1
|
Castro-Aldrete L, Einsiedler M, Novakova Martinkova J, Depypere H, Alvin Ang TF, Mielke MM, Sindi S, Eyre HA, Au R, Schumacher Dimech AM, Dé A, Szoeke C, Tartaglia MC, Santuccione Chadha A. Alzheimer disease seen through the lens of sex and gender. Nat Rev Neurol 2025:10.1038/s41582-025-01071-0. [PMID: 40229578 DOI: 10.1038/s41582-025-01071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer disease (AD) is a life-limiting neurodegenerative disorder that disproportionately affects women. Indeed, sex and gender are emerging as crucial modifiers of diagnostic and therapeutic pathways in AD. This Review provides an overview of the interactions of sex and gender with important developments in AD and offers insights into priorities for future research to facilitate the development and implementation of personalized approaches in the shifting paradigm of AD care. In particular, this Review focuses on the influence of sex and gender on important advances in the treatment and diagnosis of AD, including disease-modifying therapies, fluid-based biomarkers, cognitive assessment tools and multidomain lifestyle interventional studies.
Collapse
Affiliation(s)
| | | | - Julie Novakova Martinkova
- Women's Brain Foundation, Basel, Switzerland
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Herman Depypere
- Department of Gynecology, Breast and Menopause Clinic, University Hospital, Coupure Menopause Centre, Ghent, Belgium
| | - Ting Fang Alvin Ang
- Department of Anatomy and Neurobiology and Slone Center of Epidemiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shireen Sindi
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- The Ageing Epidemiology Research Unit, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Harris A Eyre
- Neuro-Policy Program, Center for Health and Biosciences, The Baker Institute for Public Policy, Rice University, Houston, TX, USA
- Euro-Mediterranean Economists Association, Barcelona, Spain
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Neurology, Medicine and Epidemiology, Boston University Chobanian and Avedisian School of Medicine and School of Public Health, Boston, MA, USA
| | - Anne Marie Schumacher Dimech
- Women's Brain Foundation, Basel, Switzerland
- Faculty of Health Sciences and Medicine, University of Lucerne, Lucerne, Switzerland
| | - Anna Dé
- Women's Brain Foundation, Basel, Switzerland
| | | | - Maria Carmela Tartaglia
- Women's Brain Foundation, Basel, Switzerland
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
2
|
Depypere H, Mosconi L, Brinton RD, Hampel H. Dementia prevention, intervention, and care: Comments on the 2024 report of the Lancet standing Commission. Maturitas 2025; 195:108217. [PMID: 40050160 DOI: 10.1016/j.maturitas.2025.108217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/15/2025]
Affiliation(s)
- Herman Depypere
- Menopause and Breast Clinic, Gent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Lisa Mosconi
- The Alzheimer's Prevention Program and Women's Brain Initiative, Departments of Neuroscience, Neurology and Radiology, Weill Cornell Medicine, 402 East 70th Street, New York, NY 10021, USA
| | - Roberta Diaz Brinton
- Department of Pharmacology, Neuroscience and Neurology, University of Arizona, College of Medicine, 1230 N. Cherry Avenue, Room 470, Tucson, AZ 85724-5126, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine, AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, F-75013 Paris, France
| |
Collapse
|
3
|
Li R, Miao Z, Liu Y, Chen X, Wang H, Su J, Chen J. The Brain-Gut-Bone Axis in Neurodegenerative Diseases: Insights, Challenges, and Future Prospects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307971. [PMID: 39120490 PMCID: PMC11481201 DOI: 10.1002/advs.202307971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 06/04/2024] [Indexed: 08/10/2024]
Abstract
Neurodegenerative diseases are global health challenges characterized by the progressive degeneration of nerve cells, leading to cognitive and motor impairments. The brain-gut-bone axis, a complex network that modulates multiple physiological systems, has gained increasing attention owing to its profound effects on the occurrence and development of neurodegenerative diseases. No comprehensive review has been conducted to clarify the triangular relationship involving the brain-gut-bone axis and its potential for innovative therapies for neurodegenerative disorders. In light of this, a new perspective is aimed to propose on the interplay between the brain, gut, and bone systems, highlighting the potential of their dynamic communication in neurodegenerative diseases, as they modulate multiple physiological systems, including the nervous, immune, endocrine, and metabolic systems. Therapeutic strategies for maintaining the balance of the axis, including brain health regulation, intestinal microbiota regulation, and improving skeletal health, are also explored. The intricate physiological interactions within the brain-gut-bone axis pose a challenge in the development of effective treatments that can comprehensively target this system. Furthermore, the safety of these treatments requires further evaluation. This review offers a novel insights and strategies for the prevention and treatment of neurodegenerative diseases, which have important implications for clinical practice and patient well-being.
Collapse
Affiliation(s)
- Rong Li
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| | - Zong Miao
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| | - Yu'e Liu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Xiao Chen
- Department of OrthopedicsXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
- Institute of Translational MedicineShanghai UniversityShanghai200444China
- Organoid Research CenterShanghai UniversityShanghai200444China
| | - Hongxiang Wang
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| | - Jiacan Su
- Department of OrthopedicsXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
- Institute of Translational MedicineShanghai UniversityShanghai200444China
- Organoid Research CenterShanghai UniversityShanghai200444China
| | - Juxiang Chen
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| |
Collapse
|
4
|
Wang YT, Therriault J, Tissot C, Servaes S, Rahmouni N, Macedo AC, Fernandez-Arias J, Mathotaarachchi SS, Stevenson J, Lussier FZ, Benedet AL, Pascoal TA, Ashton NJ, Zetterberg H, Blennow K, Gauthier S, Rosa-Neto P. Hormone therapy is associated with lower Alzheimer's disease tau biomarkers in post-menopausal females -evidence from two independent cohorts. Alzheimers Res Ther 2024; 16:162. [PMID: 39034389 PMCID: PMC11265084 DOI: 10.1186/s13195-024-01509-5] [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: 10/11/2023] [Accepted: 06/20/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Females represent approximately 70% of the Alzheimer's disease (AD) cases and the literature has proposed a connection between the decreased estrogen levels during menopause and an increased AD risk. Previous investigations have predominantly focused on assessing how hormone therapy (HT) affects the likelihood of AD development and cognitive deterioration. However, as the research framework has shifted toward a biomarker-defined AD and alterations in specific biomarkers could take place years before cognitive decline becomes discernible, it is crucial to examine how HT influences AD biomarkers. The main goal of this study was to evaluate the impact of HT on AD biomarker-informed pathophysiology in both cognitively unimpaired (CU) and cognitively impaired (CI) post-menopausal females across the aging and AD spectrum. METHODS This cross-sectional study included post-menopausal females without HT history (HT-) and with HT (HT+) at the time of PET imaging assessment from two cohorts: the Translational Biomarkers in Aging and Dementia (TRIAD) cohort, and the Alzheimer's Disease Neuroimaging Initiative (ADNI). Participants underwent magnetic resonance imaging (MRI), positron emission tomography (PET) and biofluid collection. Voxel-based t-tests were performed to assess the differences in amyloid-β (Aβ) and tau neurofibrillary tangles (NFTs) loads between HT- and HT + females. Linear regression models with interaction terms were also conducted to examine the interactive effects of HT and Aβ-PET on regional tau-PET. RESULTS HT + females demonstrated significantly lower tau-PET standardized uptake value ratio (SUVR) in Braak I-II ROIs (P < 0.05, Hedges' g = 0.73), Braak III-IV ROIs (P < 0.0001, Hedges' g = 0.74) and Braak V-VI ROIs (P < 0.0001, Hedges' g = 0.69) compared to HT- females. HT + females also showed significantly lower CSF p-tau181 (P < 0.001) and plasma p-tau181 (P < 0.0001) concentrations. Additionally, results from multivariate linear regression models indicated that HT interacts with cortical Aβ and is associated with lower regional NFT load. CONCLUSIONS Overall, findings from this observational study suggest that HT is associated with lower tau neuroimaging and fluid biomarkers in postmenopausal females. Due to the close link between tau and cognition, this study highlights the need for large randomized controlled trials designed to systemically study the influences of HT on AD biomarkers and disease progression.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Sulantha S Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Firoza Z Lussier
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada.
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada.
- Montreal Neurological Institute, Montreal, QC, Canada.
- The McGill University Research Centre for Studies in Aging, 6875 LaSalle Boulevard, H4H 1R3, Montreal, QC, Canada.
| |
Collapse
|
5
|
German‐Castelan L, Shanks HRC, Gros R, Saito T, Saido TC, Saksida LM, Bussey TJ, Prado MAM, Schmitz TW, Prado VF. Sex-dependent cholinergic effects on amyloid pathology: A translational study. Alzheimers Dement 2024; 20:995-1012. [PMID: 37846816 PMCID: PMC10916951 DOI: 10.1002/alz.13481] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/05/2023] [Accepted: 08/17/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION About two-thirds of Alzheimer's Disease (AD) patients are women, who exhibit more severe pathology and cognitive decline than men. Whether biological sex causally modulates the relationship between cholinergic signaling and amyloid pathology remains unknown. METHODS We quantified amyloid beta (Aβ) in male and female App-mutant mice with either decreased or increased cholinergic tone and examined the impact of ovariectomy and estradiol replacement in this relationship. We also investigated longitudinal changes in basal forebrain (cholinergic function) and Aβ in elderly individuals. RESULTS We show a causal relationship between cholinergic tone and amyloid pathology in males and ovariectomized female mice, which is decoupled in ovary-intact and ovariectomized females receiving estradiol. In elderly humans, cholinergic loss exacerbates Aβ. DISCUSSION Our findings emphasize the importance of reflecting human menopause in mouse models. They also support a role for therapies targeting estradiol and cholinergic signaling to reduce Aβ. HIGHLIGHTS Cholinergic tone regulates amyloid beta (Aβ) pathology in males and ovariectomized female mice. Estradiol uncouples the relationship between cholinergic tone and Aβ. In elderly humans, cholinergic loss correlates with increased Aβ in both sexes.
