1
|
O'Mahony C, Hidalgo-Lanussa O, Barreto GE. Unveiling FOXO3's metabolic contribution to menopause and Alzheimer's disease. Exp Gerontol 2025; 200:112679. [PMID: 39778695 DOI: 10.1016/j.exger.2025.112679] [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: 07/22/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
The increasing prevalence of Alzheimer's disease (AD) calls for a comprehensive exploration of its complex etiology, with a focus on sex-specific vulnerability, particularly the heightened susceptibility observed in postmenopausal women. Neurometabolic alterations during the endocrine transition emerge as early indicators of AD pathology, including reduced glucose metabolism and increased amyloid-beta (Aβ) deposition. The fluctuating endocrine environment, marked by declining estradiol levels and reduced estrogen receptor beta (ERβ) activity, further exacerbates this process. In this context, here we explore the potential of forkhead box O3 (FOXO3) as a critical mediator linking metabolic disturbances to hormonal decline. We propose that FOXO3 plays a key role in the intersection of menopause and AD, given its dysregulation in both AD patients and postmenopausal women, modulating cellular metabolism through interactions with the AMPK/AKT/PI3K pathways. This relationship highlights the intersection between hormonal changes and increased AD susceptibility. This review aims to open a discussion on FOXO3's contribution to the metabolic dysregulation seen in menopause and its impact on the progression of AD. Understanding the functional role of FOXO3 in menopause-associated metabolic changes could lead to targeted therapeutic strategies, offering novel insights for managing for this condition.
Collapse
Affiliation(s)
- Christopher O'Mahony
- Department of Biological Sciences, University of Limerick, Limerick V94 T9PX, Ireland
| | - Oscar Hidalgo-Lanussa
- Department of Biological Sciences, University of Limerick, Limerick V94 T9PX, Ireland
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick V94 T9PX, Ireland.
| |
Collapse
|
2
|
Sasmita AO, Ong EC, Nazarenko T, Mao S, Komarek L, Thalmann M, Hantakova V, Spieth L, Berghoff SA, Barr HJ, Hingerl M, Börensen F, Hirrlinger J, Simons M, Stevens B, Depp C, Nave KA. Parental origin of transgene modulates amyloid-β plaque burden in the 5xFAD mouse model of Alzheimer's disease. Neuron 2025:S0896-6273(24)00925-5. [PMID: 39837326 DOI: 10.1016/j.neuron.2024.12.025] [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: 07/09/2024] [Revised: 11/18/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025]
Abstract
In Alzheimer's disease (AD) research, the 5xFAD mouse model is commonly used as a heterozygote crossed with other genetic models to study AD pathology. We investigated whether the parental origin of the 5xFAD transgene affects plaque deposition. Using quantitative light-sheet microscopy, we found that paternal inheritance of the transgene led to a 2-fold higher plaque burden compared with maternal inheritance, a finding consistent across multiple 5xFAD colonies. This effect was not due to gestation in or rearing by 5xFAD females. Immunoblotting suggested that transgenic inheritance modulates transgenic protein expression, potentially due to genomic imprinting of the Thy1.2 promoter. Surprisingly, fewer than 20% of 5xFAD studies report breeding schemes, suggesting that this factor might confound previous findings. Our data highlight a significant determinant of plaque burden in 5xFAD mice and underscore the importance of reporting the parental origin of the transgene to improve scientific rigor and reproducibility in AD research.
Collapse
Affiliation(s)
- Andrew Octavian Sasmita
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; International Max Planck Research School for Neurosciences, Göttingen, Germany.
| | - Erinne Cherisse Ong
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; International Max Planck Research School for Neurosciences, Göttingen, Germany
| | - Taisiia Nazarenko
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; International Max Planck Research School for Neurosciences, Göttingen, Germany; Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Shuying Mao
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lina Komarek
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Maik Thalmann
- Department of English Philology, Georg-August University, Göttingen, Germany
| | - Veronika Hantakova
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lena Spieth
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Stefan A Berghoff
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Helena J Barr
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maximilian Hingerl
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Friederike Börensen
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Johannes Hirrlinger
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Constanze Depp
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
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
|
Jauregi-Zinkunegi A, Gleason CE, Bendlin B, Okonkwo O, Hermann BP, Blennow K, Zetterberg H, Hogervorst E, Johnson SC, Langhough R, Mueller KD, Bruno D. Menopausal hormone therapy is associated with worse levels of Alzheimer's disease biomarkers in APOE ε4-carrying women: An observational study. Alzheimers Dement 2025. [PMID: 39783876 DOI: 10.1002/alz.14456] [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: 09/19/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/12/2025]
Abstract
INTRODUCTION Menopausal hormone therapy (MHT), along with the apolipoprotein E (APOE) ε4 allele, has been suggested as a possible risk factor for Alzheimer's disease (AD). However, the relationship between MHT and cerebrospinal fluid (CSF) biomarkers is unknown: we investigated this association, and whether APOE ε4 carrier status moderates it. METHODS In an observational study of 136 cognitively unimpaired female participants (Mage = 66.0; standard deviation = 6.3), we examined whether MHT use alone or in interaction with APOE ε4 carrier status was associated with CSF levels of phosphorylated tau (p-tau), amyloid beta (Aβ)40, Aβ42, p-tau/Aβ42, and Aβ42/40 ratios. RESULTS Significant interactions were found between APOE ε4 and MHT use for CSF biomarkers. APOE ε4 carriers who were MHT users showed worse levels of CSF p-tau/Aβ42 and Aβ42/40 ratios than all other users and non-users. DISCUSSION The presence of both APOE ε4 and MHT may be associated with elevated amyloid deposition and AD pathology in this sample of participants who demonstrated high familial AD risk. HIGHLIGHTS Significant interactions were found between apolipoprotein E (APOE) ε4 and menopausal hormone therapy (MHT) use for cerebrospinal fluid (CSF) phosphorylated tau (p-tau)/amyloid beta (Aβ)42 and Aβ42/40 ratios. APOE ε4 carriers who were MHT users showed worse levels of CSF biomarkers than non-users and non-carriers, both users and non-users. Younger age at MHT initiation was associated with worse levels of the p-tau/Aβ42 and Aβ42/40 ratios in carriers only. The presence of both APOE ε4 carriage and MHT use may be associated with elevated amyloid deposition and AD pathology. Further studies with larger sample sizes are necessary to confirm the differences observed in the current study.
Collapse
Affiliation(s)
| | - Carey E Gleason
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Barbara Bendlin
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ozioma Okonkwo
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Bruce P Hermann
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kaj Blennow
- 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, Göteborg, Sweden
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- 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, Göteborg, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, UCL, London, UK
- UK Dementia Research Institute, UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Science Park, Hong Kong, China
| | - Eef Hogervorst
- School of Sports Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Rebecca Langhough
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kimberly D Mueller
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Davide Bruno
- School of Psychology, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
5
|
Wright RS, Allan AC, Gamaldo AA, Morgan AA, Lee AK, Erus G, Davatzikos C, Bygrave DC. Neighborhood disadvantage is associated with working memory and hippocampal volumes among older adults. NEUROPSYCHOLOGY, DEVELOPMENT, AND COGNITION. SECTION B, AGING, NEUROPSYCHOLOGY AND COGNITION 2025; 32:55-68. [PMID: 38656243 PMCID: PMC11499292 DOI: 10.1080/13825585.2024.2345926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
It is not well understood how neighborhood disadvantage is associated with specific domains of cognitive function and underlying brain health within older adults. Thus, the objective was to examine associations between neighborhood disadvantage, brain health, and cognitive performance, and examine whether associations were more pronounced among women. The study included 136 older adults who underwent cognitive testing and MRI. Neighborhood disadvantage was characterized using the Area Deprivation Index (ADI). Descriptive statistics, bivariate correlations, and multiple regressions were run. Multiple regressions, adjusted for age, sex, education, and depression, showed that higher ADI state rankings (greater disadvantage) were associated with poorer working memory performance (p < .01) and lower hippocampal volumes (p < .01), but not total, frontal, and white matter lesion volumes, nor visual and verbal memory performance. There were no significant sex interactions. Findings suggest that greater neighborhood disadvantage may play a role in working memory and underlying brain structure.
Collapse
Affiliation(s)
| | - Alexa C Allan
- Department of Human Development and Family Studies, The Pennsylvania State University, State College, PA, USA
| | | | | | - Anna K Lee
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Desirée C Bygrave
- Department of Psychology, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| |
Collapse
|
6
|
Suganya S, Ashok BS, Ajith TA. A Recent Update on the Role of Estrogen and Progesterone in Alzheimer's Disease. Cell Biochem Funct 2024; 42:e70025. [PMID: 39663597 DOI: 10.1002/cbf.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/23/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
Alzheimer's disease (AD), one of the most prevalent neurodegenerative disease responsible for 60%-80% dementia cases globally. The disease is more prevalent among elder females. Female reproductive hormones are found to be essential for cellular activities in brain. The physiological role of neurotrophins and sex hormones in hippocampal region during neurogenesis and neuron differentiation was studied as well. In addition to triggering cellular pathways, estrogen and progesterone carry out a number of biological processes that lead to neuroprotection. They might have an impact on learning and memory. One of estrogen's modest antioxidant properties is its direct scavenging of free radicals. The neurotrophic effect of estrogen and progesterone can be explained by their ability to rise the expression of the brain-derived neurotrophic factor (BDNF) mRNA. Additionally, they have the ability to degrade beta-amyloid and stop inflammation, apoptotic neuronal cell death, and tau protein phosphorylation. To enhance their neuroprotective action, various cross-talking pathways in cells that are mediated by estrogen, progesterone, and BDNF receptors. This include signaling by mitogen-activated protein kinase/extracellular regulated kinase, phosphatidylinositol 3-kinase/protein kinase B, and phospholipase/protein kinase C. Clinical research to establish the significance of these substances are fragmented, despite publications claiming a lower prevalence of AD when medication is started before menopause. This review article emphasizes an update on the role of estrogen, and progesterone in AD.
Collapse
Affiliation(s)
- S Suganya
- Department of Biochemistry, Sri Ramachandra Medical College and Research Institute, Chennai, Tamil Nadu, India
| | - Ben Sundra Ashok
- Department of Biochemistry, Sri Ramachandra Medical College and Research Institute, Chennai, Tamil Nadu, India
| | - Thekkuttuparambil Ananthanarayanan Ajith
- Department of Biochemistry, Amala Institute of Medical Sciences, Thrissur, Kerala, India
- Amala Integrated Medical Research Department, Amala Institute of Medical Sciences, Thrissur, Kerala, India
| |
Collapse
|
7
|
Bale LK, West SA, Gades NM, Baker DJ, Conover CA. Gene deletion of Pregnancy-associated Plasma Protein-A (PAPP-A) improves pathology and cognition in an Alzheimer's disease mouse model. Exp Neurol 2024; 382:114976. [PMID: 39349117 PMCID: PMC11502239 DOI: 10.1016/j.expneurol.2024.114976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/13/2024] [Accepted: 09/22/2024] [Indexed: 10/02/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease of age with no effective preventative or treatment approaches. Deeper understanding of the mechanisms underlying the accumulation of toxic β-amyloid oligopeptides and the formation of amyloid plaque in AD has the potential to identify new therapeutic targets. Prior research links the insulin-like growth factor (IGF) system to pathologic mechanisms underlying AD. Suppression of local IGF-I receptor (IGFIR) signaling in AD mice has been shown to reduce plaque formation in the brain and delay neurodegeneration and behavioral changes. However, direct inhibitors of IGFIR signaling are not a viable treatment option for AD due to the essentiality of the IGFIR in physiological growth and metabolism. We have previously demonstrated a more selective means to reduce local IGFIR signaling through inhibition of PAPP-A, a novel zinc metalloprotease that regulates local IGF-I bioavailability through cleavage of inhibitory IGF binding proteins. Here we tested if deletion of PAPP-A in a mouse model of AD provides protection against pathology and behavioral changes. We show that compared to AD mice, AD/PAPP-A KO mice had significantly less plaque burden, reduced astrocytic activation, decreased IGF-IR activity, and improved cognition. Human senile AD plaques showed specific immunostaining for PAPP-A. Thus, inhibition of PAPP-A expression or activity may represent a novel treatment strategy for AD.
Collapse
Affiliation(s)
- Laurie K Bale
- Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Sally A West
- Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Naomi M Gades
- Department of Comparative Medicine, Mayo Clinic, Scottsdale, AZ 85259, United States of America.
