1
|
Shang Y, Torrandell‐Haro G, Vitali F, Brinton RD. Combination therapy targeting Alzheimer's disease risk factors is associated with a significant delay in Alzheimer's disease-related cognitive decline. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2025; 11:e70074. [PMID: 40151397 PMCID: PMC11947753 DOI: 10.1002/trc2.70074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) cognitive decline can be a major contributor to loss of independent living. Therapeutic strategies that alter the course of cognitive deterioration have the potential to sustain activities of daily living, promote quality of life, and delay transition to nursing-home care. METHODS We performed longitudinal linear regression analysis of National Alzheimer's Coordinating Center (NACC) cognitive data from 7653 mild dementia AD participants at baseline with at least one medication for diabetes (DBMD), lipid-lowering (LIPL), anti-hypertensive (AHTN), and non-steroidal anti-inflammatory (NSD) medications or any combination in 5684 (74%) participants and in 1969 (26%) participants with no study-relevant prescriptions over 10 years. Change in cognitive function was determined by Mini-Mental State Examination (MMSE) and CDR® Dementia Staging Instrument Sum of Boxes (CDR-SB) scores relative to non-treated participants stratified by sex and apolipoprotein E (APOE) genotype. Validation analysis was performed using Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset. RESULTS Combination of DBMD+LIPL+AHTN+NSD (QuadRx) resulted in a significant 46% MMSE and 32% CDR-SB delay in cognitive decline at 5 years, which was sustained at 10 years with a delay in decline of 47% MMSE and 33% CDR-SB. QuadRx was equally effective for the delay of cognitive decline in both females and males at 5 and 10 years. QuadRx mitigated the impact of the APOE ε4 genotype. Findings were validated in ADNI AD participants in which QuadRx was associated with a significant 60% MMSE delay in cognitive decline at 1 and 2 years. CONCLUSIONS Combination therapy was associated with a significant delay in cognitive decline in NACC AD participants at a magnitude comparable to or greater than amyloid beta immunomodulators. Further, the delay in decline was sustained for 10 years. The impact of QuadRx to delay cognitive decline was validated in deeply characterized ADNI participants. These data support combination therapy in persons with AD risk factors to alter the course of AD that persists for a decade, enabling cognitive function at a magnitude associated with independent living. Highlights QuadRx slowed Alzheimer's disease (AD) cognitive decline by 47% in the National Alzheimer's Coordinating Center NACC and 60% in Alzheimer's Disease Neuroimaging Initiative ADNI participants.Combination therapy exhibited additive and synergistic slowing of cognitive decline.QuadRx was equally effective in females and males at 5 and 10 years.QuadRx mitigated the impact of the apolipoprotein E ε4 genotype.QuadRx was effective in AD participants reporting drug use for their AD risk factor.
Collapse
Affiliation(s)
- Yuan Shang
- Center for Innovation in Brain ScienceUniversity of Arizona Health SciencesTucsonArizonaUSA
| | - Georgina Torrandell‐Haro
- Center for Innovation in Brain ScienceUniversity of Arizona Health SciencesTucsonArizonaUSA
- Department of PharmacologyCollege of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Francesca Vitali
- Center for Innovation in Brain ScienceUniversity of Arizona Health SciencesTucsonArizonaUSA
- Department of NeurologyCollege of MedicineUniversity of ArizonaTucsonArizonaUSA
- Center for Biomedical Informatics and BiostatisticsUniversity of ArizonaTucsonArizonaUSA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain ScienceUniversity of Arizona Health SciencesTucsonArizonaUSA
- Department of PharmacologyCollege of MedicineUniversity of ArizonaTucsonArizonaUSA
- Department of NeurologyCollege of MedicineUniversity of ArizonaTucsonArizonaUSA
| | | |
Collapse
|
2
|
Salinero AE, Venkataganesh H, Abi-Ghanem C, Riccio D, Kelly RD, Gannon OJ, Sura A, Brooks HL, Zuloaga DG, Zuloaga KL. Effects of high fat diet on metabolic health vary by age of menopause onset. Int J Obes (Lond) 2024; 48:1839-1843. [PMID: 39152337 PMCID: PMC11585410 DOI: 10.1038/s41366-024-01618-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Menopause accelerates metabolic dysfunction, including (pre-)diabetes, obesity and visceral adiposity. However, the effects of endocrine vs. chronological aging in this progression are poorly understood. We hypothesized that menopause, especially in the context of middle-age, would exacerbate the metabolic effects of a high fat diet. Using young-adult and middle-aged C57BL/6J female mice, we modeled diet-induced obesity via chronic administration of high fat (HF) diet vs. control diet. We modeled peri-menopause/menopause via injections of 4-vinylcyclohexene diepoxide, which accelerates ovarian failure vs. vehicle. We performed glucose tolerance tests 2.5 and 7 months after diet onset, during the peri-menopausal and menopausal phases, respectively. Peri-menopause increased the severity of glucose intolerance and weight gain in middle-aged, HF-fed mice. Menopause increased weight gain in all mice regardless of age and diet, while chronological aging drove changes in adipose tissue distribution towards more visceral vs. subcutaneous adiposity. These data are in line with clinical data showing that post-menopausal women are more susceptible to metabolic dysfunction and suggest that greater chronological age exacerbates the effects of endocrine aging (menopause). This work highlights the importance of considering both chronological and endocrine aging in studies of metabolic health.