Collapse
Affiliation(s)
- Liliana German‐Castelan
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Hayley R. C. Shanks
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Robert Gros
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of MedicineSchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Takashi Saito
- Department of Neurocognitive ScienceInstitute of Brain ScienceNagoya City University Graduate School of Medical SciencesNagoyaJapan
- Laboratory for Proteolytic NeuroscienceRIKEN Center for Brain ScienceWako, SaitamaJapan
| | - Takaomi C. Saido
- Laboratory for Proteolytic NeuroscienceRIKEN Center for Brain ScienceWako, SaitamaJapan
| | - Lisa M. Saksida
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
| | - Timothy J. Bussey
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
| | - Marco A. M. Prado
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Taylor W. Schmitz
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
- Lawson Health Research InstituteSt. Joseph's HospitalLondonOntarioCanada
| | - Vania F. Prado
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
| | | |
Collapse
|
6
|
Wong GRM, Lee EJA, Liaw QY, Rajaram H. The role of oestrogen therapy in reducing risk of Alzheimer's disease: systematic review. BJPsych Open 2023; 9:e194. [PMID: 37846476 PMCID: PMC10594166 DOI: 10.1192/bjo.2023.579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 08/07/2023] [Accepted: 08/29/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Studies have shown a relationship between oestrogen and Alzheimer's disease. However, there is neither clear nor strong evidence on the use of oestrogen-only therapy in reducing the risk of Alzheimer's disease. AIMS To assess the effects of oestrogen-only therapy on reducing the risk of Alzheimer's disease. METHOD Inclusion criteria was determined with the PICO framework. Outcome was cognitive function measured by neuropsychological tests and strict protocols. Exclusion criteria included non-Alzheimer's dementia, progesterone-only therapy and pre-menopausal women. Searches were conducted in nine electronic healthcare databases, last searched in July 2022. Quality assessments conducted on randomised controlled trials (RCTs) were performed with the GRADE assessment, and cohort studies and case-control studies were assessed with the Newcastle-Ottawa Scale. Extracted data were used to analyse participants, interventions and outcomes. RESULTS Twenty-four studies satisfied the search criteria (four RCTs, nine cohort studies, 11 case-control studies). Fifteen studies showed positive associations for oestrogen-only therapy reducing the risk of Alzheimer's disease, and the remaining nine found no evidence of association. CONCLUSIONS Fifteen studies showed that oestrogen-only therapy effectively reduced the risk of Alzheimer's disease, whereas nine showed no correlation. Studies also investigated oestrogen-related variables such as length of oestrogen exposure, being an apolipoprotein E ε4 carrier and concomitant use of non-steroidal anti-inflammatory drugs, and their role in neuroprotection. This review was limited by the limited ranges of duration of oestrogen treatment and type of oestrogen-only therapy used. In conclusion, oestrogen-only therapy has potential for use in preventing Alzheimer's disease, although current evidence is inconclusive and requires further study.
Collapse
|
7
|
Ali N, Sohail R, Jaffer SR, Siddique S, Kaya B, Atowoju I, Imran A, Wright W, Pamulapati S, Choudhry F, Akbar A, Khawaja UA. The Role of Estrogen Therapy as a Protective Factor for Alzheimer's Disease and Dementia in Postmenopausal Women: A Comprehensive Review of the Literature. Cureus 2023; 15:e43053. [PMID: 37680393 PMCID: PMC10480684 DOI: 10.7759/cureus.43053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023] Open
Abstract
The complete cessation of menstruation for 12 months with associated vasomotor symptoms is termed menopause. Apart from playing a role in reproduction, estrogen significantly affects the central nervous system (CNS). Population-based studies highlighted a substantial difference in the prevalence of dementia between men and women, with Alzheimer-associated dementia being more prevalent in women, indicating that estrogen deficiency might be a risk factor for neurodegenerative diseases. Patients with dementia experience a progressive decline in neurocognitive function, beginning with short-term memory loss that progresses to long-term memory loss and the inability to perform everyday activities, leading ultimately to death. There is currently no cure for dementia, so preventing or slowing the disease's progression is paramount. Accordingly, researchers have widely studied the role of estrogen as a neuroprotective agent. Estrogen prevents dementia by augmenting Hippocampal and prefrontal cortex function, reducing neuroinflammation, preventing degradation of estrogen receptors, decreasing oxidative damage to the brain, and increasing cholinergic and serotonergic function. According to the window phase hypothesis, estrogen's effect on preventing dementia is more pronounced if therapy is started early, during the first five years of menopause. Other studies like The Woman's Health Initiative Memory Study (WHIMS) showed unfavorable effects of estrogen on the brain. This review aims to establish an understanding of the currently available data on estrogen's effect on neurodegeneration, namely, dementia and Alzheimer's disease.
Collapse
Affiliation(s)
- Noor Ali
- Obstetrics and Gynecology, Thumbay University Hospital, Ajman, ARE
- General Physician, Dubai Medical College, DXB, ARE
| | - Rohab Sohail
- Internal Medicine, Quaid-e-Azam Medical College, Bahawalpur, PAK
| | | | - Sadia Siddique
- Gastroenterology, Blackpool Victoria Hospital National Health Services (NHS) Foundation Trust, Blackpool, GBR
| | - Berfin Kaya
- Obstetrics and Gynaecology, Izmir Ataturk Research and Training Hospital, Izmir, TUR
- Obstetrics and Gynaecology, Izmir Kâtip Celebi University, Faculty of Medicine, Izmir, TUR
| | - Inioluwa Atowoju
- Obstetrics and Gynecology, Kharkiv National Medical University, Kharkiv, UKR
| | - Alizay Imran
- Surgery, Windsor University School of Medicine, Chicago, USA
| | - Whitney Wright
- Obstetrics and Gynecology, Texila American University, Georgetown, GUY
| | - Spandana Pamulapati
- Obstetrics and Gynecology, Alluri Sita Rama Raju Academy of Medical Sciences, Eluru, IND
| | - Faiza Choudhry
- Medicine and Surgery, Liaquat University of Medical and Health Sciences, Sindh, PAK
| | - Anum Akbar
- Pediatrics, University of Nebraska Medical Center, Omaha, USA
| | - Uzzam Ahmed Khawaja
- Pulmonary and Critical Care Medicine, Jinnah Medical and Dental College, Karachi, PAK
- Clinical and Translational Research, Dr Ferrer BioPharma, South Miami, USA
| |
Collapse
|
8
|
Yin Z, Wang Z, Li Y, Zhou J, Chen Z, Xia M, Zhang X, Wu J, Zhao L, Liang F. Neuroimaging studies of acupuncture on Alzheimer's disease: a systematic review. BMC Complement Med Ther 2023; 23:63. [PMID: 36823586 PMCID: PMC9948384 DOI: 10.1186/s12906-023-03888-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Acupuncture effectively improves cognitive function in Alzheimer's disease (AD). Many neuroimaging studies have found significant brain alterations after acupuncture treatment of AD, but the underlying central modulation mechanism is unclear. OBJECTIVE This review aims to provide neuroimaging evidence to understand the central mechanisms of acupuncture in patients with AD. METHODS Relevant neuroimaging studies about acupuncture for AD were retrieved from eight English and Chinese medicine databases (PubMed, Embase, Cochrane Library, Web of Science, SinoMed, CNKI, WF, VIP) and other resources from inception of databases until June 1, 2022, and their methodological quality was assessed using RoB 2.0 and ROBINS - I. Brain neuroimaging information was extracted to investigate the potential neural mechanism of acupuncture for AD. Descriptive statistics were used for data analysis. RESULTS Thirteen neuroimaging studies involving 275 participants were included in this review, and the overall methodological quality of included studies was moderate. The approaches applied included task-state functional magnetic resonance imaging (ts-fMRI; n = 9 studies) and rest-state functional magnetic resonance imaging (rs-fMRI; n = 4 studies). All studies focused on the instant effect of acupuncture on the brains of AD participants, including the cingulate gyrus, middle frontal gyrus, and cerebellum, indicating that acupuncture may regulate the default mode, central executive, and frontoparietal networks. CONCLUSION This study provides evidence of the neural mechanisms underlying the effect of acupuncture on AD involving cognitive- and motor-associated networks. However, this evidence is still in the preliminary investigation stage. Large-scale, well-designed, multimodal neuroimaging trials are still required to provide comprehensive insight into the central mechanism underlying the effect of acupuncture on AD. (Systematic review registration at PROSPERO, No. CRD42022331527).
Collapse
Affiliation(s)
- Zihan Yin
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Ziqi Wang
- the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Yaqin Li
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Zhou
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenghong Chen
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Manze Xia
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Xinyue Zhang
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Jiajing Wu
- School of Nursing, Zunyi Medical University, Zunyi, China
| | - Ling Zhao
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China.
| | - Fanrong Liang
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China.
| |
Collapse
|
9
|
Herman D, Andrea V, Pablo L, Simone L, Andrea B, Nicholas A, Enrica C, Henrik Z, Kaj B, Eugeen V, Harald H. Menopause hormone therapy significantly alters pathophysiological biomarkers of Alzheimer's disease. Alzheimers Dement 2022; 19:1320-1330. [PMID: 36218064 DOI: 10.1002/alz.12759] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 11/09/2022]
Abstract
INTRODUCTION This increasing body of literature indicates that menopause hormonal replacement therapy (MHT) may substantially mitigate the risk of developing late-life cognitive decline due to progressive Alzheimer's disease (AD) pathophysiology. For the first time, we investigated the question whether MHT impacts AD biomarker-informed pathophysiological dynamics in de-novo diagnosed menopausal women. METHODS We analyzed baseline and longitudinal differences between MHT-taking and -not women in terms of concentrations of core pathophysiological AD plasma biomarkers, validated in symptomatic and cognitively healthy individuals, including biomarkers of (1) the amyloid-β (Aβ) pathway, (2) tau pathophysiology, (3) neuronal loss, and (4) axonal damage and neurodegeneration. RESULTS We report a prominent and significant treatment response at the Aβ pathway biomarker level. Women at genetic risk for AD (APOE e4 allele carriers) have particularly shown favorable results from treatment. DISCUSSION To our knowledge, we present first prospective clinical evidence on effects of MHT on AD pathophysiology during menopause.