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, United States of America; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Cheryl A Conover
- Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, United States of America.
| |
Collapse
|
8
|
Kim SM, Sultana F, Korkmaz F, Rojekar S, Pallapati A, Ryu V, Lizneva D, Yuen T, Rosen CJ, Zaidi M. Neuroendocrinology of bone. Pituitary 2024; 27:761-777. [PMID: 39096452 DOI: 10.1007/s11102-024-01437-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/05/2024]
Abstract
The past decade has witnessed significant advances in our understanding of skeletal homeostasis and the mechanisms that mediate the loss of bone in primary and secondary osteoporosis. Recent breakthroughs have primarily emerged from identifying disease-causing mutations and phenocopying human bone disease in rodents. Notably, using genetically-modified rodent models, disrupting the reciprocal relationship with tropic pituitary hormone and effector hormones, we have learned that pituitary hormones have independent roles in skeletal physiology, beyond their effects exerted through target endocrine glands. The rise of follicle-stimulating hormone (FSH) in the late perimenopause may account, at least in part, for the rapid bone loss when estrogen is normal, while low thyroid-stimulating hormone (TSH) levels may contribute to the bone loss in thyrotoxicosis. Admittedly speculative, suppressed levels of adrenocorticotropic hormone (ACTH) may directly exacerbate bone loss in the setting of glucocorticoid-induced osteoporosis. Furthermore, beyond their established roles in reproduction and lactation, oxytocin and prolactin may affect intergenerational calcium transfer and therefore fetal skeletal mineralization, whereas elevated vasopressin levels in chronic hyponatremic states may increase the risk of bone loss.. Here, we discuss the interaction of each pituitary hormone in relation to its role in bone physiology and pathophysiology.
Collapse
Affiliation(s)
- Se-Min Kim
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Farhath Sultana
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Funda Korkmaz
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Satish Rojekar
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anusha Pallapati
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vitaly Ryu
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Daria Lizneva
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tony Yuen
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Mone Zaidi
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
9
|
Korkmaz F, Sims S, Sen F, Sultana F, Laurencin V, Cullen L, Pallapati A, Liu A, Chen R, Rojekar S, Pevnev G, Cheliadinova U, Vasilyeva D, Burganova G, Macdonald A, Saxena M, Goosens K, Rosen CJ, Barak O, Lizneva D, Gumerova A, Ye K, Ryu V, Yuen T, Frolinger T, Zaidi M. Gene-dose-dependent reduction of Fshr expression improves spatial memory deficits in Alzheimer's mice. Mol Psychiatry 2024:10.1038/s41380-024-02824-x. [PMID: 39548323 DOI: 10.1038/s41380-024-02824-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
High post-menopausal levels of the pituitary gonadotropin follicle-stimulating hormone (FSH) are strongly associated with the onset of Alzheimer's disease (AD). We have shown recently that FSH directly activates the hippocampal FSH receptors (FSHRs) to drive AD-like pathology and memory loss in mice. To unequivocally establish a role for FSH in memory loss, we depleted the Fshr on a 3xTg background and utilized Morris Water Maze to study deficits in spatial memory. 3xTg;Fshr+/+ mice displayed impaired spatial memory at 5 months of age. The loss of memory acquisition and retrieval were both rescued in 3xTg;Fshr-/- mice and, to a lesser extent, in 3xTg;Fshr+/- mice-documenting clear gene-dose-dependent prevention of spatial memory loss. Furthermore, at 5 and 8 months, sham-operated 3xTg;Fshr-/- mice showed better memory performance during the learning and/or retrieval phases, further suggesting that Fshr deletion prevents age-related progression of memory deficits. This prevention was not seen when mice were ovariectomized, except in the 8-month-old 3xTg;Fshr-/- mice. There was also a gene-dose-dependent reduction mainly in the amyloid β40 isoform in whole brain extracts. Finally, serum FSH levels <8 ng/mL in 16-month-old APP/PS1 mice were associated with better retrieval of spatial memory. Collectively, the data provide compelling genetic evidence for a protective effect of inhibiting FSH signaling on the progression of spatial memory deficits in mice and lay a firm foundation for the use of an FSH-blocking agent for the early prevention of memory loss in post-menopausal women.
Collapse
Affiliation(s)
- Funda Korkmaz
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven Sims
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fazilet Sen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Farhath Sultana
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Victoria Laurencin
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Liam Cullen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anusha Pallapati
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Avi Liu
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Chen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Satish Rojekar
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgii Pevnev
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Uliana Cheliadinova
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Darya Vasilyeva
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guzel Burganova
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anne Macdonald
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mansi Saxena
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ki Goosens
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Orly Barak
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daria Lizneva
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anisa Gumerova
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, University of Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Vitaly Ryu
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tony Yuen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Tal Frolinger
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Mone Zaidi
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Li Y, Yu X, Ma Z, Liu Q, Li M, Tian X, Li B, Zhang R, Gu P, Bai F, Luo G, Li M, Sun D. Nuclear Magnetic Resonance Analysis Implicates Sex-Specific Dysregulation of the Blood Lipids in Alzheimer's Disease: A Retrospective Health-Controlled Study. Psychiatry Investig 2024; 21:1211-1220. [PMID: 39610232 DOI: 10.30773/pi.2024.0164] [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: 05/11/2024] [Accepted: 08/29/2024] [Indexed: 11/30/2024] Open
Abstract
OBJECTIVE The aging demographic landscape worldwide portends a heightened prevalence of neurodegenerative disorders. Foremost among these is Alzheimer's disease (AD), the foremost cause of dementia in older adults. The shortage of efficacious therapies and early diagnostic indicators underscores the imperative to identify non-invasive biomarkers for early detection and disease monitoring. Recently, blood metabolites have emerged as promising candidates for AD biomarkers. METHODS Leveraging nuclear magnetic resonance (NMR) spectroscopy on plasma specimens, we conducted a cross-sectional study encompassing 35 AD patients and 35 age-matched healthy controls. Cognitive function was evaluated using the mini-mental state examination in all participants, followed by peripheral blood sample collection. We utilized univariate and multivariate analyses to perform targeted lipidomic profiling via NMR spectroscopy. RESULTS Our study revealed significant differences in the expression profiles of low-density lipoprotein-associated subfractions in females and high-density lipoprotein-associated subfractions in males between AD patients and healthy controls (all p<0.05). However, there was no significant metabolite overlap between males and females. Furthermore, receiver operating characteristic curve analysis demonstrated that the combination of lipid metabolites had good diagnostic values (all area under the curve>0.70; p<0.05). CONCLUSION Our findings suggest that the blood plasma samples using NMR hold promise in distinguishing between AD patients and healthy controls, with significant clinical implications for advancing AD diagnostic methodologies.
Collapse
Affiliation(s)
- Yanzhe Li
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Xue Yu
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Zhonghui Ma
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Qinghe Liu
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Min Li
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, China
| | - Xue Tian
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Baozhu Li
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Ran Zhang
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Pei Gu
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Fengfeng Bai
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Guoshuai Luo
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Meijuan Li
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Daliang Sun
- Institute of Mental Health, Tianjin Mental Health Center, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| |
Collapse
|
11
|
Kazemeini S, Nadeem-Tariq A, Shih R, Rafanan J, Ghani N, Vida TA. From Plaques to Pathways in Alzheimer's Disease: The Mitochondrial-Neurovascular-Metabolic Hypothesis. Int J Mol Sci 2024; 25:11720. [PMID: 39519272 PMCID: PMC11546801 DOI: 10.3390/ijms252111720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) presents a public health challenge due to its progressive neurodegeneration, cognitive decline, and memory loss. The amyloid cascade hypothesis, which postulates that the accumulation of amyloid-beta (Aβ) peptides initiates a cascade leading to AD, has dominated research and therapeutic strategies. The failure of recent Aβ-targeted therapies to yield conclusive benefits necessitates further exploration of AD pathology. This review proposes the Mitochondrial-Neurovascular-Metabolic (MNM) hypothesis, which integrates mitochondrial dysfunction, impaired neurovascular regulation, and systemic metabolic disturbances as interrelated contributors to AD pathogenesis. Mitochondrial dysfunction, a hallmark of AD, leads to oxidative stress and bioenergetic failure. Concurrently, the breakdown of the blood-brain barrier (BBB) and impaired cerebral blood flow, which characterize neurovascular dysregulation, accelerate neurodegeneration. Metabolic disturbances such as glucose hypometabolism and insulin resistance further impair neuronal function and survival. This hypothesis highlights the interconnectedness of these pathways and suggests that therapeutic strategies targeting mitochondrial health, neurovascular integrity, and metabolic regulation may offer more effective interventions. The MNM hypothesis addresses these multifaceted aspects of AD, providing a comprehensive framework for understanding disease progression and developing novel therapeutic approaches. This approach paves the way for developing innovative therapeutic strategies that could significantly improve outcomes for millions affected worldwide.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (S.K.); (A.N.-T.); (R.S.); (J.R.); (N.G.)
| |
Collapse
|
12
|
Martínez-Drudis L, Bérard M, Musiol D, Rivest S, Oueslati A. Pharmacological inhibition of PLK2 kinase activity mitigates cognitive decline but aggravates APP pathology in a sex-dependent manner in APP/PS1 mouse model of Alzheimer's disease. Heliyon 2024; 10:e39571. [PMID: 39498012 PMCID: PMC11532864 DOI: 10.1016/j.heliyon.2024.e39571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Converging evidence from clinical and experimental studies suggest the potential significance of Polo-like kinase 2 (PLK2) in regulating the phosphorylation and toxicity of the Alzheimer's disease (AD)-related protein, amyloid precursor protein (APP). These findings have prompted various experimental trials aimed at inhibiting PLK2 kinase activity in different transgenic mouse models of AD. While positive impacts on cognitive decline were reported in these studies, the cellular effects remained controversial. In the present study, we sought to assess the cognitive and cellular consequences of chronic PLK2 inhibitor treatment in the APP/PS1 transgenic mouse model of AD. First, we confirmed that inhibiting PLK2 prevented cognitive decline in a sex-dependent manner, particularly by enhancing working memory in male APP/PS1 mice. Surprisingly, cellular analysis revealed that treatment with PLK2 inhibitor increased the load of amyloid plaques and elevated levels of soluble amyloid β (Aβ) 40 and Aβ42 in the cortex, as well as insoluble Aβ42 in the hippocampus of female mice, without affecting APP pathology in males. These results underscore the potential of PLK2 inhibition to mitigate cognitive symptoms in males. However, paradoxically, it intensifies amyloid pathology in females by enhancing APP amyloidogenic processing, creating a controversial aspect to its therapeutic impact. Overall, these data highlight the sex-dependent nature of the effects of PLK2 inhibition, which may also be influenced by the genetic background of the transgenic mouse model utilized.
Collapse
Affiliation(s)
- Laura Martínez-Drudis
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Morgan Bérard
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Dylan Musiol
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Serge Rivest
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Abid Oueslati
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| |
Collapse
|
13
|
Saha P, Sisodia SS. Role of the gut microbiome in mediating sex-specific differences in the pathophysiology of Alzheimer's disease. Neurotherapeutics 2024; 21:e00426. [PMID: 39054179 PMCID: PMC11585881 DOI: 10.1016/j.neurot.2024.e00426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease (AD) presents distinct pathophysiological features influenced by biological sex, with women disproportionately affected due to sex-specific genetic, hormonal, and epigenetic factors. This review delves into three critical areas of sex differences in AD: First, we explore how genetic predisposition and hormonal changes, particularly those involving sex-specific modifications, influence susceptibility and progression of the disease. Second, we examine the neuroimmune dynamics in AD, emphasizing variations in microglial activity between sexes during crucial developmental stages and the effects of hormonal interventions on disease outcomes. Crucially, this review highlights the significant role of gut microbiome perturbations in shaping AD pathophysiology in a sex-specific manner, suggesting that these alterations can further influence microglial activity and overall disease trajectory. Third, we provide a viewpoint that advocates for personalized therapeutic strategies that integrate the understanding of hormonal fluctuations and microbiome dynamics into treatment plans in order to optimize patient outcomes.
