic risk scores (PRS) to investigate sex-specific genetic correlations between left amygdala volume and mental disorders (N = 25,576~105,318) of Psychiatric Genomics Consortium and other traits of UKB (N = 347,996). Although there were pronounced sex differences in brain volumes, there was no difference in the heritability between sexes. There was a significant sex-specific genetic correlation between male and female left amygdala. We identified sex-differentiated genetic effects of PRSs for schizophrenia on left amygdala volume, as well as significant sex-differentiated genetic correlations between PRSs of left amygdala and six traits in UKB. We also found several sex-differentially expressed genes in the amygdala. These findings not only advanced the current knowledge of genetic basis of sex differences in brain anatomy, but also presented an important clue for future research on the mechanism of sex differences in mental disorders and targeted treatments.
Collapse
Affiliation(s)
- Yuanyuan Gui
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Geyu Zhou
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Shuya Cui
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Hongyu Li
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Hui Lu
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Hongyu Zhao
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA.
| |
Collapse
|
2
|
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] [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
|
3
|
Biel D, Suárez-Calvet M, Dewenter A, Steward A, Roemer SN, Dehsarvi A, Zhu Z, Pescoller J, Frontzkowski L, Kreuzer A, Haass C, Schöll M, Brendel M, Franzmeier N. Female sex is linked to a stronger association between sTREM2 and CSF p-tau in Alzheimer's disease. EMBO Mol Med 2025:10.1038/s44321-024-00190-3. [PMID: 39794447 DOI: 10.1038/s44321-024-00190-3] [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: 08/02/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
In Alzheimer's disease (AD), Aβ triggers p-tau secretion, which drives tau aggregation. Therefore, it is critical to characterize modulators of Aβ-related p-tau increases which may alter AD trajectories. Here, we assessed whether factors known to alter tau levels in AD modulate the association between fibrillar Aβ and secreted p-tau181 determined in the cerebrospinal fluid (CSF). To assess potentially modulating effects of female sex, younger age, and ApoE4, we included 322 ADNI participants with cross-sectional/longitudinal p-tau181. To determine effects of microglial activation on p-tau181, we included 454 subjects with cross-sectional CSF sTREM2. Running ANCOVAs for nominal and linear regressions for metric variables, we found that women had higher Aβ-related p-tau181 levels. Higher sTREM2 was associated with elevated p-tau181, with stronger associations in women. Similarly, ApoE4 was related to higher p-tau181 levels and faster p-tau181 increases, with stronger effects in female ApoE4 carriers. Our results show that sex alone modulates the Aβ to p-tau axis, where women show higher Aβ-dependent p-tau secretion, potentially driven by elevated sTREM2-related microglial activation and stronger effects of ApoE4 carriership in women.
Collapse
Affiliation(s)
- Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei de Neurologia, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Anna Dewenter
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Sebastian N Roemer
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Zeyu Zhu
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Julia Pescoller
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Lukas Frontzkowski
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Annika Kreuzer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Schöll
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| |
Collapse
|
4
|
Shvetcov A, Thomson S, Cho AN, Wilkins HM, Reed JH, Swerdlow RH, Brown DA, Finney CA. Proteome profiling of cerebrospinal fluid using machine learning shows a unique protein signature associated with APOE4 genotype. Aging Cell 2024:e14439. [PMID: 39722190 DOI: 10.1111/acel.14439] [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: 06/16/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Proteome changes associated with APOE4 variant carriage that are independent of Alzheimer's disease (AD) pathology and diagnosis are unknown. This study investigated APOE4 proteome changes in people with AD, mild cognitive impairment, and no impairment. Clinical, APOE genotype, and cerebrospinal fluid (CSF) proteome and AD biomarker data was sourced from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Proteome profiling was done using supervised machine learning. We found an APOE4-specific proteome signature that was independent of cognitive diagnosis and AD pathological biomarkers, and increased the risk of progression to cognitive impairment. Proteins were enriched in brain regions including the caudate and cortex and cells including endothelial cells, oligodendrocytes, and astrocytes. Enriched peripheral immune cells included T cells, macrophages, and B cells. APOE4 carriers have a unique CSF proteome signature associated with a strong brain and peripheral immune and inflammatory phenotype that likely underlies APOE4 carriers' vulnerability to cognitive decline and AD as they age.
Collapse
Affiliation(s)
- Artur Shvetcov
- Translational Dementia Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
- Department of Psychological Medicine, Sydney Children's Hospital Network, Sydney, NSW, Australia
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Shannon Thomson
- Translational Dementia Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ann-Na Cho
- Human Brain Microphysiology Systems Group, School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Research Centre, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Centre, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Centre, Kansas City, KS, USA
| | - Joanne H Reed
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Autoimmunity and Amyloidosis Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Centre, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Centre, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Centre, Kansas City, KS, USA
- Department of Molecular and Integrative Physiology, University of Kansas Medical Centre, Kansas City, KS, USA
| | - David A Brown
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
- Department of Immunopathology, Institute for Clinical Pathology and Medical Research-New South Wales Health Pathology, Sydney, NSW, Australia
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Caitlin A Finney
- Translational Dementia Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
5
|
Alonaizan R, K Alotaibi W, Alsulami A, M Alkhulaifi F, Alomar S. Sex-Differences Influence Depressive-Like Behaviour via Alterations in Microglial Expression of GIF-1, TREM2, and IL-1β in an Acute Lipopolysaccharide-Induced Murine Neuroinflammation Model. Immunol Invest 2024:1-17. [PMID: 39701694 DOI: 10.1080/08820139.2024.2440006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
BACKGROUND Neurodegenerative diseases (NDs) have caused serious health issues worldwide. A growing body of evidence suggests a correlation between neuroinflammation and abnormal microglial activity with ND symptoms. Microglia survey play crucial roles in CNS during health and the injury. It is proposed that sex affects microglial roles during inflammation, resulting in mouse behavioural changes and expression alterations in key markers related to microglia functions. METHODS Male and female C57BL/6 mice were injected with a single dose of LPS (5 mg/kg, i.p.) or saline. After 48 h, an open field test was conducted, followed by brain tissues collection for measuring the expression of IGF-1, IL-1β and TREM2 and Immunohistochemistry (IHC) analysis for NLRP3 level. RESULTS Males displayed greater depressive-like behaviour in the OFT, with lower levels of IGF-1, IL-1β, and NLRP3 and high TREM2 expression. Female mice did not exhibit this behaviour, in contrast to male mice, they exhibited increased IL-1β and NLRP3 expression. DISCUSSION This study revealed that LPS-induced sex-specific changes in genes involved in neuronal cell survival caused behavioural alterations in male mice. Moreover, females had observed inflammatory responses that had no impact on behavioural alterations. Overall, both sexes exhibited sex-specific microglial activation states.
Collapse
Affiliation(s)
- Rasha Alonaizan
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Wafa K Alotaibi
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Asma Alsulami
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Fadwa M Alkhulaifi
- Biology Department, College of Science, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Suliman Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
6
|
Chen LX, Zhang MD, Xu HF, Ye HQ, Chen DF, Wang PS, Bao ZW, Zou SM, Lv YT, Wu ZY, Li HF. Single-Nucleus RNA Sequencing Reveals the Spatiotemporal Dynamics of Disease-Associated Microglia in Amyotrophic Lateral Sclerosis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0548. [PMID: 39664295 PMCID: PMC11632836 DOI: 10.34133/research.0548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/13/2024]
Abstract
Disease-associated microglia (DAM) are observed in neurodegenerative diseases, demyelinating disorders, and aging. However, the spatiotemporal dynamics and evolutionary trajectory of DAM during the progression of amyotrophic lateral sclerosis (ALS) remain unclear. Using a mouse model of ALS that expresses a human SOD1 gene mutation, we found that the microglia subtype DAM begins to appear following motor neuron degeneration, primarily in the brain stem and spinal cord. Using reverse transcription quantitative polymerase chain reaction, RNAscope in situ hybridization, and flow cytometry, we found that DAM increased in number as the disease progressed, reaching their peak in the late disease stage. DAM responded to disease progression in both SOD1G93A mice and sporadic ALS and C9orf72-mutated patients. Motor neuron loss in SOD1G93A mice exhibited 2 accelerated phases: P90 to P110 (early stage) and P130 to P150 (late stage). Some markers were synchronized with the accelerated phase of motor neuron loss, suggesting that these proteins may be particularly responsive to disease progression. Through pseudotime trajectory analysis, we tracked the dynamic transition of homeostatic microglia into DAM and cluster 6 microglia. Interestingly, we used the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 to deplete microglia in SOD1G93A mice and observed that DAM survival is independent of CSF1R. An in vitro phagocytosis assay directly confirmed that DAM could phagocytose more beads than other microglia subtypes. These findings reveal that the induction of the DAM phenotype is a shared cross-species and cross-subtype characteristic in ALS. Inducing the DAM phenotype and enhancing its function during the early phase of disease progression, or the time window between P130 and P150 where motor neuron loss slows, could serve as a neuroprotective strategy for ALS.
Collapse
Affiliation(s)
- Lu-Xi Chen
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Zhejiang Key Laboratory of Rare Diseases for Precision Medicine and Clinical Translation, Hangzhou, Zhejiang, China
- Department of Neurology, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, China
| | - Mei-Di Zhang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Feng Xu
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Zhejiang Key Laboratory of Rare Diseases for Precision Medicine and Clinical Translation, Hangzhou, Zhejiang, China
| | - Hai-Qin Ye
- Institute of Developmental and Regenerative Biology, Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences,
Hangzhou Normal University, Hangzhou, China
| | - Dian-Fu Chen
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Zhejiang Key Laboratory of Rare Diseases for Precision Medicine and Clinical Translation, Hangzhou, Zhejiang, China
| | - Pei-Shan Wang
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Zhejiang Key Laboratory of Rare Diseases for Precision Medicine and Clinical Translation, Hangzhou, Zhejiang, China
- Department of Neurology, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi-Wei Bao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng-Mei Zou
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Zhejiang Key Laboratory of Rare Diseases for Precision Medicine and Clinical Translation, Hangzhou, Zhejiang, China
| | - Yong-Ting Lv
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Zhejiang Key Laboratory of Rare Diseases for Precision Medicine and Clinical Translation, Hangzhou, Zhejiang, China
- Department of Neurology, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi-Ying Wu
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Zhejiang Key Laboratory of Rare Diseases for Precision Medicine and Clinical Translation, Hangzhou, Zhejiang, China
- Department of Neurology, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine,
Zhejiang University, Hangzhou, China
| | - Hong-Fu Li
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Zhejiang Key Laboratory of Rare Diseases for Precision Medicine and Clinical Translation, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Chaumeil J, Morey C. [X chromosome regulation and female functional specificities: Are two Xs better than one?]. Med Sci (Paris) 2024; 40:935-946. [PMID: 39705564 DOI: 10.1051/medsci/2024179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024] Open
Abstract
What if the presence of two X chromosomes confers functional specificities on female cells and contributes to the different susceptibilites of men and women to certain diseases? One of the X chromosomes is randomly silenced in each female cell from the embryonic stage, theoretically making the sexes equal. This silencing of the X chromosome is a unique epigenetic process, affecting an entire chromosome and resulting in mosaic expression of X-linked genes throughout the body. However, some genes escape this process and X-inactivation appears to be somewhat labile in certain cell types. What are the physiological implications of these observations? This question is beginning to be explored, particularly in the immune and nervous systems, where several pathologies have sexual bias.
Collapse
Affiliation(s)
- Julie Chaumeil
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Céline Morey
- UMR7216 - Épigénétique et destin cellulaire, CNRS, Université Paris Cité, Paris, France
| |
Collapse
|
8
|
Kodosaki E, Bell R, Sogorb-Esteve A, Wiltshire K, Zetterberg H, Heslegrave A. More than microglia: myeloid cells and biomarkers in neurodegeneration. Front Neurosci 2024; 18:1499458. [PMID: 39544911 PMCID: PMC11560917 DOI: 10.3389/fnins.2024.1499458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024] Open
Abstract
The role of myeloid cells (granulocytes and monocytes) in neurodegeneration and neurodegenerative disorders (NDD) is indisputable. Here we discuss the roles of myeloid cells in neurodegenerative diseases, and the recent advances in biofluid and imaging myeloid biomarker research with a focus on methods that can be used in the clinic. For this review, evidence from three neurodegenerative diseases will be included, Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). We discuss the potential for these biomarkers to be used in humans with suspected NDD as prognostic, diagnostic, or monitoring tools, identify knowledge gaps in literature, and propose potential approaches to further elucidate the role of myeloid cells in neurodegeneration and better utilize myeloid biomarkers in the understanding and treatment of NDD.