Collapse
Affiliation(s)
- Depypere Herman
- Department of Gynecology, Breast and Menopause Clinic University Hospital, Coupure Menopause Centre Ghent Belgium
| | - Vergallo Andrea
- Sorbonne University, Alzheimer Precision Medicine (APM), AP‐HP, Pitié‐Salpêtrière Hospital Boulevard de l'hôpital Paris France
| | - Lemercier Pablo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP‐HP, Pitié‐Salpêtrière Hospital Boulevard de l'hôpital Paris France
| | - Lista Simone
- Sorbonne University, Alzheimer Precision Medicine (APM), AP‐HP, Pitié‐Salpêtrière Hospital Boulevard de l'hôpital Paris France
| | - Benedet Andrea
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology the Sahlgrenska Academy, University of Gothenburg Gothenburg Sweden
| | - Ashton Nicholas
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology the Sahlgrenska Academy, University of Gothenburg Gothenburg Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology the Sahlgrenska Academy, University of Gothenburg Gothenburg Sweden
- King's College London, Institute of Psychiatry, Psychology & Neuroscience Maurice Wohl Clinical Neuroscience Institute London UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation London UK
| | - Cavedo Enrica
- Sorbonne University, Alzheimer Precision Medicine (APM), AP‐HP, Pitié‐Salpêtrière Hospital Boulevard de l'hôpital Paris France
| | - Zetterberg Henrik
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology the Sahlgrenska Academy, University of Gothenburg Gothenburg Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Mölndal Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology Queen Square London UK
- UK Dementia Research Institute at UCL London UK
- Hong Kong Center for Neurodegenerative Diseases Hong Kong China
| | - Blennow Kaj
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology the Sahlgrenska Academy, University of Gothenburg Gothenburg Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Mölndal Sweden
| | | | - Hampel Harald
- Sorbonne University, Alzheimer Precision Medicine (APM), AP‐HP, Pitié‐Salpêtrière Hospital Boulevard de l'hôpital Paris France
| | | |
Collapse
|
10
|
Jett S, Schelbaum E, Jang G, Boneu Yepez C, Dyke JP, Pahlajani S, Diaz Brinton R, Mosconi L. Ovarian steroid hormones: A long overlooked but critical contributor to brain aging and Alzheimer's disease. Front Aging Neurosci 2022; 14:948219. [PMID: 35928995 PMCID: PMC9344010 DOI: 10.3389/fnagi.2022.948219] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/28/2022] [Indexed: 01/19/2023] Open
Abstract
Ovarian hormones, particularly 17β-estradiol, are involved in numerous neurophysiological and neurochemical processes, including those subserving cognitive function. Estradiol plays a key role in the neurobiology of aging, in part due to extensive interconnectivity of the neural and endocrine system. This aspect of aging is fundamental for women's brains as all women experience a drop in circulating estradiol levels in midlife, after menopause. Given the importance of estradiol for brain function, it is not surprising that up to 80% of peri-menopausal and post-menopausal women report neurological symptoms including changes in thermoregulation (vasomotor symptoms), mood, sleep, and cognitive performance. Preclinical evidence for neuroprotective effects of 17β-estradiol also indicate associations between menopause, cognitive aging, and Alzheimer's disease (AD), the most common cause of dementia affecting nearly twice more women than men. Brain imaging studies demonstrated that middle-aged women exhibit increased indicators of AD endophenotype as compared to men of the same age, with onset in perimenopause. Herein, we take a translational approach to illustrate the contribution of ovarian hormones in maintaining cognition in women, with evidence implicating menopause-related declines in 17β-estradiol in cognitive aging and AD risk. We will review research focused on the role of endogenous and exogenous estrogen exposure as a key underlying mechanism to neuropathological aging in women, with a focus on whether brain structure, function and neurochemistry respond to hormone treatment. While still in development, this research area offers a new sex-based perspective on brain aging and risk of AD, while also highlighting an urgent need for better integration between neurology, psychiatry, and women's health practices.
Collapse
Affiliation(s)
- Steven Jett
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Eva Schelbaum
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Grace Jang
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Camila Boneu Yepez
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Jonathan P. Dyke
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| | - Roberta Diaz Brinton
- Department of Pharmacology, University of Arizona, Tucson, AZ, United States
- Department of Neurology, University of Arizona, Tucson, AZ, United States
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
11
|
Xu W, Jiang Y, Wang N, Bai H, Xu S, Xia T, Xin H. Traditional Chinese Medicine as a Promising Strategy for the Treatment of Alzheimer's Disease Complicated With Osteoporosis. Front Pharmacol 2022; 13:842101. [PMID: 35721142 PMCID: PMC9198449 DOI: 10.3389/fphar.2022.842101] [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: 12/23/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) and osteoporosis (OP) are progressive degenerative diseases caused by multiple factors, placing a huge burden on the world. Much evidence indicates that OP is a common complication in AD patients. In addition, there is also evidence to show that patients with OP have a higher risk of AD than those without OP. This suggests that the association between the two diseases may be due to a pathophysiological link rather than one disease causing the other. Several in vitro and in vivo studies have also proved their common pathogenesis. Based on the theory of traditional Chinese medicine, some classic and specific natural Chinese medicines are widely used to effectively treat AD and OP. Current evidence also shows that these treatments can ameliorate both brain damage and bone metabolism disorder and further alleviate AD complicated with OP. These valuable therapies might provide effective and safe alternatives to major pharmacological strategies.
Collapse
Affiliation(s)
- Weifan Xu
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China.,Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yiping Jiang
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Nani Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Huanhuan Bai
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Shengyan Xu
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Tianshuang Xia
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Hailiang Xin
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| |
Collapse
|
12
|
Javed MA, Ashraf N, Saeed Jan M, Mahnashi MH, Alqahtani YS, Alyami BA, Alqarni AO, Asiri YI, Ikram M, Sadiq A, Rashid U. Structural Modification, In Vitro, In Vivo, Ex Vivo, and In Silico Exploration of Pyrimidine and Pyrrolidine Cores for Targeting Enzymes Associated with Neuroinflammation and Cholinergic Deficit in Alzheimer's Disease. ACS Chem Neurosci 2021; 12:4123-4143. [PMID: 34643082 DOI: 10.1021/acschemneuro.1c00507] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
To obtain a multipotent framework that can target simultaneously COX-2, 5-LOX, acetylcholinesterase (AChE), and butyrylcholinesterase (BChE) to treat neuroinflammation, a series of derivatives containing pyrimidine and pyrrolidine cores were rationally synthesized and evaluated. Pyrazoline-pyrimidine hybrid (23g), (3-acetylcoumarin derivative of pyrrolidin-1-yl)benzenesulfonamide (27), and tacrine derivatives of (pyrrolidin-1-yl)benzenesulfonamide (31, 38) displayed excellent in vitro COX-2 inhibition having IC50 value in the nanomolar range. Tacrine-pyrrolidine hybrids 36 and 38, and tacrine-pyrimidine hybrid (46) emerged as the most potent eeAChE inhibitors with IC50 values of 23, 16, and 2 nM, respectively. However, compounds 27, 31, and 38 possessed excellent simultaneous and balanced inhibitory activity against all of the four tested targets and thus emerged as optimal multipotent hybrid compounds among all of the synthesized series of the compounds. In the ex vivo, transgenic animal models treated with compounds 36 and 46 displayed a significant decline in both AChE and BChE potentials in the hippocampus and cortical tissues. In anti-inflammatory activities, animals treated with compounds 36 and 46 displayed a significant % inhibition of edema induced by carrageenan and arachidonic acid. Biochemical analysis and histopathological examination of mice liver indicate that tacrine derivatives are devoid of hepatotoxicity and neurotoxicity against SH-SY5Y neuroblastoma cell lines. In vivo acute toxicity study showed the safety of synthesized compounds up to 1000 mg/kg dose. The inhibitory manner of interaction of these potent drugs on all of the studied in vitro targets was confirmed by molecular docking investigations.
Collapse
Affiliation(s)
- Muhammad Aamir Javed
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, Pakistan
| | - Nighat Ashraf
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, Pakistan
| | | | - Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, 1988 Najran, Saudi Arabia
| | - Yahya S. Alqahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, 1988 Najran, Saudi Arabia
| | - Bandar A. Alyami
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, 1988 Najran, Saudi Arabia
| | - Ali O. Alqarni
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, 1988 Najran, Saudi Arabia
| | - Yahya I. Asiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, 1882 Abha, Saudi Arabia
| | - Muhammad Ikram
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, KP, Pakistan
| | - Abdul Sadiq
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, 18000 Chakdara, Dir (L), KP, Pakistan
| | - Umer Rashid
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, Pakistan
| |
Collapse
|
13
|
Gao X, Li S, Liu X, Cong C, Zhao L, Liu H, Xu L. Neuroprotective effects of Tiaogeng decoction against H 2O 2-induced oxidative injury and apoptosis in PC12 cells via Nrf2 and JNK signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114379. [PMID: 34216727 DOI: 10.1016/j.jep.2021.114379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tiaogeng decoction (TGD), a mixture of 10 traditional Chinese herbs, has been used clinically for over 30 years in treating menopause-related symptoms such as cognitive changes, mood disorders, vasomotor symptoms, and sleep disorders. These central nervous system symptoms are closely associated with declined ovarian function, which dramatically increases the risk of neurodegenerative disease. Previous studies revealed that TGD may have anti-oxidative and anti-apoptotic properties, potentially preventing neurodegenerative conditions; however, the underlying pharmacological mechanism remains unclear. AIM OF THE STUDY This study aimed to examine whether TGD could activate the Nrf2 and C-Jun N-terminal kinase (JNK) signaling pathways to effectively reduce oxidative injury and apoptosis in PC12 cells and elucidate the mechanism by which this medicine may prevent neurodegenerative disease. MATERIALS AND METHODS PC12 cells were exposed to different concentrations of TGD (125, 250, 500 μg/mL) and H2O2 (150 μM). 17β-estradiol (0.05 μg/mL) was used as the positive control. A cell counting kit-8 (CCK-8) and a lactate dehydrogenase (LDH) assay were used to detect cell viability and cytotoxicity, while Hoechst and flow cytometry were performed to evaluate apoptosis levels. Mitochondrial function was assessed by measuring mitochondrial membrane potential (MMP), and superoxide dismutase (SOD), and reactive oxygen species (ROS) levels were used to measure oxidative stress (OS). Western blot analysis was used to identify the levels of Nrf2, phospho-JNK (p-JNK), phospho-mitogen-activated protein kinase kinase 7 (p-MKK7), Kelch-like ECH-associated protein 1 (Keap1), heme oxygenase-1 (HO-1), Caspase3 (Casp3), Caspase9 (Casp9), Bax, and Bcl-2 proteins. Moreover, JNK agonist anisomycin and Nrf2 inhibitor ML385 were used to validate pathways. RESULTS TGD pretreatment significantly alleviated H2O2-induced cytotoxicity, apoptosis, MMP, and OS levels. H2O2 stimulated the activation of Nrf2 and JNK signaling pathways, but TGD increased the extent of Nrf2 antioxidant activation, decreased the activation of JNK, and eventually reversed the H2O2-induced protein expression of p-MKK7, Keap1, HO-1, Cleaved Caspase3 (CL-Casp3), Cleaved Caspase9 (CL-Casp9), Bax, and Bcl-2. CONCLUSIONS This study's findings suggest that TGD may attenuate oxidative injury and apoptosis via the Nrf2 and JNK signaling pathways, making it a potential therapeutic candidate for neurodegenerative diseases.
