1
|
Abu Raya M, Zeltzer E, Blazhenets G, Schonhaut DR, Allen IE, Carrillo MC, Gatsonis C, Hanna L, Hillner BE, Iaccarino L, March A, Mundada NS, Perez JM, Smith K, Yballa C, Siegel BA, Windon C, Whitmer RA, Kaitlin C, La Joie R, Rabinovici GD. Sex differences in amyloid PET in a large, real-world sample from the Imaging Dementia-Evidence for Amyloid Scanning (IDEAS) Study. Alzheimers Dement 2025; 21:e70304. [PMID: 40415175 DOI: 10.1002/alz.70304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/27/2025]
Abstract
INTRODUCTION We examined sex effects on amyloid positron emission tomography (PET) in a large cohort of patients evaluated for cognitive complaints in a "real-world" specialty setting. METHODS We analyzed 10,361 amyloid PET scans (51% females) from the Imaging Dementia-Evidence for Amyloid Scanning Study. Amyloid positivity was defined by either local visual read or central PET processing and quantification (≥ 24.4 Centiloids). Sex differences were examined using multilinear regression and logistic regression adjusted for age, comorbidities, and other demographic and clinical covariates. RESULTS Females had higher rates of positive amyloid PET visual reads (63% vs. 59%, P < 0.001) and higher Centiloids (CLs; median 48.7 vs. 36.8, p < 0.001). On logistic regression, females had higher odds ratios (ORs) for positive amyloid PET (visual read OR 1.20, 95% confidence interval [CI]: 1.11-1.31; CL threshold-based OR 1.37, 95% CI: 1.26-1.49; both p < 0.001). DISCUSSION Females with cognitive impairment showed higher amyloid PET positivity and greater amyloid burden. Further research is needed to explore mechanisms and treatment implications. HIGHLIGHTS Females exhibited higher rates of amyloid positron emission tomography (PET) positivity and higher amyloid burden than males. These sex effects were found in patients with both mild cognitive impairment (MCI) and dementia. Females also had higher rates of dementia and amnestic MCI, while males had higher rates of non-amnestic MCI and more cholinesterase inhibitor use.
Collapse
Affiliation(s)
- Maison Abu Raya
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, California, USA
| | - Ehud Zeltzer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Ganna Blazhenets
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Daniel R Schonhaut
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Isabel Elaine Allen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, California, USA
| | - Maria C Carrillo
- Medical & Scientific Relations Division, Alzheimer's Association, Chicago, Illinois, USA
| | | | - Lucy Hanna
- Center for Statistical Sciences, Brown University, Providence, Rhode Island, USA
| | - Bruce E Hillner
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Andrew March
- Center for Research and Innovation, American College of Radiology, Reston, Virginia, USA
| | - Nidhi S Mundada
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Jhony Mejia Perez
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Karen Smith
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Claire Yballa
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Barry A Siegel
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Charles Windon
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, California, USA
| | - Rachel A Whitmer
- Division of Research, Kaiser Permanente, Division of Research, Pleasanton, California, USA
- Department of Public Health Sciences, University of California Davis, Davis, California, USA
| | - Casaletto Kaitlin
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, California, USA
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
2
|
Lee BH, Eid RS, Hodges TE, Barth C, Galea LAM. Leveraging research into sex differences and steroid hormones to improve brain health. Nat Rev Endocrinol 2025; 21:214-229. [PMID: 39587332 DOI: 10.1038/s41574-024-01061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
Sex differences, driven in part by steroid hormones, shape the structure and function of the brain throughout the lifespan and manifest across brain health and disease. The influence of steroid hormones on neuroplasticity, particularly in the adult hippocampus, differs between the sexes, which has important implications for disorders and diseases that compromise hippocampus integrity, such as depression and Alzheimer disease. This Review outlines the intricate relationship between steroid hormones and hippocampal neuroplasticity across the adult lifespan and explores how the unique physiology of male and female individuals can affect health and disease. Despite calls to include sex and gender in research, only 5% of neuroscience studies published in 2019 directly investigated the influence of sex. Drawing on insights from depression, Alzheimer disease and relevant hippocampal plasticity, this Review underscores the importance of considering sex and steroid hormones to achieve a comprehensive understanding of disease susceptibility and mechanisms. Such consideration will enable the discovery of personalized treatments, ultimately leading to improved health outcomes for all.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Rand S Eid
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Travis E Hodges
- Department of Psychology and Education, Mount Holyoke College, South Hadley, MA, USA
| | - Claudia Barth
- Division for Mental Health and Substance Abuse, Diakonhjemmet Hospital, Oslo, Norway
| | - Liisa A M Galea
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Nerattini M, Williams S, Andy C, Carlton C, Zarate C, Boneu C, Fauci F, Ajila T, Jett S, Battista M, Pahlajani S, Berti V, Andrews R, Matthews DC, Dyke JP, Brinton RD, Mosconi L. Sex-specific associations of serum testosterone with gray matter volume and cerebral blood flow in midlife individuals at risk for Alzheimer's disease. PLoS One 2025; 20:e0317303. [PMID: 39804890 PMCID: PMC11729972 DOI: 10.1371/journal.pone.0317303] [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: 06/04/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
Testosterone, an essential sex steroid hormone, influences brain health by impacting neurophysiology and neuropathology throughout the lifespan in both genders. However, human research in this area is limited, particularly in women. This study examines the associations between testosterone levels, gray matter volume (GMV) and cerebral blood flow (CBF) in midlife individuals at risk for Alzheimer's disease (AD), according to sex and menopausal status. A cohort of 294 cognitively normal midlife participants, 83% female, ages 35-65 years, with an AD family history and/or Apolipoprotein E epsilon 4 (APOE-4) genotype, underwent volumetric Magnetic Resonance Imaging (MRI) to measure GMV and MR-Arterial Spin Labeling (ASL) for measurement of CBF. We used voxel-based analysis and volumes of interest to test for associations between testosterone (both total and free testosterone) and brain imaging outcomes, stratified by sex and menopausal status. Higher total and free testosterone levels were associated with larger GMV in men, with peak effects in frontal and temporal regions. Conversely, in women, higher testosterone levels correlated with higher CBF, with peak effects in frontal and limbic regions, subcortical areas and hypothalamus. Among women, associations between testosterone and GMV were observed at the premenopausal and perimenopausal stages, but not postmenopause, whereas associations of testosterone with CBF were significant starting at the perimenopausal stage and were more pronounced among hormone therapy non-users. Results were independent of age, APOE-4 status, midlife health indicators, and sex hormone-binding globulin levels. These findings indicate sex-specific neurophysiological effects of testosterone in AD-vulnerable regions in midlife individuals at risk for AD, with variations observed across sex and menopausal status. This underscores the need for further research focusing on the neuroprotective potential of testosterone in both sexes.
Collapse
Affiliation(s)
- Matilde Nerattini
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
- Department of Experimental and Clinical Biomedical Sciences, Nuclear Medicine Unit, University of Florence, Florence, Italy
| | - Schantel Williams
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States of America
| | - Caroline Carlton
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
| | - Camila Zarate
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
| | - Camila Boneu
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
| | - Francesca Fauci
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
| | - Trisha Ajila
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
| | - Steven Jett
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
| | - Michael Battista
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States of America
| | - Valentina Berti
- Department of Experimental and Clinical Biomedical Sciences, Nuclear Medicine Unit, University of Florence, Florence, Italy
| | | | | | - Jonathan P. Dyke
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States of America
| | - Roberta Diaz Brinton
- Department of Neurology and Pharmacology, University of Arizona, Tucson, AZ, United States of America
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States of America
| |
Collapse
|
4
|
Park J, Byun MS, Yi D, Ahn H, Jung JH, Kong N, Chang YY, Jung G, Lee JY, Kim YK, Lee YS, Kang KM, Sohn CH, Lee DY, for the KBASE Research Group. The Moderating Effect of Serum Vitamin D on the Relationship between Beta-amyloid Deposition and Neurodegeneration. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2024; 22:646-654. [PMID: 39420611 PMCID: PMC11494430 DOI: 10.9758/cpn.24.1189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 10/19/2024]
Abstract
Objective Previous studies have reported that vitamin D deficiency increased the risk of Alzheimer's disease (AD) dementia in older adults. However, little is known about how vitamin D is involved in the pathophysiology of AD. Thus, this study aimed to examine the association and interaction of serum vitamin D levels with in vivo AD pathologies including cerebral beta-amyloid (Aβ) deposition and neurodegeneration in nondemented older adults. Methods 428 Nondemented older adults were recruited from the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease, a prospective cohort that began in 2014. All participants underwent comprehensive clinical assessments, measurement of serum 25-hydroxyvitamin D (25[OH]D), and multimodal brain imaging including Pittsburgh compound B (PiB) positron emission tomography and magnetic resonance imaging. Global PiB deposition was measured for the Aβ biomarker. Intracranial volume-adjusted hippocampal volume (HVa) was used as a neurodegeneration biomarker. Results Overall, serum 25(OH)D level was not associated with either Aβ deposition or HVa after controlling for age, sex, apolipoprotein E ε4 positivity, and vascular risk factors. However, serum 25(OH)D level had a significant moderating effect on the association between Aβ and neurodegeneration, with lower serum 25(OH)D level significantly exacerbating cerebral Aβ-associated hippocampal volume loss (B = 34.612, p = 0.008). Conclusion Our findings indicate that lower serum vitamin D levels may contribute to AD by exacerbating Aβ-associated neurodegeneration in nondemented older adults. Further studies to explore the potential therapeutic effect of vitamin D supplementation on the progression of AD pathology will be necessary.
Collapse
Affiliation(s)
- Junha Park
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Min Soo Byun
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Science, College of Humanities, Seoul National University, Seoul, Korea
| | - Joon Hyung Jung
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, Korea
| | - Nayeong Kong
- Department of Psychiatry, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Yoon Young Chang
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Psychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Jun-Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - Dong Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, Korea
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
- Interdisciplinary Program of Cognitive Science, College of Humanities, Seoul National University, Seoul, Korea
| | | |
Collapse
|
5
|
Espinoza MT, Blümel JE, Chedraui P, Vallejo MS, Ñañez M, Ojeda E, Rey C, Rodríguez D, Rodrigues MA, Salinas C, Tserotas K, Calle A, Dextre M, Elizalde A, Escalante C, Gómez-Tabares G, Monterrosa-Castro Á. Association between type of menopause and mild cognitive impairment: The REDLINC XII study. Maturitas 2024; 189:108110. [PMID: 39226623 DOI: 10.1016/j.maturitas.2024.108110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/13/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE To evaluate the association between type of menopause (spontaneous or surgical) and mild cognitive impairment (MCI). STUDY DESIGN This study was a cross-sectional, observational, and sub-analytical investigation conducted within gynecological consultations across nine Latin American countries. METHOD We assessed sociodemographic, clinical, and anthropometric data, family history of dementia, and the presence of MCI using the Montreal Cognitive Assessment (MoCA) tool. RESULTS The study involved 1185 postmenopausal women with a mean age of 55.3 years and a body mass index of 26.4 kg/m2. They had an average of 13.3 years of education, and 37 % were homemakers. Three hundred ninety-nine experienced menopause before 40, including 136 with surgical menopause (bilateral oophorectomy). Out of the 786 women who experienced menopause at 40 or more years, 110 did so due to bilateral oophorectomy. There were no differences in MoCA scores among women who experienced menopause before or after the age of 40. However, lower MoCA scores were observed in women with surgical menopause than in those with spontaneous menopause (23.8 ± 4.9 vs. 25.0 ± 4.3 points, respectively, p < 0.001). Our logistic regression model with clustering of patients within countries found a significant association between MCI and surgical menopause (OR 1.47, 95 % CI: 1.01-2.16), use (ever) of menopausal hormone therapy (OR 0.33, 95 % CI: 0.21-0.50), and having >12 years of education (OR 0.21, 95 % CI: 0.14-0.30). CONCLUSION When comparing women who experience spontaneous menopause over the age of 40 with those who undergo it before this age, there was no observed increased risk of developing MCI, while those with surgical menopause, independent of age, are more prone to cognitive decline. Women who have ever used menopausal hormone therapy have a lower MCI risk. Further research is warranted to delve deeper into this topic.