Collapse
Affiliation(s)
- Piyali Saha
- Department of Neurobiology, The University of Chicago, Chicago, IL, USA.
| | - Sangram S Sisodia
- Department of Neurobiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
14
|
Rosenzweig N, Kleemann KL, Rust T, Carpenter M, Grucci M, Aronchik M, Brouwer N, Valenbreder I, Cooper-Hohn J, Iyer M, Krishnan RK, Sivanathan KN, Brandão W, Yahya T, Durao A, Yin Z, Chadarevian JP, Properzi MJ, Nowarski R, Davtyan H, Weiner HL, Blurton-Jones M, Yang HS, Eggen BJL, Sperling RA, Butovsky O. Sex-dependent APOE4 neutrophil-microglia interactions drive cognitive impairment in Alzheimer's disease. Nat Med 2024; 30:2990-3003. [PMID: 38961225 DOI: 10.1038/s41591-024-03122-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 06/11/2024] [Indexed: 07/05/2024]
Abstract
APOE4 is the strongest genetic risk factor for Alzheimer's disease (AD), with increased odds ratios in female carriers. Targeting amyloid plaques shows modest improvement in male non-APOE4 carriers. Leveraging single-cell transcriptomics across APOE variants in both sexes, multiplex flow cytometry and validation in two independent cohorts of APOE4 female carriers with AD, we identify a new subset of neutrophils interacting with microglia associated with cognitive impairment. This phenotype is defined by increased interleukin (IL)-17 and IL-1 coexpressed gene modules in blood neutrophils and in microglia of cognitively impaired female APOE ε4 carriers, showing increased infiltration to the AD brain. APOE4 female IL-17+ neutrophils upregulated the immunosuppressive cytokines IL-10 and TGFβ and immune checkpoints, including LAG3 and PD-1, associated with accelerated immune aging. Deletion of APOE4 in neutrophils reduced this immunosuppressive phenotype and restored the microglial response to neurodegeneration, limiting plaque pathology in AD mice. Mechanistically, IL-17F upregulated in APOE4 neutrophils interacts with microglial IL-17RA to suppress the induction of the neurodegenerative phenotype, and blocking this axis supported cognitive improvement in AD mice. These findings provide a translational basis to target IL-17F in APOE ε4 female carriers with cognitive impairment.
Collapse
Affiliation(s)
- Neta Rosenzweig
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kilian L Kleemann
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Thomas Rust
- Department of Biomedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Madison Carpenter
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Madeline Grucci
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Aronchik
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nieske Brouwer
- Department of Biomedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Isabel Valenbreder
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joya Cooper-Hohn
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Malvika Iyer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rajesh K Krishnan
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kisha N Sivanathan
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Wesley Brandão
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Taha Yahya
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana Durao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhuoran Yin
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jean Paul Chadarevian
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Roni Nowarski
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Hayk Davtyan
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bart J L Eggen
- Department of Biomedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Krishnan P, Bobak CA, Hill JE. Sex-specific blood-derived RNA biomarkers for childhood tuberculosis. Sci Rep 2024; 14:16859. [PMID: 39039071 PMCID: PMC11263679 DOI: 10.1038/s41598-024-66946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
Confirmatory diagnosis of childhood tuberculosis (TB) remains a challenge mainly due to its dependence on sputum samples and the paucibacillary nature of the disease. Thus, only ~ 30% of suspected cases in children are diagnosed and the need for minimally invasive, non-sputum-based biomarkers remains unmet. Understanding host molecular changes by measuring blood-based transcriptomic markers has shown promise as a diagnostic tool for TB. However, the implication of sex contributing to disease heterogeneity and therefore diagnosis remains to be understood. Using publicly available gene expression data (GSE39939, GSE39940; n = 370), we report a sex-specific RNA biomarker signature that could improve the diagnosis of TB disease in children. We found four gene biomarker signatures for male (SLAMF8, GBP2, WARS, and FCGR1C) and female pediatric patients (GBP6, CELSR3, ALDH1A1, and GBP4) from Kenya, South Africa, and Malawi. Both signatures achieved a sensitivity of 85% and a specificity of 70%, which approaches the WHO-recommended target product profile for a triage test. Our gene signatures outperform most other gene signatures reported previously for childhood TB diagnosis.
Collapse
Affiliation(s)
- Preethi Krishnan
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Carly A Bobak
- Department of Biomedical Data Science, Dartmouth College, Hanover, NH, 03755, USA
| | - Jane E Hill
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
16
|
Dunham SJB, Avelar‐Barragan J, Rothman JA, Adams ED, Faraci G, Forner S, Kawauchi S, Tenner AJ, Green KN, LaFerla FM, MacGregor GR, Mapstone M, Whiteson KL. Sex-specific associations between AD genotype and the microbiome of human amyloid beta knock-in (hAβ-KI) mice. Alzheimers Dement 2024; 20:4935-4950. [PMID: 38572865 PMCID: PMC11247698 DOI: 10.1002/alz.13794] [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: 10/24/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Emerging evidence links changes in the gut microbiome to late-onset Alzheimer's disease (LOAD), necessitating examination of AD mouse models with consideration of the microbiome. METHODS We used shotgun metagenomics and untargeted metabolomics to study the human amyloid beta knock-in (hAβ-KI) murine model for LOAD compared to both wild-type (WT) mice and a model for early-onset AD (3xTg-AD). RESULTS Eighteen-month female (but not male) hAβ-KI microbiomes were distinct from WT microbiomes, with AD genotype accounting for 18% of the variance by permutational multivariate analysis of variance (PERMANOVA). Metabolomic diversity differences were observed in females, however no individual metabolites were differentially abundant. hAβ-KI mice microbiomes were distinguishable from 3xTg-AD animals (81% accuracy by random forest modeling), with separation primarily driven by Romboutsia ilealis and Turicibacter species. Microbiomes were highly cage specific, with cage assignment accounting for more than 40% of the PERMANOVA variance between the groups. DISCUSSION These findings highlight a sex-dependent variation in the microbiomes of hAβ-KI mice and underscore the importance of considering the microbiome when designing studies that use murine models for AD. HIGHLIGHTS Microbial diversity and the abundance of several species differed in human amyloid beta knock-in (hAβ-KI) females but not males. Correlations to Alzheimer's disease (AD) genotype were stronger for the microbiome than the metabolome. Microbiomes from hAβ-KI mice were distinct from 3xTg-AD mice. Cage effects accounted for most of the variance in the microbiome and metabolome.
Collapse
Affiliation(s)
- Sage J. B. Dunham
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Julio Avelar‐Barragan
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Jason A. Rothman
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Eric D. Adams
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Gina Faraci
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND)University of California IrvineIrvineCaliforniaUSA
| | - Shimako Kawauchi
- Department of Anatomy and NeurobiologyDevelopmental Biology CenterUniversity of California IrvineCollege of MedicineIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Molecular Biology & BiochemistryDepartment of Neurobiology and BehaviorDepartment of Pathology and Laboratory MedicineSchool of MedicineInstitute for Memory Impairments and Neurological Disorders (UCI MIND)University of California IrvineIrvineCaliforniaUSA
| | - Kim N. Green
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Department of Neurobiology and BehaviorSchool of Biological SciencesCenter for Neural Circuit MappingUniversity of California IrvineIrvineCaliforniaUSA
| | - Frank M. LaFerla
- Institute for Memory Impairments and Neurological Disorders (UCI MIND)Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Grant R. MacGregor
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCaliforniaUSA
| | - Mark Mapstone
- Department of NeurologyUniversity of California IrvineIrvineCaliforniaUSA
| | - Katrine L. Whiteson
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| |
Collapse
|
17
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of pathology in a mouse model of Alzheimer's disease. Nat Commun 2024; 15:5217. [PMID: 38890307 PMCID: PMC11189507 DOI: 10.1038/s41467-024-49589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and dietary protein restriction extends the lifespan and healthspan of mice. In this study, we examined the effect of protein restriction (PR) on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. Here, we show that PR promotes leanness and glycemic control in 3xTg mice, specifically rescuing the glucose intolerance of 3xTg females. PR induces sex-specific alterations in circulating and brain metabolites, downregulating sphingolipid subclasses in 3xTg females. PR also reduces AD pathology and mTORC1 activity, increases autophagy, and improves the cognition of 3xTg mice. Finally, PR improves the survival of 3xTg mice. Our results suggest that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jessica H Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dominique A Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Natalie M Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
18
|
Wang D, Hendrix CC, Lee Y, Noval C, Crego N. Characteristics of Older Adults with Alzheimer's Disease Who Were Hospitalized during the COVID-19 Pandemic: A Secondary Data Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:703. [PMID: 38928949 PMCID: PMC11203573 DOI: 10.3390/ijerph21060703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024]
Abstract
We aim to investigate the relationships between the population characteristics of patients with Alzheimer's Disease (AD) and their Healthcare Utilization (HU) during the COVID-19 pandemic. Electronic health records (EHRs) were utilized. The study sample comprised those with ICD-10 codes G30.0, G30.1, G30.8, and G30.9 between 1 January 2020 and 31 December 2021. Pearson's correlation and multiple regression were used. The analysis utilized 1537 patient records with an average age of 82.20 years (SD = 7.71); 62.3% were female. Patients had an average of 1.64 hospitalizations (SD = 1.18) with an average length of stay (ALOS) of 7.45 days (SD = 9.13). Discharge dispositions were primarily home (55.1%) and nursing facilities (32.4%). Among patients with multiple hospitalizations, a negative correlation was observed between age and both ALOS (r = -0.1264, p = 0.0030) and number of hospitalizations (r = -0.1499, p = 0.0004). Predictors of longer ALOS included male gender (p = 0.0227), divorced or widowed (p = 0.0056), and the use of Medicare Advantage and other private insurance (p = 0.0178). Male gender (p = 0.0050) and Black race (p = 0.0069) were associated with a higher hospitalization frequency. We recommend future studies including the co-morbidities of AD patients, larger samples, and longitudinal data.
Collapse
Affiliation(s)
- Dingyue Wang
- School of Nursing, Duke University, Durham, NC 27710, USA; (C.C.H.); (Y.L.); (C.N.); (N.C.)
| | - Cristina C. Hendrix
- School of Nursing, Duke University, Durham, NC 27710, USA; (C.C.H.); (Y.L.); (C.N.); (N.C.)
- GRECC Durham Veterans Affairs Medical Center, Durham, NC 27705, USA
| | - Youran Lee
- School of Nursing, Duke University, Durham, NC 27710, USA; (C.C.H.); (Y.L.); (C.N.); (N.C.)
| | - Christian Noval
- School of Nursing, Duke University, Durham, NC 27710, USA; (C.C.H.); (Y.L.); (C.N.); (N.C.)
| | - Nancy Crego
- School of Nursing, Duke University, Durham, NC 27710, USA; (C.C.H.); (Y.L.); (C.N.); (N.C.)
| |
Collapse
|
19
|
Zhang T, Chu Y, Wang Y, Wang Y, Wang J, Ji X, Zhang G, Shi G, Cui R, Kang Y. Testosterone deficiency worsens mitochondrial dysfunction in APP/PS1 mice. Front Aging Neurosci 2024; 16:1390915. [PMID: 38752208 PMCID: PMC11094339 DOI: 10.3389/fnagi.2024.1390915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 04/12/2024] [Indexed: 05/18/2024] Open
Abstract
Background Recent studies show testosterone (T) deficiency worsens cognitive impairment in Alzheimer's disease (AD) patients. Mitochondrial dysfunction, as an early event of AD, is becoming critical hallmark of AD pathogenesis. However, currently, whether T deficiency exacerbates mitochondrial dysfunction of men with AD remains unclear. Objective The purpose of this study is to explore the effects of T deficiency on mitochondrial dysfunction of male AD mouse models and its potential mechanisms. Methods Alzheimer's disease animal model with T deficiency was performed by castration to 3-month-old male APP/PS1 mice. Hippocampal mitochondrial function of mice was analyzed by spectrophotometry and flow cytometry. The gene expression levels related to mitochondrial biogenesis and mitochondrial dynamics were determined through quantitative real-time PCR (qPCR) and western blot analysis. SH-SY5Y cells treated with flutamide, T and/or H2O2 were processed for analyzing the potential mechanisms of T on mitochondrial dysfunction. Results Testosterone deficiency significantly aggravated the cognitive deficits and hippocampal pathologic damage of male APP/PS1 mice. These effects were consistent with exacerbated mitochondrial dysfunction by gonadectomy to male APP/PS1 mice, reflected by further increase in oxidative damage and decrease in mitochondrial membrane potential, complex IV activity and ATP levels. More importantly, T deficiency induced the exacerbation of compromised mitochondrial homeostasis in male APP/PS1 mice by exerting detrimental effects on mitochondrial biogenesis and mitochondrial dynamics at mRNA and protein level, leading to more defective mitochondria accumulated in the hippocampus. In vitro studies using SH-SY5Y cells validated T's protective effects on the H2O2-induced mitochondrial dysfunction, mitochondrial biogenesis impairment, and mitochondrial dynamics imbalance. Administering androgen receptor (AR) antagonist flutamide weakened the beneficial effects of T pretreatment on H2O2-treated SH-SY5Y cells, demonstrating a critical role of classical AR pathway in maintaining mitochondrial function. Conclusion Testosterone deficiency exacerbates hippocampal mitochondrial dysfunction of male APP/PS1 mice by accumulating more defective mitochondria. Thus, appropriate T levels in the early stage of AD might be beneficial in delaying AD pathology by improving mitochondrial biogenesis and mitochondrial dynamics.