Collapse
Affiliation(s)
- Xianwei Gao
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Shengnan Li
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Xiaofei Liu
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Chao Cong
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Li Zhao
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Huicong Liu
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Lianwei Xu
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
14
|
The Beneficial Role of Natural Endocrine Disruptors: Phytoestrogens in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3961445. [PMID: 34527172 PMCID: PMC8437597 DOI: 10.1155/2021/3961445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with a growing incidence rate primarily among the elderly. It is a neurodegenerative, progressive disorder leading to significant cognitive loss. Despite numerous pieces of research, no cure for halting the disease has been discovered yet. Phytoestrogens are nonestradiol compounds classified as one of the endocrine-disrupting chemicals (EDCs), meaning that they can potentially disrupt hormonal balance and result in developmental and reproductive abnormalities. Importantly, phytoestrogens are structurally, chemically, and functionally akin to estrogens, which undoubtedly has the potential to be detrimental to the organism. What is intriguing, although classified as EDCs, phytoestrogens seem to have a beneficial influence on Alzheimer's disease symptoms and neuropathologies. They have been observed to act as antioxidants, improve visual-spatial memory, lower amyloid-beta production, and increase the growth, survival, and plasticity of brain cells. This review article is aimed at contributing to the collective understanding of the role of phytoestrogens in the prevention and treatment of Alzheimer's disease. Importantly, it underlines the fact that despite being EDCs, phytoestrogens and their use can be beneficial in the prevention of Alzheimer's disease.
Collapse
|
15
|
Rani R, Kumar A, Jaggi AS, Singh N. Pharmacological investigations on efficacy of Phlorizin a sodium-glucose co-transporter (SGLT) inhibitor in mouse model of intracerebroventricular streptozotocin induced dementia of AD type. J Basic Clin Physiol Pharmacol 2021; 32:1057-1064. [PMID: 33548170 DOI: 10.1515/jbcpp-2020-0330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/09/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The study has been commenced to discover the potential of Phlorizin (dual SGLT inhibitor) in streptozotocin induced dementia of Alzheimer's disease (AD) type. MATERIAL AND METHODS Injection of Streptozotocin (STZ) was given via i.c.v. route (3 mg/kg) to induce dementia of Alzheimer's type. In these animals learning and memory was evaluated using Morris water maze (MWM) test. Glutathione (GSH) and thiobarbituric acid reactive species (TBARS) level was quantified to evaluate the oxidative stress; cholinergic activity of brain was estimated in term of acetylcholinesterase (AChE) activity; and the levels of myeloperoxidase (MPO) were measured as inflammation marker. RESULTS The mice model had decreased performance in MWM, representing impairment of cognitive functions. Biochemical evaluation showed rise in TBARS level, MPO and AChE activity, and fall in GSH level. The histopathological study revealed severe infiltration of neutrophils. In the study, Phlorizin/Donepezil (serving as positive control) treatment mitigate streptozotocin induced cognitive decline, histopathological changes and biochemical alterations. CONCLUSIONS The results suggest that Phlorizin decreased cognitive function via its anticholinesterase, antioxidative, antiinflammatory effects and probably through SGLT inhibitory action. It can be conferred that SGLTs can be an encouraging target for the treatment of dementia of AD.
Collapse
Affiliation(s)
- Reena Rani
- CNS Research Lab., Pharmacology Division, Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala, Punjab, India
| | - Amit Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Amteshwar Singh Jaggi
- CNS Research Lab., Pharmacology Division, Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala, Punjab, India
| | - Nirmal Singh
- CNS Research Lab., Pharmacology Division, Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala, Punjab, India
| |
Collapse
|
16
|
Abstract
OBJECTIVE Prevalence of Alzheimer's disease (AD) is higher for women, possibly influenced by sex-dependent effects of the estrogen. We examined the association between estrogen and cognitive decline in over 2,000 older adult women in a 12-year population-based study in Cache County, Utah. METHODS The baseline sample included 2,114 women (mean age = 74.94 y, SD = 6.71) who were dementia-free at baseline and completed a women's health questionnaire, asking questions regarding reproductive history and hormone therapy (HT). Endogenous estrogen exposure (EEE) was calculated taking the reproductive window (age at menarche to age at menopause), adjusted for pregnancy and breastfeeding. HT variables included duration of use, HT type (unopposed; opposed), and time of HT initiation. A modified version of the Mini-Mental State Examination (3MS) was administered at four triennial waves to assess cognitive status. Linear mixed-effects models examined the relationship between estrogen exposure and 3MS score over time. RESULTS EEE was positively associated with cognitive status (β = 0.03, P = 0.054). In addition, longer duration of HT use was positively associated with cognitive status (β = 0.02, P = 0.046) and interacted with age; older women had greater benefit compared with younger women. The timing of HT initiation was significantly associated with 3MS (β = 0.55, P = 0.048), with higher scores for women who initiated HT within 5 years of menopause compared with those initiating HT 6-or-more years later. CONCLUSIONS Our results suggest that longer EEE and HT use, especially in older women, are associated with higher cognitive status in late life.
Collapse
|
17
|
Christensen A, Liu J, Pike CJ. Aging Reduces Estradiol Protection Against Neural but Not Metabolic Effects of Obesity in Female 3xTg-AD Mice. Front Aging Neurosci 2020; 12:113. [PMID: 32431604 PMCID: PMC7214793 DOI: 10.3389/fnagi.2020.00113] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/03/2020] [Indexed: 12/29/2022] Open
Abstract
Vulnerability to Alzheimer's disease (AD) is increased by several risk factors, including midlife obesity, female sex, and the depletion of estrogens in women as a consequence of menopause. Conversely, estrogen-based hormone therapies have been linked with protection from age-related increases in adiposity and dementia risk, although treatment efficacy appears to be affected by the age of initiation. Potential interactions between obesity, AD, aging, and estrogen treatment are likely to have significant impact on optimizing the use of hormone therapies in postmenopausal women. In the current study, we compared how treatment with the primary estrogen, 17β-estradiol (E2), affects levels of AD-like neuropathology, behavioral impairment, and other neural and systemic effects of preexisting diet-induced obesity in female 3xTg-AD mice. Importantly, experiments were conducted at chronological ages associated with both the early and late stages of reproductive senescence. We observed that E2 treatment was generally associated with significantly improved metabolic outcomes, including reductions in body weight, adiposity, and leptin, across both age groups. Conversely, neural benefits of E2 in obese mice, including decreased β-amyloid burden, improved behavioral performance, and reduced microglial activation, were observed only in the early aging group. These results are consistent with the perspective that neural benefits of estrogen-based therapies require initiation of treatment during early rather than later phases of reproductive aging. Further, the discordance between E2 protection against systemic versus neural effects of obesity across age groups suggests that pathways other than general metabolic function, perhaps including reduced microglial activation, contribute to the mechanism(s) of the observed E2 actions. These findings reinforce the potential systemic and neural benefits of estrogen therapies against obesity, while also highlighting the critical role of aging as a mediator of estrogens' protective actions.
Collapse
Affiliation(s)
| | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
18
|
Ferretti MT, Martinkova J, Biskup E, Benke T, Gialdini G, Nedelska Z, Rauen K, Mantua V, Religa D, Hort J, Santuccione Chadha A, Schmidt R. Sex and gender differences in Alzheimer's disease: current challenges and implications for clinical practice: Position paper of the Dementia and Cognitive Disorders Panel of the European Academy of Neurology. Eur J Neurol 2020; 27:928-943. [PMID: 32056347 DOI: 10.1111/ene.14174] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/11/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is characterized by high heterogeneity in disease manifestation, progression and risk factors. High phenotypic variability is currently regarded as one of the largest hurdles in early diagnosis and in the design of clinical trials; there is therefore great interest in identifying factors driving variability that can be used for patient stratification. In addition to genetic and lifestyle factors, the individual's sex and gender are emerging as crucial drivers of phenotypic variability. Evidence exists on sex and gender differences in the rate of cognitive deterioration and brain atrophy, and in the effect of risk factors as well as in the patterns of diagnostic biomarkers. Such evidence might be of high relevance and requires attention in clinical practice and clinical trials. However, sex and gender differences are currently seldom appreciated; importantly, consideration of sex and gender differences is not currently a focus in the design and analysis of clinical trials for AD. The objective of this position paper is (i) to provide an overview of known sex and gender differences that might have implications for clinical practice, (ii) to identify the most important knowledge gaps in the field (with a special regard to clinical trials) and (iii) to provide conclusions for future studies. This scientific statement is endorsed by the European Academy of Neurology.