Collapse
Affiliation(s)
- María T Espinoza
- Unidad de Ginecología Obstétrica, Clínica Los Ángeles, Cochabamba, Bolivia
| | - Juan E Blümel
- Medicina Interna Sur, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Peter Chedraui
- Escuela de Posgrado en Salud, Universidad Espíritu Santo, Samborondón, Ecuador
| | - María S Vallejo
- Obstetricia y Ginecología, Hospital Clínico, Universidad de Chile, Santiago, Chile
| | - Mónica Ñañez
- II Cátedra de Ginecología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Eliana Ojeda
- Departamento Académico de Medicina Humana, Universidad Andina del Cusco, Cusco, Peru
| | - Claudia Rey
- Asociación Argentina para el Estudio del Climaterio, Buenos Aires, Argentina
| | - Doris Rodríguez
- Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marcio A Rodrigues
- Department of Gynecology and Obstetrics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carlos Salinas
- Servicio de Obstetricia y Ginecología, Hospital Ángeles, Puebla, Mexico
| | | | - Andrés Calle
- Centro Integral de Salud Obstétrica y Femenina. Ginecología, Universidad Indoamérica, Academia Ecuatoriana de Medicina, Quito, Ecuador
| | - Maribel Dextre
- Ginecología Obstetricia, Clínica Internacional - Clínica Javier Prado, Lima, Peru
| | - Alejandra Elizalde
- Departamento de la Mujer, Niñez y Adolescencia, Facultad de Medicina de la Universidad Nacional del Nordeste, Corrientes, Argentina
| | - Carlos Escalante
- Departamento de Ginecología, Facultad de Medicina, Universidad de Costa Rica, Costa Rica
| | - Gustavo Gómez-Tabares
- Departamento de Ginecología, Escuela de Medicina, Facultad de Salud, Universidad del Valle, Cali, Colombia.
| | | |
Collapse
|
6
|
Kolahchi Z, Henkel N, Eladawi MA, Villarreal EC, Kandimalla P, Lundh A, McCullumsmith RE, Cuevas E. Sex and Gender Differences in Alzheimer's Disease: Genetic, Hormonal, and Inflammation Impacts. Int J Mol Sci 2024; 25:8485. [PMID: 39126053 PMCID: PMC11313277 DOI: 10.3390/ijms25158485] [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: 04/16/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Two-thirds of Americans with Alzheimer's disease are women, indicating a profound variance between the sexes. Variances exist between the sexes in the age and intensity of the presentation, cognitive deficits, neuroinflammatory factors, structural and functional brain changes, as well as psychosocial and cultural circumstances. Herein, we summarize the existing evidence for sexual dimorphism and present the available evidence for these distinctions. Understanding these complexities is critical to developing personalized interventions for the prevention, care, and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Zahra Kolahchi
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA; (Z.K.); (E.C.V.)
| | - Nicholas Henkel
- Department of Neurosciences and Neurological Disorders, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (N.H.); (M.A.E.); (P.K.); (A.L.); (R.E.M.)
| | - Mahmoud A. Eladawi
- Department of Neurosciences and Neurological Disorders, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (N.H.); (M.A.E.); (P.K.); (A.L.); (R.E.M.)
| | - Emma C. Villarreal
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA; (Z.K.); (E.C.V.)
| | - Prathik Kandimalla
- Department of Neurosciences and Neurological Disorders, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (N.H.); (M.A.E.); (P.K.); (A.L.); (R.E.M.)
| | - Anna Lundh
- Department of Neurosciences and Neurological Disorders, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (N.H.); (M.A.E.); (P.K.); (A.L.); (R.E.M.)
| | - Robert E. McCullumsmith
- Department of Neurosciences and Neurological Disorders, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (N.H.); (M.A.E.); (P.K.); (A.L.); (R.E.M.)
- ProMedica Neurosciences Center, Toledo, OH 43606, USA
| | - Elvis Cuevas
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA; (Z.K.); (E.C.V.)
| |
Collapse
|
7
|
Duggan MR, Gomez GT, Joynes CM, Bilgel M, Chen J, Fattorelli N, Hohman TJ, Mancuso R, Cordon J, Castellano T, Koran MEI, Candia J, Lewis A, Moghekar A, Ashton NJ, Kac PR, Karikari TK, Blennow K, Zetterberg H, Martinez-Muriana A, De Strooper B, Thambisetty M, Ferrucci L, Gottesman RF, Coresh J, Resnick SM, Walker KA. Proteome-wide analysis identifies plasma immune regulators of amyloid-beta progression. Brain Behav Immun 2024; 120:604-619. [PMID: 38977137 PMCID: PMC11682725 DOI: 10.1016/j.bbi.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/07/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024] Open
Abstract
While immune function is known to play a mechanistic role in Alzheimer's disease (AD), whether immune proteins in peripheral circulation influence the rate of amyloid-β (Aβ) progression - a central feature of AD - remains unknown. In the Baltimore Longitudinal Study of Aging, we quantified 942 immunological proteins in plasma and identified 32 (including CAT [catalase], CD36 [CD36 antigen], and KRT19 [keratin 19]) associated with rates of cortical Aβ accumulation measured with positron emission tomography (PET). Longitudinal changes in a subset of candidate proteins also predicted Aβ progression, and the mid- to late-life (20-year) trajectory of one protein, CAT, was associated with late-life Aβ-positive status in the Atherosclerosis Risk in Communities (ARIC) study. Genetic variation that influenced plasma levels of CAT, CD36 and KRT19 predicted rates of Aβ accumulation, including causal relationships with Aβ PET levels identified with two-sample Mendelian randomization. In addition to associations with tau PET and plasma AD biomarker changes, as well as expression patterns in human microglia subtypes and neurovascular cells in AD brain tissue, we showed that 31 % of candidate proteins were related to mid-life (20-year) or late-life (8-year) dementia risk in ARIC. Our findings reveal plasma proteins associated with longitudinal Aβ accumulation, and identify specific peripheral immune mediators that may contribute to the progression of AD pathophysiology.
Collapse
Affiliation(s)
- Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Gabriela T Gomez
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cassandra M Joynes
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicola Fattorelli
- VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders Laboratory, Center for Molecular Neurology, Flanders Institute for Biotechnology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jenifer Cordon
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Tonnar Castellano
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mary Ellen I Koran
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julián Candia
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Alexandria Lewis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK; NIHR Biomedical Research Center for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK; Center for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Przemysław R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; ICM Institute, Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France; First Affiliated Hospital, University of Science and Technology of China, Anhui, PR China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, University College London Institute of Neurology, London, UK; UK Dementia Research Institute, University College London, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong Special Administrative Region; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Martinez-Muriana
- VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium; UK Dementia Research Institute, University College London, London, UK
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rebecca F Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Josef Coresh
- Departments of Population Health and Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
8
|
Sobczuk J, Paczkowska K, Andrusiów S, Bolanowski M, Daroszewski J. Are Women with Polycystic Ovary Syndrome at Increased Risk of Alzheimer Disease? Lessons from Insulin Resistance, Tryptophan and Gonadotropin Disturbances and Their Link with Amyloid-Beta Aggregation. Biomolecules 2024; 14:918. [PMID: 39199306 PMCID: PMC11352735 DOI: 10.3390/biom14080918] [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: 06/29/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Alzheimer disease, the leading cause of dementia, and polycystic ovary syndrome, one of the most prevalent female endocrine disorders, appear to be unrelated conditions. However, studies show that both disease entities have common risk factors, and the amount of certain protein marker of neurodegeneration is increased in PCOS. Reports on the pathomechanism of both diseases point to the possibility of common denominators linking them. Dysregulation of the kynurenine pathway, insulin resistance, and impairment of the hypothalamic-pituitary-gonadal axis, which are correlated with amyloid-beta aggregation are these common areas. This article discusses the relationship between Alzheimer disease and polycystic ovary syndrome, with a particular focus on the role of disorders of tryptophan metabolism in both conditions. Based on a review of the available literature, we concluded that systemic changes occurring in PCOS influence the increased risk of neurodegeneration.
Collapse
Affiliation(s)
- Joachim Sobczuk
- Department of Endocrinology, Diabetes and Isotope Therapy, University Clinical Hospital, 50-367 Wroclaw, Poland
| | | | - Szymon Andrusiów
- Department of Neurology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Marek Bolanowski
- Department of Endocrinology, Diabetes and Isotope Therapy, University Clinical Hospital, 50-367 Wroclaw, Poland
- Department of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Jacek Daroszewski
- Department of Endocrinology, Diabetes and Isotope Therapy, University Clinical Hospital, 50-367 Wroclaw, Poland
- Department of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
9
|
Xu Q, Shen H, Zhu Y, Zhang J, Shen Z, Jiang J, Zhou J. Causal effects of genetically predicted testosterone on Alzheimer's disease: a two-sample mendelian randomization study. Acta Neurol Belg 2024; 124:591-601. [PMID: 38007406 DOI: 10.1007/s13760-023-02426-4] [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: 06/14/2023] [Accepted: 10/30/2023] [Indexed: 11/27/2023]
Abstract
OBJECTIVE Although several studies have reported that testosterone may protect against Alzheimer's disease, no evidence of a causal relationship has been demonstrated. METHODS A Mendelian randomization (MR) study was performed to determine the causal role of testosterone in Alzheimer's disease. The study utilized public databases obtained from separately published genome-wide associationstudies (GWAS). Single-nucleotide polymorphisms (SNPs) for testosterone were extracted from the most recent and largest published GWAS meta-analysis (178,782 participants), and SNPs for Alzheimer's disease were extracted from UK Biobank (954 AD cases and 487,331 controls). The odds ratio (OR) of the inverse variance weighting (IVW) approach was the primary outcome, and the weighted median and MR Egger regression were used for sensitivity analysis. RESULTS Through IVW, we observed a causal association between genetically predicted testosterone and the risk of Alzheimer's disease, with an OR of 0.99 (95% confidence interval [CI] = 0.998-0.999, p = 0.047). In the sensitivity analyses, the weighted median regression showed directionally similar estimates (OR = 0.99, 95% CI = 0.998-0.999, p = 0.048). The MR Egger regression showed similar estimates (OR = 0.99, 95% CI = 0.998-1.00, p = 0.35), but with lower precision. Funnel plots, MR Egger intercepts, and Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) analysis indicated the absence of directional pleiotropy effects. CONCLUSION In conclusion, our MR study provides evidence of a causal relationship between testosterone levels and Alzheimer's disease; however, this relationship must be validated in future studies with larger sample sizes. Early testosterone monitoring may enable the prevention of Alzheimer's and related diseases.