Collapse
Affiliation(s)
- Eleftheria Kodosaki
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Rosie Bell
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Aitana Sogorb-Esteve
- UK Dementia Research Institute at UCL, London, United Kingdom
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Katharine Wiltshire
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong SAR, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Amanda Heslegrave
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| |
Collapse
|
9
|
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
|
10
|
Yeung J, DeYoung T, Spring S, de Guzman AE, Elder MW, Beauchamp A, Wong CS, Palmert MR, Lerch JP, Nieman BJ. Sex chromosomes and hormones independently influence healthy brain development but act similarly after cranial radiation. Proc Natl Acad Sci U S A 2024; 121:e2404042121. [PMID: 39207735 PMCID: PMC11388377 DOI: 10.1073/pnas.2404042121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The course of normal development and response to pathology are strongly influenced by biological sex. For instance, female childhood cancer survivors who have undergone cranial radiation therapy (CRT) tend to display more pronounced cognitive deficits than their male counterparts. Sex effects can be the result of sex chromosome complement (XX vs. XY) and/or gonadal hormone influence. The contributions of each can be separated using the four-core genotype mouse model (FCG), where sex chromosome complement and gonadal sex are decoupled. While studies of FCG mice have evaluated brain differences in adulthood, it is still unclear how sex chromosome and sex hormone effects emerge through development in both healthy and pathological contexts. Our study utilizes longitudinal MRI with the FCG model to investigate sex effects in healthy development and after CRT in wildtype and immune-modified Ccl2-knockout mice. Our findings in normally developing mice reveal a relatively prominent chromosome effect prepubertally, compared to sex hormone effects which largely emerge later. Spatially, sex chromosome and hormone influences were independent of one another. After CRT in Ccl2-knockout mice, both male chromosomes and male hormones similarly improved brain outcomes but did so more separately than in combination. Our findings highlight the crucial role of sex chromosomes in early development and identify roles for sex chromosomes and hormones after CRT-induced inflammation, highlighting the influences of biological sex in both normal brain development and pathology.
Collapse
Affiliation(s)
- Jonas Yeung
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
| | - Taylor DeYoung
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
| | - Shoshana Spring
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
| | - A Elizabeth de Guzman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Rovereto TN 38068, Italy
| | - Madeline W Elder
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
| | - Antoine Beauchamp
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
| | - C Shun Wong
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Odette Cancer Centre, Toronto ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto ON M5T 1P5, Canada
| | - Mark R Palmert
- Division of Endocrinology, The Hospital for Sick Children, University of Toronto, Toronto ON M5G 1X8, Canada
- Department of Pediatrics, University of Toronto, Toronto ON M5S 1A8, Canada
- Department of Physiology, University of Toronto, Toronto ON M5S 1A8, Canada
- Genetics and Genome Biology, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Wellcome Centre for Integrative Neuroimaging, Medical Sciences Division, University of Oxford, Oxford, OXF OX3 9DU, United Kingdom
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, OXF OX3 9DU, United Kingdom
| | - Brian J Nieman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| |
Collapse
|
11
|
Anwar MM, Pérez-Martínez L, Pedraza-Alva G. Exploring the Significance of Microglial Phenotypes and Morphological Diversity in Neuroinflammation and Neurodegenerative Diseases: From Mechanisms to Potential Therapeutic Targets. Immunol Invest 2024; 53:891-946. [PMID: 38836373 DOI: 10.1080/08820139.2024.2358446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Studying various microglial phenotypes and their functions in neurodegenerative diseases is crucial due to the intricate nature of their phenomics and their vital immunological role. Microglia undergo substantial phenomic changes, encompassing morphological, transcriptional, and functional aspects, resulting in distinct cell types with diverse structures, functions, properties, and implications. The traditional classification of microglia as ramified, M1 (proinflammatory), or M2 (anti-inflammatory) phenotypes is overly simplistic, failing to capture the wide range of recently identified microglial phenotypes in various brain regions affected by neurodegenerative diseases. Altered and activated microglial phenotypes deviating from the typical ramified structure are significant features of many neurodegenerative conditions. Understanding the precise role of each microglial phenotype is intricate and sometimes contradictory. This review specifically focuses on elucidating recent modifications in microglial phenotypes within neurodegenerative diseases. Recognizing the heterogeneity of microglial phenotypes in diseased states can unveil novel therapeutic strategies for targeting microglia in neurodegenerative diseases. Moreover, the exploration of the use of healthy isolated microglia to mitigate disease progression has provided an innovative perspective. In conclusion, this review discusses the dynamic landscape of mysterious microglial phenotypes, emphasizing the need for a nuanced understanding to pave the way for innovative therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Leonor Pérez-Martínez
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| | - Gustavo Pedraza-Alva
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| |
Collapse
|
12
|
Saika F, Fukazawa Y, Hatano Y, Kishioka S, Hino Y, Hino S, Suzuki K, Kiguchi N. Sexually dimorphic effects of pexidartinib on nerve injury-induced neuropathic pain in mice. Glia 2024; 72:1402-1417. [PMID: 38591338 DOI: 10.1002/glia.24535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024]
Abstract
It is well-established that spinal microglia and peripheral macrophages play critical roles in the etiology of neuropathic pain; however, growing evidence suggests sex differences in pain hypersensitivity owing to microglia and macrophages. Therefore, it is crucial to understand sex- and androgen-dependent characteristics of pain-related myeloid cells in mice with nerve injury-induced neuropathic pain. To deplete microglia and macrophages, pexidartinib (PLX3397), an inhibitor of the colony-stimulating factor 1 receptor, was orally administered, and mice were subjected to partial sciatic nerve ligation (PSL). Following PSL induction, healthy male and female mice and male gonadectomized (GDX) mice exhibited similar levels of spinal microglial activation, peripheral macrophage accumulation, and mechanical allodynia. Treatment with PLX3397 significantly suppressed mechanical allodynia in normal males; this was not observed in female and GDX male mice. Sex- and androgen-dependent differences in the PLX3397-mediated preventive effects were observed on spinal microglia and dorsal root ganglia (DRG) macrophages, as well as in expression patterns of pain-related inflammatory mediators in these cells. Conversely, no sex- or androgen-dependent differences were detected in sciatic nerve macrophages, and inhibition of peripheral CC-chemokine receptor 5 prevented neuropathic pain in both sexes. Collectively, these findings demonstrate the presence of considerable sex- and androgen-dependent differences in the etiology of neuropathic pain in spinal microglia and DRG macrophages but not in sciatic nerve macrophages. Given that the mechanisms of neuropathic pain may differ among experimental models and clinical conditions, accumulating several lines of evidence is crucial to comprehensively clarifying the sex-dependent regulatory mechanisms of pain.
Collapse
Affiliation(s)
- Fumihiro Saika
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
- Faculty of Wakayama Health Care Sciences, Takarazuka University of Medical and Health Care, Wakayama, Japan
| | - Yohji Fukazawa
- Department of Anatomy, Kansai University of Health Sciences, Osaka, Japan
| | - Yu Hatano
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, Japan
| | - Shiroh Kishioka
- Faculty of Wakayama Health Care Sciences, Takarazuka University of Medical and Health Care, Wakayama, Japan
| | - Yuko Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Kentaro Suzuki
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, Japan
| | - Norikazu Kiguchi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
13
|
Jacob SM, Lee S, Kim SH, Sharkey KA, Pfeffer G, Nguyen MD. Brain-body mechanisms contribute to sexual dimorphism in amyotrophic lateral sclerosis. Nat Rev Neurol 2024; 20:475-494. [PMID: 38965379 DOI: 10.1038/s41582-024-00991-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2024] [Indexed: 07/06/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common form of human motor neuron disease. It is characterized by the progressive degeneration of upper and lower motor neurons, leading to generalized motor weakness and, ultimately, respiratory paralysis and death within 3-5 years. The disease is shaped by genetics, age, sex and environmental stressors, but no cure or routine biomarkers exist for the disease. Male individuals have a higher propensity to develop ALS, and a different manifestation of the disease phenotype, than female individuals. However, the mechanisms underlying these sex differences remain a mystery. In this Review, we summarize the epidemiology of ALS, examine the sexually dimorphic presentation of the disease and highlight the genetic variants and molecular pathways that might contribute to sex differences in humans and animal models of ALS. We advance the idea that sexual dimorphism in ALS arises from the interactions between the CNS and peripheral organs, involving vascular, metabolic, endocrine, musculoskeletal and immune systems, which are strikingly different between male and female individuals. Finally, we review the response to treatments in ALS and discuss the potential to implement future personalized therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Sarah M Jacob
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sukyoung Lee
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University Hospital, Seoul, South Korea
| | - Keith A Sharkey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gerald Pfeffer
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
14
|
Grassano M, Moglia C, Palumbo F, Koumantakis E, Cugnasco P, Callegaro S, Canosa A, Manera U, Vasta R, De Mattei F, Matteoni E, Fuda G, Salamone P, Marchese G, Casale F, De Marchi F, Mazzini L, Mora G, Calvo A, Chiò A. Sex Differences in Amyotrophic Lateral Sclerosis Survival and Progression: A Multidimensional Analysis. Ann Neurol 2024; 96:159-169. [PMID: 38568048 DOI: 10.1002/ana.26933] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 06/20/2024]
Abstract
OBJECTIVE To investigate sex-related differences in amyotrophic lateral sclerosis (ALS) prognosis and their contributing factors. METHODS Our primary cohort was the Piemonte and Aosta Register for ALS (PARALS); the Pooled Resource Open-Access ALS Clinical Trials (PRO-ACT) and the Answer ALS databases were used for validation. Survival analyses were conducted accounting for age and onset site. The roles of forced vital capacity and weight decline were explored through a causal mediation analysis. Survival and disease progression rates were also evaluated after propensity score matching. RESULTS The PARALS cohort included 1,890 individuals (44.8% women). Men showed shorter survival when stratified by onset site (spinal onset HR 1.20, 95% CI 1.00-1.44, p = 0.0439; bulbar onset HR 1.36, 95% CI 1.09-1.70, p = 0.006917), although women had a steeper functional decline (+0.10 ALSFRS-R points/month, 95% CI 0.07-0.15, p < 0.00001) regardless of onset site. Instead, men showed worse respiratory decline (-4.2 forced vital capacity%/month, 95% CI -6.3 to -2.2, p < 0.0001) and faster weight loss (-0.15 kg/month, 95% CI -0.25 to -0.05, p = 0.0030). Causal mediation analysis showed that respiratory function and weight loss were pivotal in sex-related survival differences. Analysis of patients from PRO-ACT (n = 1,394, 40.9% women) and Answer ALS (n = 849, 37.2% women) confirmed these trends. INTERPRETATION The shorter survival in men is linked to worse respiratory function and weight loss rather than a faster disease progression. These findings emphasize the importance of considering sex-specific factors in understanding ALS pathophysiology and designing tailored therapeutic strategies. ANN NEUROL 2024;96:159-169.
Collapse
Affiliation(s)
- Maurizio Grassano
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Cristina Moglia
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Neurology Unit 1U, "City of Health and Science" University Hospital, Turin, Italy
| | - Francesca Palumbo
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | | | - Paolo Cugnasco
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Stefano Callegaro
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Antonio Canosa
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Neurology Unit 1U, "City of Health and Science" University Hospital, Turin, Italy
| | - Umberto Manera
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Neurology Unit 1U, "City of Health and Science" University Hospital, Turin, Italy
| | - Rosario Vasta
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Filippo De Mattei
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Enrico Matteoni
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Giuseppe Fuda
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Paolina Salamone
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Giulia Marchese
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Federico Casale
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Fabiola De Marchi
- ALS Center, Department of Neurology, Azienda Ospedaliera Universitaria Maggiore della Carità, Novara, Italy
| | - Letizia Mazzini
- ALS Center, Department of Neurology, Azienda Ospedaliera Universitaria Maggiore della Carità, Novara, Italy
| | - Gabriele Mora
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Andrea Calvo
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Neurology Unit 1U, "City of Health and Science" University Hospital, Turin, Italy
| | - Adriano Chiò
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Neurology Unit 1U, "City of Health and Science" University Hospital, Turin, Italy
- Institute of Cognitive Sciences and Technologies, National Council of Research, Rome, Italy
| |
Collapse
|
15
|
Ugursu B, Sah A, Sartori S, Popp O, Mertins P, Dunay IR, Kettenmann H, Singewald N, Wolf SA. Microglial sex differences in innate high anxiety and modulatory effects of minocycline. Brain Behav Immun 2024; 119:465-481. [PMID: 38552926 DOI: 10.1016/j.bbi.2024.03.035] [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: 09/12/2023] [Revised: 02/08/2024] [Accepted: 03/26/2024] [Indexed: 04/18/2024] Open
Abstract
Microglia modulate synaptic refinement in the central nervous system (CNS). We have previously shown that a mouse model with innate high anxiety-related behavior (HAB) displays higher CD68+ microglia density in the key regions of anxiety circuits compared to mice with normal anxiety-related behavior (NAB) in males, and that minocycline treatment attenuated the enhanced anxiety of HAB male. Given that a higher prevalence of anxiety is widely reported in females compared to males, little is known concerning sex differences at the cellular level. Herein, we address this by analyzing microglia heterogeneity and function in the HAB and NAB brains of both sexes. Single-cell RNA sequencing revealed ten distinct microglia clusters varied by their frequency and gene expression profile. We report striking sex differences, especially in the major microglia clusters of HABs, indicating a higher expression of genes associated with phagocytosis and synaptic engulfment in the female compared to the male. On a functional level, we show that female HAB microglia engulfed a greater amount of hippocampal vGLUT1+ excitatory synapses compared to the male. We moreover show that female HAB microglia engulfed more synaptosomes compared to the male HAB in vitro. Due to previously reported effects of minocycline on microglia, we finally administered oral minocycline to HABs of both sexes and showed a significant reduction in the engulfment of synapses by female HAB microglia. In parallel to our microglia-specific findings, we further showed an anxiolytic effect of minocycline on female HABs, which is complementary to our previous findings in the male HABs. Our study, therefore, identifies the altered function of synaptic engulfment by microglia as a potential avenue to target and resolve microglia heterogeneity in mice with innate high anxiety.