Collapse
Affiliation(s)
- M T Ferretti
- Institute for Regenerative Medicine - IREM, University of Zurich, Zurich, Switzerland.,Women's Brain Project, Guntershausen, Switzerland
| | - J Martinkova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - E Biskup
- College of Fundamental Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China.,Division of Internal Medicine, University Hospital of Basel, Basel, Switzerland
| | - T Benke
- Neurology Clinic, Medical University Innsbruck, Innsbruck, Austria
| | - G Gialdini
- Neurology - Private Practice, Lucca, Italy
| | - Z Nedelska
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St Anne's University Hospital Brno, Brno, Czech Republic
| | - K Rauen
- Institute for Regenerative Medicine - IREM, University of Zurich, Zurich, Switzerland.,Women's Brain Project, Guntershausen, Switzerland.,Department of Geriatric Psychiatry, University Hospital of Psychiatry Zurich, Zurich, Switzerland
| | - V Mantua
- Italian Medicines Agency, Rome, Italy
| | - D Religa
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - J Hort
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St Anne's University Hospital Brno, Brno, Czech Republic
| | - A Santuccione Chadha
- Women's Brain Project, Guntershausen, Switzerland.,Global Medical and Scientific Affairs, Roche Diagnostics International Ltd, Rotkreuz, Switzerland
| | - R Schmidt
- Department of Neurogeriatrics, University Clinic of Neurology, Medical University Graz, Graz, Austria
| |
Collapse
|
19
|
Deems NP, Leuner B. Pregnancy, postpartum and parity: Resilience and vulnerability in brain health and disease. Front Neuroendocrinol 2020; 57:100820. [PMID: 31987814 PMCID: PMC7225072 DOI: 10.1016/j.yfrne.2020.100820] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/25/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Risk and resilience in brain health and disease can be influenced by a variety of factors. While there is a growing appreciation to consider sex as one of these factors, far less attention has been paid to sex-specific variables that may differentially impact females such as pregnancy and reproductive history. In this review, we focus on nervous system disorders which show a female bias and for which there is data from basic research and clinical studies pointing to modification in disease risk and progression during pregnancy, postpartum and/or as a result of parity: multiple sclerosis (MS), depression, stroke, and Alzheimer's disease (AD). In doing so, we join others (Shors, 2016; Galea et al., 2018a) in aiming to illustrate the importance of looking beyond sex in neuroscience research.
Collapse
Affiliation(s)
- Nicholas P Deems
- The Ohio State University, Department of Psychology, Columbus, OH, USA
| | - Benedetta Leuner
- The Ohio State University, Department of Psychology, Columbus, OH, USA.
| |
Collapse
|
20
|
Pu Z, Tang X, Fei Y, Hou Q, Lin Y, Zha X. Bone metabolic biomarkers and bone mineral density in male patients with early-stage Alzheimer's disease. Eur Geriatr Med 2020; 11:403-408. [PMID: 32297255 DOI: 10.1007/s41999-020-00289-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 01/07/2020] [Indexed: 02/01/2023]
Abstract
PURPOSE Alzheimer's disease (AD), osteoporosis, and osteopenia are the most common diseases in older individuals and share some similar pathophysiological processes of degeneration. The aim of this study is to investigate the association between bone metabolic biomarkers, bone mineral density (BMD), and early-stage AD in men. METHODS Forty-two male early-stage AD patients and 40 age-matched healthy older volunteers were enrolled. Serum calcium, osteocalcin, 1,25(OH)2D3, urine deoxypyridinoline/creatinine (DPD/Cr) ratio, urine calcium/creatinine (Ca/Cr) ratio, and BMD were measured. The correlation between early-stage AD and bone quality was evaluated. RESULTS The urine DPD/Cr, urine Ca/Cr, and serum osteocalcin levels in the early-stage AD patients were significantly higher than those in the healthy control (HC) group (P < 0.05). The BMD data showed that the cortical and total BMD at 38% of the tibial length in the early-stage AD patients were lower than those in the HC group (P < 0.05). Furthermore, there was a negative correlation between the Montreal Cognitive Assessment score and serum osteocalcin or urine DPD/Cr levels. Abnormal urine DPD/Cr, urine Ca/Cr, and cortical BMD levels were independent risk factors in male patients with early-stage AD. CONCLUSION Bone metabolic biomarkers and BMD are closely associated with early-stage AD in male patients. Our data indicated that the measurement of bone metabolic biomarkers and BMD may provide an alternative approach for screening AD patients at the early stage.
Collapse
Affiliation(s)
- Zhengping Pu
- Department of Psychogeriatrics, Kangci Hospital of Jiaxing, No. 3118 Huancheng North Road, Tongxiang, 314500, Zhejiang, China
| | - Xiaoqing Tang
- Orthopedics Department, Tongxiang Hospital of Traditional Chinese Medicine, Tongxiang, 314500, Zhejiang, China
| | - Yu'e Fei
- Department of Psychogeriatrics, Kangci Hospital of Jiaxing, No. 3118 Huancheng North Road, Tongxiang, 314500, Zhejiang, China.
| | - Qingmei Hou
- Department of Clinical Psychology, The 2nd Specialized Hospital of Hegang, Hegang, 154102, Heilongjiang, China
| | - Yong Lin
- Department of Psychogeriatrics, Kangci Hospital of Jiaxing, No. 3118 Huancheng North Road, Tongxiang, 314500, Zhejiang, China
| | - Xianyou Zha
- Department of Psychogeriatrics, Kangci Hospital of Jiaxing, No. 3118 Huancheng North Road, Tongxiang, 314500, Zhejiang, China
| |
Collapse
|
21
|
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]
|
22
|
Pertesi S, Coughlan G, Puthusseryppady V, Morris E, Hornberger M. Menopause, cognition and dementia - A review. Post Reprod Health 2019; 25:200-206. [PMID: 31690174 DOI: 10.1177/2053369119883485] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is increasing evidence that menopausal changes can have an impact on women’s cognition and potentially, the future development of dementia. In particular, the role of reduced levels of estrogen in postmenopausal changes has been linked to an increased risk of developing dementia in observational studies. Not surprisingly, this has led to several clinical trials investigating whether postmenopausal hormone replacement therapy can potentially delay/avoid cognitive changes and subsequently, the onset of dementia. However, the evidence of these trials has been mixed, with some showing positive effects while others show no or even negative effects. In the current review, we investigate this controversy further by reviewing the existing studies and trials in cognition and dementia. Based on the current evidence, we conclude that previous approaches may have used a mixture of women with different genetic risk factors for dementia which might explain these contradicting findings. Therefore, it is recommended that future interventional studies take a more personalised approach towards hormone replacement therapy use in postmenopausal women, by taking into account the women’s genetic status for dementia risk.
Collapse
Affiliation(s)
- S Pertesi
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - G Coughlan
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | | - E Morris
- Norfolk and Norwich University Hospital, Norwich, UK
| | - M Hornberger
- Norwich Medical School, University of East Anglia, Norwich, UK.,Norfolk and Suffolk Foundation Trust, Norwich, UK
| |
Collapse
|
23
|
Marongiu R. Accelerated Ovarian Failure as a Unique Model to Study Peri-Menopause Influence on Alzheimer's Disease. Front Aging Neurosci 2019; 11:242. [PMID: 31551757 PMCID: PMC6743419 DOI: 10.3389/fnagi.2019.00242] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
Despite decades of extensive research efforts, efficacious therapies for Alzheimer's disease (AD) are lacking. The multi-factorial nature of AD neuropathology and symptomatology has taught us that a single therapeutic approach will most likely not fit all. Women constitute ~70% of the affected AD population, and pathology and rate of symptoms progression are 2-3 times higher in women than men. Epidemiological data suggest that menopausal estrogen loss may be causative of the more severe symptoms observed in AD women, however, results from clinical trials employing estrogen replacement therapy are inconsistent. AD pathological hallmarks-amyloid β (Aβ), neurofibrillary tangles (NFTs), and chronic gliosis-are laid down during a 20-year prodromal period before clinical symptoms appear, which coincides with the menopause transition (peri-menopause) in women (~45-54-years-old). Peri-menopause is marked by widely fluctuating estrogen levels resulting in periods of irregular hormone-receptor interactions. Recent studies showed that peri-menopausal women have increased indicators of AD phenotype (brain Aβ deposition and hypometabolism), and peri-menopausal women who used hormone replacement therapy (HRT) had a reduced AD risk. This suggests that neuroendocrine changes during peri-menopause may be a trigger that increases risk of AD in women. Studies on sex differences have been performed in several AD rodent models over the years. However, it has been challenging to study the menopause influence on AD due to lack of optimal models that mimic the human process. Recently, the rodent model of accelerated ovarian failure (AOF) was developed, which uniquely recapitulates human menopause, including a transitional peri-AOF period with irregular estrogen fluctuations and a post-AOF stage with low estrogen levels. This model has proven useful in hypertension and cognition studies with wild type animals. This review article will highlight the molecular mechanisms by which peri-menopause may influence the female brain vulnerability to AD and AD risk factors, such as hypertension and apolipoprotein E (APOE) genotype. Studies on these biological mechanisms together with the use of the AOF model have the potential to shed light on key molecular pathways underlying AD pathogenesis for the development of precision medicine approaches that take sex and hormonal status into account.
Collapse
Affiliation(s)
- Roberta Marongiu
- Laboratory of Molecular Neurosurgery, Weill Cornell Medicine, Department of Neurosurgery, Cornell University, New York, NY, United States
| |
Collapse
|
24
|
Neues und praktisch Relevantes der gynäkologischen Endokrinologie, Reproduktionsmedizin und Pränatalmedizin – Teil 2. GYNAKOLOGISCHE ENDOKRINOLOGIE 2019. [DOI: 10.1007/s10304-019-0253-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Abstract
Alzheimer disease (AD) is characterized by wide heterogeneity in cognitive and behavioural syndromes, risk factors and pathophysiological mechanisms. Addressing this phenotypic variation will be crucial for the development of precise and effective therapeutics in AD. Sex-related differences in neural anatomy and function are starting to emerge, and sex might constitute an important factor for AD patient stratification and personalized treatment. Although the effects of sex on AD epidemiology are currently the subject of intense investigation, the notion of sex-specific clinicopathological AD phenotypes is largely unexplored. In this Review, we critically discuss the evidence for sex-related differences in AD symptomatology, progression, biomarkers, risk factor profiles and treatment. The cumulative evidence reviewed indicates sex-specific patterns of disease manifestation as well as sex differences in the rates of cognitive decline and brain atrophy, suggesting that sex is a crucial variable in disease heterogeneity. We discuss critical challenges and knowledge gaps in our current understanding. Elucidating sex differences in disease phenotypes will be instrumental in the development of a 'precision medicine' approach in AD, encompassing individual, multimodal, biomarker-driven and sex-sensitive strategies for prevention, detection, drug development and treatment.