Collapse
Affiliation(s)
- Qian Xu
- Department of Neurology, Suzhou Wujiang District Hospital of Traditional Chinese Medicine (Suzhou Wujiang District Second People's Hospital), Wujiang District, Suzhou, 215200, China
| | - Hong Shen
- Department of Neurology, Suzhou Wujiang District Hospital of Traditional Chinese Medicine (Suzhou Wujiang District Second People's Hospital), Wujiang District, Suzhou, 215200, China
| | - Yifan Zhu
- Department of Neurology, Suzhou Wujiang District Hospital of Traditional Chinese Medicine (Suzhou Wujiang District Second People's Hospital), Wujiang District, Suzhou, 215200, China
| | - Junlei Zhang
- Department of Neurology, Suzhou Wujiang District Hospital of Traditional Chinese Medicine (Suzhou Wujiang District Second People's Hospital), Wujiang District, Suzhou, 215200, China
| | - Zhongmei Shen
- Department of Neurology, Suzhou Wujiang District Hospital of Traditional Chinese Medicine (Suzhou Wujiang District Second People's Hospital), Wujiang District, Suzhou, 215200, China
| | - Jianming Jiang
- Department of Neurology, Suzhou Wujiang District Hospital of Traditional Chinese Medicine (Suzhou Wujiang District Second People's Hospital), Wujiang District, Suzhou, 215200, China
| | - Jie Zhou
- Department of Neurology, Suzhou Wujiang District Hospital of Traditional Chinese Medicine (Suzhou Wujiang District Second People's Hospital), Wujiang District, Suzhou, 215200, China.
| |
Collapse
|
10
|
Majeed J, Sabbagh MN, Kang MH, Lawrence JJ, Pruitt K, Bacus S, Reyna E, Brown M, Decourt B. Cancer drugs with high repositioning potential for Alzheimer's disease. Expert Opin Emerg Drugs 2023; 28:311-332. [PMID: 38100555 PMCID: PMC10877737 DOI: 10.1080/14728214.2023.2296079] [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: 08/25/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Despite the recent full FDA approval of lecanemab, there is currently no disease modifying therapy (DMT) that can efficiently slow down the progression of Alzheimer's disease (AD) in the general population. This statement emphasizes the need to identify novel DMTs in the shortest time possible to prevent a global epidemic of AD cases as the world population experiences an increase in lifespan. AREAS COVERED Here, we review several classes of anti-cancer drugs that have been or are being investigated in Phase II/III clinical trials for AD, including immunomodulatory drugs, RXR agonists, sex hormone therapies, tyrosine kinase inhibitors, and monoclonal antibodies. EXPERT OPINION Given the overall course of brain pathologies during the progression of AD, we express a great enthusiasm for the repositioning of anti-cancer drugs as possible AD DMTs. We anticipate an increasing number of combinatorial therapy strategies to tackle AD symptoms and their underlying pathologies. However, we strongly encourage improvements in clinical trial study designs to better assess target engagement and possible efficacy over sufficient periods of drug exposure.
Collapse
Affiliation(s)
- Jad Majeed
- University of Arizona Honors College, Tucson, Arizona, USA
| | - Marwan N. Sabbagh
- Alzheimer’s and Memory Disorders Division, Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Min H. Kang
- Department of Pediatrics, Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - J. Josh Lawrence
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kevin Pruitt
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Ellie Reyna
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Maddy Brown
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
- Roseman University of Health Sciences, Las Vegas, Nevada, USA
| |
Collapse
|
11
|
Saleem A, Shah SIA, Mangar SA, Coello C, Wall MB, Rizzo G, Jones T, Price PM. Cognitive Dysfunction in Patients Treated with Androgen Deprivation Therapy: A Multimodality Functional Imaging Study to Evaluate Neuroinflammation. Prostate Cancer 2023; 2023:6641707. [PMID: 37885823 PMCID: PMC10599921 DOI: 10.1155/2023/6641707] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 07/14/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
Background Androgen deprivation therapy (ADT) for prostate cancer is implicated as a possible cause of cognitive impairment (CI). CI in dementia and Alzheimer's disease is associated with neuroinflammation. In this study, we investigated a potential role of neuroinflammation in ADT-related CI. Methods Patients with prostate cancer on ADT for ≥3 months were categorized as having ADT-emergent CI or normal cognition (NC) based on self-report at interview. Neuroinflammation was evaluated using positron emission tomography (PET) with the translocator protein (TSPO) radioligand [11C]-PBR28. [11C]-PBR28 uptake in various brain regions was quantified as standardized uptake value (SUVR, normalized to cerebellum) and related to blood oxygen level-dependent functional magnetic resonance imaging (BOLD-fMRI) choice-reaction time task (CRT) activation maps. Results Eleven patients underwent PET: four with reported CI (rCI), six with reported NC (rNC), and one status unrecorded. PET did not reveal any between-group differences in SUVR regionally or globally. There was no difference between groups on brain activation to the CRT. Regardless of the reported cognitive status, there was strong correlation between PET-TSPO signal and CRT activation in the hippocampus, amygdala, and medial cortex. Conclusions We found no difference in neuroinflammation measured by PET-TSPO between patients with rCI and rNC. However, we speculate that the strong correlation between TSPO uptake and BOLD-fMRI activation in brain regions involved in memory and known to have high androgen-receptor expression mediating plasticity (hippocampus and amygdala) might reflect inflammatory effects of ADT with compensatory upregulated/increased synaptic functions. Further studies of this imaging readout are warranted to investigate ADT-related CI.
Collapse
Affiliation(s)
- Azeem Saleem
- Invicro, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
- Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Syed Imran Ali Shah
- Department of Surgery and Cancer, Imperial College, London, UK
- Department of Biochemistry, CMH Lahore Medical College & Institute of Dentistry, Lahore, Pakistan
| | | | - Christopher Coello
- Invicro, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - Matthew B. Wall
- Invicro, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - Gaia Rizzo
- Invicro, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
- Division of Brain Sciences, Imperial College London, London, UK
| | - Terry Jones
- Department of Radiology, University of California Davis Medical Center, Davis, California, USA
| | | |
Collapse
|
12
|
Bini J. The historical progression of positron emission tomography research in neuroendocrinology. Front Neuroendocrinol 2023; 70:101081. [PMID: 37423505 PMCID: PMC10530506 DOI: 10.1016/j.yfrne.2023.101081] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
The rapid and continual development of a number of radiopharmaceuticals targeting different receptor, enzyme and small molecule systems has fostered Positron Emission Tomography (PET) imaging of endocrine system actions in vivo in the human brain for several decades. PET radioligands have been developed to measure changes that are regulated by hormone action (e.g., glucose metabolism, cerebral blood flow, dopamine receptors) and actions within endocrine organs or glands such as steroids (e.g., glucocorticoids receptors), hormones (e.g., estrogen, insulin), and enzymes (e.g., aromatase). This systematic review is targeted to the neuroendocrinology community that may be interested in learning about positron emission tomography (PET) imaging for use in their research. Covering neuroendocrine PET research over the past half century, researchers and clinicians will be able to answer the question of where future research may benefit from the strengths of PET imaging.
Collapse
Affiliation(s)
- Jason Bini
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
13
|
Wrigglesworth J, Harding IH, Islam RM, Ward PGD, Woods RL, Bell RJ, McNeil JJ, Storey E, Egan G, Murray AM, Trevaks RE, Ward SA, Davis SR, Ryan J. The association between sex hormones and the change in brain-predicted age difference in older women. Clin Endocrinol (Oxf) 2023; 98:692-699. [PMID: 36807922 PMCID: PMC10073334 DOI: 10.1111/cen.14898] [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: 06/23/2022] [Revised: 01/17/2023] [Accepted: 02/19/2023] [Indexed: 02/22/2023]
Abstract
OBJECTIVE The role of circulating sex hormones on structural brain ageing is yet to be established. This study explored whether concentrations of circulating sex hormones in older women are associated with the baseline and longitudinal changes in structural brain ageing, defined by the brain-predicted age difference (brain-PAD). DESIGN Prospective cohort study using data from NEURO and Sex Hormones in Older Women; substudies of the ASPirin in Reducing Events in the Elderly clinical trial. PATIENTS Community-dwelling older women (aged 70+ years). MEASUREMENTS Oestrone, testosterone, dehydroepiandrosterone (DHEA), and sex-hormone binding globulin (SHBG) were quantified from plasma samples collected at baseline. T1-weighted magnetic resonance imaging was performed at baseline, 1 and 3 years. Brain age was derived from whole brain volume using a validated algorithm. RESULTS The sample comprised of 207 women not taking medications known to influence sex hormone concentrations. A statistically higher baseline brain-PAD (older brain age relative to chronological age) was seen for women in the highest DHEA tertile compared with the lowest in the unadjusted analysis (p = .04). This was not significant when adjusted for chronological age, and potential confounding health and behavioural factors. Oestrone, testosterone and SHBG were not associated with brain-PAD cross-sectionally, nor were any of the examined sex hormones or SHBG associated with brain-PAD longitudinally. CONCLUSION No strong evidence of an association between circulating sex hormones and brain-PAD. Given there is prior evidence to suggests sex hormones may be important for brain ageing, further studies of circulating sex hormones and brain health in postmenopausal women are warranted.
Collapse
Affiliation(s)
- Jo Wrigglesworth
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Ian H. Harding
- Monash Biomedical Imaging, Monash University, Melbourne, 3800, Victoria, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne Victoria, Australia
| | - Rakibul M. Islam
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Phillip G. D. Ward
- Monash Biomedical Imaging, Monash University, Melbourne, 3800, Victoria, Australia
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, 3800, Victoria Australia
- Australian Research Council Centre of Excellence for Integrative Brain Function, Australia
| | - Robyn L. Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Robin J. Bell
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - John J. McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Elsdon Storey
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Gary Egan
- Monash Biomedical Imaging, Monash University, Melbourne, 3800, Victoria, Australia
- Australian Research Council Centre of Excellence for Integrative Brain Function, Australia
| | - Anne M. Murray
- Berman Centre for Outcomes & Clinical Research, Hennepin Healthcare Research Institute, Hennepin, Minneapolis, MN, 55404, USA
- Department of Medicine, Division of Geriatrics, Hennepin Healthcare, University of Minnesota, Minneapolis, MN, 55404, USA
| | - Ruth E. Trevaks
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Stephanie A. Ward
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, New South Wales, Australia
- Department of Geriatric Medicine, Prince of Wales Hospital, Randwick, 2031, New South Wales, Australia
| | - Susan R. Davis
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
- Department of Endocrinology and Diabetes, Alfred Health, Melbourne, 3004, Victoria, Australia
| | - Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| |
Collapse
|
14
|
Niu PP, Wang X, Xu YM. Causal effects of serum testosterone levels on brain volume: a sex-stratified Mendelian randomization study. J Endocrinol Invest 2023:10.1007/s40618-023-02028-0. [PMID: 36780066 DOI: 10.1007/s40618-023-02028-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/28/2023] [Indexed: 02/14/2023]
Abstract
PURPOSE To assess the causal effects of serum testosterone and sex hormone-binding globulin levels on brain volumetric measurements in women and men. METHODS We performed a sex-stratified two-sample Mendelian randomization study using the random-effects inverse variance-weighted method as the primary analysis method. Sex-specific genetic instruments were obtained from a study with up to 194,453 men and 230,454 women. For testosterone, variants with dominant effects on both total and bioavailable testosterone but no aggregate effect on sex hormone-binding globulin were used as the main genetic instruments. Sex-specific summary-level data for magnetic resonance imaging brain volumetric measurements were obtained from a study with 11,624 women and 10,514 men. RESULTS Analyses showed per standard deviation (approximately 3.7 nmol/L) higher testosterone levels in men were suggestively associated with larger gray matter volume (beta = 0.208, 95% confidence interval = 0.067 to 0.349, p = 0.004). The association remained in sensitivity analyses and multivariable analyses. Further analyses showed the effect was mainly act on peripheral cortical gray matter, but not on subcortical gray matter. Testosterone in men was not associated with hippocampal volume. Testosterone in women and sex hormone binding globulin in both sexes had no effect on all outcomes. CONCLUSION Our findings overall support previous evidence that testosterone might have neuroprotective properties in elderly men. Future larger trials with long duration of intervention are warranted to assess the efficacy of testosterone for elderly men with cognitive impairment, especially in those with hypoandrogenism.