Collapse
Affiliation(s)
- Bilge Ugursu
- Psychoneuroimmunology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Experimental Ophthalmology, ChariteUniversitätsmedizin Berlin, Germany
| | - Anupam Sah
- Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Austria
| | - Simone Sartori
- Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Austria
| | - Oliver Popp
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute of Health, Berlin, Germany
| | - Philip Mertins
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute of Health, Berlin, Germany
| | - Ildiko R Dunay
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Germany
| | - Helmut Kettenmann
- Shenzhen Key Laboratory of Immunomodulation for Neurological Diseases, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Nicolas Singewald
- Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Austria
| | - Susanne A Wolf
- Psychoneuroimmunology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Experimental Ophthalmology, ChariteUniversitätsmedizin Berlin, Germany.
| |
Collapse
|
16
|
Cleland NRW, Potter GJ, Buck C, Quang D, Oldham D, Neal M, Saviola A, Niemeyer CS, Dobrinskikh E, Bruce KD. Altered metabolism and DAM-signatures in female brains and microglia with aging. Brain Res 2024; 1829:148772. [PMID: 38244754 DOI: 10.1016/j.brainres.2024.148772] [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: 07/13/2023] [Revised: 12/21/2023] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Despite Alzheimer's disease (AD) disproportionately affecting women, the mechanisms remain elusive. In AD, microglia undergo 'metabolic reprogramming', which contributes to microglial dysfunction and AD pathology. However, how sex and age contribute to metabolic reprogramming in microglia is understudied. Here, we use metabolic imaging, transcriptomics, and metabolic assays to probe age- and sex-associated changes in brain and microglial metabolism. Glycolytic and oxidative metabolism in the whole brain was determined using Fluorescence Lifetime Imaging Microscopy (FLIM). Young female brains appeared less glycolytic than male brains, but with aging, the female brain became 'male-like.' Transcriptomic analysis revealed increased expression of disease-associated microglia (DAM) genes (e.g., ApoE, Trem2, LPL), and genes involved in glycolysis and oxidative metabolism in microglia from aged females compared to males. To determine whether estrogen can alter the expression of these genes, BV-2 microglia-like cell lines, which abundantly express DAM genes, were supplemented with 17β-estradiol (E2). E2 supplementation resulted in reduced expression of DAM genes, reduced lipid and cholesterol transport, and substrate-dependent changes in glycolysis and oxidative metabolism. Consistent with the notion that E2 may suppress DAM-associated factors, LPL activity was elevated in the brains of aged female mice. Similarly, DAM gene and protein expression was higher in monocyte-derived microglia-like (MDMi) cells derived from middle-aged females compared to age-matched males and was responsive to E2 supplementation. FLIM analysis of MDMi from young and middle-aged females revealed reduced oxidative metabolism and FAD+ with age. Overall, our findings show that altered metabolism defines age-associated changes in female microglia and suggest that estrogen may inhibit the expression and activity of DAM-associated factors, which may contribute to increased AD risk, especially in post-menopausal women.
Collapse
Affiliation(s)
- Nicholas R W Cleland
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Garrett J Potter
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Courtney Buck
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Daphne Quang
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dean Oldham
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mikaela Neal
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anthony Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christy S Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Evgenia Dobrinskikh
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Kimberley D Bruce
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
17
|
Zhao J, Jiao Y, Wang H, Song P, Gao Z, Bing X, Zhang C, Ouyang A, Yao J, Wang S, Jiang H. Radiomic features of the hippocampal based on magnetic resonance imaging in the menopausal mouse model linked to neuronal damage and cognitive deficits. Brain Imaging Behav 2024; 18:368-377. [PMID: 38102441 PMCID: PMC11156756 DOI: 10.1007/s11682-023-00808-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] [Accepted: 10/01/2023] [Indexed: 12/17/2023]
Abstract
Estrogen deficiency in the early postmenopausal phase is associated with an increased long-term risk of cognitive decline or dementia. Non-invasive characterization of the pathological features of the pathological hallmarks in the brain associated with postmenopausal women (PMW) could enhance patient management and the development of therapeutic strategies. Radiomics is a means to quantify the radiographic phenotype of a diseased tissue via the high-throughput extraction and mining of quantitative features from images acquired from modalities such as CT and magnetic resonance imaging (MRI). This study set out to explore the correlation between radiomics features based on MRI and pathological features of the hippocampus and cognitive function in the PMW mouse model. Ovariectomized (OVX) mice were used as PWM models. MRI scans were performed two months after surgery. The brain's hippocampal region was manually annotated, and the radiomic features were extracted with PyRadiomics. Chemiluminescence was used to evaluate the peripheral blood estrogen level of mice, and the Morris water maze test was used to evaluate the cognitive ability of mice. Nissl staining and immunofluorescence were used to quantify neuronal damage and COX1 expression in brain sections of mice. The OVX mice exhibited marked cognitive decline, brain neuronal damage, and increased expression of mitochondrial complex IV subunit COX1, which are pathological phenomena commonly observed in the brains of AD patients, and these phenotypes were significantly correlated with radiomics features (p < 0.05, |r|>0.5), including Original_firstorder_Interquartile Range, Original_glcm_Difference Average, Original_glcm_Difference Average and Wavelet-LHH_glszm_Small Area Emphasis. Meanwhile, the above radiomics features were significantly different between the sham-operated and OVX groups (p < 0.01) and were associated with decreased serum estrogen levels (p < 0.05, |r|>0.5). This initial study indicates that the above radiomics features may have a role in the assessment of the pathology of brain damage caused by estrogen deficiency using routinely acquired structural MR images.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Jiao
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hui Wang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Peiji Song
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhen Gao
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xue Bing
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chunling Zhang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Aimei Ouyang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jian Yao
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Song Wang
- Department of Radiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725, South Wanping Road, Shanghai, 200032, China.
| | - Huijie Jiang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
18
|
Nordengen K, Cappelletti C, Bahrami S, Frei O, Pihlstrøm L, Henriksen SP, Geut H, Rozemuller AJM, van de Berg WDJ, Andreassen OA, Toft M. Pleiotropy with sex-specific traits reveals genetic aspects of sex differences in Parkinson's disease. Brain 2024; 147:858-870. [PMID: 37671566 PMCID: PMC10907091 DOI: 10.1093/brain/awad297] [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: 03/01/2023] [Revised: 08/01/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
Parkinson's disease is an age-related neurodegenerative disorder with a higher incidence in males than females. The causes for this sex difference are unknown. Genome-wide association studies (GWAS) have identified 90 Parkinson's disease risk loci, but the genetic studies have not found sex-specific differences in allele frequency on autosomal chromosomes or sex chromosomes. Genetic variants, however, could exert sex-specific effects on gene function and regulation of gene expression. To identify genetic loci that might have sex-specific effects, we studied pleiotropy between Parkinson's disease and sex-specific traits. Summary statistics from GWASs were acquired from large-scale consortia for Parkinson's disease (n cases = 13 708; n controls = 95 282), age at menarche (n = 368 888 females) and age at menopause (n = 69 360 females). We applied the conditional/conjunctional false discovery rate (FDR) method to identify shared loci between Parkinson's disease and these sex-specific traits. Next, we investigated sex-specific gene expression differences in the superior frontal cortex of both neuropathologically healthy individuals and Parkinson's disease patients (n cases = 61; n controls = 23). To provide biological insights to the genetic pleiotropy, we performed sex-specific expression quantitative trait locus (eQTL) analysis and sex-specific age-related differential expression analysis for genes mapped to Parkinson's disease risk loci. Through conditional/conjunctional FDR analysis we found 11 loci shared between Parkinson's disease and the sex-specific traits age at menarche and age at menopause. Gene-set and pathway analysis of the genes mapped to these loci highlighted the importance of the immune response in determining an increased disease incidence in the male population. Moreover, we highlighted a total of nine genes whose expression or age-related expression in the human brain is influenced by genetic variants in a sex-specific manner. With these analyses we demonstrated that the lack of clear sex-specific differences in allele frequencies for Parkinson's disease loci does not exclude a genetic contribution to differences in disease incidence. Moreover, further studies are needed to elucidate the role that the candidate genes identified here could have in determining a higher incidence of Parkinson's disease in the male population.
Collapse
Affiliation(s)
- Kaja Nordengen
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| | - Chiara Cappelletti
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Department of Mechanical, Electronics and Chemical Engineering, Faculty of Technology, Art and Design, OsloMet—Oslo Metropolitan University, 0130 Oslo, Norway
- Department of Research, Innovation and Education, Oslo University Hospital, 0424 Oslo, Norway
| | - Shahram Bahrami
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Oleksandr Frei
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | | | - Hanneke Geut
- Section of Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Section of Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Ole A Andreassen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
19
|
Van Steenwinckel J, Bokobza C, Laforge M, Shearer IK, Miron VE, Rua R, Matta SM, Hill‐Yardin EL, Fleiss B, Gressens P. Key roles of glial cells in the encephalopathy of prematurity. Glia 2024; 72:475-503. [PMID: 37909340 PMCID: PMC10952406 DOI: 10.1002/glia.24474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 11/03/2023]
Abstract
Across the globe, approximately one in 10 babies are born preterm, that is, before 37 weeks of a typical 40 weeks of gestation. Up to 50% of preterm born infants develop brain injury, encephalopathy of prematurity (EoP), that substantially increases their risk for developing lifelong defects in motor skills and domains of learning, memory, emotional regulation, and cognition. We are still severely limited in our abilities to prevent or predict preterm birth. No longer just the "support cells," we now clearly understand that during development glia are key for building a healthy brain. Glial dysfunction is a hallmark of EoP, notably, microgliosis, astrogliosis, and oligodendrocyte injury. Our knowledge of glial biology during development is exponentially expanding but hasn't developed sufficiently for development of effective neuroregenerative therapies. This review summarizes the current state of knowledge for the roles of glia in infants with EoP and its animal models, and a description of known glial-cell interactions in the context of EoP, such as the roles for border-associated macrophages. The field of perinatal medicine is relatively small but has worked passionately to improve our understanding of the etiology of EoP coupled with detailed mechanistic studies of pre-clinical and human cohorts. A primary finding from this review is that expanding our collaborations with computational biologists, working together to understand the complexity of glial subtypes, glial maturation, and the impacts of EoP in the short and long term will be key to the design of therapies that improve outcomes.
Collapse
Affiliation(s)
| | - Cindy Bokobza
- NeuroDiderot, INSERMUniversité Paris CitéParisFrance
| | | | - Isabelle K. Shearer
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Veronique E. Miron
- Barlo Multiple Sclerosis CentreSt. Michael's HospitalTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
- College of Medicine and Veterinary MedicineThe Dementia Research Institute at The University of EdinburghEdinburghUK
| | - Rejane Rua
- CNRS, INSERM, Centre d'Immunologie de Marseille‐Luminy (CIML), Turing Centre for Living SystemsAix‐Marseille UniversityMarseilleFrance
| | - Samantha M. Matta
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Elisa L. Hill‐Yardin
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Bobbi Fleiss
- NeuroDiderot, INSERMUniversité Paris CitéParisFrance
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | | |
Collapse
|
20
|
Hou Y, Caldwell JZK, Lathia JD, Leverenz JB, Pieper AA, Cummings J, Cheng F. Microglial immunometabolism endophenotypes contribute to sex difference in Alzheimer's disease. Alzheimers Dement 2024; 20:1334-1349. [PMID: 37985399 PMCID: PMC10916937 DOI: 10.1002/alz.13546] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/20/2023] [Accepted: 10/05/2023] [Indexed: 11/22/2023]
Abstract
INTRODUCTION The molecular mechanisms that contribute to sex differences, in particular female predominance, in Alzheimer's disease (AD) prevalence, symptomology, and pathology, are incompletely understood. METHODS To address this problem, we investigated cellular metabolism and immune responses ("immunometabolism endophenotype") across AD individuals as a function of sex with diverse clinical diagnosis of cognitive status at death (cogdx), Braak staging, and Consortium to Establish a Registry for AD (CERAD) scores using human cortex metabolomics and transcriptomics data from the Religious Orders Study / Memory and Aging Project (ROSMAP) cohort. RESULTS We identified sex-specific metabolites, immune and metabolic genes, and pathways associated with the AD diagnosis and progression. We identified female-specific elevation in glycerophosphorylcholine and N-acetylglutamate, which are AD inflammatory metabolites involved in interleukin (IL)-17 signaling, C-type lectin receptor, interferon signaling, and Toll-like receptor pathways. We pinpointed distinct microglia-specific immunometabolism endophenotypes (i.e., lipid- and amino acid-specific IL-10 and IL-17 signaling pathways) between female and male AD subjects. In addition, female AD subjects showed evidence of diminished excitatory neuron and microglia communications via glutamate-mediated immunometabolism. DISCUSSION Our results point to new understanding of the molecular basis for female predominance in AD, and warrant future independent validations with ethnically diverse patient cohorts to establish a likely causal relationship of microglial immunometabolism in the sex differences in AD. HIGHLIGHTS Sex-specific immune metabolites, gene networks and pathways, are associated with Alzheimer's disease pathogenesis and disease progression. Female AD subjects exhibit microglial immunometabolism endophenotypes characterized by decreased glutamate metabolism and elevated interleukin-10 pathway activity. Female AD subjects showed a shift in glutamate-mediated cell-cell communications between excitatory neurons to microglia and astrocyte.