Collapse
|
26
|
Abstract
There are 3 common physiological estrogens, of which estradiol (E2) is seen to decline rapidly over the menopausal transition. This decline in E2 has been associated with a number of changes in the brain, including cognitive changes, effects on sleep, and effects on mood. These effects have been demonstrated in both rodent and non-human preclinical models. Furthermore, E2 interactions have been indicated in a number of neuropsychiatric disorders, including Alzheimer's disease, schizophrenia, and depression. In normal brain aging, there are a number of systems that undergo changes and a number of these show interactions with E2, particularly the cholinergic system, the dopaminergic system, and mitochondrial function. E2 treatment has been shown to ameliorate some of the behavioral and morphological changes seen in preclinical models of menopause; however, in clinical populations, the effects of E2 treatment on cognitive changes after menopause are mixed. The future use of sex hormone treatment will likely focus on personalized or precision medicine for the prevention or treatment of cognitive disturbances during aging, with a better understanding of who may benefit from such treatment.
Collapse
Affiliation(s)
- Jason K Russell
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
| | - Paul A Newhouse
- Center for Cognitive Medicine, Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, 37212, USA.
- Geriatric Research, Education, and Clinical Center (GRECC), Tennessee VA Health Systems, Nashville, TN, 37212, USA.
| |
Collapse
|
27
|
Li Z, Yang N, Lei X, Lin C, Li N, Jiang X, Wei X, Xu B. The association between the ApoE polymorphisms and the MRI-defined intracranial lesions in a cohort of southern China population. J Clin Lab Anal 2019; 33:e22950. [PMID: 31199015 PMCID: PMC6757122 DOI: 10.1002/jcla.22950] [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] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 05/13/2019] [Accepted: 05/23/2019] [Indexed: 11/20/2022] Open
Abstract
Background The apolipoprotein E (APOE) ε4 allele is considered as a risk factor for Alzheimer's disease (AD). However, the association of APOE allele with MRI evidence of intracranial lesions has not been well understood. Methods Quantitative real‐time PCR was performed to detect the APOE genotype; MRI was examined for intracranial lesions. Their association was evaluated in a cohort of 226 AD patients and 2607 healthy individuals in southern China. Results The frequencies of ε2, ε3, and ε4 alleles were 8.0%, 82.9%, and 9.1% in the whole study population. The frequency of APOE‐ε4 allele was significantly higher in the AD subjects than that in the control group (14.4% vs 8.6%, P < 0.001). We found that brain atrophy occurred at a rate of 12.3% in ε4 allele group vs 8.5% in non‐ε4 genotype group, with a significance of P = 0.008. Severe brain atrophy occurred at a rate of 1.0% in ε4 allele group vs 0.2% in non‐ε4 genotype group (P = 0.011). The individuals carrying APOE ε4/ε4 had an odds ratio (OR) of 7.64 (P < 0.01) for developing AD, while the APOE ε3/ε4 gene carriers had an OR of 1.47 (P = 0.031) and the OR in APOE ε2/ε3 carriers is 0.81 (P = 0.372). Interestingly, we found that the risk of ε4/ε4 allele carrier developing AD was significantly higher in male (P < 0.001) than female (P = 0.478). Conclusion Compared to ε2 and ε3 alleles, the presence of APOE‐ε4 allele might increase the risk for AD in a dose‐dependent manner in southern China. Moreover, the presence of APOE‐ε4 allele results in a higher incidence of brain atrophy.
Collapse
Affiliation(s)
- Zhuoran Li
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Na Yang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiuxia Lei
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chuying Lin
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Nan Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinqing Jiang
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xinhua Wei
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
28
|
Silva MVF, Loures CDMG, Alves LCV, de Souza LC, Borges KBG, Carvalho MDG. Alzheimer's disease: risk factors and potentially protective measures. J Biomed Sci 2019; 26:33. [PMID: 31072403 PMCID: PMC6507104 DOI: 10.1186/s12929-019-0524-y] [Citation(s) in RCA: 479] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/18/2019] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia and typically manifests through a progressive loss of episodic memory and cognitive function, subsequently causing language and visuospatial skills deficiencies, which are often accompanied by behavioral disorders such as apathy, aggressiveness and depression. The presence of extracellular plaques of insoluble β-amyloid peptide (Aβ) and neurofibrillary tangles (NFT) containing hyperphosphorylated tau protein (P-tau) in the neuronal cytoplasm is a remarkable pathophysiological cause in patients' brains. Approximately 70% of the risk of developing AD can be attributed to genetics. However, acquired factors such as cerebrovascular diseases, diabetes, hypertension, obesity and dyslipidemia increase the risk of AD development. The aim of the present minireview was to summarize the pathophysiological mechanism and the main risk factors for AD. As a complement, some protective factors associated with a lower risk of disease incidence, such as cognitive reserve, physical activity and diet will also be addressed.
Collapse
Affiliation(s)
- Marcos Vinícius Ferreira Silva
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| | - Cristina de Mello Gomide Loures
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Luan Carlos Vieira Alves
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Leonardo Cruz de Souza
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190 - Santa Efigênia, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Karina Braga Gomes Borges
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Maria das Graças Carvalho
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| |
Collapse
|
29
|
Savolainen-Peltonen H, Rahkola-Soisalo P, Hoti F, Vattulainen P, Gissler M, Ylikorkala O, Mikkola TS. Use of postmenopausal hormone therapy and risk of Alzheimer's disease in Finland: nationwide case-control study. BMJ 2019; 364:l665. [PMID: 30842086 PMCID: PMC6402043 DOI: 10.1136/bmj.l665] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To compare the use of hormone therapy between Finnish postmenopausal women with and without a diagnosis for Alzheimer's disease. DESIGN Nationwide case-control study. SETTING Finnish national population and drug register, between 1999 and 2013. PARTICIPANTS All postmenopausal women (n=84 739) in Finland who, between 1999 and 2013, received a diagnosis of Alzheimer's disease from a neurologist or geriatrician, and who were identified from a national drug register. Control women without a diagnosis (n=84 739), matched by age and hospital district, were traced from the Finnish national population register. INTERVENTIONS Data on hormone therapy use were obtained from the Finnish national drug reimbursement register. MAIN OUTCOME MEASURES Odds ratios and 95% confidence intervals for Alzheimer's disease, calculated with conditional logistic regression analysis. RESULTS In 83 688 (98.8%) women, a diagnosis for Alzheimer's disease was made at the age of 60 years or older, and 47 239 (55.7%) women had been over 80 years of age at diagnosis. Use of systemic hormone therapy was associated with a 9-17% increased risk of Alzheimer's disease. The risk of the disease did not differ significantly between users of estradiol only (odds ratio 1.09, 95% confidence interval 1.05 to 1.14) and those of oestrogen-progestogen (1.17, 1.13 to 1.21). The risk increases in users of oestrogen-progestogen therapy were not related to different progestogens (norethisterone acetate, medroxyprogesterone acetate, or other progestogens); but in women younger than 60 at hormone therapy initiation, these risk increases were associated with hormone therapy exposure over 10 years. Furthermore, the age at initiation of systemic hormone therapy was not a decisive determinant for the increase in risk of Alzheimer's disease. The exclusive use of vaginal estradiol did not affect the risk of the disease (0.99, 0.96 to 1.01). CONCLUSIONS Long term use of systemic hormone therapy might be accompanied with an overall increased risk of Alzheimer's disease, which is not related to the type of progestogen or the age at initiation of systemic hormone therapy. By contrast, use of vaginal estradiol shows no such risk. Even though the absolute risk increase for Alzheimer's disease is small, our data should be implemented into information for present and future users of hormone therapy.
Collapse
Affiliation(s)
- Hanna Savolainen-Peltonen
- University of Helsinki and Helsinki University Hospital, Obstetrics and Gynecology, Haartmaninkatu 2, PO Box 140, FIN-00029 HUS, 00029 Helsinki, Finland
- Folkhälsan Research Center, Biomedicum, Helsinki, Finland
| | - Päivi Rahkola-Soisalo
- University of Helsinki and Helsinki University Hospital, Obstetrics and Gynecology, Haartmaninkatu 2, PO Box 140, FIN-00029 HUS, 00029 Helsinki, Finland
| | | | | | - Mika Gissler
- National Institute for Health and Welfare, Helsinki, Finland
- Karolinska Institute, Department of Neurobiology, Care Sciences and Society, Division of Family Medicine, Huddinge, Sweden
- University of Turku, Research Centre for Child Psychiatry, Turku, Finland
| | - Olavi Ylikorkala
- University of Helsinki and Helsinki University Hospital, Obstetrics and Gynecology, Haartmaninkatu 2, PO Box 140, FIN-00029 HUS, 00029 Helsinki, Finland
| | - Tomi S Mikkola
- University of Helsinki and Helsinki University Hospital, Obstetrics and Gynecology, Haartmaninkatu 2, PO Box 140, FIN-00029 HUS, 00029 Helsinki, Finland
- Folkhälsan Research Center, Biomedicum, Helsinki, Finland
| |
Collapse
|
30
|
Rezzani R, Franco C, Rodella LF. Sex differences of brain and their implications for personalized therapy. Pharmacol Res 2019; 141:429-442. [PMID: 30659897 DOI: 10.1016/j.phrs.2019.01.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 01/06/2023]
Abstract
Nowadays, it is known that the sex differences regard many organs, e.g., liver, vessels, pancreas, lungs, bronchi and also the brain. Sex differences are not just a matter of ethical and moral principles, as they are central to explain many still unknown diseases and their understanding is a prerequisite to develop an effective therapy for each individual. This review reports on those sex differences that are not only macroscopic and morphological, but also involve molecular and functional dimorphism in the brain. It will recapitulate the main structural differences between male and female brain including the neurotransmission systems; in particular, the main objective is to identify a correlation, already known or to be investigated in the future, between the differences that characterize male and female brains from a morphological and biochemical point of view and neurological syndromes. This correlation could provide a starting point for future scientific research aimed to investigate and define a personalized therapy.