Collapse
Affiliation(s)
- P-P Niu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Jian She Road 1#, Zhengzhou, 450000, China.
| | - X Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Jian She Road 1#, Zhengzhou, 450000, China
| | - Y-M Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Jian She Road 1#, Zhengzhou, 450000, China
| |
Collapse
|
15
|
Abstract
As men grow older, circulating testosterone concentrations decline, while prevalence of cognitive impairment and dementia increase. Epidemiological studies of middle-aged and older men have demonstrated associations of lower testosterone concentrations with higher prevalence and incidence of cognitive decline and dementia, including Alzheimer's disease. In observational studies, men with prostate cancer treated by androgen deprivation therapy had a higher risk of dementia. Small intervention studies of testosterone using different measures of cognitive function have provided inconsistent results, with some suggesting improvement. A randomised placebo-controlled trial of one year's testosterone treatment conducted in 788 men aged ≥ 65 years, baseline testosterone < 9.54 nmol/L, showed an improvement in sexual function, but no improvement in cognitive function. There is a known association between diabetes and dementia risk. A randomised placebo-controlled trial of two year's testosterone treatment in 1,007 men aged 50-74 years, waist circumference ≥ 95 cm, baseline testosterone ≤ 14 nmol/L, showed an effect of testosterone in reducing type 2 diabetes risk. There were no cognitive endpoints in that trial. Additional research is warranted but at this stage lower testosterone concentrations in ageing men should be regarded as a biomarker rather than a proven therapeutic target for risk reduction of cognitive decline and dementia, including Alzheimer's disease.
Collapse
Affiliation(s)
- Bu B Yeap
- Medical School, University of Western Australia, Perth, Australia.
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Australia.
| | - Leon Flicker
- Medical School, University of Western Australia, Perth, Australia
- Western Australian Centre for Health and Ageing, University of Western Australia, Perth, Australia
- Department of Geriatric Medicine, Royal Perth Hospital, Perth, Australia
| |
Collapse
|
16
|
Kim JW, Byun MS, Lee JH, Yi D, Kim MJ, Jung G, Lee JY, Lee YS, Kim YK, Kang KM, Sohn CH, Lee DY. Spouse bereavement and brain pathologies: A propensity score matching study. Psychiatry Clin Neurosci 2022; 76:490-504. [PMID: 35751876 PMCID: PMC9796777 DOI: 10.1111/pcn.13439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/25/2022] [Accepted: 06/16/2022] [Indexed: 01/07/2023]
Abstract
AIM Spouse bereavement is one of life's greatest stresses and has been suggested to trigger or accelerate cognitive decline and dementia. However, little information is available about the potential brain pathologies underlying the association between spouse bereavement and cognitive decline. We aimed to investigate that lifetime spouse bereavement is associated with in vivo human brain pathologies underlying cognitive decline. METHODS A total of 319 ever-married older adults between the ages of 61 and 90 years underwent comprehensive clinical assessments and multimodal brain imaging including [11 C] Pittsburgh compound B-positron emission tomography (PET), AV-1451 PET, [18 F] fluorodeoxyglucose-PET, and magnetic resonance imaging. Participants were classified as experiencing no spouse bereavement or spouse bereavement, and comparisons using propensity score matching (59 cases and 59 controls) were performed. RESULTS Spouse bereavement was significantly associated with higher cerebral white matter hyperintensity (WMH) volume compared with no spouse bereavement. Interaction and subsequent subgroup analyses showed that spouse bereavement was significantly associated with higher WMH in the older (>75 years) subgroup and among those with no- or low-skill occupations. In addition, spouse bereavement at 60 years or older affects WMH volume compared with no spouse bereavement, whereas spouse bereavement at younger than 60 years did not. No group differences were observed in other brain pathologies between spouse bereavement categories. CONCLUSIONS The findings suggest that the spouse bereavement may contribute to dementia or cognitive decline by increasing cerebrovascular injury, particularly in older individuals and those with no- or low-skill occupations.
Collapse
Affiliation(s)
- Jee Wook Kim
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Republic of Korea.,Department of Psychiatry, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jun Ho Lee
- Department of Geriatric Psychiatry, National Center for Mental Health, Seoul, Republic of Korea
| | - Dahyun Yi
- Medical Research Center Seoul National University, Institute of Human Behavioral Medicine, Seoul, Republic of Korea
| | - Min Jung Kim
- Department of Psychiatry, Eulji University Nowon Eulji Medical Center, Seoul, Republic of Korea
| | - Gijung Jung
- Medical Research Center Seoul National University, Institute of Human Behavioral Medicine, Seoul, Republic of Korea
| | - Jun-Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.,Medical Research Center Seoul National University, Institute of Human Behavioral Medicine, Seoul, Republic of Korea
| | | |
Collapse
|
17
|
Xiang X, Yu Z, Liu Y, Huang Y, Wang J, Chen L, Ma M. Differential proteomics between unhatched male and female egg yolks reveal the molecular mechanisms of sex-allocation and sex-determination in chicken. Poult Sci 2022; 101:101906. [PMID: 35696754 PMCID: PMC9198474 DOI: 10.1016/j.psj.2022.101906] [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: 12/07/2021] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 12/09/2022] Open
Abstract
There is a huge demand to identify the sex of unhatched fertilized eggs for laying industry and to understand the differences between male and female eggs as early as possible. Then the molecular mechanisms of sex determination and sex allocation in chicken were revealed. Therefore, TMT proteomic was applied to characterize the variation of molecular matrix between unhatched male and female egg yolks. A total of 411 proteins were identified and 35 differentially expressed proteins (DEPs), including 375332005, 015809562, 763550308 (upregulated, UPs) and 1337178851, 89000557, 89000581 (downregulated, DPs), etc. were confirmed between them. Gene ontology analyses showed that DEPs were mainly involved in response to stimulus, distributed in the extracellular region and participated in binding; KEGG analyses showed that few DPs were participated in cell growth and death, transport and catabolism, signaling molecules, interaction and were enriched in ubiquitin mediated proteolysis, endocytosis, ferroptosis, etc. metabolic pathways. Moreover, most of the DEPs and related metabolic pathways were associated with sex hormones. More importantly, this study supports maternal sex-allocation theory and extends our understanding of the molecular mechanism of sex determination and differentiation in avian. Which also provides a powerful evidence for ovo sexing of unhatched fertilized domestic chicken eggs by nondestructive approach and will be of great significance to eggs processing and production.
Collapse
Affiliation(s)
- Xiaole Xiang
- School of Food Science and Bioengineering, Changsha University of Science and Technology, Changsha, Hunan, China; National Research and Development Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Zhuosi Yu
- School of Food Science and Bioengineering, Changsha University of Science and Technology, Changsha, Hunan, China
| | - Yongle Liu
- School of Food Science and Bioengineering, Changsha University of Science and Technology, Changsha, Hunan, China
| | - Yiqun Huang
- School of Food Science and Bioengineering, Changsha University of Science and Technology, Changsha, Hunan, China
| | - Jingjing Wang
- Department of Food Science, Foshan University, Foshan, 528000, China
| | - Lei Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Meihu Ma
- National Research and Development Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China.
| |
Collapse
|
18
|
Кузнецов КО, Хайдарова РР, Хабибуллина РХ, Стыценко ЕС, Философова ВИ, Нуриахметова ИР, Хисамеева ЭМ, Важоров ГС, Хайбуллин ФР, Иванова ЕА, Горбатова КВ. [Testosterone and Alzheimer's disease]. PROBLEMY ENDOKRINOLOGII 2022; 68:97-107. [PMID: 36337024 PMCID: PMC9762454 DOI: 10.14341/probl13136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/24/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that causes dementia in half of the cases. Asthma is usually found in people over 65 years of age. The etiopathogenesis of the disease is multifactorial and includes genetic factors, nutritional disorders, mitochondrial dysfunction, oxidative stress, and aging. Sex hormones have an important influence on the development of AD, as evidenced by a higher incidence in women than in men. Considering the significant influence of T on the maintenance of normal brain function, the present study is aimed at evaluating the impact of androgen deprivation therapy (ADT), as well as testosterone therapy, on the risk of AD development and progression. Although there is some clinical inconsistency between studies, androgens have a significant effect on brain function and are beneficial for AD patients. Low levels of circulating androgens should be considered as a significant risk factor for the development of AD and memory loss. With a reduced level of T in the plasma of men, its administration improves cognitive performance and memory, treatment should be started at an early stage of the disease. In men and women with AD, androgens improve mental state and slow the progression of the disease, providing a protective effect. In the future, it is necessary to conduct studies on a large population, taking into account personality factors and a more specific approach to assessing cognitive functions and the causal relationship of T administration in AD.