Collapse
Affiliation(s)
- Yuan Hou
- Genomic Medicine InstituteCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - Jessica Z. K. Caldwell
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Lou Ruvo Center for Brain HealthCleveland ClinicNeurological InstituteLas VegasNevadaUSA
| | - Justin D. Lathia
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Department of Cardiovascular & Metabolic ScienceCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - James B. Leverenz
- Lou Ruvo Center for Brain HealthCleveland ClinicNeurological InstituteClevelandOhioUSA
| | - Andrew A. Pieper
- Brain Health Medicines CenterHarrington Discovery InstituteUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Department of PsychiatryCase Western Reserve UniversityClevelandOhioUSA
- Geriatric PsychiatryGRECCLouis Stokes Cleveland VA Medical CenterClevelandOhioUSA
- Institute for Transformative Molecular MedicineSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Department of NeurosciencesSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Department of PathologySchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Jeffrey Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of Nevada Las VegasLas VegasNevadaUSA
| | - Feixiong Cheng
- Genomic Medicine InstituteCleveland ClinicLerner Research InstituteClevelandOhioUSA
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
21
|
Biechele G, Rauchmann BS, Janowitz D, Buerger K, Franzmeier N, Weidinger E, Guersel S, Schuster S, Finze A, Harris S, Lindner S, Albert NL, Wetzel C, Rupprecht R, Rominger A, Palleis C, Katzdobler S, Burow L, Kurz C, Zaganjori M, Trappmann LK, Goldhardt O, Grimmer T, Haeckert J, Keeser D, Stoecklein S, Morenas-Rodriguez E, Bartenstein P, Levin J, Höglinger GU, Simons M, Perneczky R, Brendel M. Associations between sex, body mass index and the individual microglial response in Alzheimer's disease. J Neuroinflammation 2024; 21:30. [PMID: 38263017 PMCID: PMC10804830 DOI: 10.1186/s12974-024-03020-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVES 18-kDa translocator protein position-emission-tomography (TSPO-PET) imaging emerged for in vivo assessment of neuroinflammation in Alzheimer's disease (AD) research. Sex and obesity effects on TSPO-PET binding have been reported for cognitively normal humans (CN), but such effects have not yet been systematically evaluated in patients with AD. Thus, we aimed to investigate the impact of sex and obesity on the relationship between β-amyloid-accumulation and microglial activation in AD. METHODS 49 patients with AD (29 females, all Aβ-positive) and 15 Aβ-negative CN (8 female) underwent TSPO-PET ([18F]GE-180) and β-amyloid-PET ([18F]flutemetamol) imaging. In 24 patients with AD (14 females), tau-PET ([18F]PI-2620) was additionally available. The brain was parcellated into 218 cortical regions and standardized-uptake-value-ratios (SUVr, cerebellar reference) were calculated. Per region and tracer, the regional increase of PET SUVr (z-score) was calculated for AD against CN. The regression derived linear effect of regional Aβ-PET on TSPO-PET was used to determine the Aβ-plaque-dependent microglial response (slope) and the Aβ-plaque-independent microglial response (intercept) at the individual patient level. All read-outs were compared between sexes and tested for a moderation effect of sex on associations with body mass index (BMI). RESULTS In AD, females showed higher mean cortical TSPO-PET z-scores (0.91 ± 0.49; males 0.30 ± 0.75; p = 0.002), while Aβ-PET z-scores were similar. The Aβ-plaque-independent microglial response was stronger in females with AD (+ 0.37 ± 0.38; males with AD - 0.33 ± 0.87; p = 0.006), pronounced at the prodromal stage. On the contrary, the Aβ-plaque-dependent microglial response was not different between sexes. The Aβ-plaque-independent microglial response was significantly associated with tau-PET in females (Braak-II regions: r = 0.757, p = 0.003), but not in males. BMI and the Aβ-plaque-independent microglial response were significantly associated in females (r = 0.44, p = 0.018) but not in males (BMI*sex interaction: F(3,52) = 3.077, p = 0.005). CONCLUSION While microglia response to fibrillar Aβ is similar between sexes, women with AD show a stronger Aβ-plaque-independent microglia response. This sex difference in Aβ-independent microglial activation may be associated with tau accumulation. BMI is positively associated with the Aβ-plaque-independent microglia response in females with AD but not in males, indicating that sex and obesity need to be considered when studying neuroinflammation in AD.
Collapse
Affiliation(s)
- Gloria Biechele
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Institute of Neuroradiology, LMU University Hospital, LMU Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Gothenburg, Sweden
| | - Endy Weidinger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Selim Guersel
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Anika Finze
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Christian Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Department of Nuclear Medicine, University of Bern, Inselspital, Bern, Switzerland
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Carolin Kurz
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena-Katharina Trappmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Jan Haeckert
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sophia Stoecklein
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuronal Cell Biology, TU Munich, Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
22
|
Tewari M, Michalski S, Egan TM. Modulation of Microglial Function by ATP-Gated P2X7 Receptors: Studies in Rat, Mice and Human. Cells 2024; 13:161. [PMID: 38247852 PMCID: PMC10814008 DOI: 10.3390/cells13020161] [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: 11/08/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
P2X receptors are a family of seven ATP-gated ion channels that trigger physiological and pathophysiological responses in a variety of cells. Five of the family members are sensitive to low concentrations of extracellular ATP, while the P2X6 receptor has an unknown affinity. The last subtype, the P2X7 receptor, is unique in requiring millimolar concentrations to fully activate in humans. This low sensitivity imparts the agonist with the ability to act as a damage-associated molecular pattern that triggers the innate immune response in response to the elevated levels of extracellular ATP that accompany inflammation and tissue damage. In this review, we focus on microglia because they are the primary immune cells of the central nervous system, and they activate in response to ATP or its synthetic analog, BzATP. We start by introducing purinergic receptors and then briefly consider the roles that microglia play in neurodevelopment and disease by referencing both original works and relevant reviews. Next, we move to the role of extracellular ATP and P2X receptors in initiating and/or modulating innate immunity in the central nervous system. While most of the data that we review involve work on mice and rats, we highlight human studies of P2X7R whenever possible.
Collapse
|
23
|
Velikic G, Maric DM, Maric DL, Supic G, Puletic M, Dulic O, Vojvodic D. Harnessing the Stem Cell Niche in Regenerative Medicine: Innovative Avenue to Combat Neurodegenerative Diseases. Int J Mol Sci 2024; 25:993. [PMID: 38256066 PMCID: PMC10816024 DOI: 10.3390/ijms25020993] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Regenerative medicine harnesses the body's innate capacity for self-repair to restore malfunctioning tissues and organs. Stem cell therapies represent a key regenerative strategy, but to effectively harness their potential necessitates a nuanced understanding of the stem cell niche. This specialized microenvironment regulates critical stem cell behaviors including quiescence, activation, differentiation, and homing. Emerging research reveals that dysfunction within endogenous neural stem cell niches contributes to neurodegenerative pathologies and impedes regeneration. Strategies such as modifying signaling pathways, or epigenetic interventions to restore niche homeostasis and signaling, hold promise for revitalizing neurogenesis and neural repair in diseases like Alzheimer's and Parkinson's. Comparative studies of highly regenerative species provide evolutionary clues into niche-mediated renewal mechanisms. Leveraging endogenous bioelectric cues and crosstalk between gut, brain, and vascular niches further illuminates promising therapeutic opportunities. Emerging techniques like single-cell transcriptomics, organoids, microfluidics, artificial intelligence, in silico modeling, and transdifferentiation will continue to unravel niche complexity. By providing a comprehensive synthesis integrating diverse views on niche components, developmental transitions, and dynamics, this review unveils new layers of complexity integral to niche behavior and function, which unveil novel prospects to modulate niche function and provide revolutionary treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Gordana Velikic
- Department for Research and Development, Clinic Orto MD-Parks Dr. Dragi Hospital, 21000 Novi Sad, Serbia
- Hajim School of Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Dusan M. Maric
- Department for Research and Development, Clinic Orto MD-Parks Dr. Dragi Hospital, 21000 Novi Sad, Serbia
- Faculty of Stomatology Pancevo, University Business Academy, 26000 Pancevo, Serbia;
| | - Dusica L. Maric
- Department of Anatomy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Gordana Supic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Miljan Puletic
- Faculty of Stomatology Pancevo, University Business Academy, 26000 Pancevo, Serbia;
| | - Oliver Dulic
- Department of Surgery, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia;
| | - Danilo Vojvodic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| |
Collapse
|
24
|
Reed EG, Keller-Norrell PR. Minding the Gap: Exploring Neuroinflammatory and Microglial Sex Differences in Alzheimer's Disease. Int J Mol Sci 2023; 24:17377. [PMID: 38139206 PMCID: PMC10743742 DOI: 10.3390/ijms242417377] [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: 11/20/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Research into Alzheimer's Disease (AD) describes a link between AD and the resident immune cells of the brain, the microglia. Further, this suspected link is thought to have underlying sex effects, although the mechanisms of these effects are only just beginning to be understood. Many of these insights are the result of policies put in place by funding agencies such as the National Institutes of Health (NIH) to consider sex as a biological variable (SABV) and the move towards precision medicine due to continued lackluster therapeutic options. The purpose of this review is to provide an updated assessment of the current research that summarizes sex differences and the research pertaining to microglia and their varied responses in AD.
Collapse
Affiliation(s)
- Erin G. Reed
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44242, USA
| | | |
Collapse
|
25
|
Daniels MJD, Lefevre L, Szymkowiak S, Drake A, McCulloch L, Tzioras M, Barrington J, Dando OR, He X, Mohammad M, Sasaguri H, Saito T, Saido TC, Spires-Jones TL, McColl BW. Cystatin F ( Cst7) drives sex-dependent changes in microglia in an amyloid-driven model of Alzheimer's disease. eLife 2023; 12:e85279. [PMID: 38085657 PMCID: PMC10715728 DOI: 10.7554/elife.85279] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 11/04/2023] [Indexed: 12/18/2023] Open
Abstract
Microglial endolysosomal (dys)function is strongly implicated in neurodegenerative disease. Transcriptomic studies show that a microglial state characterised by a set of genes involved in endolysosomal function is induced in both mouse Alzheimer's disease (AD) models and human AD brain, and that the emergence of this state is emphasised in females. Cst7 (encoding cystatin F) is among the most highly upregulated genes in these microglia. However, despite such striking and robust upregulation, the function of Cst7 in neurodegenerative disease is not understood. Here, we crossed Cst7-/- mice with the AppNL-G-F mouse to test the role of Cst7 in a model of amyloid-driven AD. Surprisingly, we found that Cst7 plays a sexually dimorphic role regulating microglia in this model. In females, Cst7-/-AppNL-G-F microglia had greater endolysosomal gene expression, lysosomal burden, and amyloid beta (Aβ) burden in vivo and were more phagocytic in vitro. However, in males, Cst7-/-AppNL-G-F microglia were less inflammatory and had a reduction in lysosomal burden but had no change in Aβ burden. Overall, our study reveals functional roles for one of the most commonly upregulated genes in microglia across disease models, and the sex-specific profiles of Cst7-/--altered microglial disease phenotypes. More broadly, the findings raise important implications for AD including crucial questions on sexual dimorphism in neurodegenerative disease and the interplay between endolysosomal and inflammatory pathways in AD pathology.