Collapse
Affiliation(s)
- Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| | - Caterina Franco
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Luigi F Rodella
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
31
|
Speth RC, D'Ambra M, Ji H, Sandberg K. A heartfelt message, estrogen replacement therapy: use it or lose it. Am J Physiol Heart Circ Physiol 2018; 315:H1765-H1778. [PMID: 30216118 PMCID: PMC6336974 DOI: 10.1152/ajpheart.00041.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 12/24/2022]
Abstract
The issue of cardiovascular and cognitive health in women is complex. During the premenopausal phase of life, women have healthy blood pressure levels that are lower than those of age-matched men, and they have less cardiovascular disease. However, in the postmenopausal stage of life, blood pressure in women increases, and they are increasingly susceptible to cardiovascular disease, cognitive impairments, and dementia, exceeding the incidence in men. The major difference between pre- and postmenopausal women is the loss of estrogen. Thus, it seemed logical that postmenopausal estrogen replacement therapy, with or without progestin, generally referred to as menopausal hormone treatment (MHT), would prevent these adverse sequelae. However, despite initially promising results, a major randomized clinical trial refuted the benefits of MHT, leading to its falling from favor. However, reappraisal of this study in the framework of a "critical window," or "timing hypothesis," has changed our perspective on the benefit-to-risk ratio of MHT, and this review discusses the historical, current, and future approaches to MHT.
Collapse
Affiliation(s)
- Robert C Speth
- College of Pharmacy, Nova Southeastern University , Fort Lauderdale, Florida
- Department of Pharmacology and Physiology, College of Medicine, Georgetown University , Washington, District of Columbia
| | | | - Hong Ji
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University , Washington, District of Columbia
| | | |
Collapse
|
32
|
The effect of APOE genotype on Alzheimer's disease risk is influenced by sex and docosahexaenoic acid status. Neurobiol Aging 2018; 69:209-220. [DOI: 10.1016/j.neurobiolaging.2018.05.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/23/2018] [Accepted: 05/14/2018] [Indexed: 01/21/2023]
|
33
|
Precision medicine and drug development in Alzheimer's disease: the importance of sexual dimorphism and patient stratification. Front Neuroendocrinol 2018; 50:31-51. [PMID: 29902481 DOI: 10.1016/j.yfrne.2018.06.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/29/2018] [Accepted: 06/07/2018] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases (ND) are among the leading causes of disability and mortality. Considerable sex differences exist in the occurrence of the various manifestations leading to cognitive decline. Alzheimer's disease (AD) exhibits substantial sexual dimorphisms and disproportionately affects women. Women have a higher life expectancy compared to men and, consequently, have more lifespan to develop AD. The emerging precision medicine and pharmacology concepts - taking into account the individual genetic and biological variability relevant for disease risk, prevention, detection, diagnosis, and treatment - are expected to substantially enhance our knowledge and management of AD. Stratifying the affected individuals by sex and gender is an important basic step towards personalization of scientific research, drug development, and care. We hypothesize that sex and gender differences, extending from genetic to psychosocial domains, are highly relevant for the understanding of AD pathophysiology, and for the conceptualization of basic/translational research and for clinical therapy trial design.
Collapse
|
34
|
Hampel H, Toschi N, Babiloni C, Baldacci F, Black KL, Bokde AL, Bun RS, Cacciola F, Cavedo E, Chiesa PA, Colliot O, Coman CM, Dubois B, Duggento A, Durrleman S, Ferretti MT, George N, Genthon R, Habert MO, Herholz K, Koronyo Y, Koronyo-Hamaoui M, Lamari F, Langevin T, Lehéricy S, Lorenceau J, Neri C, Nisticò R, Nyasse-Messene F, Ritchie C, Rossi S, Santarnecchi E, Sporns O, Verdooner SR, Vergallo A, Villain N, Younesi E, Garaci F, Lista S. Revolution of Alzheimer Precision Neurology. Passageway of Systems Biology and Neurophysiology. J Alzheimers Dis 2018; 64:S47-S105. [PMID: 29562524 PMCID: PMC6008221 DOI: 10.3233/jad-179932] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The Precision Neurology development process implements systems theory with system biology and neurophysiology in a parallel, bidirectional research path: a combined hypothesis-driven investigation of systems dysfunction within distinct molecular, cellular, and large-scale neural network systems in both animal models as well as through tests for the usefulness of these candidate dynamic systems biomarkers in different diseases and subgroups at different stages of pathophysiological progression. This translational research path is paralleled by an "omics"-based, hypothesis-free, exploratory research pathway, which will collect multimodal data from progressing asymptomatic, preclinical, and clinical neurodegenerative disease (ND) populations, within the wide continuous biological and clinical spectrum of ND, applying high-throughput and high-content technologies combined with powerful computational and statistical modeling tools, aimed at identifying novel dysfunctional systems and predictive marker signatures associated with ND. The goals are to identify common biological denominators or differentiating classifiers across the continuum of ND during detectable stages of pathophysiological progression, characterize systems-based intermediate endophenotypes, validate multi-modal novel diagnostic systems biomarkers, and advance clinical intervention trial designs by utilizing systems-based intermediate endophenotypes and candidate surrogate markers. Achieving these goals is key to the ultimate development of early and effective individualized treatment of ND, such as Alzheimer's disease. The Alzheimer Precision Medicine Initiative (APMI) and cohort program (APMI-CP), as well as the Paris based core of the Sorbonne University Clinical Research Group "Alzheimer Precision Medicine" (GRC-APM) were recently launched to facilitate the passageway from conventional clinical diagnostic and drug development toward breakthrough innovation based on the investigation of the comprehensive biological nature of aging individuals. The APMI movement is gaining momentum to systematically apply both systems neurophysiology and systems biology in exploratory translational neuroscience research on ND.
Collapse
Affiliation(s)
- Harald Hampel
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Department of Radiology, “Athinoula A. Martinos” Center for Biomedical Imaging, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Claudio Babiloni
- Department of Physiology and Pharmacology “Vittorio Erspamer”, University of Rome “La Sapienza”, Rome, Italy
- Institute for Research and Medical Care, IRCCS “San Raffaele Pisana”, Rome, Italy
| | - Filippo Baldacci
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Keith L. Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Arun L.W. Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - René S. Bun
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Francesco Cacciola
- Unit of Neurosurgery, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Enrica Cavedo
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
- IRCCS “San Giovanni di Dio-Fatebenefratelli”, Brescia, Italy
| | - Patrizia A. Chiesa
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Olivier Colliot
- Inserm, U1127, Paris, France; CNRS, UMR 7225 ICM, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Paris, France; Institut du Cerveau et de la Moelle Épinière (ICM) Paris, France; Inria, Aramis project-team, Centre de Recherche de Paris, France; Department of Neuroradiology, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France; Department of Neurology, AP-HP, Hôpital de la Pitié-Salpêtrière, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Paris, France
| | - Cristina-Maria Coman
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Bruno Dubois
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
| | - Andrea Duggento
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Stanley Durrleman
- Inserm, U1127, Paris, France; CNRS, UMR 7225 ICM, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Paris, France; Institut du Cerveau et de la Moelle Épinière (ICM) Paris, France; Inria, Aramis project-team, Centre de Recherche de Paris, France
| | - Maria-Teresa Ferretti
- IREM, Institute for Regenerative Medicine, University of Zurich, Zürich, Switzerland
- ZNZ Neuroscience Center Zurich, Zürich, Switzerland
| | - Nathalie George
- Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle Épinière, ICM, Ecole Normale Supérieure, ENS, Centre MEG-EEG, F-75013, Paris, France
| | - Remy Genthon
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
| | - Marie-Odile Habert
- Département de Médecine Nucléaire, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
- Laboratoire d’Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, Inserm U 1146, CNRS UMR 7371, Paris, France
| | - Karl Herholz
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Foudil Lamari
- AP-HP, UF Biochimie des Maladies Neuro-métaboliques, Service de Biochimie Métabolique, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | | | - Stéphane Lehéricy
- Centre de NeuroImagerie de Recherche - CENIR, Institut du Cerveau et de la Moelle Épinière - ICM, F-75013, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM, F-75013, Paris, France
| | - Jean Lorenceau
- Institut de la Vision, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR_S968, CNRS UMR7210, Paris, France
| | - Christian Neri
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, CNRS UMR 8256, Institut de Biologie Paris-Seine (IBPS), Place Jussieu, F-75005, Paris, France
| | - Robert Nisticò
- Department of Biology, University of Rome “Tor Vergata” & Pharmacology of Synaptic Disease Lab, European Brain Research Institute (E.B.R.I.), Rome, Italy
| | - Francis Nyasse-Messene
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
| | - Craig Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Simone Rossi
- Department of Medicine, Surgery and Neurosciences, Unit of Neurology and Clinical Neurophysiology, Brain Investigation & Neuromodulation Lab. (Si-BIN Lab.), University of Siena, Siena, Italy
- Department of Medicine, Surgery and Neurosciences, Section of Human Physiology University of Siena, Siena, Italy
| | - Emiliano Santarnecchi
- Department of Medicine, Surgery and Neurosciences, Unit of Neurology and Clinical Neurophysiology, Brain Investigation & Neuromodulation Lab. (Si-BIN Lab.), University of Siena, Siena, Italy
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Olaf Sporns
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
- IU Network Science Institute, Indiana University, Bloomington, IN, USA
| | | | - Andrea Vergallo
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Nicolas Villain
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | | | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Casa di Cura “San Raffaele Cassino”, Cassino, Italy
| | - Simone Lista
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| |
Collapse
|
35
|
Huang X, Dong S, Zhen J, Zhang H, Lin T, Zeng Y, Van Halm-Lutterodt N, Yuan L. The Role of ApoE Polymorphism in the Relationship between Serum Steroid Hormone Levels and Cognition in Older Chinese Adults: A Cross-Sectional Study. Front Endocrinol (Lausanne) 2018; 9:71. [PMID: 29559956 PMCID: PMC5845719 DOI: 10.3389/fendo.2018.00071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/16/2018] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Epidemiology studies have indicated an association of apolipoprotein E (ApoE) genetic polymorphism and circulating steroid hormone levels with the risk of Alzheimer's disease. The established physiologic relationship between apolipoproteins and steroid hormone indicate an important role of ApoE polymorphism in impacting the relationship between serum steroid hormones and cognition in the elderly. STUDY DESIGN A total of 500 Chinese adults aged between 50 and 75 participated in this community-based cross-sectional study. Blood samples were collected in the morning for ApoE genotyping and serum parameter assessment. Cognitive performance of participants was evaluated by Montreal Cognitive Assessment test. RESULTS Age, gender, educational level, smoking, and physical activity levels are factors associated with cognitive performance in this older Chinese adults. Compared to the control subjects, MCI subjects demonstrated higher serum total cholesterol, HDL-C, and estradiol status (P < 0.05). ApoE genotype difference of serum lipid profile was observed with a relatively higher mean serum triglyceride levels in ApoE2 and ApoE4 carriers (P < 0.05), and lower mean serum HDL-C level in ApoE4 carriers (P < 0.05). Memory and delayed recall ability was serum estradiol level related; and subjects with higher circulating estradiol concentration exhibited lower memory and delayed recall ability (P < 0.05). The association of serum estradiol and cortisol concentration with cognitive performance was ApoE genotypes dependent. Poor cognitive performance was observed in ApoE2 and ApoE4 carriers with higher serum estradiol level (P < 0.05). Moreover, ApoE2 and ApoE4 carriers with higher serum cortisol status demonstrated decreased language ability (P < 0.05). Multiple logistic regression analysis indicates that subjects with higher serum estradiol status may have an increased risk for MCI [OR = 2.004, 95% confidence interval (CI): 1.135, 3.540; P = 0.017]. ApoE2 carriers with higher serum steroid levels may be potentially predisposed to an increased risk of MCI (OR = 3.353; 95% CI: 1.135, 9.907; P = 0.029). CONCLUSION Cognitive outcomes in older Chinese adults are associated with serum steroid hormone status. Higher serum steroid levels in ApoE2 carriers might pose an increased risk of MCI in the elderly.