Collapse
Affiliation(s)
- К. О. Кузнецов
- Российский национальный исследовательский медицинский университет им. Н.И. Пирогова
| | | | - Р. Х. Хабибуллина
- Первый Санкт-Петербургский государственный медицинский университет им. акад. И.П. Павлова
| | - Е. С. Стыценко
- Санкт-Петербургский государственный педиатрический медицинский университет
| | - В. И. Философова
- Первый Санкт-Петербургский государственный медицинский университет им. акад. И.П. Павлова
| | | | | | - Г. С. Важоров
- Чувашский государственный университет им. И.Н. Ульянова
| | | | | | | |
Collapse
|
19
|
Wrigglesworth J, Harding IH, Ward P, Woods RL, Storey E, Fitzgibbon B, Egan G, Murray A, Shah RC, Trevaks RE, Ward S, McNeil JJ, Ryan J, on behalf of the ASPREE investigator group. Factors Influencing Change in Brain-Predicted Age Difference in a Cohort of Healthy Older Individuals. J Alzheimers Dis Rep 2022; 6:163-176. [PMID: 35591948 PMCID: PMC9108625 DOI: 10.3233/adr-220011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/09/2022] [Indexed: 12/11/2022] Open
Abstract
Background There is considerable variability in the rate at which we age biologically, and the brain is particularly susceptible to the effects of aging. Objective We examined the test-retest reliability of brain age at one- and three-year intervals and identified characteristics that predict the longitudinal change in brain-predicted age difference (brain-PAD, defined by deviations of brain age from chronological age). Methods T1-weighted magnetic resonance images were acquired at three timepoints from 497 community-dwelling adults (73.8±3.5 years at baseline, 48% were female). Brain age was estimated from whole brain volume, using a publicly available algorithm trained on an independent dataset. Linear mixed models were used, adjusting for sex, age, and age2. Results Excellent retest reliability of brain age was observed over one and three years. We identified a significant sex difference in brain-PAD, where a faster rate of brain aging (worsening in brain age relative to chronological age) was observed in men, and this finding replicated in secondary analyses. The effect size, however, was relatively weak, equivalent to 0.16 years difference per year. A higher score in physical health related quality of life and verbal fluency were associated with a faster rate of brain aging, while depression was linked to a slower rate of brain aging, but these findings were not robust. Conclusion Our study provides consistent evidence that older men have slightly faster brain atrophy than women. Given the sparsity of longitudinal research on brain age in older populations, future prospective studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Jo Wrigglesworth
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Ian H. Harding
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Phillip Ward
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
- Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
- Australian Research Council Centre of Excellence for Integrative Brain Function, Clayton, VIC, Australia
| | - Robyn L. Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Elsdon Storey
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Bernadette Fitzgibbon
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Gary Egan
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
- Australian Research Council Centre of Excellence for Integrative Brain Function, Clayton, VIC, Australia
| | - Anne Murray
- Berman Center for Outcomes & Clinical Research, Hennepin Healthcare Research Institute, Minneapolis, MN, USA
- Department of Medicine, Division of Geriatrics, Hennepin Healthcare, University of Minnesota, Minneapolis, MN, USA
| | - Raj C. Shah
- Department of Family Medicine and the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Ruth E. Trevaks
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Stephanie Ward
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, NSW, Australia
- Department of Geriatric Medicine, Prince of Wales Hospital, Randwick, NSW, Australia
| | - John J. McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - on behalf of the ASPREE investigator group
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
- Australian Research Council Centre of Excellence for Integrative Brain Function, Clayton, VIC, Australia
- Berman Center for Outcomes & Clinical Research, Hennepin Healthcare Research Institute, Minneapolis, MN, USA
- Department of Medicine, Division of Geriatrics, Hennepin Healthcare, University of Minnesota, Minneapolis, MN, USA
- Department of Family Medicine and the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, NSW, Australia
- Department of Geriatric Medicine, Prince of Wales Hospital, Randwick, NSW, Australia
| |
Collapse
|
20
|
Bianchi VE. Impact of Testosterone on Alzheimer's Disease. World J Mens Health 2022; 40:243-256. [PMID: 35021306 PMCID: PMC8987133 DOI: 10.5534/wjmh.210175] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 11/15/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease responsible for almost half of all dementia cases in the world and progressively increasing. The etiopathology includes heritability, genetic factors, aging, nutrition, but sex hormones play a relevant role. Animal models demonstrated that testosterone (T) exerted a neuroprotective effect reducing the production of amyloid-beta (Aβ), improving synaptic signaling, and counteracting neuronal death. This study aims to evaluate the impact of T deprivation and T administration in humans on the onset of dementia and AD. A search was conducted on MEDLINE and Scopus for the “androgen deprivation therapy” and “testosterone therapy” with “dementia” and “Alzheimer’s.” Studies lasting twenty years with low risk of bias, randomized clinical trial, and case-controlled studies were considered. Twelve articles on the effect of androgen deprivation therapy (ADT) and AD and seventeen on T therapy and AD were retrieved. Men with prostate cancer under ADT showed a higher incidence of dementia and AD. The effect of T administration in hypogonadal men with AD and cognitive impairment has evidenced some positive results. The majority of studies showed the T administration improved memory and cognition in AD while others did not find any benefit. Although some biases in the studies are evident, T therapy for AD patients may represent an essential clinical therapy to reduce dementia incidence and AD progression. However, more specific case-controlled trials on the effect of androgens therapy in men and women to reducing the onset of AD are necessary.
Collapse
Affiliation(s)
- Vittorio Emanuele Bianchi
- Department of Endocrinology and Metabolism, Clinical Research Center Stella Maris, Falciano, San Marino, Italy.
| |
Collapse
|
21
|
Bianchi VE, Bresciani E, Meanti R, Rizzi L, Omeljaniuk RJ, Torsello A. The role of androgens in women's health and wellbeing. Pharmacol Res 2021; 171:105758. [PMID: 34242799 DOI: 10.1016/j.phrs.2021.105758] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/29/2022]
Abstract
Androgens in women, as well as in men, are intrinsic to maintenance of (i) reproductive competency, (ii) cardiac health, (iii) appropriate bone remodeling and mass retention, (iii) muscle tone and mass, and (iv) brain function, in part, through their mitigation of neurodegenerative disease effects. In recognition of the pluripotency of endogenous androgens, exogenous androgens, and selected congeners, have been prescribed off-label for several decades to treat low libido and sexual dysfunction in menopausal women, as well as, to improve physical performance. However, long-term safety and efficacy of androgen administration has yet to be fully elucidated. Side effects often observed include (i) hirsutism, (ii) acne, (iii) deepening of the voice, and (iv) weight gain but are associated most frequently with supra-physiological doses. By contrast, short-term clinical trials suggest that the use of low-dose testosterone therapy in women appears to be effective, safe and economical. There are, however, few clinical studies, which have focused on effects of androgen therapy on pre- and post-menopausal women; moreover, androgen mechanisms of action have not yet been thoroughly explained in these subjects. This review considers clinical effects of androgens on women's health in order to prevent chronic diseases and reduce cancer risk in gynecological tissues.
Collapse
Affiliation(s)
- Vittorio E Bianchi
- Endocrinology and Metabolism, Clinical Center Stella Maris, Strada Rovereta 42, Falciano 47891, San Marino.
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Robert J Omeljaniuk
- Department of Biology, Lakehead University, 955 Oliver Rd, Thunder Bay, Ontario P7B 5E1, Canada.
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| |
Collapse
|
22
|
Gadomsky L, Dos Santos Guilherme M, Winkler J, van der Kooij MA, Hartmann T, Grimm M, Endres K. Elevated Testosterone Level and Urine Scent Marking in Male 5xFAD Alzheimer Model Mice. Curr Alzheimer Res 2021; 17:80-92. [PMID: 32065104 DOI: 10.2174/1567205017666200217105537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Function of the Amyloid Precursor Protein (AβPP) and its various cleavage products still is not unraveled down to the last detail. While its role as a source of the neurotoxic Amyloid beta (Aβ) peptides in Alzheimer's Disease (AD) is undisputed and its property as a cell attachment protein is intriguing, while functions outside the neuronal context are scarcely investigated. This is particularly noteworthy because AβPP has a ubiquitous expression profile and its longer isoforms, AβPP750 and 770, are found in various tissues outside the brain and in non-neuronal cells. OBJECTIVE Here, we aimed at analyzing the 5xFAD Alzheimer's disease mouse model in regard to male sexual function. The transgenes of this mouse model are regulated by Thy1 promoter activity and Thy1 is expressed in testes, e.g. by Sertoli cells. This allows speculation about an influence on sexual behavior. METHODS We analyzed morphological as well as biochemical properties of testicular tissue from 5xFAD mice and wild type littermates and testosterone levels in serum, testes and the brain. Sexual behavior was assessed by a urine scent marking test at different ages for both groups. RESULTS While sperm number, testes weight and morphological phenotypes of sperms were nearly indistinguishable from those of wild type littermates, testicular testosterone levels were significantly increased in the AD model mice. This was accompanied by elevated and prolonged sexual interest as displayed within the urine scent marking test. CONCLUSION We suggest that overexpression of AβPP, which mostly is used to mimic AD in model mice, also affects male sexual behavior as assessed additional by the Urine Scent Marking (USM) test. The elevated testosterone levels might have an additional impact on central nervous system androgen receptors and also have to be considered when assessing learning and memory capabilities.
Collapse
Affiliation(s)
- Lisa Gadomsky
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg- University, Mainz, Germany
| | - Malena Dos Santos Guilherme
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg- University, Mainz, Germany
| | - Jakob Winkler
- German Institute for Dementia Prevention (GIDP), Neurodegeneration and Neurobiology, Saarland University, Homburg/Saar, Germany and Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Michael A van der Kooij
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg- University, Mainz, Germany
| | - Tobias Hartmann
- German Institute for Dementia Prevention (GIDP), Neurodegeneration and Neurobiology, Saarland University, Homburg/Saar, Germany and Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Marcus Grimm
- German Institute for Dementia Prevention (GIDP), Neurodegeneration and Neurobiology, Saarland University, Homburg/Saar, Germany and Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg- University, Mainz, Germany
| |
Collapse
|
23
|
Qu P, Yu JX, Chen GH. Neuroendocrine Modulation of Cognitive Performance in the Patients with Fibromyalgia. Eur Neurol 2021; 84:254-264. [PMID: 33975317 DOI: 10.1159/000514756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 01/24/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Fibromyalgia (FM) is a chronic widespread pain disorder associated with fatigue, tender points, sleep disturbances, and mood disorders. Symptoms associated with FM also include decreased cognitive function in which the neural basis is poorly understood. Neuroendocrine hormones may be correlated with cognitive performance under some ill conditions. However, we are unaware of current evidence on neuroendocrine hormones as factors influencing cognitive function in adults with FM. OBJECTIVES The aim of the study was to assess whether neuroendocrine hormones could affect cognition in the patients with FM. STUDY DESIGN This study used a case-control trial design. SETTING Study patients were recruited from the neurological outpatient clinics in the Second Affiliated Hospital and Affiliated Chaohu Hospital of Anhui Medical University and met the American College of Rheumatology criteria for FM. METHODS Forty-six patients with FM were compared with twenty-nine healthy controls (HCs). Several measures of cognitive performance and serum levels of neuroendocrine hormones were used to make these comparisons, and the patients were also asked to complete questionnaires on depression and sleep quality. Partial correlation analysis was performed to control the confounders and linear regression analysis was used to examine the effects of neuroendocrine hormones on cognitive measures. RESULTS The FM patients had worse performance in attention, short-term memory, orientation, object working memory and spatial reference memory, higher depression scores, and worse sleep quality than HCs. The raised level of cortisol and gonadotropin-releasing hormone (GnRH) can protect general cognition, whereas the raised level of cortisol and thyroid-stimulating hormone (TSH) will damage spatial memory. LIMITATIONS We did not study the sex hormones comprehensively. CONCLUSIONS The FM patients showed significant cognitive impairment in several domains. The altered levels of cortisol, thyrotrophin-releasing hormone (TRH), and GnRH may mediate cognitive changes in FM.