Collapse
Affiliation(s)
- Tianyun Zhang
- Postdoctoral Research Station of Biology, Hebei Medical University, Shijiazhuang, China
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Yun Chu
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Yue Wang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Yu Wang
- Postdoctoral Research Station of Biology, Hebei Medical University, Shijiazhuang, China
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Jinyang Wang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoming Ji
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Guoliang Zhang
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Geming Shi
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Rui Cui
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Yunxiao Kang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of Alzheimer's disease in mice. RESEARCH SQUARE 2024:rs.3.rs-3342413. [PMID: 37790423 PMCID: PMC10543316 DOI: 10.21203/rs.3.rs-3342413/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and many independent groups of researchers have shown that dietary protein restriction (PR) extends the lifespan and healthspan of mice. Here, we examined the effect of PR on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. We found that PR has metabolic benefits for 3xTg mice and non-transgenic controls of both sexes, promoting leanness and glycemic control in 3xTg mice and rescuing the glucose intolerance of 3xTg females. We found that PR induces sex-specific alterations in circulating metabolites and in the brain metabolome and lipidome, downregulating sphingolipid subclasses including ceramides, glucosylceramides, and sphingomyelins in 3xTg females. Consumption of a PR diet starting at 6 months of age reduced AD pathology in conjunction with reduced mTORC1 activity, increased autophagy, and had cognitive benefits for 3xTg mice. Finally, PR improved the survival of 3xTg mice. Our results demonstrate that PR slows the progression of AD at molecular and pathological levels, preserves cognition in this mouse model of AD, and suggests that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M. Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J. Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dominique A. Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Natalie M. Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
21
|
Fernández A, Cuesta P, Marcos A, Montenegro-Peña M, Yus M, Rodríguez-Rojo IC, Bruña R, Maestú F, López ME. Sex differences in the progression to Alzheimer's disease: a combination of functional and structural markers. GeroScience 2024; 46:2619-2640. [PMID: 38105400 PMCID: PMC10828170 DOI: 10.1007/s11357-023-01020-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023] Open
Abstract
Mild cognitive impairment (MCI) has been frequently interpreted as a transitional phase between healthy cognitive aging and dementia, particularly of the Alzheimer's disease (AD) type. Of note, few studies explored that transition from a multifactorial perspective, taking into consideration the effect of basic factors such as biological sex. In the present study 96 subjects with MCI (37 males and 59 females) were followed-up and divided into two subgroups according to their clinical outcome: "progressive" MCI (pMCI = 41), if they fulfilled the diagnostic criteria for AD at the end of follow-up; and "stable" MCI (sMCI = 55), if they remained with the initial diagnosis. Different markers were combined to characterize sex differences between groups, including magnetoencephalography recordings, cognitive performance, and brain volumes derived from magnetic resonance imaging. Results indicated that the pMCI group exhibited higher low-frequency activity, lower scores in neuropsychological tests and reduced brain volumes than the sMCI group, being these measures significantly correlated. When sex was considered, results revealed that this pattern was mainly due to the influence of the females' sample. Overall, females exhibited lower cognitive scores and reduced brain volumes. More interestingly, females in the pMCI group showed an increased theta activity that correlated with a more abrupt reduction of cognitive and volumetric scores as compared with females in the sMCI group and with males in the pMCI group. These findings suggest that females' brains might be more vulnerable to the effects of AD pathology, since regardless of age, they showed signs of more pronounced deterioration than males.
Collapse
Affiliation(s)
- Alberto Fernández
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid, Spain
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
| | - Pablo Cuesta
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
- Department of Radiology, Rehabilitation and Physiotherapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Alberto Marcos
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
- Neurology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Mercedes Montenegro-Peña
- Centre for the Prevention of Cognitive Impairment, Madrid Salud, Madrid City Council, Madrid, Spain
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Miguel Yus
- Radiology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Inmaculada Concepción Rodríguez-Rojo
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Department of Nursing and Psysiotherapy, Universidad de Alcalá, Madrid, Spain
| | - Ricardo Bruña
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
- Department of Radiology, Rehabilitation and Physiotherapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Fernando Maestú
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| | - María Eugenia López
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain.
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain.
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
22
|
Niedowicz DM, Gollihue JL, Weekman EM, Phe P, Wilcock DM, Norris CM, Nelson PT. Using digital pathology to analyze the murine cerebrovasculature. J Cereb Blood Flow Metab 2024; 44:595-610. [PMID: 37988134 PMCID: PMC10981399 DOI: 10.1177/0271678x231216142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/22/2023]
Abstract
Research on the cerebrovasculature may provide insights into brain health and disease. Immunohistochemical staining is one way to visualize blood vessels, and digital pathology has the potential to revolutionize the measurement of blood vessel parameters. These tools provide opportunities for translational mouse model research. However, mouse brain tissue presents a formidable set of technical challenges, including potentially high background staining and cross-reactivity of endogenous IgG. Formalin-fixed paraffin-embedded (FFPE) and fixed frozen sections, both of which are widely used, may require different methods. In this study, we optimized blood vessel staining in mouse brain tissue, testing both FFPE and frozen fixed sections. A panel of immunohistochemical blood vessel markers were tested (including CD31, CD34, collagen IV, DP71, and VWF), to evaluate their suitability for digital pathological analysis. Collagen IV provided the best immunostaining results in both FFPE and frozen fixed murine brain sections, with highly-specific staining of large and small blood vessels and low background staining. Subsequent analysis of collagen IV-stained sections showed region and sex-specific differences in vessel density and vessel wall thickness. We conclude that digital pathology provides a useful tool for relatively unbiased analysis of the murine cerebrovasculature, provided proper protein markers are used.
Collapse
Affiliation(s)
- Dana M Niedowicz
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Jenna L Gollihue
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Erica M Weekman
- Stark Neurosciences Research Institute, Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Panhavuth Phe
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Donna M Wilcock
- Stark Neurosciences Research Institute, Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christopher M Norris
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Pharmacology, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
23
|
Do AN, Ali M, Timsina J, Wang L, Western D, Liu M, Sanford J, Rosende-Roca M, Boada M, Puerta R, Wilson T, Ruiz A, Pastor P, Wyss-Coray T, Cruchaga C, Sung YJ. CSF proteomic profiling with amyloid/tau positivity identifies distinctive sex-different alteration of multiple proteins involved in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.15.24304164. [PMID: 38559166 PMCID: PMC10980123 DOI: 10.1101/2024.03.15.24304164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
In Alzheimer's disease (AD), the most common cause of dementia, females have higher prevalence and faster progression, but sex-specific molecular findings in AD are limited. Here, we comprehensively examined and validated 7,006 aptamers targeting 6,162 proteins in cerebral spinal fluid (CSF) from 2,077 amyloid/tau positive cases and controls to identify sex-specific proteomic signatures of AD. In discovery (N=1,766), we identified 330 male-specific and 121 female-specific proteomic alternations in CSF (FDR <0.05). These sex-specific proteins strongly predicted amyloid/tau positivity (AUC=0.98 in males; 0.99 in females), significantly higher than those with age, sex, and APOE-ε4 (AUC=0.85). The identified sex-specific proteins were well validated (r≥0.5) in the Stanford study (N=108) and Emory study (N=148). Biological follow-up of these proteins led to sex differences in cell-type specificity, pathways, interaction networks, and drug targets. Male-specific proteins, enriched in astrocytes and oligodendrocytes, were involved in postsynaptic and axon-genesis. The male network exhibited direct connections among 152 proteins and highlighted PTEN, NOTCH1, FYN, and MAPK8 as hubs. Drug target suggested melatonin (used for sleep-wake cycle regulation), nabumetone (used for pain), daunorubicin, and verteporfin for treating AD males. In contrast, female-specific proteins, enriched in neurons, were involved in phosphoserine residue binding including cytokine activities. The female network exhibits strong connections among 51 proteins and highlighted JUN and 14-3-3 proteins (YWHAG and YWHAZ) as hubs. Drug target suggested biperiden (for muscle control of Parkinson's disease), nimodipine (for cerebral vasospasm), quinostatin and ethaverine for treating AD females. Together, our findings provide mechanistic understanding of sex differences for AD risk and insights into clinically translatable interventions.
Collapse
Affiliation(s)
- Anh N Do
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Muhammad Ali
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Jigyasha Timsina
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Lihua Wang
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel Western
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Menghan Liu
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Jessie Sanford
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Matitee Rosende-Roca
- Research Center and Memory clinic Fundació ACE. Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Merce Boada
- Research Center and Memory clinic Fundació ACE. Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Raquel Puerta
- Research Center and Memory clinic Fundació ACE. Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Ted Wilson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Agustin Ruiz
- Research Center and Memory clinic Fundació ACE. Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Pau Pastor
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Carlos Cruchaga
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
24
|
Justice MJ. Sex matters in preclinical research. Dis Model Mech 2024; 17:dmm050759. [PMID: 38450661 PMCID: PMC10941654 DOI: 10.1242/dmm.050759] [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] [Indexed: 03/08/2024] Open
Abstract
International Women's Day 2024 has a theme of inclusion. As publishers of preclinical research, we aim to show how inclusion of females in research advances scientific rigor and improves treatment reliability. Sexual reproduction is key to all life across the plant and animal kingdoms. Biological sex takes many forms that are morphologically differentiated during development: stamens versus pistils in plants; color and plumage in birds; fallopian tubes versus vas deferens in mammals; and differences in size, for instance, males are smaller in the fruit fly Drosophila melanogaster. Physical differences may be obvious, but many traits may be more obscure, including hormonal, physiological and metabolic factors. These traits have a big influence on disease and responses to treatment. Thus, we call for improved inclusion, analysis and reporting of sex as a biological variable in preclinical animal modeling research.
Collapse
Affiliation(s)
- Monica J. Justice
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| |
Collapse
|
25
|
Frolinger T, Korkmaz F, Sims S, Sen F, Sultana F, Laurencin V, Cullen L, Pallapati AR, Liu A, Rojekar S, Pevnev G, Cheliadinova U, Vasilyeva D, Burganova G, Macdonald A, Saxena M, Goosens K, Rosen C, Barak O, Lizneva D, Gumerova A, Ye K, Ryu V, Yuen T, Zaidi M. Gene-Dose-Dependent Reduction Fshr Expression Improves Spatial Memory Deficits in Alzheimer's Mice. RESEARCH SQUARE 2024:rs.3.rs-3964789. [PMID: 38463956 PMCID: PMC10925392 DOI: 10.21203/rs.3.rs-3964789/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Alzheimer's disease (AD) is a major progressive neurodegenerative disorder of the aging population. High post-menopausal levels of the pituitary gonadotropin follicle-stimulating hormone (FSH) are strongly associated with the onset of AD, and we have shown recently that FSH directly activates the hippocampal Fshr to drive AD-like pathology and memory loss in mice. To establish a role for FSH in memory loss, we used female 3xTg;Fshr+/+, 3xTg;Fshr+/- and 3xTg;Fshr-/- mice that were either left unoperated or underwent sham surgery or ovariectomy at 8 weeks of age. Unoperated and sham-operated 3xTg;Fshr-/- mice were implanted with 17β-estradiol pellets to normalize estradiol levels. Morris Water Maze and Novel Object Recognition behavioral tests were performed to study deficits in spatial and recognition memory, respectively, and to examine the effects of Fshr depletion. 3xTg;Fshr+/+ mice displayed impaired spatial memory at 5 months of age; both the acquisition and retrieval of the memory were ameliorated in 3xTg;Fshr-/- mice and, to a lesser extent, in 3xTg;Fshr+/- mice- -thus documenting a clear gene-dose-dependent prevention of hippocampal-dependent spatial memory impairment. At 5 and 10 months, sham-operated 3xTg;Fshr-/- mice showed better memory performance during the acquasition and/or retrieval phases, suggesting that Fshr deletion prevented the progression of spatial memory deficits with age. However, this prevention was not seen when mice were ovariectomized, except in the 10-month-old 3xTg;Fshr-/- mice. In the Novel Object Recognition test performed at 10 months, all groups of mice, except ovariectomized 3xTg;Fshr-/- mice showed a loss of recognition memory. Consistent with the neurobehavioral data, there was a gene-dose-dependent reduction mainly in the amyloid β40 isoform in whole brain extracts. Finally, serum FSH levels < 8 ng/mL in 16-month-old APP/PS1 mice were associated with better retrieval of spatial memory. Collectively, the data provide compelling genetic evidence for a protective effect of inhibiting FSH signaling on the progression of spatial and recognition memory deficits in mice, and lay a firm foundation for the use of an FSH-blocking agent for the early prevention of cognitive decline in postmenopausal women.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Avi Liu
- Icahn School of Medicine at Mount Sinai
| | | | | | | | | | | | | | | | | | | | | | | | | | - Keqiang Ye
- Shenzhen Institute of Advanced Technology
| | | | | | | |
Collapse
|
26
|
Geleta U, Prajapati P, Bachstetter A, Nelson PT, Wang WX. Sex-Biased Expression and Response of microRNAs in Neurological Diseases and Neurotrauma. Int J Mol Sci 2024; 25:2648. [PMID: 38473893 PMCID: PMC10931569 DOI: 10.3390/ijms25052648] [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: 01/27/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Neurological diseases and neurotrauma manifest significant sex differences in prevalence, progression, outcome, and therapeutic responses. Genetic predisposition, sex hormones, inflammation, and environmental exposures are among many physiological and pathological factors that impact the sex disparity in neurological diseases. MicroRNAs (miRNAs) are a powerful class of gene expression regulator that are extensively involved in mediating biological pathways. Emerging evidence demonstrates that miRNAs play a crucial role in the sex dimorphism observed in various human diseases, including neurological diseases. Understanding the sex differences in miRNA expression and response is believed to have important implications for assessing the risk of neurological disease, defining therapeutic intervention strategies, and advancing both basic research and clinical investigations. However, there is limited research exploring the extent to which miRNAs contribute to the sex disparities observed in various neurological diseases. Here, we review the current state of knowledge related to the sexual dimorphism in miRNAs in neurological diseases and neurotrauma research. We also discuss how sex chromosomes may contribute to the miRNA sexual dimorphism phenomenon. We attempt to emphasize the significance of sexual dimorphism in miRNA biology in human diseases and to advocate a gender/sex-balanced science.