Collapse
Affiliation(s)
- Michael JD Daniels
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| | - Lucas Lefevre
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| | - Stefan Szymkowiak
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| | - Alice Drake
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| | - Laura McCulloch
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of EdinburghEdinburghUnited Kingdom
| | - Makis Tzioras
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| | - Jack Barrington
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| | - Owen R Dando
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain, University of EdinburghEdinburghUnited Kingdom
| | - Xin He
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain, University of EdinburghEdinburghUnited Kingdom
| | - Mehreen Mohammad
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| | - Hiroki Sasaguri
- Department of Neurology and Neurological Science, Graduate School of Medicine, Tokyo Medical and Dental UniversityTokyoJapan
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science InstituteWakoJapan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science InstituteWakoJapan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya UniversityNagoyaJapan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science InstituteWakoJapan
| | - Tara L Spires-Jones
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| | - Barry W McColl
- UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
26
|
Ortholand J, Pradat PF, Tezenas du Montcel S, Durrleman S. Interaction of sex and onset site on the disease trajectory of amyotrophic lateral sclerosis. J Neurol 2023; 270:5903-5912. [PMID: 37615751 DOI: 10.1007/s00415-023-11932-7] [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: 06/27/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Studies showed the impact of sex and onset site (spinal or bulbar) on disease onset and survival in ALS. However, they mainly result from cross-sectional or survival analysis, and the interaction of sex and onset site on the different proxies of disease trajectory has not been fully investigated. METHODS We selected all patients with repeated observations in the PRO-ACT database. We divided them into four groups depending on their sex and onset site. We estimated a multivariate disease progression model, named ALS Course Map, to investigate the combined temporal changes of the four sub-scores of the revised ALS functional rating scale (ALSFRSr), the forced vital capacity (FVC), and the body mass index (BMI). We then compared the progression rate, the estimated age at onset, and the relative progression of the outcomes across each group. RESULTS We included 1438 patients from the PRO-ACT database. They were 51% men with spinal onset, 12% men with bulbar onset, 26% women with spinal onset, and 11% women with bulbar onset. We showed a significant influence of both sex and onset site on the ALSFRSr progression. The BMI decreased 8.9 months earlier (95% CI [3.9, 13.8]) in women than men, after correction for the onset site. Among patients with bulbar onset, FVC was impaired 2.6 months earlier (95% CI [0.6, 4.6]) in women. CONCLUSION Using a multivariable disease modelling approach, we showed that sex and onset site are important drivers of the progression of motor function, BMI, and FVC decline.
Collapse
Affiliation(s)
- Juliette Ortholand
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, ICM, CNRS, InriaInserm, AP-HP, Hôpital de La Pitié Salpêtrière, 75013, Paris, France.
| | - Pierre-François Pradat
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, Paris, France
- APHP, Département de Neurologie, Hôpital Pitié-Salpêtrière, Centre Référent SLA, Paris, France
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute Ulster University, C-TRIC, Altnagelvin Hospital, Derry, Londonderry, UK
| | - Sophie Tezenas du Montcel
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, ICM, CNRS, InriaInserm, AP-HP, Hôpital de La Pitié Salpêtrière, 75013, Paris, France
| | - Stanley Durrleman
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, ICM, CNRS, InriaInserm, AP-HP, Hôpital de La Pitié Salpêtrière, 75013, Paris, France
| |
Collapse
|
27
|
Cleland NRW, Potter GJ, Buck C, Quang D, Oldham D, Neal M, Saviola A, Niemeyer CS, Dobrinskikh E, Bruce KD. Altered Metabolism and DAM-signatures in Female Brains and Microglia with Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.28.569104. [PMID: 38076915 PMCID: PMC10705419 DOI: 10.1101/2023.11.28.569104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Despite Alzheimer's disease (AD) disproportionately affecting women, the mechanisms remain elusive. In AD, microglia undergo 'metabolic reprogramming', which contributes to microglial dysfunction and AD pathology. However, how sex and age contribute to metabolic reprogramming in microglia is understudied. Here, we use metabolic imaging, transcriptomics, and metabolic assays to probe age-and sex-associated changes in brain and microglial metabolism. Glycolytic and oxidative metabolism in the whole brain was determined using Fluorescence Lifetime Imaging Microscopy (FLIM). Young female brains appeared less glycolytic than male brains, but with aging, the female brain became 'male-like.' Transcriptomic analysis revealed increased expression of disease-associated microglia (DAM) genes (e.g., ApoE, Trem2, LPL), and genes involved in glycolysis and oxidative metabolism in microglia from aged females compared to males. To determine whether estrogen can alter the expression of these genes, BV-2 microglia-like cell lines, which abundantly express DAM genes, were supplemented with 17β-estradiol (E2). E2 supplementation resulted in reduced expression of DAM genes, reduced lipid and cholesterol transport, and substrate-dependent changes in glycolysis and oxidative metabolism. Consistent with the notion that E2 may suppress DAM-associated factors, LPL activity was elevated in the brains of aged female mice. Similarly, DAM gene and protein expression was higher in monocyte-derived microglia-like (MDMi) cells derived from middle-aged females compared to age-matched males and was responsive to E2 supplementation. FLIM analysis of MDMi from young and middle-aged females revealed reduced oxidative metabolism and FAD+ with age. Overall, our findings show that altered metabolism defines age-associated changes in female microglia and suggest that estrogen may inhibit the expression and activity of DAM-associated factors, which may contribute to increased AD risk, especially in post-menopausal women.
Collapse
Affiliation(s)
- Nicholas R W Cleland
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Garrett J Potter
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Courtney Buck
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Daphne Quang
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Dean Oldham
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Mikaela Neal
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Anthony Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Christy S. Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Evgenia Dobrinskikh
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Kimberley D. Bruce
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| |
Collapse
|
28
|
Bobotis BC, Braniff O, Gargus M, Akinluyi ET, Awogbindin IO, Tremblay MÈ. Sex differences of microglia in the healthy brain from embryonic development to adulthood and across lifestyle influences. Brain Res Bull 2023; 202:110752. [PMID: 37652267 DOI: 10.1016/j.brainresbull.2023.110752] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Microglia, the central nervous system innate immune cells, play a critical role in maintaining a homeostatic environment in the brain throughout life. These cells exhibit an impressive range of functions and characteristics that help to ensure proper functioning of the brain. Notably, microglia can present differences in their genetic and physical traits, which can be influenced by a range of factors, including age, environmental exposures, disease, and sex. Remarkably, microglia have been found to express receptors for sex hormones, suggesting that these hormones may play a role in modulating microglial behavior and potentially contribute to sex differences. Additionally, sex-chromosomal factors were shown to impact microglial genetics and functioning. In this review, we will examine how microglial responses in homeostasis are impacted by their interaction with sex hormones and sex chromosomes. Specifically, our investigation will focus on examining this interaction from embryonic development to adulthood, and the influence of lifestyle elements on various microglial features, including density and distribution, morphology, transcriptome, and proteome.
Collapse
Affiliation(s)
| | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Makenna Gargus
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Elizabeth Toyin Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Pharmacology and Therapeutics, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Ifeoluwa Oluleke Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neurosciences Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada; Department of Molecular Medicine, Université Laval, Québec, QC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
| |
Collapse
|
29
|
Pan K, Garaschuk O. The role of intracellular calcium-store-mediated calcium signals in in vivo sensor and effector functions of microglia. J Physiol 2023; 601:4203-4215. [PMID: 35315518 DOI: 10.1113/jp279521] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/09/2022] [Indexed: 11/08/2022] Open
Abstract
Under physiological conditions microglia, the immune sentinels of the brain, constantly monitor their microenvironment. In the case of danger, damage or cell/tissue dyshomeostasis, they react with changes in process motility, polarization, directed process movement, morphology and gene expression profile; release pro- and anti-inflammatory mediators; proliferate; and clean brain parenchyma by means of phagocytosis. Based on recent transcriptomic and in vivo Ca2+ imaging data, we argue that the local cell/tissue dyshomeostasis is sensed by microglia via intracellular Ca2+ signals, many of which are mediated by Ca2+ release from the intracellular Ca2+ stores. These signals encode the strength, duration and spatiotemporal pattern of the stimulus and, at the same time, relay this information further to trigger the respective Ca2+ -dependent effector pathways. We also point to the fact that microglial Ca2+ signalling is sexually dimorphic and undergoes profound changes across the organism's lifespan. Interestingly, the first changes in microglial Ca2+ signalling are visible already in 9- to 11-month-old mice, roughly corresponding to 40-year-old humans.
Collapse
Affiliation(s)
- Kuang Pan
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
30
|
Thaker AA, McConnell BV, Rogers DM, Carlson NE, Coughlan C, Jensen AM, Lopez-Paniagua D, Holden SK, Pressman PS, Pelak VS, Filley CM, Potter H, Solano DA, Heffernan KS, Bettcher BM. Astrogliosis, neuritic microstructure, and sex effects: GFAP is an indicator of neuritic orientation in women. Brain Behav Immun 2023; 113:124-135. [PMID: 37394144 PMCID: PMC10584366 DOI: 10.1016/j.bbi.2023.06.026] [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: 12/08/2022] [Revised: 06/14/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND Data from human studies suggest that immune dysregulation is associated with Alzheimer's disease (AD) pathology and cognitive decline and that neurites may be affected early in the disease trajectory. Data from animal studies further indicate that dysfunction in astrocytes and inflammation may have a pivotal role in facilitating dendritic damage, which has been linked with negative cognitive outcomes. To elucidate these relationships further, we have examined the relationship between astrocyte and immune dysregulation, AD-related pathology, and neuritic microstructure in AD-vulnerable regions in late life. METHODS We evaluated panels of immune, vascular, and AD-related protein markers in blood and conducted in vivo multi-shell neuroimaging using Neurite Orientation Dispersion and Density Imaging (NODDI) to assess indices of neuritic density (NDI) and dispersion (ODI) in brain regions vulnerable to AD in a cohort of older adults (n = 109). RESULTS When examining all markers in tandem, higher plasma GFAP levels were strongly related to lower neurite dispersion (ODI) in grey matter. No biomarker associations were found with higher neuritic density. Associations between GFAP and neuritic microstructure were not significantly impacted by symptom status, APOE status, or plasma Aβ42/40 ratio; however, there was a large sex effect observed for neurite dispersion, wherein negative associations between GFAP and ODI were only observed in females. DISCUSSION This study provides a comprehensive, concurrent appraisal of immune, vascular, and AD-related biomarkers in the context of advanced grey matter neurite orientation and dispersion methodology. Sex may be an important modifier of the complex associations between astrogliosis, immune dysregulation, and brain microstructure in older adults.
Collapse
Affiliation(s)
- Ashesh A Thaker
- Department of Radiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brice V McConnell
- Department of Neurology, Behavioral Neurology Section, University of Colorado Alzheimer's & Cognition Center, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Dustin M Rogers
- Department of Biostatistics and Informatics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Nichole E Carlson
- Department of Biostatistics and Informatics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Christina Coughlan
- Department of Neurology, University of Colorado Alzheimer's & Cognition Center, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexandria M Jensen
- Quantitative Sciences Unit, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Dan Lopez-Paniagua
- Department of Radiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Samantha K Holden
- Department of Neurology, Behavioral Neurology Section, University of Colorado Alzheimer's & Cognition Center, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Peter S Pressman
- Department of Neurology, Behavioral Neurology Section, University of Colorado Alzheimer's & Cognition Center, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Victoria S Pelak
- Department of Neurology, Behavioral Neurology Section, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA; Department of Ophthalmology, Sue Anschutz-Rodgers University of Colorado Eye Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Christopher M Filley
- Behavioral Neurology Section, Departments of Neurology and Psychiatry, University of Colorado Alzheimer's & Cognition Center, Marcus Institute for Brain Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Huntington Potter
- Department of Neurology, University of Colorado Alzheimer's & Cognition Center, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - D Adriana Solano
- Department of Neurology, University of Colorado Alzheimer's & Cognition Center, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kate S Heffernan
- Division of Neuropharmacology and Neurological Disorders, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Brianne M Bettcher
- Department of Neurology, Behavioral Neurology Section, University of Colorado Alzheimer's & Cognition Center, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
31
|
Bhatia TN, Jamenis AS, Abbas M, Clark RN, Miner KM, Chandwani MN, Kim RE, Hilinski W, O'Donnell LA, Luk KC, Shi Y, Hu X, Chen J, Brodsky JL, Leak RK. A 14-day pulse of PLX5622 modifies α-synucleinopathy in preformed fibril-infused aged mice of both sexes. Neurobiol Dis 2023; 184:106196. [PMID: 37315905 PMCID: PMC10528721 DOI: 10.1016/j.nbd.2023.106196] [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: 12/01/2022] [Revised: 05/16/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023] Open
Abstract
Reactive microglia are observed with aging and in Lewy body disorders, including within the olfactory bulb of men with Parkinson's disease. However, the functional impact of microglia in these disorders is still debated. Resetting these reactive cells by a brief dietary pulse of the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 may hold therapeutic potential against Lewy-related pathologies. To our knowledge, withdrawal of PLX5622 after short-term exposure has not been tested in the preformed α-synuclein fibril (PFF) model, including in aged mice of both sexes. Compared to aged female mice, we report that aged males on the control diet showed higher numbers of phosphorylated α-synuclein+ inclusions in the limbic rhinencephalon after PFFs were injected in the posterior olfactory bulb. However, aged females displayed larger inclusion sizes compared to males. Short-term (14-day) dietary exposure to PLX5622 followed by control chow reduced inclusion numbers and levels of insoluble α-synuclein in aged males-but not females-and unexpectedly raised inclusion sizes in both sexes. Transient delivery of PLX5622 also improved spatial reference memory in PFF-infused aged mice, as evidenced by an increase in novel arm entries in a Y-maze. Superior memory was positively correlated with inclusion sizes but negatively correlated with inclusion numbers. Although we caution that PLX5622 delivery must be tested further in models of α-synucleinopathy, our data suggest that larger-sized-but fewer-α-synucleinopathic structures are associated with better neurological outcomes in PFF-infused aged mice.