Collapse
Affiliation(s)
- Ping Qu
- Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jin-Xia Yu
- Official Hospital of the People's Government of Anhui Province, Hefei, China
| | - Gui-Hai Chen
- Departments of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
24
|
Byeon G, Byun MS, Yi D, Lee JH, Jeon SY, Ko K, Jung G, Lee JY, Kim YK, Lee YS, Kang KM, Sohn CH, Lee DY. Synergistic Effect of Serum Homocysteine and Diabetes Mellitus on Brain Alterations. J Alzheimers Dis 2021; 81:287-295. [PMID: 33749655 DOI: 10.3233/jad-210036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Both elevated blood homocysteine and diabetes mellitus (DM) are related to cognitive impairments or dementia. A previous study also demonstrated that the association between homocysteine and cognitive decline was much stronger in individuals with DM than in those without DM. OBJECTIVE This study aimed to examine the interactive effect of blood homocysteine and DM on brain pathological changes including brain atrophy, amyloid-β and tau deposition, and small vessel disease (SVD) related to cognitive impairments. METHODS A total of 430 non-demented older adults underwent comprehensive clinical assessment, measurement of serum homocysteine level, [11C] Pittsburgh Compound B (PiB) PET, [18F] AV-1451 PET, and brain MRI. RESULTS The interactive effect of homocysteine with the presence of DM on brain atrophy, especially in aging-related brain regions, was significant. Higher homocysteine concentration was associated with more prominent brain atrophy in individuals with DM, but not in those without DM. In contrast, interaction effect of homocysteine and DM was found neither on Alzheimer's disease (AD) pathologies, including amyloid-β and tau deposition, nor white matter hyperintensity volume as a measure of SVD. CONCLUSION The present findings suggest that high blood homocysteine level and DM synergistically aggravate brain damage independently of AD and cerebrovascular disease. With regard to preventing dementia or cognitive decline in older adults, these results support the importance of strictly controlling blood glucose in individuals with hyperhomocysteinemia and lowering blood homocysteine level in those with DM.
Collapse
Affiliation(s)
- Gihwan Byeon
- Department of Neuropsychiatry, Kangwon National University Hospital, Chuncheon, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Jun Ho Lee
- Department of Psychiatry, National Center for Mental Health, Seoul, Republic of Korea
| | - So Yeon Jeon
- Department of Neuropsychiatry, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Kang Ko
- Department of Psychiatry, National Center for Mental Health, Seoul, Republic of Korea
| | - Gijung Jung
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong Young Lee
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea.,Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | |
Collapse
|
25
|
Carraro U. 30 Years of Translational Mobility Medicine: 2020 Padua Muscle Days go virtual from Euganean Hills, November 19th to 21st. Eur J Transl Myol 2020; 30:9437. [PMID: 33520146 PMCID: PMC7844408 DOI: 10.4081/ejtm.2020.9437] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
In the autumn of 2019, the 2020 Padua Muscle Days (PMDs) were planned to be held from March 18 to March 21, 2020. The program listed Scientific Sessions to occur over three full days at either Padova University or the Hotel Augustus on Euganei Hills (Padova), Italy. Abruptly, however, in early January the Coronavirus COVID-19 outbreak started in China and changed the world perspectives. In Italy, the epidemia had the first Italian cases and victims in an area south of Milan and in a Village of the Euganei Hills (Vo Euganeo, Padova). Thus, it was a mandatory decision to post-pone the PMDs meeting to 19-21 November, 2020. Luckily, almost all chairs, speakers, and attendees accepted the decision and have assured their presence in late November by long-distance communications. Thus, the Collection of Abstracts were e-published in 30(1) 2020 Issue of the European Journal of Translational Myology (EJTM) together with the many EJTM Communications submitted by speakers and attendees of the 2020 PMDs Here we add a few new entries and the detailed Program of the 2020 Virtual PMDs to be organized November 19-21, 2020 from the Hotel Petrarca of Euganei Hills (Padova), Italy. The Program of the 2020 Virtual PMDs ends with invitation by Zipora Yablonka-Reuveni and myself to the 2021 (Virtual) Padua Muscle Days, March 25-27, Euganei Hills (Padova), Italy.
Collapse
Affiliation(s)
- Ugo Carraro
- Department of Biomedical Sciences, University of Padova, Italy.,Armando & Carmela Mioni-Carraro Foundation for Translational Myology Padova, Italy
| |
Collapse
|
26
|
Li Y, Li S, Xu S, Yu H, Tang L, Liu X, Wang X, Zhang Y, Zhang K, Mi S, Chen M, Cui H. Association of Androgens and Gonadotropins with Amnestic Mild Cognitive Impairment and Probable Alzheimer’s Disease in Chinese Elderly Men. J Alzheimers Dis 2020; 78:277-290. [DOI: 10.3233/jad-200233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Age-related hormone changes play important roles in cognitive decline in older men, and apolipoprotein E ɛ4 (APOE ɛ4) is a risk factor for Alzheimer’s disease (AD). Objective: This study aimed to investigate the interactive role of androgen decline and APOE ɛ4 genotype in the pathogenesis of amnestic mild cognitive impairment (aMCI) and AD. Methods: In total, 576 elderly men over 65 years old from communities in Shijiazhuang were enrolled in this study, including 243 with normal cognition (NC), 271 with aMCI, and 62 with probable AD. Cognitive function was evaluated with a battery of neuropsychological tests. The serum levels of androgen and gonadotropin were detected by ELISA and chemiluminescence immunoassay. Results: The levels of free testosterone (FT) and dihydrotestosterone (DHT) were lower in the aMCI group (p < 0.05), and even lower in the AD group (p < 0.001), but the levels of follicle stimulating hormone (FSH) and luteinizing hormone (LH) were higher in AD group (p < 0.01), comparing with that in NC or aMCI group. The interaction of lower FT or DHT levels with APOE ɛ4 had a risk role in global cognitive impairment (p < 0.05). The area under the curve (AUC) of the ROC curve for predicting aMCI by serum FT levels was 0.745. Conclusion: These results indicated that the interaction of androgen decline and APOE ɛ4 genotype play a role in aMCI and AD. Serum FT levels have a predictive value for aMCI and might be a potential biomarker for prodromal AD.
Collapse
Affiliation(s)
- Yan Li
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, P. R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P. R. China
- College of Nursing, Hebei Medical University, Shijiazhuang, P. R. China
| | - Sha Li
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, P. R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P. R. China
| | - Shunjiang Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Hong Yu
- College of Nursing, Hebei Medical University, Shijiazhuang, P. R. China
| | - Longmei Tang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang, P. R. China
| | - Xiaoyun Liu
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P. R. China
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Xuemei Wang
- College of Nursing, Hebei Medical University, Shijiazhuang, P. R. China
| | - Yuanyuan Zhang
- College of Nursing, Hebei Medical University, Shijiazhuang, P. R. China
| | - Kaixia Zhang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Shixiong Mi
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, P. R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P. R. China
| | - Meiqin Chen
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, P. R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P. R. China
| | - Huixian Cui
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, P. R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P. R. China
| |
Collapse
|
27
|
Exploring a Cost-Efficient Model for Predicting Cerebral Aβ Burden Using MRI and Neuropsychological Markers in the ADNI-2 Cohort. J Pers Med 2020; 10:jpm10040197. [PMID: 33121011 PMCID: PMC7712671 DOI: 10.3390/jpm10040197] [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: 08/30/2020] [Revised: 10/02/2020] [Accepted: 10/22/2020] [Indexed: 11/17/2022] Open
Abstract
Many studies have focused on the early detection of Alzheimer’s disease (AD). Cerebral amyloid beta (Aβ) is a hallmark of AD and can be observed in vivo via positron emission tomography imaging using an amyloid tracer or cerebrospinal fluid assessment. However, these methods are expensive. The current study aimed to identify and compare the ability of magnetic resonance imaging (MRI) markers and neuropsychological markers to predict cerebral Aβ status in an AD cohort using machine learning (ML) approaches. The prediction ability of candidate markers for cerebral Aβ status was examined by analyzing 724 participants from the ADNI-2 cohort. Demographic variables, structural MRI markers, and neuropsychological test scores were used as input in several ML algorithms to predict cerebral Aβ positivity. Out of five combinations of candidate markers, neuropsychological markers with demographics showed the most cost-efficient result. The selected model could distinguish abnormal levels of Aβ with a prediction ability of 0.85, which is the same as that for MRI-based models. In this study, we identified the prediction ability of MRI markers using ML approaches and showed that the neuropsychological model with demographics can predict Aβ positivity, suggesting a more cost-efficient method for detecting cerebral Aβ status compared to MRI markers.
Collapse
|
28
|
Kang KM, Byun MS, Lee JH, Yi D, Choi HJ, Lee E, Lee Y, Lee JY, Kim YK, Sohn BK, Sohn CH, Lee DY. Association of carotid and intracranial stenosis with Alzheimer's disease biomarkers. ALZHEIMERS RESEARCH & THERAPY 2020; 12:106. [PMID: 32912336 PMCID: PMC7488394 DOI: 10.1186/s13195-020-00675-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022]
Abstract
Background To clarify whether atherosclerosis of the carotid and intracranial arteries is related to Alzheimer’s disease (AD) pathology in vivo, we investigated the associations of carotid and intracranial artery stenosis with cerebral beta-amyloid (Aβ) deposition and neurodegeneration in middle- and old-aged individuals. Given different variations of the pathologies between cognitive groups, we focused separately on cognitively normal (CN) and cognitively impaired (CI) groups. Methods A total of 281 CN and 199 CI (mild cognitive impairment and AD dementia) subjects underwent comprehensive clinical assessment, [11C] Pittsburgh compound B-positron emission tomography, and magnetic resonance (MR) imaging including MR angiography. We evaluated extracranial carotid and intracranial arteries for the overall presence, severity (i.e., number and degree of narrowing), and location of stenosis. Results We found no associations between carotid and intracranial artery stenosis and cerebral Aβ burden in either the CN or the CI group. In terms of neurodegeneration, exploratory univariable analyses showed associations between the presence and severity of stenosis and regional neurodegeneration biomarkers (i.e., reduced hippocampal volume [HV] and cortical thickness in the AD-signature regions) in both the CN and CI groups. In confirmatory multivariable analyses controlling for demographic covariates and diagnosis, the association between number of stenotic intracranial arteries ≥ 2 and reduced HV in the CI group remained significant. Conclusions Neither carotid nor intracranial artery stenosis appears to be associated with brain Aβ burden, while intracranial artery stenosis is related to amyloid-independent neurodegeneration, particularly hippocampal atrophy.
Collapse
Affiliation(s)
- Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jun Ho Lee
- Department of Neuropsychiatry, National Center for Mental Health, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Hye Jeong Choi
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Eunjung Lee
- Department of Radiology, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Younghwa Lee
- Department of Neuropsychiatry, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea.,Department of Psychiatry, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Bo Kyung Sohn
- Department of Psychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Department of Radiology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Dong Young Lee
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea. .,Department of Neuropsychiatry, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Department of Psychiatry, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | | |
Collapse
|
29
|
Bianchi VE, Rizzi L, Bresciani E, Omeljaniuk RJ, Torsello A. Androgen Therapy in Neurodegenerative Diseases. J Endocr Soc 2020; 4:bvaa120. [PMID: 33094209 PMCID: PMC7568521 DOI: 10.1210/jendso/bvaa120] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative diseases, including Alzheimer disease (AD), Parkinson disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington disease, are characterized by the loss of neurons as well as neuronal function in multiple regions of the central and peripheral nervous systems. Several studies in animal models have shown that androgens have neuroprotective effects in the brain and stimulate axonal regeneration. The presence of neuronal androgen receptors in the peripheral and central nervous system suggests that androgen therapy might be useful in the treatment of neurodegenerative diseases. To illustrate, androgen therapy reduced inflammation, amyloid-β deposition, and cognitive impairment in patients with AD. As well, improvements in remyelination in MS have been reported; by comparison, only variable results are observed in androgen treatment of PD. In ALS, androgen administration stimulated motoneuron recovery from progressive damage and regenerated both axons and dendrites. Only a few clinical studies are available in human individuals despite the safety and low cost of androgen therapy. Clinical evaluations of the effects of androgen therapy on these devastating diseases using large populations of patients are strongly needed.