Collapse
Affiliation(s)
- Urim Geleta
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
| | - Paresh Prajapati
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
| | - Adam Bachstetter
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Neuroscience, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Peter T. Nelson
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Wang-Xia Wang
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
27
|
Mensah-Kane P, Davis DL, Shi HS, Trinh OT, Vann PH, Dory L, Sumien N. Hyperbaric oxygen alleviates selective domains of cognitive and motor deficits in female 5xFAD mice. GeroScience 2024; 46:517-530. [PMID: 38153668 PMCID: PMC10828284 DOI: 10.1007/s11357-023-01047-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/14/2023] [Indexed: 12/29/2023] Open
Abstract
Treatment of Alzheimer's disease (AD) has been limited to managing of symptoms or anti-amyloid therapy with limited results and uncertainty. Seeking out new therapies that can reverse the effects of this devastating disease is important. Hyperbaric oxygen (HBO) therapy could be such a candidate as it has been shown to improve brain function in certain neurological conditions. Furthermore, the role sex plays in the vulnerability/resilience to AD remains equivocal. An understanding of what makes one sex more vulnerable to AD could unveil new pathways for therapy development. In this study, we investigated the effects of HBO on cognitive, motor, and affective function in a mouse model of AD (5xFAD) and assessed protein oxidation in peripheral tissues as a safety indicator. The motor and cognitive abilities of 5xFAD mice were significantly impaired. HBO therapy improved cognitive flexibility and associative learning of 5xFAD females but not males, but HBO had no effect other aspects of cognition. HBO also reversed AD-related declines in balance but had no impact on gait and anxiety-like behavior. HBO did not affect body weights or oxidative stress in peripheral tissues. Our study provides further support for HBO therapy as a potential treatment for AD and emphasizes the importance of considering sex as a biological variable in therapeutic development. Further investigations into the underlying mechanisms of HBO's sex-specific responses are warranted, as well as optimizing treatment protocols for maximum benefits.
Collapse
Affiliation(s)
- Paapa Mensah-Kane
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Delaney L Davis
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Helen S Shi
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Oanh T Trinh
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Philip H Vann
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Ladislav Dory
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Nathalie Sumien
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA.
| |
Collapse
|
28
|
Karnik SJ, Margetts TJ, Wang HS, Movila A, Oblak AL, Fehrenbacher JC, Kacena MA, Plotkin LI. Mind the Gap: Unraveling the Intricate Dance Between Alzheimer's Disease and Related Dementias and Bone Health. Curr Osteoporos Rep 2024; 22:165-176. [PMID: 38285083 PMCID: PMC10912190 DOI: 10.1007/s11914-023-00847-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/30/2024]
Abstract
PURPOSE OF REVIEW This review examines the linked pathophysiology of Alzheimer's disease/related dementia (AD/ADRD) and bone disorders like osteoporosis. The emphasis is on "inflammaging"-a low-level inflammation common to both, and its implications in an aging population. RECENT FINDINGS Aging intensifies both ADRD and bone deterioration. Notably, ADRD patients have a heightened fracture risk, impacting morbidity and mortality, though it is uncertain if fractures worsen ADRD. Therapeutically, agents targeting inflammation pathways, especially Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) and TNF-α, appear beneficial for both conditions. Additionally, treatments like Sirtuin 1 (SIRT-1), known for anti-inflammatory and neuroprotective properties, are gaining attention. The interconnectedness of AD/ADRD and bone health necessitates a unified treatment approach. By addressing shared mechanisms, we can potentially transform therapeutic strategies, enriching our understanding and refining care in our aging society. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Sonali J Karnik
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tyler J Margetts
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hannah S Wang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alexandru Movila
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adrian L Oblak
- Department of Radiology & Imaging Sciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jill C Fehrenbacher
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
| | - Lilian I Plotkin
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
29
|
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
|
30
|
Saha P, Weigle IQ, Slimmon N, Poli PB, Patel P, Zhang X, Cao Y, Michalkiewicz J, Gomm A, Zhang C, Tanzi RE, Dylla N, Al-Hendy A, Sisodia SS. Early modulation of the gut microbiome by female sex hormones alters amyloid pathology and microglial function. Sci Rep 2024; 14:1827. [PMID: 38246956 PMCID: PMC10800351 DOI: 10.1038/s41598-024-52246-6] [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: 10/18/2023] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
It is well-established that women are disproportionately affected by Alzheimer's disease. The mechanisms underlying this sex-specific disparity are not fully understood, but several factors that are often associated-including interactions of sex hormones, genetic factors, and the gut microbiome-likely contribute to the disease's etiology. Here, we have examined the role of sex hormones and the gut microbiome in mediating Aβ amyloidosis and neuroinflammation in APPPS1-21 mice. We report that postnatal gut microbiome perturbation in female APPPS1-21 mice leads to an elevation in levels of circulating estradiol. Early stage ovariectomy (OVX) leads to a reduction of plasma estradiol that is correlated with a significant alteration of gut microbiome composition and reduction in Aβ pathology. On the other hand, supplementation of OVX-treated animals with estradiol restores Aβ burden and influences gut microbiome composition. The reduction of Aβ pathology with OVX is paralleled by diminished levels of plaque-associated microglia that acquire a neurodegenerative phenotype (MGnD-type) while estradiol supplementation of OVX-treated animals leads to a restoration of activated microglia around plaques. In summary, our investigation elucidates the complex interplay between sex-specific hormonal modulations, gut microbiome dynamics, metabolic perturbations, and microglial functionality in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Piyali Saha
- Department of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Ian Q Weigle
- Department of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Nicholas Slimmon
- School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, TX, USA
| | - Pedro Blauth Poli
- Department of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Priyam Patel
- Center for Genetic Medicine, Northwestern University, Chicago, IL, USA
| | - Xiaoqiong Zhang
- Department of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Yajun Cao
- Genomic Facility, The University of Chicago, Chicago, IL, USA
| | - Julia Michalkiewicz
- Department of Physiology and Biophysics, The University of Illinois, Chicago, IL, USA
| | - Ashley Gomm
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Can Zhang
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Rudolph E Tanzi
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Nicholas Dylla
- Duchossois Family Institute, The University of Chicago, Chicago, IL, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL, USA
| | - Sangram S Sisodia
- Department of Neurobiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
31
|
Wu D, Bi X, Chow KHM. Identification of female-enriched and disease-associated microglia (FDAMic) contributes to sexual dimorphism in late-onset Alzheimer's disease. J Neuroinflammation 2024; 21:1. [PMID: 38178204 PMCID: PMC10765928 DOI: 10.1186/s12974-023-02987-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Late-onset Alzheimer's disease (LOAD) is the most common form of dementia; it disproportionally affects women in terms of both incidence rates and severity of progression. The cellular and molecular mechanisms underlying this clinical phenomenon remain elusive and ill-defined. METHODS In-depth analyses were performed with multiple human LOAD single-nucleus transcriptome datasets to thoroughly characterize cell populations in the cerebral cortex. ROSMAP bulk human brain tissue transcriptome and DNA methylome datasets were also included for validation. Detailed assessments of microglial cell subpopulations and their relevance to sex-biased changes at the tissue level were performed. Clinical trait associations, cell evolutionary trajectories, and transcription regulon analyses were conducted. RESULTS The relative numbers of functionally defective microglia were aberrantly increased uniquely among affected females. Substratification of the microglia into different subtypes according to their transcriptomic signatures identified a group of female-enriched and disease-associated microglia (FDAMic), the numbers of which were positively associated with disease severity. Phenotypically, these cells exhibit transcriptomic signatures that support active proliferation, MHC class II autoantigen presentation and amyloid-β binding, but they are also likely defective in phagocytosis. FDAMic are likely evolved from female activated response microglia (ARMic) with an APOE4 background and compromised estrogen receptor (ER) signaling that is deemed to be active among most subtypes of microglia. CONCLUSION This study offered important insights at both the cellular and molecular levels into how ER signaling affects microglial heterogeneity and function. FDAMic are associated with more advanced pathologies and severe trends of cognitive decline. Their emergence could, at least in part, explain the phenomenon of greater penetrance of the APOE4 genotype found in females. The biases of FDAMic emergence toward female sex and APOE4 status may also explain why hormone replacement therapy is more effective in APOE4 carriers. The pathologic nature of FDAMic suggests that selective modulations of these cells may help to regain brain neuroimmune homeostasis, serving as a new target for future drug development.
Collapse
Affiliation(s)
- Deng Wu
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Xiaoman Bi
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Kim Hei-Man Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China.
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China.
- Nexus of Rare Neurodegenerative Diseases, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China.
| |
Collapse
|
32
|
Merrick R, Brayne C. Sex Differences in Dementia, Cognition, and Health in the Cognitive Function and Ageing Studies (CFAS). J Alzheimers Dis 2024; 100:S3-S12. [PMID: 39121118 DOI: 10.3233/jad-240358] [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: 08/11/2024]
Abstract
Background There is renewed interest in whether sex differences in dementia risk exist, and what influence social and biological factors have. Objective To review evidence from the Cognitive Function and Ageing Studies (CFAS), a multi-center population-representative cohort study in the UK; focusing on dementia and cognition, incorporating findings on participants' health and social circumstances. Methods After identifying all CFAS publications, the results of all sex-stratified primary analyses of CFAS data were narratively reviewed. Results Of 337 publications, 94 report results by sex (including null findings), which are summarized by theme: dementia epidemiology, cognition, mental health, health expectancy, social context and biological resource (including neuropathology). Conclusions Where differences are found they most commonly favor men; however, greater mortality in men may confound associations with age-related outcomes. This 'survival bias' may explain findings of greater risk of dementia and faster cognitive decline in women. Age-specific dementia incidence was similar between sexes, although reduced incidence across study generations was more pronounced in men. Mood disorders were more prevalent in women, but adjusting for disability and deprivation attenuated the association. Prominent findings from other cohorts that women have more Alzheimer's disease pathology and greater risk of dementia from the Apolipoprotein E ɛ4 allele were not observed, warranting further investigation. The 'male-female health-survival paradox' is demonstrated whereby women live longer but with more comorbidity and disability. Examining why health expectancies changed differently over two decades for each sex (interacting with deprivation) may inform population interventions to improve cognitive, mental and physical health in later life.