Collapse
Affiliation(s)
- Tarun N Bhatia
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Anuj S Jamenis
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Muslim Abbas
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Rachel N Clark
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Kristin M Miner
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Manisha N Chandwani
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Roxanne E Kim
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | | | - Lauren A O'Donnell
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Kelvin C Luk
- Dept. of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yejie Shi
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jun Chen
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Dept. of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA.
| |
Collapse
|
32
|
Gancz NN, Levinson JA, Callaghan BL. Sex and gender as critical and distinct contributors to the human brain-gut-microbiome axis. Brain Res Bull 2023; 199:110665. [PMID: 37192716 PMCID: PMC11149430 DOI: 10.1016/j.brainresbull.2023.110665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/25/2023] [Accepted: 05/13/2023] [Indexed: 05/18/2023]
Abstract
The brain-gut-microbiome axis (BGMA) is a pivotal contributor to human health. A large body of research, especially from animal models, has revealed bidirectional, causal relationships between the BGMA and sex. In particular, sex steroids appear to be affected by the BGMA, to influence the BGMA, and to moderate environmental effects on the BGMA. However, animal research on the relationship between sex and the BGMA has not translated well to human models. We contend that this is due in part to an oversimplified approach to sex: although BGMA researchers have traditionally approached sex as a unidimensional, dichotomous variable, it is in fact multidimensional and is comprised of both multi-categorical and continuous dimensions. We also contend that research on the BGMA in humans should approach gender as a variable that is distinct from sex and that gender may influence the BGMA through pathways that are independent from the effects of sex alone. Research practices that consider the complexity and distinctiveness of sex and gender in relation to the human BGMA will not only yield improved understanding of this consequential system, but will also enhance the development of treatments for adverse health outcomes with BGMA-related etiologies. We conclude with recommendations for the implementation of such practices.
Collapse
Affiliation(s)
- N N Gancz
- University of California, Los Angeles, Department of Psychology, 502 Portola Plaza, Los Angeles, CA 90095, USA.
| | - J A Levinson
- University of California, Los Angeles, Department of Psychology, 502 Portola Plaza, Los Angeles, CA 90095, USA
| | - B L Callaghan
- University of California, Los Angeles, Department of Psychology, 502 Portola Plaza, Los Angeles, CA 90095, USA
| |
Collapse
|
33
|
Yeung CHC, Au Yeung SL, Kwok MK, Zhao JV, Schooling CM. The influence of growth and sex hormones on risk of alzheimer's disease: a mendelian randomization study. Eur J Epidemiol 2023:10.1007/s10654-023-01015-2. [PMID: 37253999 DOI: 10.1007/s10654-023-01015-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/04/2023] [Indexed: 06/01/2023]
Abstract
Alzheimer's disease is more prevalent in women, possibly due to sex or growth hormones but existing evidence is inconclusive. We investigated whether genetically predicted sex and growth hormones are associated with risk of Alzheimer's disease. Genetic variants strongly and independently predicting insulin-like growth factor 1 (IGF-1), testosterone and sex hormone-binding globulin (SHBG) were obtained from large, published genome wide associations studies (GWAS) and applied to GWAS of Alzheimer's disease based on clinical diagnosis (cases = 21,982, control = 41,944) from the International Genomics of Alzheimer's Project and the UK Biobank parental (maternal cases = 27,696; paternal cases = 14,338) and siblings' diagnosis (cases = 2,171) as proxy cases. Published GWAS summary statistics were used in our analyses. Estimates were obtained from inverse variance weighting with sensitivity analysis (i.e., MR-Egger, weighted median and MR-PRESSO). Multivariable analyses adjusted for pleiotropic effects and possible sources of selection bias were also performed. Genetically predicted higher total testosterone may reduce the risk of paternal Alzheimer's disease (odds ratio (OR) 0.86, 95% confidence interval (CI) 0.76 to 0.97, per SD increase in testosterone) and in meta-analysis for women (OR 0.92, 95% CI 0.87, 0.98) with directionally similar results from other analyses. SHBG were not associated with Alzheimer's disease. IGF-1 in women was inversely associated with risk of clinical Alzheimer's disease in sensitivity analysis but not in the main analysis. These results suggest genetically predicted higher total testosterone may lower risk of Alzheimer's disease. The role of testosterone and the immune system in Alzheimer's disease could be further investigated.
Collapse
Affiliation(s)
- Chris Ho Ching Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Shiu Lun Au Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Man Ki Kwok
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jie V Zhao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - C Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Graduate School of Public Health and Health Policy, City University of New York, New York, USA
| |
Collapse
|
34
|
Terrin F, Tesoriere A, Plotegher N, Dalla Valle L. Sex and Brain: The Role of Sex Chromosomes and Hormones in Brain Development and Parkinson's Disease. Cells 2023; 12:1486. [PMID: 37296608 PMCID: PMC10252697 DOI: 10.3390/cells12111486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Sex hormones and genes on the sex chromosomes are not only key factors in the regulation of sexual differentiation and reproduction but they are also deeply involved in brain homeostasis. Their action is crucial for the development of the brain, which presents different characteristics depending on the sex of individuals. The role of these players in the brain is fundamental in the maintenance of brain function during adulthood as well, thus being important also with respect to age-related neurodegenerative diseases. In this review, we explore the role of biological sex in the development of the brain and analyze its impact on the predisposition toward and the progression of neurodegenerative diseases. In particular, we focus on Parkinson's disease, a neurodegenerative disorder that has a higher incidence in the male population. We report how sex hormones and genes encoded by the sex chromosomes could protect from the disease or alternatively predispose toward its development. We finally underline the importance of considering sex when studying brain physiology and pathology in cellular and animal models in order to better understand disease etiology and develop novel tailored therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Nicoletta Plotegher
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| | - Luisa Dalla Valle
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| |
Collapse
|
35
|
Seeker LA, Bestard-Cuche N, Jäkel S, Kazakou NL, Bøstrand SMK, Wagstaff LJ, Cholewa-Waclaw J, Kilpatrick AM, Van Bruggen D, Kabbe M, Baldivia Pohl F, Moslehi Z, Henderson NC, Vallejos CA, La Manno G, Castelo-Branco G, Williams A. Brain matters: unveiling the distinct contributions of region, age, and sex to glia diversity and CNS function. Acta Neuropathol Commun 2023; 11:84. [PMID: 37217978 PMCID: PMC10204264 DOI: 10.1186/s40478-023-01568-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 04/15/2023] [Indexed: 05/24/2023] Open
Abstract
The myelinated white matter tracts of the central nervous system (CNS) are essential for fast transmission of electrical impulses and are often differentially affected in human neurodegenerative diseases across CNS region, age and sex. We hypothesize that this selective vulnerability is underpinned by physiological variation in white matter glia. Using single nucleus RNA sequencing of human post-mortem white matter samples from the brain, cerebellum and spinal cord and subsequent tissue-based validation we found substantial glial heterogeneity with tissue region: we identified region-specific oligodendrocyte precursor cells (OPCs) that retain developmental origin markers into adulthood, distinguishing them from mouse OPCs. Region-specific OPCs give rise to similar oligodendrocyte populations, however spinal cord oligodendrocytes exhibit markers such as SKAP2 which are associated with increased myelin production and we found a spinal cord selective population particularly equipped for producing long and thick myelin sheaths based on the expression of genes/proteins such as HCN2. Spinal cord microglia exhibit a more activated phenotype compared to brain microglia, suggesting that the spinal cord is a more pro-inflammatory environment, a difference that intensifies with age. Astrocyte gene expression correlates strongly with CNS region, however, astrocytes do not show a more activated state with region or age. Across all glia, sex differences are subtle but the consistent increased expression of protein-folding genes in male donors hints at pathways that may contribute to sex differences in disease susceptibility. These findings are essential to consider for understanding selective CNS pathologies and developing tailored therapeutic strategies.
Collapse
Affiliation(s)
- Luise A Seeker
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Nadine Bestard-Cuche
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sarah Jäkel
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
- Institute for Stroke and Dementia Research, Klinikum Der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nina-Lydia Kazakou
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sunniva M K Bøstrand
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Laura J Wagstaff
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Justyna Cholewa-Waclaw
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - David Van Bruggen
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Mukund Kabbe
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Fabio Baldivia Pohl
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Zahra Moslehi
- Laboratory of Neurodevelopmental Systems Biology, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Catalina A Vallejos
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh, EH4 2XU, UK
- The Alan Turing Institute, 96 Euston Road, London, NW1 2DB, UK
| | - Gioele La Manno
- Laboratory of Neurodevelopmental Systems Biology, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Goncalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Stockholm Node, 171 77, Stockholm, Sweden
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
36
|
Bourque M, Morissette M, Soulet D, Di Paolo T. Impact of Sex on Neuroimmune contributions to Parkinson's disease. Brain Res Bull 2023:110668. [PMID: 37196734 DOI: 10.1016/j.brainresbull.2023.110668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/27/2023] [Accepted: 05/13/2023] [Indexed: 05/19/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease. Inflammation has been observed in both the idiopathic and familial forms of PD. Importantly, PD is reported more often in men than in women, men having at least 1.5- fold higher risk to develop PD than women. This review summarizes the impact of biological sex and sex hormones on the neuroimmune contributions to PD and its investigation in animal models of PD. Innate and peripheral immune systems participate in the brain neuroinflammation of PD patients and is reproduced in neurotoxin, genetic and alpha-synuclein based models of PD. Microglia and astrocytes are the main cells of the innate immune system in the central nervous system and are the first to react to restore homeostasis in the brain. Analysis of serum immunoprofiles in female and male control and PD patients show that a great proportion of these markers differ between male and female. The relationship between CSF inflammatory markers and PD clinical characteristics or PD biomarkers shows sex differences. Conversely, in animal models of PD, sex differences in inflammation are well documented and the beneficial effects of endogenous and exogenous estrogenic modulation in inflammation have been reported. Targeting neuroinflammation in PD is an emerging therapeutic option but gonadal drugs have not yet been investigated in this respect, thus offering new opportunities for sex specific treatments.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada.
| | - Marc Morissette
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada.
| | - Denis Soulet
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada; Faculté de Pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec (Québec) G1V 0A6, Canada.
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada; Faculté de Pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec (Québec) G1V 0A6, Canada.
| |
Collapse
|
37
|
Prengel TM, Brunne B, Habiballa M, Rune GM. Sexually differentiated microglia and CA1 hippocampal synaptic connectivity. J Neuroendocrinol 2023; 35:e13276. [PMID: 37170708 DOI: 10.1111/jne.13276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/14/2023] [Accepted: 03/31/2023] [Indexed: 05/13/2023]
Abstract
Microglia have been shown to sculpt postnatal circuitry from birth up to adulthood due to their role in both synapse formation, synaptic pruning, and the elimination of weak, redundant synapses. Microglia are differentiated in a sex-dependent manner. In this study, we tested whether sexual differentiation of microglia results in sex-dependent postnatal reorganization of CA1 synaptic connectivity in the hippocampus. The stereological counting of synapses in mice using electron microscopy showed a continuous rise in synapse density until the fourth week, followed by a plateau phase and loss of synapses from the eighth week onwards, with no difference between sexes. This course of alteration in synapse numbers did not differ between sexes. However, selectively, on postnatal day (P) 14 the density of synapses was significantly higher in the female than in the male hippocampus. Higher synapse density in females was paralleled by higher activity of microglia, as indicated by morphological changes, CD68 expression, and proximity of microglia to synaptic sites. In Thy1-GFP mice, consistent with increased synapse numbers, bouton density was also clearly increased in females at P14. At this time point, CD47 expression, the "don't eat me" signal of neurons, was similar in males and females. The decrease in bouton density thereafter in conjunction with increased synapse numbers argues for a role of microglia in the formation of multispine boutons (MSB). Our data in females at P14 support the regulatory role of microglia in synapse density. Sexual differentiation of microglia, however, does not substantially affect long-term synaptic reorganization in the hippocampus.
Collapse
Affiliation(s)
- Tim M Prengel
- Institute of Neuroanatomy, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Bianka Brunne
- Institute of Neuroanatomy, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Moataz Habiballa
- Institute of Neuroanatomy, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Gabriele M Rune
- Institute of Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
38
|
Ocañas SR, Ansere VA, Kellogg CM, Isola JVV, Chucair-Elliott AJ, Freeman WM. Chromosomal and gonadal factors regulate microglial sex effects in the aging brain. Brain Res Bull 2023; 195:157-171. [PMID: 36804773 PMCID: PMC10810555 DOI: 10.1016/j.brainresbull.2023.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
Biological sex contributes to phenotypic sex effects through genetic (sex chromosomal) and hormonal (gonadal) mechanisms. There are profound sex differences in the prevalence and progression of age-related brain diseases, including neurodegenerative diseases. Inflammation of neural tissue is one of the most consistent age-related phenotypes seen with healthy aging and disease. The pro-inflammatory environment of the aging brain has primarily been attributed to microglial reactivity and adoption of heterogeneous reactive states dependent upon intrinsic (i.e., sex) and extrinsic (i.e., age, disease state) factors. Here, we review sex effects in microglia across the lifespan, explore potential genetic and hormonal molecular mechanisms of microglial sex effects, and discuss currently available models and methods to study sex effects in the aging brain. Despite recent attention to this area, significant further research is needed to mechanistically understand the regulation of microglial sex effects across the lifespan, which may open new avenues for sex informed prevention and treatment strategies.