Collapse
Affiliation(s)
- Vittorio Emanuele Bianchi
- Department of Endocrinology and Metabolism, Clinical Center Stella Maris, Strada Rovereta, Falciano, San Marino
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
30
|
Jung JH, Lee GW, Lee JH, Byun MS, Yi D, Jeon SY, Jung GJ, Joung H, Shin SA, Kim YK, Kang KM, Sohn CH, Lee DY. Multiparity, Brain Atrophy, and Cognitive Decline. Front Aging Neurosci 2020; 12:159. [PMID: 32581769 PMCID: PMC7291884 DOI: 10.3389/fnagi.2020.00159] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/08/2020] [Indexed: 12/21/2022] Open
Abstract
Background Multiparity – grand multiparity (i.e., five or more childbirths) in particular – has been reported to have an association with increased risk of Alzheimer’s disease (AD) dementia or related cognitive decline in women. However, the pathological links underlying this relationship are still unknown. This study was conducted to examine the relationships of multiparity with cerebral amyloid-beta (Aβ) deposition, brain atrophy, and white matter hyperintensities (WMHs). Methods In this study, total of 237 older women with 148 cognitively normal and 89 mild cognitive impairment from the Korean Brain Aging Study for Early Diagnosis and Prediction of Alzheimer’s Disease (KBASE) were included. Participants underwent clinical and neuropsychological assessments in addition to 11C-labeled Pittsburgh Compound B positron emission tomography, and magnetic resonance imaging. The associations of parity with Aβ deposition, hippocampal volume, cortical volume, WMH volume and mini-mental status examination (MMSE) score were examined. Results Participants with grand multiparity showed significantly reduced adjusted hippocampal volume, spatial pattern of atrophy for recognition of AD volume and spatial pattern of atrophy for recognition of brain aging volume even after controlling for potential confounders. Furthermore, MMSE score was also significantly lower in this group. In contrast, grand multiparity did not show any association with global Aβ retention, Aβ positivity rate, or WMH volume, regardless of covariates. Conclusion Our findings suggest that grand multiparity contributes to cognitive decline or increased dementia risk in older women by aggravating amyloid-independent hippocampal or cortical atrophy.
Collapse
Affiliation(s)
- Joon Hyung Jung
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| | - Ga Won Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Jun Ho Lee
- Department of Neuropsychiatry, National Center for Mental Health, Seoul, South Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, South Korea
| | - So Yeon Jeon
- Department of Neuropsychiatry, Chungnam National University Hospital, Daejeon, South Korea
| | - Gi Jung Jung
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Haejung Joung
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Seong A Shin
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Dong Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea.,Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea.,Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, South Korea
| |
Collapse
|
31
|
Giatti S, Diviccaro S, Falvo E, Garcia-Segura LM, Melcangi RC. Physiopathological role of the enzymatic complex 5α-reductase and 3α/β-hydroxysteroid oxidoreductase in the generation of progesterone and testosterone neuroactive metabolites. Front Neuroendocrinol 2020; 57:100836. [PMID: 32217094 DOI: 10.1016/j.yfrne.2020.100836] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/04/2020] [Accepted: 03/18/2020] [Indexed: 12/22/2022]
Abstract
The enzymatic complex 5α-reductase (5α-R) and 3α/3β-hydroxysteroid oxidoreductase (HSOR) is expressed in the nervous system, where it transforms progesterone (PROG) and testosterone (T) into neuroactive metabolites. These metabolites regulate myelination, brain maturation, neurotransmission, reproductive behavior and the stress response. The expression of 5α-R and 3α-HSOR and the levels of PROG and T reduced metabolites show regional and sex differences in the nervous system and are affected by changing physiological conditions as well as by neurodegenerative and psychiatric disorders. A decrease in their nervous tissue levels may negatively impact the course and outcome of some pathological events. However, in other pathological conditions their increased levels may have a negative impact. Thus, the use of synthetic analogues of these steroids or 5α-R modulation have been proposed as therapeutic approaches for several nervous system pathologies. However, further research is needed to fully understand the consequences of these manipulations, in particular with 5α-R inhibitors.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Eva Falvo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
32
|
Jeon SY, Byun MS, Yi D, Lee JH, Ko K, Sohn BK, Lee JY, Ryu SH, Lee DW, Shin SA, Kim YK, Kang KM, Sohn CH, Lee DY. Midlife Lifestyle Activities Moderate APOE ε4 Effect on in vivo Alzheimer's Disease Pathologies. Front Aging Neurosci 2020; 12:42. [PMID: 32256335 PMCID: PMC7093017 DOI: 10.3389/fnagi.2020.00042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/07/2020] [Indexed: 12/24/2022] Open
Abstract
This study aimed to investigate whether the midlife cognitive activity and physical activity moderate the relationship between apolipoprotein Eε4 (APOE4) and in vivo Alzheimer's disease (AD) pathologies. In total, 287 non-demented older adults (mean age 72 years) from the Korean Brain Aging Study for the Early diagnosis and prediction of Alzheimer's disease cohort were included. Participants underwent a comprehensive clinical assessment including the evaluation for midlife CA and physical activity, [11C]-Pittsburgh-Compound-B-positron emission tomography (PET), [18F]-fluorodeoxyglucose PET, structural magnetic resonance imaging (MRI), and APOE genotyping. We used linear regression and regression-based mediated-moderation models for statistical analyses. Neither midlife cognitive activity nor physical activity moderated the effect of APOE4 on β-amyloid (Aβ) retention itself. Midlife cognitive activity significantly moderated the effect of APOE4 on hippocampal volume [B (SE) = - 627.580 (252.327), t = -2.488, p = 0.014]: APOE4 carriers had smaller hippocampal volume than non-carriers at relatively high cognitive activity state (p = 0.004), but not at relatively low cognitive activity condition (p = 0.937). Midlife physical activity significantly moderated the effect of Aβ retention, which was closely related to APOE4, on AD-signature region cerebral glucose metabolism [AD-CM; B (SE) = 0.004 (0.002), t = 2.030, p = 0.043]: higher Aβ accumulation was associated with lower AD-CM in relatively low physical activity condition (p < 0.001), whereas no such association was observed in relatively high physical activity state (p = 0.791). The findings suggest that high midlife cognitive activity may accelerate hippocampal atrophy induced by APOE4, whereas high midlife physical activity may delay AD-related cerebral hypometabolism by weakening the influence of APOE4-associated Aβ retention.
Collapse
Affiliation(s)
- So Yeon Jeon
- Department of Psychiatry, Chungnam National University Hospital, Daejeon, South Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Jun-Ho Lee
- Department of Psychiatry, National Center for Mental Health, Seoul, South Korea
| | - Kang Ko
- Department of Psychiatry, National Center for Mental Health, Seoul, South Korea
| | - Bo Kyung Sohn
- Department of Neuropsychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Seung-Ho Ryu
- Department of Psychiatry, School of Medicine, Konkuk University Medical Center, Konkuk University, Seoul, South Korea
| | - Dong Woo Lee
- Department of Neuropsychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Seoung A Shin
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea.,Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, South Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
33
|
Cherrier MM, Higano CS. Impact of androgen deprivation therapy on mood, cognition, and risk for AD. Urol Oncol 2020; 38:53-61. [DOI: 10.1016/j.urolonc.2019.01.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 01/11/2019] [Accepted: 01/18/2019] [Indexed: 01/14/2023]
|
34
|
Chowen JA, Garcia-Segura LM. Microglia, neurodegeneration and loss of neuroendocrine control. Prog Neurobiol 2020; 184:101720. [PMID: 31715222 DOI: 10.1016/j.pneurobio.2019.101720] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/19/2019] [Accepted: 11/02/2019] [Indexed: 02/07/2023]
|
35
|
Giatti S, Diviccaro S, Serafini MM, Caruso D, Garcia-Segura LM, Viviani B, Melcangi RC. Sex differences in steroid levels and steroidogenesis in the nervous system: Physiopathological role. Front Neuroendocrinol 2020; 56:100804. [PMID: 31689419 DOI: 10.1016/j.yfrne.2019.100804] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The nervous system, in addition to be a target for steroid hormones, is the source of a variety of neuroactive steroids, which are synthesized and metabolized by neurons and glial cells. Recent evidence indicates that the expression of neurosteroidogenic proteins and enzymes and the levels of neuroactive steroids are different in the nervous system of males and females. We here summarized the state of the art of neuroactive steroids, particularly taking in consideration sex differences occurring in the synthesis and levels of these molecules. In addition, we discuss the consequences of sex differences in neurosteroidogenesis for the function of the nervous system under healthy and pathological conditions and the implications of neuroactive steroids and neurosteroidogenesis for the development of sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Melania Maria Serafini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Barbara Viviani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
36
|
Hillerer KM, Slattery DA, Pletzer B. Neurobiological mechanisms underlying sex-related differences in stress-related disorders: Effects of neuroactive steroids on the hippocampus. Front Neuroendocrinol 2019; 55:100796. [PMID: 31580837 PMCID: PMC7115954 DOI: 10.1016/j.yfrne.2019.100796] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don't model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.
Collapse
Affiliation(s)
- Katharina M Hillerer
- Department of Obstetrics and Gynaecology, Salzburger Landeskrankenhaus (SALK), Paracelsus Medical University (PMU), Clinical Research Center Salzburg (CRCS), Salzburg, Austria.
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Belinda Pletzer
- Department of Psychology, University of Salzburg, Salzburg, Austria; Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| |
Collapse
|
37
|
Fiechter M, Roggo A, Haider A, Bengs S, Burger IA, Marędziak M, Portmann A, Treyer V, Becker AS, Messerli M, Mühlematter UJ, Kudura K, von Felten E, Benz DC, Fuchs TA, Gräni C, Pazhenkottil AP, Buechel RR, Kaufmann PA, Gebhard C. Metabolic Activity in Central Neural Structures of Patients With Myocardial Injury. J Am Heart Assoc 2019; 8:e013070. [PMID: 31566462 PMCID: PMC6806042 DOI: 10.1161/jaha.119.013070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background Increasing evidence suggests a psychosomatic link between neural systems and the heart. In light of the growing burden of ischemic cardiovascular disease across the globe, a better understanding of heart‐brain interactions and their implications for cardiovascular treatment strategies is needed. Thus, we sought to investigate the interaction between myocardial injury and metabolic alterations in central neural areas in patients with suspected or known coronary artery disease. Methods and Results The association between resting metabolic activity in distinct neural structures and cardiac function was analyzed in 302 patients (aged 66.8±10.2 years; 70.9% men) undergoing fluor‐18‐deoxyglucose positron emission tomography and 99mTc‐tetrofosmin single‐photon emission computed tomography myocardial perfusion imaging. There was evidence for reduction of callosal, caudate, and brainstem fluor‐18‐deoxyglucose uptake in patients with impaired left ventricular ejection fraction (<55% versus ≥55%: P=0.047, P=0.022, and P=0.013, respectively) and/or in the presence of myocardial ischemia (versus normal perfusion: P=0.010, P=0.013, and P=0.016, respectively). In a sex‐stratified analysis, these differences were observed in men, but not in women. A first‐order interaction term consisting of sex and impaired left ventricular ejection fraction or myocardial ischemia was identified as predictor of metabolic activity in these neural regions (left ventricular ejection fraction: P=0.015 for brainstem; myocardial ischemia: P=0.004, P=0.018, and P=0.003 for callosal, caudate, or brainstem metabolism, respectively). Conclusions Myocardial dysfunction and injury are associated with reduced resting metabolic activity of central neural structures, including the corpus callosum, the caudate nucleus, and the brainstem. These associations differ in women and men, suggesting sex differences in the pathophysiological interplay of the nervous and cardiovascular systems.