Collapse
Affiliation(s)
- Richard Merrick
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| |
Collapse
|
33
|
Shaik SM, Cao Y, Gogola JV, Dodiya HB, Zhang X, Boutej H, Han W, Kriz J, Sisodia SS. Translational profiling identifies sex-specific metabolic and epigenetic reprogramming of cortical microglia/macrophages in APPPS1-21 mice with an antibiotic-perturbed-microbiome. Mol Neurodegener 2023; 18:95. [PMID: 38104136 PMCID: PMC10725591 DOI: 10.1186/s13024-023-00668-7] [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: 04/19/2023] [Accepted: 10/14/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Microglia, the brain-resident macrophages perform immune surveillance and engage with pathological processes resulting in phenotype changes necessary for maintaining homeostasis. In preceding studies, we showed that antibiotic-induced perturbations of the gut microbiome of APPPS1-21 mice resulted in significant attenuation in Aβ amyloidosis and altered microglial phenotypes that are specific to male mice. The molecular events underlying microglial phenotypic transitions remain unclear. Here, by generating 'APPPS1-21-CD11br' reporter mice, we investigated the translational state of microglial/macrophage ribosomes during their phenotypic transition and in a sex-specific manner. METHODS Six groups of mice that included WT-CD11br, antibiotic (ABX) or vehicle-treated APPPS1-21-CD11br males and females were sacrificed at 7-weeks of age (n = 15/group) and used for immunoprecipitation of microglial/macrophage polysomes from cortical homogenates using anti-FLAG antibody. Liquid chromatography coupled to tandem mass spectrometry and label-free quantification was used to identify newly synthesized peptides isolated from polysomes. RESULTS We show that ABX-treatment leads to decreased Aβ levels in male APPPS1-21-CD11br mice with no significant changes in females. We identified microglial/macrophage polypeptides involved in mitochondrial dysfunction and altered calcium signaling that are associated with Aβ-induced oxidative stress. Notably, female mice also showed downregulation of newly-synthesized ribosomal proteins. Furthermore, male mice showed an increase in newly-synthesized polypeptides involved in FcγR-mediated phagocytosis, while females showed an increase in newly-synthesized polypeptides responsible for actin organization associated with microglial activation. Next, we show that ABX-treatment resulted in substantial remodeling of the epigenetic landscape, leading to a metabolic shift that accommodates the increased bioenergetic and biosynthetic demands associated with microglial polarization in a sex-specific manner. While microglia in ABX-treated male mice exhibited a metabolic shift towards a neuroprotective phenotype that promotes Aβ clearance, microglia in ABX-treated female mice exhibited loss of energy homeostasis due to persistent mitochondrial dysfunction and impaired lysosomal clearance that was associated with inflammatory phenotypes. CONCLUSIONS Our studies provide the first snapshot of the translational state of microglial/macrophage cells in a mouse model of Aβ amyloidosis that was subject to ABX treatment. ABX-mediated changes resulted in metabolic reprogramming of microglial phenotypes to modulate immune responses and amyloid clearance in a sex-specific manner. This microglial plasticity to support neuro-energetic homeostasis for its function based on sex paves the path for therapeutic modulation of immunometabolism for neurodegeneration.
Collapse
Affiliation(s)
- Shabana M Shaik
- Dept. of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Yajun Cao
- Dept. of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Joseph V Gogola
- Dept. of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Hemraj B Dodiya
- Dept. of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Xulun Zhang
- Dept. of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Hejer Boutej
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Laval University, Québec, QC, Canada
| | - Weinong Han
- Dept. of Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Jasna Kriz
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Laval University, Québec, QC, Canada
| | | |
Collapse
|
34
|
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
|
35
|
Gilmer G, Hettinger ZR, Tuakli-Wosornu Y, Skidmore E, Silver JK, Thurston RC, Lowe DA, Ambrosio F. Female aging: when translational models don't translate. NATURE AGING 2023; 3:1500-1508. [PMID: 38052933 PMCID: PMC11099540 DOI: 10.1038/s43587-023-00509-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/25/2023] [Indexed: 12/07/2023]
Abstract
For many pathologies associated with aging, female patients present with higher morbidity and more frequent adverse events from treatments compared to male patients. While preclinical models are the foundation of our mechanistic understanding of age-related diseases, the most common models fail to recapitulate archetypical female aging trajectories. For example, while over 70% of the top age-related diseases are influenced by the systemic effects of reproductive senescence, we found that preclinical studies that include menopausal phenotypes modeling those seen in humans make up <1% of published aging biology research. The long-term impacts of pregnancy, birthing and breastfeeding are also typically omitted from preclinical work. In this Perspective, we summarize limitations in the most commonly used aging models, and we provide recommendations for better incorporating menopause, pregnancy and other considerations of sex in vivo and in vitro. Lastly, we outline action items for aging biology researchers, journals, funding agencies and animal providers to address this gap.
Collapse
Affiliation(s)
- Gabrielle Gilmer
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding Rehabilitation, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA, USA
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Cellular and Molecular Pathology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary R Hettinger
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding Rehabilitation, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
- Department of Geriatric Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yetsa Tuakli-Wosornu
- Department of Social and Behavioral Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elizabeth Skidmore
- Department of Occupational Therapy, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julie K Silver
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Massachusetts General Hospital, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Brigham and Women's Hospital, Boston, MA, USA
| | - Rebecca C Thurston
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dawn A Lowe
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Fabrisia Ambrosio
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding Rehabilitation, Boston, MA, USA.
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA, USA.
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Samson AD, Rajagopal S, Pasvanis S, Villeneuve S, McIntosh AR, Rajah MN. Sex differences in longitudinal changes of episodic memory-related brain activity and cognition in cognitively unimpaired older adults with a family history of Alzheimer's disease. Neuroimage Clin 2023; 40:103532. [PMID: 37931333 PMCID: PMC10652211 DOI: 10.1016/j.nicl.2023.103532] [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: 07/10/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
Episodic memory decline is an early symptom of Alzheimer's disease (AD) - a neurodegenerative disease that has a higher prevalence rate in older females compared to older males. However, little is known about why these sex differences in prevalence rate exist. In the current longitudinal task fMRI study, we explored whether there were sex differences in the patterns of memory decline and brain activity during object-location (spatial context) encoding and retrieval in a large sample of cognitively unimpaired older adults from the Pre-symptomatic Evaluation of Novel or Experimental Treatments for Alzheimer's Disease (PREVENT-AD) program who are at heightened risk of developing AD due to having a family history (+FH) of the disease. The goal of the study was to gain insight into whether there are sex differences in the neural correlates of episodic memory decline, which may advance knowledge about sex-specific patterns in the natural progression to AD. Our results indicate that +FH females performed better than +FH males at both baseline and follow-up on neuropsychological and task fMRI measures of episodic memory. Moreover, multivariate data-driven task fMRI analysis identified generalized patterns of longitudinal decline in medial temporal lobe activity that was paralleled by longitudinal increases in lateral prefrontal cortex, caudate and midline cortical activity during successful episodic retrieval and novelty detection in +FH males, but not females. Post-hoc analyses indicated that higher education had a stronger effect on +FH females neuropsychological scores compared to +FH males. We conclude that higher educational attainment may have a greater neuroprotective effect in older +FH females compared to +FH males.
Collapse
Affiliation(s)
- Alexandria D Samson
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Ontario M6A 2E1, Canada; Department of Psychology, University of Toronto, Toronto, Ontario M5S 3G3, Canada
| | - Sricharana Rajagopal
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Quebec H4H 1R3, Canada
| | - Stamatoula Pasvanis
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Quebec H4H 1R3, Canada
| | - Sylvia Villeneuve
- Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Hospital Research Centre, Montreal, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Anthony R McIntosh
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Ontario M6A 2E1, Canada; Department of Psychology, University of Toronto, Toronto, Ontario M5S 3G3, Canada; Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - M Natasha Rajah
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada; Department of Psychology, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada.
| |
Collapse
|
37
|
Israel LL, Braubach O, Shatalova ES, Chepurna O, Sharma S, Klymyshyn D, Galstyan A, Chiechi A, Cox A, Herman D, Bliss B, Hasen I, Ting A, Arechavala R, Kleinman MT, Patil R, Holler E, Ljubimova JY, Koronyo-Hamaoui M, Sun T, Black KL. Exposure to environmental airborne particulate matter caused wide-ranged transcriptional changes and accelerated Alzheimer's-related pathology: A mouse study. Neurobiol Dis 2023; 187:106307. [PMID: 37739136 DOI: 10.1016/j.nbd.2023.106307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023] Open
Abstract
Air pollution poses a significant threat to human health, though a clear understanding of its mechanism remains elusive. In this study, we sought to better understand the effects of various sized particulate matter from polluted air on Alzheimer's disease (AD) development using an AD mouse model. We exposed transgenic Alzheimer's mice in their prodromic stage to different sized particulate matter (PM), with filtered clean air as control. After 3 or 6 months of exposure, mouse brains were harvested and analyzed. RNA-seq analysis showed that various PM have differential effects on the brain transcriptome, and these effects seemed to correlate with PM size. Many genes and pathways were affected after PM exposure. Among them, we found a strong activation in mRNA Nonsense Mediated Decay pathway, an inhibition in pathways related to transcription, neurogenesis and survival signaling as well as angiogenesis, and a dramatic downregulation of collagens. Although we did not detect any extracellular Aβ plaques, immunostaining revealed that both intracellular Aβ1-42 and phospho-Tau levels were increased in various PM exposure conditions compared to the clean air control. NanoString GeoMx analysis demonstrated a remarkable activation of immune responses in the PM exposed mouse brain. Surprisingly, our data also indicated a strong activation of various tumor suppressors including RB1, CDKN1A/p21 and CDKN2A/p16. Collectively, our data demonstrated that exposure to airborne PM caused a profound transcriptional dysregulation and accelerated Alzheimer's-related pathology.
Collapse
Affiliation(s)
- Liron L Israel
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Oliver Braubach
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Ekaterina S Shatalova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Oksana Chepurna
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Sachin Sharma
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Dmytro Klymyshyn
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Anna Galstyan
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Antonella Chiechi
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Alysia Cox
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - David Herman
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Bishop Bliss
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Irene Hasen
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Amanda Ting
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Rebecca Arechavala
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Michael T Kleinman
- Department of Environmental and Occupational Health, University of California, Irvine 92697, United States of America
| | - Rameshwar Patil
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Eggehard Holler
- Terasaki Institute, Los Angeles, CA 90024, United States of America
| | | | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America.
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America.
| |
Collapse
|
38
|
Wei L, Yang X, Wang J, Wang Z, Wang Q, Ding Y, Yu A. H3K18 lactylation of senescent microglia potentiates brain aging and Alzheimer's disease through the NFκB signaling pathway. J Neuroinflammation 2023; 20:208. [PMID: 37697347 PMCID: PMC10494370 DOI: 10.1186/s12974-023-02879-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023] Open
Abstract
Cellular senescence serves as a fundamental and underlying activity that drives the aging process, and it is intricately associated with numerous age-related diseases, including Alzheimer's disease (AD), a neurodegenerative aging-related disorder characterized by progressive cognitive impairment. Although increasing evidence suggests that senescent microglia play a role in the pathogenesis of AD, their exact role remains unclear. In this study, we quantified the levels of lactic acid in senescent microglia, and hippocampus tissues of naturally aged mice and AD mice models (FAD4T and APP/PS1). We found lactic acid levels were significantly elevated in these cells and tissues compared to their corresponding counterparts, which increased the level of pan histone lysine lactylation (Kla). We aslo identified all histone Kla sites in senescent microglia, and found that both the H3K18 lactylation (H3K18la) and Pan-Kla were significantly up-regulated in senescent microglia and hippocampus tissues of naturally aged mice and AD modeling mice. We demonstrated that enhanced H3K18la directly stimulates the NFκB signaling pathway by increasing binding to the promoter of Rela (p65) and NFκB1(p50), thereby upregulating senescence-associated secretory phenotype (SASP) components IL-6 and IL-8. Our study provides novel insights into the physiological function of Kla and the epigenetic regulatory mechanism that regulates brain aging and AD. Specifically, we have identified the H3K18la/NFκB axis as a critical player in this process by modulating IL-6 and IL-8. Targeting this axis may be a potential therapeutic strategy for delaying aging and AD by blunting SASP.