Collapse
Affiliation(s)
- Sarah R Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Victor A Ansere
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Collyn M Kellogg
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jose V V Isola
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ana J Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
39
|
Bianco A, Antonacci Y, Liguori M. Sex and Gender Differences in Neurodegenerative Diseases: Challenges for Therapeutic Opportunities. Int J Mol Sci 2023; 24:6354. [PMID: 37047320 PMCID: PMC10093984 DOI: 10.3390/ijms24076354] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
The term "neurodegenerative diseases" (NDs) identifies a group of heterogeneous diseases characterized by progressive loss of selectively vulnerable populations of neurons, which progressively deteriorates over time, leading to neuronal dysfunction. Protein aggregation and neuronal loss have been considered the most characteristic hallmarks of NDs, but growing evidence confirms that significant dysregulation of innate immune pathways plays a crucial role as well. NDs vary from multiple sclerosis, in which the autoimmune inflammatory component is predominant, to more "classical" NDs, such as Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, and spinal muscular atrophy. Of interest, many of the clinical differences reported in NDs seem to be closely linked to sex, which may be justified by the significant changes in immune mechanisms between affected females and males. In this review, we examined some of the most studied NDs by looking at their pathogenic and phenotypical features to highlight sex-related discrepancies, if any, with particular interest in the individuals' responses to treatment. We believe that pointing out these differences in clinical practice may help achieve more successful precision and personalized care.
Collapse
Affiliation(s)
| | | | - Maria Liguori
- National Research Council (CNR), Institute of Biomedical Technologies, Bari Unit, 70125 Bari, Italy
| |
Collapse
|
40
|
Novakova Martinkova J, Ferretti MT, Ferrari A, Lerch O, Matuskova V, Secnik J, Hort J. Longitudinal progression of choroid plexus enlargement is associated with female sex, cognitive decline and ApoE E4 homozygote status. Front Psychiatry 2023; 14:1039239. [PMID: 36970283 PMCID: PMC10031049 DOI: 10.3389/fpsyt.2023.1039239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/27/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction Choroid plexus (CP)-related mechanisms have been implicated in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease. In this pilot study, we aimed to elucidate the association between longitudinal changes in CP volume, sex and cognitive impairment. Methods We assessed longitudinal changes in CP volume in a cohort of n = 613 subjects across n = 2,334 datapoints from ADNI 2 and ADNI-GO, belonging to cognitively unimpaired (CN), stable mild cognitive impairment (MCI), clinically diagnosed Alzheimer's disease dementia (AD) or convertor (to either AD or MCI) subgroups. CP volume was automatically segmented and used as a response variable in linear mixed effect models with random intercept clustered by patient identity. Temporal effects of select variables were assessed by interactions and subgroup analyses. Results We found an overall significant increase of CP volume in time (14.92 mm3 per year, 95% confidence interval, CI (11.05, 18.77), p < 0.001). Sex-disaggregated results showed an annual rate of increase 9.48 mm3 in males [95% CI (4.08, 14.87), p < 0.001], and 20.43 mm3 in females [95% CI (14.91, 25.93), p < 0.001], indicating more than double the rate of increase in females, which appeared independent of other temporal variables. The only diagnostic group with a significant CP increase as compared to CN was the convertors group, with an increase of 24.88 mm3/year [95% CI (14, 35.82), p < 0.001]. ApoE exhibited a significant temporal effect, with the E4 homozygote group's CP increasing at more than triple the rate of non-carrier or heterozygote groups [40.72, 95% CI (25.97, 55.46), p < 0.001 vs. 12.52, 95% CI (8.02, 17.02), p < 0.001 for ApoE E4 homozygotes and E4 non-carriers, respectively], and may have modified the diagnostic group relationship. Conclusion Our results contribute to potential mechanisms for sex differences in cognitive impairment with a novel finding of twice the annual choroid plexus enlargement in females and provide putative support for CP-related mechanisms of cognitive deterioration and its relationship to ApoE E4.
Collapse
Affiliation(s)
- Julie Novakova Martinkova
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | | | | | - Ondrej Lerch
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Veronika Matuskova
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Juraj Secnik
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
- Center for Alzheimer Research, Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Jakub Hort
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | | |
Collapse
|
41
|
Isenbrandt A, Coulombe K, Morissette M, Bourque M, Lamontagne-Proulx J, Di Paolo T, Soulet D. Three-Dimensional Analysis of Sex- and Gonadal Status- Dependent Microglial Activation in a Mouse Model of Parkinson's Disease. Pharmaceuticals (Basel) 2023; 16:152. [PMID: 37259303 PMCID: PMC9967417 DOI: 10.3390/ph16020152] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/03/2025] Open
Abstract
Parkinson's disease (PD) is characterized by neurodegeneration and neuroinflammation. PD prevalence and incidence are higher in men than in women and modulation of gonadal hormones could have an impact on the disease course. This was investigated in male and female gonadectomized (GDX) and SHAM operated (SHAM) mice. Dutasteride (DUT), a 5α-reductase inhibitor, was administered to these mice for 10 days to modulate their gonadal sex hormones. On the fifth day of DUT treatment, mice received 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to model PD. We have previously shown in these mice the toxic effect of MPTP in SHAM and GDX males and in GDX females on dopamine markers and astrogliosis whereas SHAM females were protected by their female sex hormones. In SHAM males, DUT protected against MPTP toxicity. In the present study, microglial density and the number of doublets, representative of a microglial proliferation, were increased by the MPTP lesion only in male mice and prevented by DUT in SHAM males. A three-dimensional morphological microglial analysis showed that MPTP changed microglial morphology from quiescent to activated only in male mice and was not prevented by DUT. In conclusion, microgliosis can be modulated by sex hormone-dependent and independent factors in a mice model of PD.
Collapse
Affiliation(s)
- Amandine Isenbrandt
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
- Faculté de Pharmacie, Pavillon Ferdinand-Vandry, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Katherine Coulombe
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Marc Morissette
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Mélanie Bourque
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Jérôme Lamontagne-Proulx
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
- Faculté de Pharmacie, Pavillon Ferdinand-Vandry, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Denis Soulet
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
- Faculté de Pharmacie, Pavillon Ferdinand-Vandry, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6, Canada
| |
Collapse
|
42
|
CSF1R inhibitors induce a sex-specific resilient microglial phenotype and functional rescue in a tauopathy mouse model. Nat Commun 2023; 14:118. [PMID: 36624100 PMCID: PMC9829908 DOI: 10.1038/s41467-022-35753-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 12/23/2022] [Indexed: 01/10/2023] Open
Abstract
Microglia are central to pathogenesis in many neurological conditions. Drugs targeting colony-stimulating factor-1 receptor (CSF1R) to block microglial proliferation in preclinical disease models have shown mixed outcomes, thus the therapeutic potential of this approach remains unclear. Here, we show that CSF1R inhibitors given by multiple dosing paradigms in the Tg2541 tauopathy mouse model cause a sex-independent reduction in pathogenic tau and reversion of non-microglial gene expression patterns toward a normal wild type signature. Despite greater drug exposure in male mice, only female mice have functional rescue and extended survival. A dose-dependent upregulation of immediate early genes and neurotransmitter dysregulation are observed in the brains of male mice only, indicating that excitotoxicity may preclude functional benefits. Drug-resilient microglia in male mice exhibit morphological and gene expression patterns consistent with increased neuroinflammatory signaling, suggesting a mechanistic basis for sex-specific excitotoxicity. Complete microglial ablation is neither required nor desirable for neuroprotection and therapeutics targeting microglia must consider sex-dependent effects.
Collapse
|
43
|
Zhuo B, Zheng D, Cai M, Wang C, Zhang S, Zhang Z, Tian F, Wang X, Lin H. Mediation Effect of Brain Volume on the Relationship Between Peripheral Inflammation and Cognitive Decline. J Alzheimers Dis 2023; 95:523-533. [PMID: 37545239 DOI: 10.3233/jad-230253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
BACKGROUND Studies have reported the associations between inflammation, brain volume, and cognition separately. It is reasonable to assume peripheral inflammation may contribute to cognitive decline through brain volume atrophy. OBJECTIVE To examine the associations between peripheral inflammation, brain volume, and cognition among adults, and to investigate whether brain volume atrophy mediates the inflammation-cognition relationshipMethods:We retrieved 20,381 participants with available data on peripheral inflammation, brain volume, and cognition from the UK Biobank cohort. Cognitive function was assessed by performance on cognitive tasks probing various cognitive domains. Brain volumes were measured by magnetic resonance imaging (MRI). Multivariable linear models were used to investigate the associations between three peripheral inflammatory indexes (C-reactive protein, systemic immune-inflammatory index, neutrophil-to-lymphocyte ratio), brain volume, and cognition. Mediation analyses were conducted to assess the potential mediating effect of brain volume atrophy. All results were corrected for multiple comparisons using the false-discovery rate (FDR). RESULTS Peripheral inflammation was inversely associated with grey matter volume (GMV), white matter volume (WMV), and cognition after adjusting for potential covariates. For instance, CRP was associated with the GMV of left parahippocampal gyrus (β= -0.05, 95% confidence interval [CI]: -0.06 to -0.04, pFDR =1.07×10-16) and general cognitive factor (β= -0.03, 95% CI: -0. -0.04 to -0.01, pFDR = 0.001). Brain volume atrophy mediated the inflammation-cognitive decline relationship, accounting for 15-29% of the overall impact. CONCLUSION In this cohort study, peripheral inflammation was associated with brain volume atrophy and cognitive decline. Brain atrophy may mediate the inflammation-cognitive decline relationship.
Collapse
Affiliation(s)
- Bingting Zhuo
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dashan Zheng
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Miao Cai
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chongjian Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Henan, China
| | - Shiyu Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zilong Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fei Tian
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaojie Wang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
44
|
Nona RJ, Xu Z, Robinson GA, Henderson RD, McCombe PA. Age of Onset and Length of Survival of Queensland Patients with Amyotrophic Lateral Sclerosis: Details of Subjects with Early Onset and Subjects with Long Survival. NEURODEGENER DIS 2022; 22:104-121. [PMID: 36587610 PMCID: PMC10627495 DOI: 10.1159/000528875] [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: 06/16/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION The aims of the study were to document the characteristics of amyotrophic lateral sclerosis (ALS) patients in Queensland, to examine factors influencing age of onset, and survival, and to study those with early-onset (<45 years) disease and those with long (>5 years) survival. METHODS We studied subjects seen at the ALS Clinic at the Royal Brisbane and Women's Hospital. We recorded sex, age of onset, region of onset, length of survival, presence of family history, type of disease, and evidence of cognitive involvement. We analysed the influence of these features on age of onset and survival. We analysed the features of patients with early onset of disease and patients with long survival. RESULTS There were 855 ALS patients (505 males) in the cohort. The age of onset was lower in males than females, in patients with a family history of ALS compared to those without, and in patients with spinal onset compared to bulbar onset. Early-onset disease was seen in 10% of patients, and had a greater proportion of males, spinal onset, and classical ALS phenotype compared to late-onset disease. Survival was shorter in females, in patients with bulbar onset, and in patients with classical ALS. Long survival was seen in 18% of patients. Patients with long survival had younger age of onset, greater proportion of males, spinal onset, and fewer patients with classical ALS. CONCLUSION Our study confirms that ALS is more prevalent in males and that spinal onset is more common than bulbar onset. Males have earlier onset but longer survival. We found that overall, patients with classical ALS have worse survival than ALS variants, but some patients who were considered to have classical ALS had long survival. This study confirms the similarity of ALS in our region to ALS in other geographical regions.
Collapse
Affiliation(s)
- Robert J. Nona
- Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
| | - Zhouwei Xu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Gail A. Robinson
- Queensland Brain Institute and School of Psychology, University of Queensland, St Lucia, Queensland, Australia
| | - Robert D. Henderson
- Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Pamela A. McCombe
- Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| |
Collapse
|
45
|
Milner TA, Chen RX, Welington D, Rubin BR, Contoreggi NH, Johnson MA, Mazid S, Marques-Lopes J, Marongiu R, Glass MJ. Angiotensin II differentially affects hippocampal glial inflammatory markers in young adult male and female mice. Learn Mem 2022; 29:265-273. [PMID: 36206386 PMCID: PMC9488028 DOI: 10.1101/lm.053507.121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/03/2022] [Indexed: 12/16/2022]
Abstract
Hypertension is a risk factor for neurodegenerative disorders involving inflammation and inflammatory cytokine-producing brain cells (microglia and astrocytes) in the hippocampus and medial prefrontal cortex (mPFC). Here we investigated the effect of slow-pressor angiotensin II (AngII) on gliosis in the hippocampus and mPFC of young adult (2-mo-old) male and female mice. In males, AngII induced hypertension, and this resulted in an increase in the density of the astrocyte marker glial fibrillary acidic protein (GFAP) in the subgranular hilus and a decrease in the density of the microglial marker ionized calcium binding adapter molecule (Iba-1) in the CA1 region. Females infused with AngII did not show hypertension but, significantly, showed alterations in hippocampal glial activation. Compared with vehicle, AngII-infused female mice had an increased density of Iba-1 in the dentate gyrus and CA2/3a region. Like males, females infused with AngII exhibited decreased Iba-1 in the CA1 region. Neither male nor female mice showed differences in GFAP or Iba-1 in the mPFC following AngII infusion. These results demonstrate that the hippocampus is particularly vulnerable to AngII in young adulthood. Differences in gonadal hormones or the sensitivity to AngII hypertension may account for divergences in GFAP and Iba-1 in males and females.