Collapse
Affiliation(s)
- Michael Fiechter
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland.,Center for Molecular Cardiology University of Zurich Switzerland.,Swiss Paraplegic Center Nottwil Switzerland
| | - Andrea Roggo
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Ahmed Haider
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland.,Center for Molecular Cardiology University of Zurich Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland.,Center for Molecular Cardiology University of Zurich Switzerland
| | - Irene A Burger
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Monika Marędziak
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland.,Center for Molecular Cardiology University of Zurich Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Anton S Becker
- Department of Diagnostic and Interventional Radiology University Hospital Zurich Zurich Switzerland
| | - Michael Messerli
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Urs J Mühlematter
- Department of Diagnostic and Interventional Radiology University Hospital Zurich Zurich Switzerland
| | - Ken Kudura
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Elia von Felten
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Dominik C Benz
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Tobias A Fuchs
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Christoph Gräni
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Aju P Pazhenkottil
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Ronny R Buechel
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Philipp A Kaufmann
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland
| | - Catherine Gebhard
- Department of Nuclear Medicine University Hospital Zurich Zurich Switzerland.,Center for Molecular Cardiology University of Zurich Switzerland
| |
Collapse
|
38
|
Honarpisheh P, McCullough LD. Sex as a biological variable in the pathology and pharmacology of neurodegenerative and neurovascular diseases. Br J Pharmacol 2019; 176:4173-4192. [PMID: 30950038 DOI: 10.1111/bph.14675] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/19/2019] [Accepted: 02/24/2019] [Indexed: 12/14/2022] Open
Abstract
The incidence of dementia, most commonly caused by cerebrovascular and neurodegenerative diseases, continues to grow as our population ages. Alzheimer disease (AD) and vascular cognitive impairment (VCI) are responsible for more than 80% of all cases of dementia. There are few effective, long-term treatments for AD and VCI-related conditions (e.g., stroke and cerebral amyloid angiopathy (CAA)). This review focuses on AD (as the most common "neurodegenerative" cause of dementia), CAA (as an "emerging" cause of dementia), and stroke (as the most common cause of "vascular" dementia). We will discuss the available literature on the pharmacological therapies that demonstrate sex differences, which refer to any combination of structural, chromosomal, gonadal, or hormonal differences between males and females. We will emphasize the importance of considering sex as a biological variable in the design of preclinical and clinical studies that investigate underlying pathologies or response to pharmacological interventions in dementia. LINKED ARTICLES: This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Pedram Honarpisheh
- Department of Neurology, University of Texas McGovern Medical School, Houston, Texas
| | - Louise D McCullough
- Department of Neurology, University of Texas McGovern Medical School, Houston, Texas
| |
Collapse
|
39
|
Ko H, Ihm JJ, Kim HG. Cognitive Profiling Related to Cerebral Amyloid Beta Burden Using Machine Learning Approaches. Front Aging Neurosci 2019; 11:95. [PMID: 31105554 PMCID: PMC6499028 DOI: 10.3389/fnagi.2019.00095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/08/2019] [Indexed: 12/31/2022] Open
Abstract
Background: Cerebral amyloid beta (Aβ) is a hallmark of Alzheimer’s disease (AD). Aβ can be detected in vivo with amyloid imaging or cerebrospinal fluid assessments. However, these technologies can be both expensive and invasive, and their accessibility is limited in many clinical settings. Hence the current study aims to identify multivariate cost-efficient markers for Aβ positivity among non-demented individuals using machine learning (ML) approaches. Methods: The relationship between cost-efficient candidate markers and Aβ status was examined by analyzing 762 participants from the Alzheimer’s Disease Neuroimaging Initiative-2 cohort at baseline visit (286 cognitively normal, 332 with mild cognitive impairment, and 144 with AD; mean age 73.2 years, range 55–90). Demographic variables (age, gender, education, and APOE status) and neuropsychological test scores were used as predictors in an ML algorithm. Cerebral Aβ burden and Aβ positivity were measured using 18F-florbetapir positron emission tomography images. The adaptive least absolute shrinkage and selection operator (LASSO) ML algorithm was implemented to identify cognitive performance and demographic variables and distinguish individuals from the population at high risk for cerebral Aβ burden. For generalizability, results were further checked by randomly dividing the data into training sets and test sets and checking predictive performances by 10-fold cross-validation. Results: Out of neuropsychological predictors, visuospatial ability and episodic memory test results were consistently significant predictors for Aβ positivity across subgroups with demographic variables and other cognitive measures considered. The adaptive LASSO model using out-of-sample classification could distinguish abnormal levels of Aβ. The area under the curve of the receiver operating characteristic curve was 0.754 in the mild change group, 0.803 in the moderate change group, and 0.864 in the severe change group, respectively. Conclusion: Our results showed that the cost-efficient neuropsychological model with demographics could predict Aβ positivity, suggesting a potential surrogate method for detecting Aβ deposition non-invasively with clinical utility. More specifically, it could be a very brief screening tool in various settings to recruit participants with potential biomarker evidence of AD brain pathology. These identified individuals would be valuable participants in secondary prevention trials aimed at detecting an anti-amyloid drug effect in the non-demented population.
Collapse
Affiliation(s)
- Hyunwoong Ko
- Interdisciplinary Program in Cognitive Science, Seoul National University, Seoul, South Korea.,Biomedical Knowledge Engineering Laboratory, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Jung-Joon Ihm
- School of Dentistry, Seoul National University, Seoul, South Korea
| | - Hong-Gee Kim
- Interdisciplinary Program in Cognitive Science, Seoul National University, Seoul, South Korea.,Biomedical Knowledge Engineering Laboratory, School of Dentistry, Seoul National University, Seoul, South Korea.,School of Dentistry, Seoul National University, Seoul, South Korea
| | | |
Collapse
|
40
|
Letra L, Rodrigues T, Matafome P, Santana I, Seiça R. Adiponectin and sporadic Alzheimer's disease: Clinical and molecular links. Front Neuroendocrinol 2019; 52:1-11. [PMID: 29038028 DOI: 10.1016/j.yfrne.2017.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/05/2017] [Accepted: 10/10/2017] [Indexed: 01/21/2023]
Abstract
Obesity has been consistently associated with Alzheimer's disease (AD) though the exact mechanisms by which it influences cognition are still elusive and subject of current research. Adiponectin, the most abundant adipokine in circulation, is inversely correlated with adipose tissue dysfunction and seems to be a central player in this association. In fact, different signalling pathways are shared by adiponectin and proteins involved in AD pathophysiology and considerable amount of evidence supports its direct and indirect influence on β-amyloid and tau aggregates formation. In this paper we present a critical review of cellular, animal and clinical studies which have contributed to a more thorough understanding of the extent to which adiponectin influences the risk of developing AD as well as its progression. Finally, the effect of acetylcholinesterase inhibitors on circulating adiponectin levels, possible therapeutic applications and future research strategies are also discussed.
Collapse
Affiliation(s)
- Liliana Letra
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Neurology Department, Centro Hospitalar do Baixo Vouga - Aveiro, Av. Artur Ravara, 3814-501 Aveiro, Portugal.
| | - Tiago Rodrigues
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| | - Paulo Matafome
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| | - Isabel Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Professor Mota Pinto, 3000-075 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| | - Raquel Seiça
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
41
|
Lei Y, Renyuan Z. Effects of Androgens on the Amyloid-β Protein in Alzheimer's Disease. Endocrinology 2018; 159:3885-3894. [PMID: 30215697 DOI: 10.1210/en.2018-00660] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 09/06/2018] [Indexed: 12/24/2022]
Abstract
Age-related androgen depletion has been implicated in compromised neuroprotection and is involved in the pathogenesis of neurodegenerative disease, including Alzheimer's disease (AD), the leading cause of dementia. Emerging data revealed that reduction of both serum and brain androgen levels in males is associated with increased amyloid-β (Aβ) accumulation, a putative cause of AD. It has been demonstrated that androgens can function as the endogenous negative regulators of Aβ. However, the mechanisms by which androgens regulate Aβ production, degradation, and clearance, as well as the Aβ-induced pathological process in AD, are still elusive. This review emphasizes the contributions of androgen to Aβ metabolism and toxicity in AD and thus may provide novel strategies for prevention and therapeutics.
Collapse
Affiliation(s)
- Yang Lei
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai, China
| | - Zhou Renyuan
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Keyvani K, Münster Y, Kurapati NK, Rubach S, Schönborn A, Kocakavuk E, Karout M, Hammesfahr P, Wang YC, Hermann DM, Teuber-Hanselmann S, Herring A. Higher levels of kallikrein-8 in female brain may increase the risk for Alzheimer's disease. Brain Pathol 2018; 28:947-964. [PMID: 29505099 DOI: 10.1111/bpa.12599] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 02/28/2018] [Indexed: 01/21/2023] Open
Abstract
Women seem to have a higher vulnerability to Alzheimer's disease (AD), but the underlying mechanisms of this sex dichotomy are not well understood. Here, we first determined the influence of sex on various aspects of Alzheimer's pathology in transgenic CRND8 mice. We demonstrate that beta-amyloid (Aβ) plaque burden starts to be more severe around P180 (moderate disease stage) in female transgenics when compared to males and that aging aggravates this sex-specific difference. Furthermore, we show that female transgenics suffer from higher levels of neurovascular dysfunction around P180, resulting in impaired Aβ peptide clearance across the blood-brain-barrier at P360. Female transgenics show also higher levels of diffuse microgliosis and inflammation, but the density of microglial cells surrounding Aβ plaques is less in females. In line with this finding, testosterone compared to estradiol was able to improve microglial viability and Aβ clearance in vitro. The spatial memory of transgenics was in general poorer than in wildtypes and at P360 worse in females irrespective of their genotype. This difference was accompanied by a slightly diminished dendritic complexity in females. While all the above-named sex-differences emerged after the onset of Aβ pathology, kallikrein-8 (KLK8) protease levels were, as an exception, higher in female than in male brains very early when virtually no plaques were detectable. In a second step, we quantified cerebral KLK8 levels in AD patients and healthy controls, and could ascertain, similar to mice, higher KLK8 levels not only in AD-affected but also in healthy brains of women. Accordingly, we could demonstrate that estradiol but not testosterone induces KLK8 synthesis in neuronal and microglial cells. In conclusion, multiple features of AD are more pronounced in females. Here, we show for the first time that this sex-specific difference may be meditated by estrogen-induced KLK8 overproduction long before AD pathology emerges.
Collapse
Affiliation(s)
- Kathy Keyvani
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Yvonne Münster
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Nirup K Kurapati
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Sebastian Rubach
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Andreas Schönborn
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Emre Kocakavuk
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Mohamed Karout
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Pia Hammesfahr
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Ya-Chao Wang
- Department of Neurology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Sarah Teuber-Hanselmann
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Arne Herring
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| |
Collapse
|