Collapse
Affiliation(s)
- Xiaochen Huang
- School of Public Health, Capital Medical University, Beijing, China
| | - Shengqi Dong
- School of Public Health, Capital Medical University, Beijing, China
| | - Jie Zhen
- School of Public Health, Capital Medical University, Beijing, China
| | - Huiqiang Zhang
- School of Public Health, Capital Medical University, Beijing, China
| | - Tong Lin
- School of Public Health, Capital Medical University, Beijing, China
| | - Yuhong Zeng
- School of Public Health, Capital Medical University, Beijing, China
| | - Nicholas Van Halm-Lutterodt
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Keck Medical Center of USC, Department of Orthopaedics and Neurosurgery, University of Southern California, Los Angeles, CA, United States
| | - Linhong Yuan
- School of Public Health, Capital Medical University, Beijing, China
- *Correspondence: Linhong Yuan,
| |
Collapse
|
36
|
Gaignard P, Liere P, Thérond P, Schumacher M, Slama A, Guennoun R. Role of Sex Hormones on Brain Mitochondrial Function, with Special Reference to Aging and Neurodegenerative Diseases. Front Aging Neurosci 2017; 9:406. [PMID: 29270123 PMCID: PMC5725410 DOI: 10.3389/fnagi.2017.00406] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/24/2017] [Indexed: 01/18/2023] Open
Abstract
The mitochondria have a fundamental role in both cellular energy supply and oxidative stress regulation and are target of the effects of sex steroids, particularly the neuroprotective ones. Aging is associated with a decline in the levels of different steroid hormones, and this decrease may underline some neural dysfunctions. Besides, modifications in mitochondrial functions associated with aging processes are also well documented. In this review, we will discuss studies that describe the modifications of brain mitochondrial function and of steroid levels associated with physiological aging and with neurodegenerative diseases. A special emphasis will be placed on describing and discussing our recent findings concerning the concomitant study of mitochondrial function (oxidative phosphorylation, oxidative stress) and brain steroid levels in both young (3-month-old) and aged (20-month-old) male and female mice.
Collapse
Affiliation(s)
- Pauline Gaignard
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Le Kremlin-Bicêtre, France
- Biochemistry Laboratory, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Philippe Liere
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Patrice Thérond
- Biochemistry Laboratory, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Michael Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Abdelhamid Slama
- Biochemistry Laboratory, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
37
|
Merlo S, Spampinato SF, Sortino MA. Estrogen and Alzheimer's disease: Still an attractive topic despite disappointment from early clinical results. Eur J Pharmacol 2017; 817:51-58. [DOI: 10.1016/j.ejphar.2017.05.059] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/13/2017] [Accepted: 05/30/2017] [Indexed: 01/06/2023]
|
38
|
Williams CJ, Williams MG, Eynon N, Ashton KJ, Little JP, Wisloff U, Coombes JS. Genes to predict VO 2max trainability: a systematic review. BMC Genomics 2017; 18:831. [PMID: 29143670 PMCID: PMC5688475 DOI: 10.1186/s12864-017-4192-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Cardiorespiratory fitness (VO2max) is an excellent predictor of chronic disease morbidity and mortality risk. Guidelines recommend individuals undertake exercise training to improve VO2max for chronic disease reduction. However, there are large inter-individual differences between exercise training responses. This systematic review is aimed at identifying genetic variants that are associated with VO2max trainability. Methods Peer-reviewed research papers published up until October 2016 from four databases were examined. Articles were included if they examined genetic variants, incorporated a supervised aerobic exercise intervention; and measured VO2max/VO2peak pre and post-intervention. Results Thirty-five articles describing 15 cohorts met the criteria for inclusion. The majority of studies used a cross-sectional retrospective design. Thirty-two studies researched candidate genes, two used Genome-Wide Association Studies (GWAS), and one examined mRNA gene expression data, in addition to a GWAS. Across these studies, 97 genes to predict VO2max trainability were identified. Studies found phenotype to be dependent on several of these genotypes/variants, with higher responders to exercise training having more positive response alleles than lower responders (greater gene predictor score). Only 13 genetic variants were reproduced by more than two authors. Several other limitations were noted throughout these studies, including the robustness of significance for identified variants, small sample sizes, limited cohorts focused primarily on Caucasian populations, and minimal baseline data. These factors, along with differences in exercise training programs, diet and other environmental gene expression mediators, likely influence the ideal traits for VO2max trainability. Conclusion Ninety-seven genes have been identified as possible predictors of VO2max trainability. To verify the strength of these findings and to identify if there are more genetic variants and/or mediators, further tightly-controlled studies that measure a range of biomarkers across ethnicities are required.
Collapse
Affiliation(s)
- Camilla J Williams
- Centre for Research on Exercise, Physical Activity and Health (CRExPAH), School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Mark G Williams
- Molecular Genetics Department, Mater Pathology, South Brisbane, Queensland, Australia
| | - Nir Eynon
- Institute of Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, 8001, Australia.
| | - Kevin J Ashton
- Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia, Okanagan, Canada
| | - Ulrik Wisloff
- Centre for Research on Exercise, Physical Activity and Health (CRExPAH), School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia.,Cardiac K.G. Jebsen Center for Exercise in Medicine at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jeff S Coombes
- Centre for Research on Exercise, Physical Activity and Health (CRExPAH), School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
39
|
Rojas-Gutierrez E, Muñoz-Arenas G, Treviño S, Espinosa B, Chavez R, Rojas K, Flores G, Díaz A, Guevara J. Alzheimer's disease and metabolic syndrome: A link from oxidative stress and inflammation to neurodegeneration. Synapse 2017; 71:e21990. [PMID: 28650104 DOI: 10.1002/syn.21990] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and one of the most important causes of morbidity and mortality among the aging population. AD diagnosis is made post-mortem, and the two pathologic hallmarks, particularly evident in the end stages of the illness, are amyloid plaques and neurofibrillary tangles. Currently, there is no curative treatment for AD. Additionally, there is a strong relation between oxidative stress, metabolic syndrome, and AD. The high levels of circulating lipids and glucose imbalances amplify lipid peroxidation that gradually diminishes the antioxidant systems, causing high levels of oxidative metabolism that affects cell structure, leading to neuronal damage. Accumulating evidence suggests that AD is closely related to a dysfunction of both insulin signaling and glucose metabolism in the brain, leading to an insulin-resistant brain state. Four drugs are currently used for this pathology: Three FDA-approved cholinesterase inhibitors and one NMDA receptor antagonist. However, wide varieties of antioxidants are promissory to delay or prevent the symptoms of AD and may help in treating the disease. Therefore, therapeutic efforts to achieve attenuation of oxidative stress could be beneficial in AD treatment, attenuating Aβ-induced neurotoxicity and improve neurological outcomes in AD. The term inflammaging characterizes a widely accepted paradigm that aging is accompanied by a low-grade chronic up-regulation of certain pro-inflammatory responses in the absence of overt infection, and is a highly significant risk factor for both morbidity and mortality in the elderly.
Collapse
Affiliation(s)
- Eduardo Rojas-Gutierrez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Guadalupe Muñoz-Arenas
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Samuel Treviño
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Blanca Espinosa
- Departamento de Bioquímica, Instituto Nacional de Enfermedades Respiratorias-INER, Ciudad de México, Mexico
| | - Raúl Chavez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Karla Rojas
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Alfonso Díaz
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Jorge Guevara
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
40
|
Chen YH, Lo RY. Alzheimer's disease and osteoporosis. Tzu Chi Med J 2017; 29:138-142. [PMID: 28974906 PMCID: PMC5615992 DOI: 10.4103/tcmj.tcmj_54_17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/13/2017] [Accepted: 03/13/2017] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) and osteoporosis are both common degenerative diseases in the elderly population. The incidence of both diseases increases with age and will be posing enormous societal burden worldwide. It may appear that AD and osteoporosis are two distinct diseases although many risk factors are shared. Previous observational studies have shown that patients with osteoporosis have higher risks of developing AD than those who do not have osteoporosis. Although osteoporosis, falls, and fractures are more often seen in patients with AD than other older adults, the association between these two diseases may be due to a pathophysiological link rather than one condition causing the other. Several in vitro and in vivo studies lend support to this notion. Patients with AD have excessive amyloid plaques in the brain, and the pathology may extend to peripheral organs and cause skeletal amyloid deposition, which would enhance receptor activator nuclear factor-kappa B ligand signaling and lead to greater osteoclast activities. Patients with osteoporosis may have Vitamin D deficiency or lower levels of Vitamin D binding protein, which protects against amyloid aggregation, thus linking Vitamin D deficiency and AD or osteoporosis and AD. Osteoporosis coexisting with AD provides a window to examine the amyloid hypothesis from peripheral tissues. Future studies are warranted to clarify the role of genetic background regarding Vitamin D levels, exposure to sunlight, estrogen replacement therapy, and physical activity in patients with both chronic diseases.
Collapse
Affiliation(s)
- Yu-Hung Chen
- Department of Nuclear Medicine, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Raymond Y. Lo
- Department of Neurology, Memory and Aging Center, Buddhist Tzu Chi General Hospital and Tzu Chi University, Hualien, Taiwan
| |
Collapse
|