Collapse
Affiliation(s)
- Lin Wei
- Department of Clinical Laboratory, Hunan Provincial People's Hospital, Central Laboratory of Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Department of Laboratory Medicine, Hubei University of Medicine, Taihe Hospital, The Affiliated Hospital of Hubei University of Medicine, Shiyan, 442000, China
| | - Xiaowen Yang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Department of Laboratory Medicine, Hubei University of Medicine, Taihe Hospital, The Affiliated Hospital of Hubei University of Medicine, Shiyan, 442000, China
| | - Jie Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Department of Laboratory Medicine, Hubei University of Medicine, Taihe Hospital, The Affiliated Hospital of Hubei University of Medicine, Shiyan, 442000, China
| | - Zhixiao Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Department of Laboratory Medicine, Hubei University of Medicine, Taihe Hospital, The Affiliated Hospital of Hubei University of Medicine, Shiyan, 442000, China
| | - Qiguang Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Department of Laboratory Medicine, Hubei University of Medicine, Taihe Hospital, The Affiliated Hospital of Hubei University of Medicine, Shiyan, 442000, China
| | - Yan Ding
- Hubei Key Laboratory of Embryonic Stem Cell Research, Department of Laboratory Medicine, Hubei University of Medicine, Taihe Hospital, The Affiliated Hospital of Hubei University of Medicine, Shiyan, 442000, China.
| | - Aiqing Yu
- Department of Clinical Laboratory, Hunan Provincial People's Hospital, Central Laboratory of Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha, 410000, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Department of Laboratory Medicine, Hubei University of Medicine, Taihe Hospital, The Affiliated Hospital of Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
39
|
Vila-Castelar C, Chen Y, Langella S, Lopera F, Zetterberg H, Hansson O, Dage JL, Janelidzde S, Su Y, Chen K, McDowell CP, Martinez JE, Ramirez-Gomez L, Garcia G, Aguillon D, Baena A, Giraldo-Chica M, Protas HD, Ghisays V, Rios-Romenets S, Tariot PN, Blennow K, Reiman EM, Quiroz YT. Sex differences in blood biomarkers and cognitive performance in individuals with autosomal dominant Alzheimer's disease. Alzheimers Dement 2023; 19:4127-4138. [PMID: 37279390 PMCID: PMC10527358 DOI: 10.1002/alz.13314] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/08/2023]
Abstract
INTRODUCTION Plasma tau phosphorylated at threonine 217 (P-tau217) and neurofilament light (NfL) have emerged as markers of Alzheimer's disease (AD) pathology. Few studies have examined the role of sex in plasma biomarkers in sporadic AD, yielding mixed findings, and none in autosomal dominant AD. METHODS We examined the effects of sex and age on plasma P-tau217 and NfL, and their association with cognitive performance in a cross-sectional study of 621 Presenilin-1 E280A mutation carriers (PSEN1) and non-carriers. RESULTS As plasma P-tau217 levels increase, cognitively unimpaired female carriers showed better cognitive performance than cognitively unimpaired male carriers. Yet, as disease progresses, female carriers had a greater plasma NfL increase than male carriers. There were no sex differences in the association between age and plasma biomarkers among non-carriers. DISCUSSION Our findings suggest that, among PSEN1 mutation carriers, females had a greater rate of neurodegeneration than males, yet it did not predict cognitive performance. HIGHLIGHTS We examined sex differences in plasma P-tau217 and NfL in Presenilin-1 E280A (PSEN1) mutation carriers and non-carriers. Female carriers had a greater plasma NfL increase, but not P-tau217, than male carriers. As plasma P-tau217 levels increase, cognitively unimpaired female carriers showed better cognitive performance than cognitively unimpaired male carriers. The interaction effect of sex by plasma NfL levels did not predict cognition among carriers.
Collapse
Affiliation(s)
- Clara Vila-Castelar
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Yinghua Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, 85718, USA
| | - Stephanie Langella
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 405 30, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 405 30, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Oskar Hansson
- Memory Clinic, Skåne University Hospital, Malmö, 214 28, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 205 02, Sweden
| | - Jeffrey L. Dage
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | | | - Yi Su
- Banner Alzheimer’s Institute, Phoenix, AZ, 85718, USA
| | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, 85718, USA
| | - Celina Pluim McDowell
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Department of Psychological and Brain Sciences, Boston University, Boston, 02215, MA
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA
| | - Jairo E. Martinez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Department of Psychological and Brain Sciences, Boston University, Boston, 02215, MA
| | | | - Gloria Garcia
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | - David Aguillon
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | - Ana Baena
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | | | | | | | - Silvia Rios-Romenets
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 405 30, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 405 30, Sweden
| | | | - Yakeel T. Quiroz
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| |
Collapse
|
40
|
Dennison J, Mendez A, Szeto A, Lohse I, Wahlestedt C, Volmar CH. Low-Dose Chidamide Treatment Displays Sex-Specific Differences in the 3xTg-AD Mouse. Biomolecules 2023; 13:1324. [PMID: 37759724 PMCID: PMC10526199 DOI: 10.3390/biom13091324] [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: 06/13/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Epigenetic compounds have become attractive small molecules for targeting the multifaceted aspects of Alzheimer's disease (AD). Although AD disproportionately affects women, most of the current literature investigating epigenetic compounds for the treatment of AD do not report sex-specific results. This is remarkable because there is rising evidence that epigenetic compounds intrinsically affect males and females differently. This manuscript explores the sexual dimorphism observed after chronic, low-dose administration of a clinically relevant histone deacetylase inhibitor, chidamide (Tucidinostat), in the 3xTg-AD mouse model. We found that chidamide treatment significantly improves glucose tolerance and increases expression of glucose transporters in the brain of males. We also report a decrease in total tau in chidamide-treated mice. Differentially expressed genes in chidamide-treated mice were much greater in males than females. Genes involved in the neuroinflammatory pathway and amyloid processing pathway were mostly upregulated in chidamide-treated males while downregulated in chidamide-treated females. This work highlights the need for drug discovery projects to consider sex as a biological variable to facilitate translation.
Collapse
Affiliation(s)
- Jessica Dennison
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.D.)
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Armando Mendez
- Diabetes Research Institute, Division of Endocrinology, Diabetes, and Metabolism, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Angela Szeto
- Diabetes Research Institute, Division of Endocrinology, Diabetes, and Metabolism, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ines Lohse
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.D.)
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Claes Wahlestedt
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.D.)
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Claude-Henry Volmar
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.D.)
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
41
|
Sims S, Barak O, Ryu V, Miyashita S, Kannangara H, Korkmaz F, Wizman S, Macdonald A, Gumerova A, Goosens K, Zaidi M, Yuen T, Lizneva D, Frolinger T. Absent LH signaling rescues the anxiety phenotype in aging female mice. Mol Psychiatry 2023; 28:3324-3331. [PMID: 37563278 DOI: 10.1038/s41380-023-02209-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023]
Abstract
Clinical studies and experimental data together support a role for pituitary gonadotropins, including luteinizing hormone (LH), otherwise considered solely as fertility hormones, in age-related cognitive decline. Furthermore, rising levels of LH in post-menopausal women have been implicated in the high prevalence of mood disorders. This study was designed to examine the effect of deficient LH signaling on both cognitive and emotional behavior in 12-month-old Lhcgr-/- mice. For this, we established and validated a battery of five tests, including Dark-Light Box (DLB), Y-Maze Spontaneous Alternation, Novel Object Recognition (NOR), and contextual and cued Fear Conditioning (FCT) tests. We found that 12-month-old female wild type mice display a prominent anxiety phenotype on DLB and FCT. This phenotype was not seen in 12-month-old female Lhcgr-/- mice, indicating full phenotypic rescue. Furthermore, there was no effect of LHCGR depletion on recognition memory or working spatial memory on NOR and Y-maze testing, respectively, in 12-month-old mice, notwithstanding the absence of a basal phenotype in wild type littermates. The latter data do not exclude an effect of LH on cognition documented in previous studies. Finally, 12-month-old male mice and 3-month-old male and female mice did not consistently display deficits on any test. The data collectively document, for the first time, that loss of LH signaling reverses age-related emotional disturbances, a prelude to future targeted therapies that block LH action.
Collapse
Affiliation(s)
- Steven Sims
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Orly Barak
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vitaly Ryu
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sari Miyashita
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hasni Kannangara
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Soleil Wizman
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anne Macdonald
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anisa Gumerova
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ki Goosens
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Tal Frolinger
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
42
|
Fernández-Moncada I, Eraso-Pichot A, Tor TD, Fortunato-Marsol B, Marsicano G. An enquiry to the role of CB1 receptors in neurodegeneration. Neurobiol Dis 2023:106235. [PMID: 37481040 DOI: 10.1016/j.nbd.2023.106235] [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: 04/03/2023] [Revised: 06/27/2023] [Accepted: 07/20/2023] [Indexed: 07/24/2023] Open
Abstract
Neurodegenerative disorders are debilitating conditions that impair patient quality of life and that represent heavy social-economic burdens to society. Whereas the root of some of these brain illnesses lies in autosomal inheritance, the origin of most of these neuropathologies is scantly understood. Similarly, the cellular and molecular substrates explaining the progressive loss of brain functions remains to be fully described too. Indeed, the study of brain neurodegeneration has resulted in a complex picture, composed of a myriad of altered processes that include broken brain bioenergetics, widespread neuroinflammation and aberrant activity of signaling pathways. In this context, several lines of research have shown that the endocannabinoid system (ECS) and its main signaling hub, the type-1 cannabinoid (CB1) receptor are altered in diverse neurodegenerative disorders. However, some of these data are conflictive or poorly described. In this review, we summarize the findings about the alterations in ECS and CB1 receptors signaling in three representative brain illnesses, the Alzheimer's, Parkinson's and Huntington's diseases, and we discuss the relevance of these studies in understanding neurodegeneration development and progression, with a special focus on astrocyte function. Noteworthy, the analysis of ECS defects in neurodegeneration warrant much more studies, as our conceptual understanding of ECS function has evolved quickly in the last years, which now include glia cells and the subcellular-specific CB1 receptors signaling as critical players of brain functions.
Collapse
Affiliation(s)
| | - Abel Eraso-Pichot
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Tommaso Dalla Tor
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France; Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania 95124, Italy
| | | | - Giovanni Marsicano
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France.
| |
Collapse
|
43
|
Barreto GE. Repurposing of Tibolone in Alzheimer's Disease. Biomolecules 2023; 13:1115. [PMID: 37509151 PMCID: PMC10377087 DOI: 10.3390/biom13071115] [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: 06/15/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disease characterised by the accumulation of amyloid-beta and tau in the brain, leading to the progressive loss of memory and cognition. The causes of its pathogenesis are still not fully understood, but some risk factors, such as age, genetics, and hormones, may play a crucial role. Studies show that postmenopausal women have a higher risk of developing AD, possibly due to the decrease in hormone levels, especially oestrogen, which may be directly related to a reduction in the activity of oestrogen receptors, especially beta (ERβ), which favours a more hostile cellular environment, leading to mitochondrial dysfunction, mainly affecting key processes related to transport, metabolism, and oxidative phosphorylation. Given the influence of hormones on biological processes at the mitochondrial level, hormone therapies are of clinical interest to reduce the risk or delay the onset of symptoms associated with AD. One drug with such potential is tibolone, which is used in clinics to treat menopause-related symptoms. It can reduce amyloid burden and have benefits on mitochondrial integrity and dynamics. Many of its protective effects are mediated through steroid receptors and may also be related to neuroglobin, whose elevated levels have been shown to protect against neurological diseases. Its importance has increased exponentially due to its implication in the pathogenesis of AD. In this review, we discuss recent advances in tibolone, focusing on its mitochondrial-protective effects, and highlight how valuable this compound could be as a therapeutic alternative to mitigate the molecular pathways characteristic of AD.
Collapse
Affiliation(s)
- George E Barreto
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland
| |
Collapse
|
44
|
Digma LA, Litvan I, Del Ser T, Bayram E. Sex differences for cognitive decline in progressive supranuclear palsy. Parkinsonism Relat Disord 2023; 112:105454. [PMID: 37301014 PMCID: PMC10911684 DOI: 10.1016/j.parkreldis.2023.105454] [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: 02/17/2023] [Revised: 04/16/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023]
Abstract
INTRODUCTION Cognitive dysfunction is a core clinical feature of progressive supranuclear palsy (PSP), with executive function being most markedly affected. In other neurodegenerative conditions, such as Alzheimer's and Parkinson's diseases, there are a growing number of reports demonstrating that cognition is differentially impacted in men and women. In PSP, however, the sex differences in cognitive decline have yet to be fully characterized. METHODS Data were obtained from the TAUROS trial for 139 participants with mild-to-moderate PSP (62 women, 77 men). Sex differences in longitudinal change in cognitive performance were evaluated with linear mixed models. Exploratory subgroup analyses assessed whether sex differences varied by baseline executive dysfunction, PSP phenotype, or baseline age. RESULTS In the primary whole group analyses, there were no sex differences for change in cognitive performance. Among participants with normal executive function at baseline, men declined more severely on executive function and language tests. Among the PSP-Parkinsonism subgroup, men declined more severely on category fluency. Across people aged≥65, men had a worse decline on category fluency, whereas across people aged <65, women had a worse decline on DRS construction. CONCLUSION In people with mild-to-moderate PSP, there are no sex differences in cognitive decline. However, the rate of cognitive decline may differ for women and men based on the level of baseline executive dysfunction, PSP-phenotype and age. Further studies are needed to clarify how sex differences in PSP clinical progression vary by disease stage and to examine the contributions of co-pathology to these observed sex differences.