Collapse
Affiliation(s)
- Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Harold and Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York 10065, USA
| | - Ryan X Chen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - Diedreanna Welington
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - Batsheva R Rubin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - Megan A Johnson
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - Sanoara Mazid
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - Roberta Marongiu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Neurological Surgery Department, Weill Cornell Medicine, New York, New York 10065, USA
| | - Michael J Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| |
Collapse
|
46
|
Otaru S, Lawrence DA. Autism: genetics, environmental stressors, maternal immune activation, and the male bias in autism. EXPLORATION OF NEUROPROTECTIVE THERAPY 2022. [DOI: 10.37349/ent.2022.00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/20/2022] [Indexed: 01/05/2025]
Abstract
Autism spectrum disorder (ASD) is a class of neurodevelopmental disorders (NDD) characterized by deficits in three domains: impairments in social interactions, language, and communication, and increased stereotyped restrictive/repetitive behaviors and interests. The exact etiology of ASD remains unknown. Genetics, gestational exposure to inflammation, and environmental stressors, which combine to affect mitochondrial dysfunction and metabolism, are implicated yet poorly understood contributors and incompletely delineated pathways toward the relative risk of ASD. Many studies have shown a clear male bias in the incidence of ASD and other NDD. In other words, being male is a significant yet poorly understood risk factor for the development of NDD. This review discusses the link between these factors by looking at the current body of evidence. Understanding the link between the multiplicity of hits—from genes to environmental stressors and possible sexual determinants, contributing to autism susceptibility is critical to developing targeted interventions to mitigate these risks.
Collapse
Affiliation(s)
- Sarah Otaru
- Department of Environmental Health Sciences, University at Albany School of Public Health, Rensselaer, New York 12144, USA
| | - David A. Lawrence
- Department of Environmental Health Sciences, University at Albany School of Public Health, Rensselaer, New York 12144, USA;Clinical and Experimental Immunology, Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| |
Collapse
|
47
|
Miller SJ, Campbell CE, Jimenez-Corea HA, Wu GH, Logan R. Neuroglial Senescence, α-Synucleinopathy, and the Therapeutic Potential of Senolytics in Parkinson’s Disease. Front Neurosci 2022; 16:824191. [PMID: 35516803 PMCID: PMC9063319 DOI: 10.3389/fnins.2022.824191] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/22/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson’s disease (PD) is the most common movement disorder and the second most prevalent neurodegenerative disease after Alzheimer’s disease. Despite decades of research, there is still no cure for PD and the complicated intricacies of the pathology are still being worked out. Much of the research on PD has focused on neurons, since the disease is characterized by neurodegeneration. However, neuroglia has become recognized as key players in the health and disease of the central nervous system. This review provides a current perspective on the interactive roles that α-synuclein and neuroglial senescence have in PD. The self-amplifying and cyclical nature of oxidative stress, neuroinflammation, α-synucleinopathy, neuroglial senescence, neuroglial chronic activation and neurodegeneration will be discussed. Finally, the compelling role that senolytics could play as a therapeutic avenue for PD is explored and encouraged.
Collapse
Affiliation(s)
- Sean J. Miller
- Pluripotent Diagnostics Corp. (PDx), Molecular Medicine Research Institute, Sunnyvale, CA, United States
| | | | | | - Guan-Hui Wu
- Department of Neurology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Robert Logan
- Pluripotent Diagnostics Corp. (PDx), Molecular Medicine Research Institute, Sunnyvale, CA, United States
- Department of Biology, Eastern Nazarene College, Quincy, MA, United States
- *Correspondence: Robert Logan,
| |
Collapse
|
48
|
Milanova IV, Korpel NL, Correa-da-Silva F, Berends E, Osman S, la Fleur SE, Fliers E, Kalsbeek A, Yi CX. Loss of Microglial Insulin Receptor Leads to Sex-Dependent Metabolic Disorders in Obese Mice. Int J Mol Sci 2022; 23:ijms23062933. [PMID: 35328354 PMCID: PMC8954452 DOI: 10.3390/ijms23062933] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/27/2022] [Accepted: 03/05/2022] [Indexed: 02/08/2023] Open
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are highly prevalent disorders, associated with insulin resistance and chronic inflammation. The brain is key for energy homeostasis and contains many insulin receptors. Microglia, the resident brain immune cells, are known to express insulin receptors (InsR) and to be activated by a hypercaloric environment. The aim of this study was to evaluate whether microglial insulin signaling is involved in the control of systemic energy homeostasis and whether this function is sex-dependent. We generated a microglia-specific knockout of the InsR gene in male and female mice and exposed them to control or obesogenic dietary conditions. Following 10 weeks of diet exposure, we evaluated insulin tolerance, energy metabolism, microglial morphology and phagocytic function, and neuronal populations. Lack of microglial InsR resulted in increased plasma insulin levels and insulin resistance in obese female mice. In the brain, loss of microglial InsR led to a decrease in microglial primary projections in both male and female mice, irrespective of the diet. In addition, in obese male mice lacking microglial InsR the number of proopiomelanocortin neurons was decreased, compared to control diet, while no differences were observed in female mice. Our results demonstrate a sex-dependent effect of microglial InsR-signaling in physiology and obesity, and stress the importance of a heterogeneous approach in the study of diseases such as obesity and T2DM.
Collapse
Affiliation(s)
- Irina V. Milanova
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.V.M.); (N.L.K.); (F.C.-d.-S.); (E.B.); (S.O.); (S.E.l.F.); (E.F.); (A.K.)
- Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), 1105 AZ Amsterdam, The Netherlands
| | - Nikita L. Korpel
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.V.M.); (N.L.K.); (F.C.-d.-S.); (E.B.); (S.O.); (S.E.l.F.); (E.F.); (A.K.)
- Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), 1105 AZ Amsterdam, The Netherlands
| | - Felipe Correa-da-Silva
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.V.M.); (N.L.K.); (F.C.-d.-S.); (E.B.); (S.O.); (S.E.l.F.); (E.F.); (A.K.)
- Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), 1105 AZ Amsterdam, The Netherlands
| | - Eline Berends
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.V.M.); (N.L.K.); (F.C.-d.-S.); (E.B.); (S.O.); (S.E.l.F.); (E.F.); (A.K.)
- Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), 1105 AZ Amsterdam, The Netherlands
| | - Samar Osman
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.V.M.); (N.L.K.); (F.C.-d.-S.); (E.B.); (S.O.); (S.E.l.F.); (E.F.); (A.K.)
- Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), 1105 AZ Amsterdam, The Netherlands
| | - Susanne E. la Fleur
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.V.M.); (N.L.K.); (F.C.-d.-S.); (E.B.); (S.O.); (S.E.l.F.); (E.F.); (A.K.)
- Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), 1105 AZ Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.V.M.); (N.L.K.); (F.C.-d.-S.); (E.B.); (S.O.); (S.E.l.F.); (E.F.); (A.K.)
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.V.M.); (N.L.K.); (F.C.-d.-S.); (E.B.); (S.O.); (S.E.l.F.); (E.F.); (A.K.)
- Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), 1105 AZ Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.V.M.); (N.L.K.); (F.C.-d.-S.); (E.B.); (S.O.); (S.E.l.F.); (E.F.); (A.K.)
- Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), 1105 AZ Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
49
|
Payne A, Nahashon S, Taka E, Adinew GM, Soliman KFA. Epigallocatechin-3-Gallate (EGCG): New Therapeutic Perspectives for Neuroprotection, Aging, and Neuroinflammation for the Modern Age. Biomolecules 2022; 12:biom12030371. [PMID: 35327563 PMCID: PMC8945730 DOI: 10.3390/biom12030371] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/28/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s and Parkinson’s diseases are the two most common forms of neurodegenerative diseases. The exact etiology of these disorders is not well known; however, environmental, molecular, and genetic influences play a major role in the pathogenesis of these diseases. Using Alzheimer’s disease (AD) as the archetype, the pathological findings include the aggregation of Amyloid Beta (Aβ) peptides, mitochondrial dysfunction, synaptic degradation caused by inflammation, elevated reactive oxygen species (ROS), and cerebrovascular dysregulation. This review highlights the neuroinflammatory and neuroprotective role of epigallocatechin-3-gallate (EGCG): the medicinal component of green tea, a known nutraceutical that has shown promise in modulating AD progression due to its antioxidant, anti-inflammatory, and anti-aging abilities. This report also re-examines the current literature and provides innovative approaches for EGCG to be used as a preventive measure to alleviate AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Ashley Payne
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (A.P.); (E.T.); (G.M.A.)
| | - Samuel Nahashon
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN 37209, USA;
| | - Equar Taka
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (A.P.); (E.T.); (G.M.A.)
| | - Getinet M. Adinew
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (A.P.); (E.T.); (G.M.A.)
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (A.P.); (E.T.); (G.M.A.)
- Correspondence: ; Tel.: +1850-322-8788
| |
Collapse
|
50
|
Austad SN, Ballinger S, Buford TW, Carter CS, Smith DL, Darley-Usmar V, Zhang J. Targeting whole body metabolism and mitochondrial bioenergetics in the drug development for Alzheimer's disease. Acta Pharm Sin B 2022; 12:511-531. [PMID: 35256932 PMCID: PMC8897048 DOI: 10.1016/j.apsb.2021.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Aging is by far the most prominent risk factor for Alzheimer's disease (AD), and both aging and AD are associated with apparent metabolic alterations. As developing effective therapeutic interventions to treat AD is clearly in urgent need, the impact of modulating whole-body and intracellular metabolism in preclinical models and in human patients, on disease pathogenesis, have been explored. There is also an increasing awareness of differential risk and potential targeting strategies related to biological sex, microbiome, and circadian regulation. As a major part of intracellular metabolism, mitochondrial bioenergetics, mitochondrial quality-control mechanisms, and mitochondria-linked inflammatory responses have been considered for AD therapeutic interventions. This review summarizes and highlights these efforts.
Collapse
Key Words
- ACE2, angiotensin I converting enzyme (peptidyl-dipeptidase A) 2
- AD, Alzheimer's disease
- ADP, adenosine diphosphate
- ADRD, AD-related dementias
- Aβ, amyloid β
- CSF, cerebrospinal fluid
- Circadian regulation
- DAMPs
- DAMPs, damage-associated molecular patterns
- Diabetes
- ER, estrogen receptor
- ETC, electron transport chain
- FCCP, trifluoromethoxy carbonylcyanide phenylhydrazone
- FPR-1, formyl peptide receptor 1
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide-1
- HBP, hexoamine biosynthesis pathway
- HTRA, high temperature requirement A
- Hexokinase biosynthesis pathway
- I3A, indole-3-carboxaldehyde
- IRF-3, interferon regulatory factor 3
- LC3, microtubule associated protein light chain 3
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- MAVS, mitochondrial anti-viral signaling
- MCI, mild cognitive impairment
- MRI, magnetic resonance imaging
- MRS, magnetic resonance spectroscopy
- Mdivi-1, mitochondrial division inhibitor 1
- Microbiome
- Mitochondrial DNA
- Mitochondrial electron transport chain
- Mitochondrial quality control
- NLRP3, leucine-rich repeat (LRR)-containing protein (NLR)-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- NeuN, neuronal nuclear protein
- PET, fluorodeoxyglucose (FDG)-positron emission tomography
- PKA, protein kinase A
- POLβ, the base-excision repair enzyme DNA polymerase β
- ROS, reactive oxygen species
- Reactive species
- SAMP8, senescence-accelerated mice
- SCFAs, short-chain fatty acids
- SIRT3, NAD-dependent deacetylase sirtuin-3
- STING, stimulator of interferon genes
- STZ, streptozotocin
- SkQ1, plastoquinonyldecyltriphenylphosphonium
- T2D, type 2 diabetes
- TCA, Tricarboxylic acid
- TLR9, toll-like receptor 9
- TMAO, trimethylamine N-oxide
- TP, tricyclic pyrone
- TRF, time-restricted feeding
- cAMP, cyclic adenosine monophosphate
- cGAS, cyclic GMP/AMP synthase
- hAPP, human amyloid precursor protein
- hPREP, human presequence protease
- i.p., intraperitoneal
- mTOR, mechanistic target of rapamycin
- mtDNA, mitochondrial DNA
- αkG, alpha-ketoglutarate
Collapse
Affiliation(s)
- Steven N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christy S. Carter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|