Collapse
Affiliation(s)
- Leonardino A Digma
- Parkinson and Other Movement Disorder Center, Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Irene Litvan
- Parkinson and Other Movement Disorder Center, Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Teodoro Del Ser
- Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, C. de Valderrebollo, 5, 28031, Madrid, Spain.
| | - Ece Bayram
- Parkinson and Other Movement Disorder Center, Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
45
|
Elsherbini A, Zhu Z, Quadri Z, Crivelli SM, Ren X, Vekaria HJ, Tripathi P, Zhang L, Zhi W, Bieberich E. Novel Isolation Method Reveals Sex-Specific Composition and Neurotoxicity of Small Extracellular Vesicles in a Mouse Model of Alzheimer's Disease. Cells 2023; 12:1623. [PMID: 37371093 PMCID: PMC10297289 DOI: 10.3390/cells12121623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
We developed a new method to isolate small extracellular vesicles (sEVs) from male and female wild-type and 5xFAD mouse brains to investigate the sex-specific functions of sEVs in Alzheimer's disease (AD). A mass spectrometric analysis revealed that sEVs contained proteins critical for EV formation and Aβ. ExoView analysis showed that female mice contained more GFAP and Aβ-labeled sEVs, suggesting that a larger proportion of sEVs from the female brain is derived from astrocytes and/or more likely to bind to Aβ. Moreover, sEVs from female brains had more acid sphingomyelinase (ASM) and ceramide, an enzyme and its sphingolipid product important for EV formation and Aβ binding to EVs, respectively. We confirmed the function of ASM in EV formation and Aβ binding using co-labeling and proximity ligation assays, showing that ASM inhibitors prevented complex formation between Aβ and ceramide in primary cultured astrocytes. Finally, our study demonstrated that sEVs from female 5xFAD mice were more neurotoxic than those from males, as determined by impaired mitochondrial function (Seahorse assays) and LDH cytotoxicity assays. Our study suggests that sex-specific sEVs are functionally distinct markers for AD and that ASM is a potential target for AD therapy.
Collapse
Affiliation(s)
- Ahmed Elsherbini
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Zhihui Zhu
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Zainuddin Quadri
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Simone M. Crivelli
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Xiaojia Ren
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA;
- Veterans Affairs Medical Center, Lexington, KY 40502, USA
| | - Priyanka Tripathi
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Liping Zhang
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Wenbo Zhi
- Department of Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA 30912, USA;
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
- Veterans Affairs Medical Center, Lexington, KY 40502, USA
| |
Collapse
|
46
|
DiMarco GM, Harris BN, Savonenko AV, Soto PL. Acute stressors do not impair short-term memory or attention in an aged mouse model of amyloidosis. Front Behav Neurosci 2023; 17:1151833. [PMID: 37250187 PMCID: PMC10213425 DOI: 10.3389/fnbeh.2023.1151833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Memory impairment in Alzheimer's disease patients is thought to be associated with the accumulation of amyloid-beta peptides and tau proteins. However, inconsistent reports of cognitive deficits in pre-clinical studies have raised questions about the link between amyloid-beta and cognitive decline. One possible explanation may be that studies reporting memory deficits often involve behavioral assessments that entail a high stress component. In contrast, in tasks without a high stress component transgenic mice do not consistently show declines in memory. The glucocorticoid cascade hypothesis of aging and the vicious cycle of stress framework suggest that stress exacerbates dementia progression by initiating a cycle of hypothalamic-pituitary-adrenal axis activation and subsequent brain deterioration. Using the APPswe/PS1dE9 mouse model of amyloidosis, we assessed whether stressor exposure prior to testing differentially impaired cognitive performance of aged male and female mice. As part of a larger study, mice performed a delayed match-to-position (DMTP) or a 3-choice serial-reaction time (3CSRT) task. Unexpectedly, these mice did not exhibit cognitive declines during aging. Therefore, at 73 and 74 weeks of age, we exposed mice to a predator odor or forced swim stressor prior to testing to determine if stress revealed cognitive deficits. We predicted stressor exposure would decrease performance accuracy more robustly in transgenic vs. non-transgenic mice. Acute stressor exposure increased accuracy in the DMTP task, but not in the 3CSRT task. Our data suggest that acute stressor exposure prior to testing does not impair cognitive performance in APPswe/PS1dE9 mice.
Collapse
Affiliation(s)
- Giuliana M. DiMarco
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
- Center for Molecular and Behavioral Neuroscience, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Breanna N. Harris
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Alena V. Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Paul L. Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
47
|
Grover L, Sklioutovskaya-Lopez K, Parkman JK, Wang K, Hendricks E, Adams-Duffield J, Kim JH. Diet, sex, and genetic predisposition to obesity and type 2 diabetes modulate motor and anxiety-related behaviors in mice, and alter cerebellar gene expression. Behav Brain Res 2023; 445:114376. [PMID: 36868363 PMCID: PMC10065959 DOI: 10.1016/j.bbr.2023.114376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/28/2022] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Obesity and type 2 diabetes (T2D) are serious health problems linked to neurobehavioral alterations. We compared motor function, anxiety-related behavior, and cerebellar gene expression in TALLYHO/Jng (TH), a polygenic model prone to insulin resistance, obesity, and T2D, and normal C57BL/6 J (B6) mice. Male and female mice were weaned onto chow or high fat (HF) diet at 4 weeks of age (wk), and experiments conducted at young (5 wk) and old (14 - 20 wk) ages. In the open field, distance traveled was significantly lower in TH (vs. B6). For old mice, anxiety-like behavior (time in edge zone) was significantly increased for TH (vs B6), females (vs males), and for both ages HF diet (vs chow). In Rota-Rod testing, latency to fall was significantly shorter in TH (vs B6). For young mice, longer latencies to fall were observed for females (vs males) and HF (vs chow). Grip strength in young mice was greater in TH (vs B6), and there was a diet-strain interaction, with TH on HF showing increased strength, whereas B6 on HF showed decreased strength. For older mice, there was a strain-sex interaction, with B6 males (but not TH males) showing increased strength compared to the same strain females. There were significant sex differences in cerebellar mRNA levels, with Tnfα higher, and Glut4 and Irs2 lower in females (vs males). There were significant strain effects for Gfap and Igf1 mRNA levels with lower in TH (vs B6). Altered cerebellar gene expression may contribute to strain differences in coordination and locomotion.
Collapse
Affiliation(s)
- Lawrence Grover
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | | | - Jacaline K Parkman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Katherine Wang
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Emily Hendricks
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jessica Adams-Duffield
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jung Han Kim
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
48
|
Anand DV, Chung MK. Hodge Laplacian of Brain Networks. IEEE TRANSACTIONS ON MEDICAL IMAGING 2023; 42:1563-1573. [PMID: 37018280 PMCID: PMC10909176 DOI: 10.1109/tmi.2022.3233876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The closed loops or cycles in a brain network embeds higher order signal transmission paths, which provide fundamental insights into the functioning of the brain. In this work, we propose an efficient algorithm for systematic identification and modeling of cycles using persistent homology and the Hodge Laplacian. Various statistical inference procedures on cycles are developed. We validate the our methods on simulations and apply to brain networks obtained through the resting state functional magnetic resonance imaging. The computer codes for the Hodge Laplacian are given in https://github.com/laplcebeltrami/hodge.
Collapse
|
49
|
Sha S, Chaigneau T, Krantic S. Pre-symptomatic synaptic dysfunction and longitudinal decay of hippocampal synaptic function in APPPS1 mouse model of Alzheimer's disease is sex-independent. Brain Res Bull 2023; 198:36-49. [PMID: 37080395 DOI: 10.1016/j.brainresbull.2023.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/19/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Alzheimer's disease (AD) is an incurable, age-related and progressive neurodegenerative disease characterized by cognitive impairments. Deficits in synaptic plasticity were reported in various models of AD-like pathology and are considered as an early contributing factor of cognitive impairment. However, the majority of previous studies were focused on overt, symptomatic stages of pathology and assessed long-term potentiation (LTP), whereas long-term depression (LTD) was much less investigated and the precise nature of its involvement remains poorly defined. To better understand the earliest synaptic dysfunctions along the pre-symptomatic stage of AD-like pathology, we performed a detailed analysis of underlying mechanisms and quantified basal synaptic activity, presynaptic release probability, and synaptic plasticity such as post-tetanic potentiation (PTP), as well as LTP and LTD. These parameters were studied in APPPS1 mouse model at two time points (early- and mid-) along the pre-symptomatic stage, which were compared with alterations monitored at two later time-points, i.e. the onset of cognitive deficits and the overt stage of full-blown pathology. Because sex is known to be an instrumental biological parameter in AD pathophysiology, all alterations were assessed in both males and females. Our data show that, as compared to wild-type (WT) littermates, initial neuronal hyperexcitability, seen at early pre-symptomatic stage shifts subsequently towards hypoexcitability at mid-pre-symptomatic stage and remains impaired at advanced stages. The pre-symptomatic changes also involve increased synaptic plasticity as assessed by paired-pulse facilitation (PPF), which returns to basal level at the onset of pathology and remains stable afterwards. Synaptic plasticity is impaired by mid-pre-symptomatic stage and manifests as lowered LTP and absence of LTD induction, the latter being reported here for the first time. Observed LTP and LTD impairments both persist in older APPPS1 mice. Remarkably, none of the observed differences was gender-dependent. Altogether, our data evidence that major impairments in basal synaptic efficacy and plasticity are detectable already during mid-pre-symptomatic stage of AD-like pathogenesis and likely involve hyperexcitability as the underlying mechanism. Our study also uncovers synaptic alterations that may become critical read-outs for testing the efficiency of novel, pre-symptomatic stage-targeted therapies for AD.
Collapse
Affiliation(s)
- Sha Sha
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, F-75012 Paris, France; Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Thomas Chaigneau
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Slavica Krantic
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, F-75012 Paris, France.
| |
Collapse
|
50
|
Noma T, Kayo G, Kabayama M, Gondo Y, Yasumoto S, Masui Y, Sugimoto K, Akasaka H, Takami Y, Takeya Y, Yamamoto K, Ikebe K, Arai Y, Ishizaki T, Rakugi H, Kamide K. Lower cognitive function as a risk factor for anemia among older Japanese women from the longitudinal observation in the SONIC study. Geriatr Gerontol Int 2023; 23:334-340. [PMID: 36958816 DOI: 10.1111/ggi.14571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/29/2023] [Accepted: 02/24/2023] [Indexed: 03/25/2023]
Abstract
AIM The aging-related increase in the incidence of anemia potentially affects the mortality risk. Lower cognitive function is common among older adults, and anemia is one of the causes of cognitive decline. However, to the best of our knowledge, no study has investigated whether cognitive decline is a risk factor for anemia in older people. Therefore, in this study, we used a 3-year longitudinal evaluation to examine the association of cognitive function with anemia in community-dwelling older adults. METHODS This longitudinal study enrolled participants without anemia (diagnosed based on the World Health Organization's criteria) at baseline. Cognitive function was assessed using the Japanese version of the Montreal Cognitive Assessment. Multiple logistic regression models were used to examine the association between cognitive function at baseline and the presence of anemia 3 years later. RESULTS Participants were in the 69-71 and 79-81 years age groups, and 974 older people (48.6% men) were enrolled, of whom 126 (12.9%) had anemia after 3 years. After adjusting, cognitive function at baseline was associated with anemia in women, but not in men. CONCLUSIONS Older Japanese women with lower cognitive function have an increased risk for anemia 3 years later. The adoption of a lifestyle that utilizes or improves cognitive function might be important to prevent anemia in older women. Geriatr Gerontol Int 2023; ••: ••-••.
Collapse
Affiliation(s)
- Tomoko Noma
- Department of Nutrition, College of Nutrition, Koshien University, Takarazuka, Japan
- Department of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Godai Kayo
- Department of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mai Kabayama
- Department of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yasuyuki Gondo
- Department of Clinical Thanatology and Geriatric Behavioral Science, Graduate School of Human Sciences, Osaka University, Osaka, Japan
| | - Saori Yasumoto
- Office of International Exchange, Graduate School of Human Sciences, Osaka University, Osaka, Japan
| | - Yukie Masui
- Research Team for Human Care, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Ken Sugimoto
- Department of General Geriatric Medicine, Kawasaki Medical School, Okayama, Japan
| | - Hiroshi Akasaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoichi Takami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasushi Takeya
- Department of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazunori Ikebe
- Department of Prosthodontics, Gerodontology and Oral Rehabilitation, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yasumichi Arai
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuro Ishizaki
- Research Team for Human Care, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kei Kamide
- Department of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|