Collapse
Affiliation(s)
- Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Harini Venkataganesh
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - David Riccio
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Richard D Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Avi Sura
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Heddwen L Brooks
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Damian G Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, 1400 Washington Ave, Biology 325, Albany, NY, 12222, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
3
|
Breeze B, Connell E, Wileman T, Muller M, Vauzour D, Pontifex MG. Menopause and Alzheimer's disease susceptibility: Exploring the potential mechanisms. Brain Res 2024; 1844:149170. [PMID: 39163895 DOI: 10.1016/j.brainres.2024.149170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024]
Abstract
Alzheimer's Disease (AD), responsible for 62% of all dementia cases, is a progressive neurodegenerative condition that leads to cognitive dysfunction. The prevalence of AD is consistently higher in women suggesting they are disproportionately affected by this disease. Despite this, our understanding of this female AD vulnerability remains limited. Menopause has been identified as a potential contributing factor to AD in women, with earlier menopause onset associated with greater AD risk. However, the underlying mechanisms responsible for this increased risk are not fully understood. This review examines the potential role of menopause in the development of Alzheimer's Disease providing a mechanistic overview of the available literature from hormones to pathology. While literature is now emerging that indicates a role of hormonal shifts, gut dysbiosis, lipid dysregulation and inflammation, more research is needed to fully elucidate the mechanisms involved.
Collapse
Affiliation(s)
- Bernadette Breeze
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - Emily Connell
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - Tom Wileman
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom; Quadram Institute Biosciences, Norwich NR4 7UQ, United Kingdom
| | - Michael Muller
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - David Vauzour
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - Matthew G Pontifex
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom.
| |
Collapse
|
4
|
Cai J, Rong H, Chen J, Deng Z, Chen S, Liao H, Pan D, Chen Y, Shi Z, Li Y, Li H, Xu Y, Tang Y. Association of immune-mediated diseases with the risk of dementia and brain structure in UK Biobank participants. Age Ageing 2024; 53:afae274. [PMID: 39714283 DOI: 10.1093/ageing/afae274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Immunity and inflammation may be essential to the pathogenesis of dementia. However, the association of immune-mediated diseases with the risk of incident dementia has not been well characterised. OBJECTIVES We aimed to investigate the prospective association of 27 immune-mediated diseases and incident dementia risk and to explore the underlying mechanisms driven by brain structures. METHODS We included 487 459 UK Biobank participants aged 37-73 years without dementia at enrolment. Immune-mediated diseases and dementia cases were ascertained according to the International Classification of Diseases codes. Time-varying Cox proportional hazards regression and general linear regression models were used to examine the association of immune-mediated disease with incident dementia risk and brain morphometric measures, respectively. RESULTS Over a median follow-up of 12.3 years, 1654 cases of incident dementia were documented in 86 243 patients with immune-mediated diseases. Overall, immune-mediated diseases were associated with a higher all-cause dementia risk (hazard ratio [HR], 1.24; 95% confidence interval, 1.17-1.32). Five out of 27 immune-mediated diseases were associated with an increased risk of dementia individually. Comorbidity of multiple immune-mediated diseases further increased the risk. Moreover, the immune-mediated disease was associated with smaller total surface areas of both left (β, -286.51; SE, 102.58; P = .014) and right hemispheres (β, -298.56; SE, 103.96; P = .016), greater white matter hyperintensities volume (β, 1.02; SE, 0.13; P < .001) and less healthy white matter microstructures. CONCLUSIONS Immune-mediated diseases were associated with an increased risk of incident dementia, and the association of those diseases with brain structural abnormalities might provide clues to the underlying mechanisms.
Collapse
Affiliation(s)
- Jinhua Cai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Heng Rong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiongxue Chen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhenhong Deng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Sitai Chen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Huanquan Liao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of Neurology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Dong Pan
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Yanting Chen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yi Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Honghong Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
5
|
Shi Y, Ma J, Li S, Liu C, Liu Y, Chen J, Liu N, Liu S, Huang H. Sex difference in human diseases: mechanistic insights and clinical implications. Signal Transduct Target Ther 2024; 9:238. [PMID: 39256355 PMCID: PMC11387494 DOI: 10.1038/s41392-024-01929-7] [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: 12/20/2023] [Revised: 06/26/2024] [Accepted: 07/23/2024] [Indexed: 09/12/2024] Open
Abstract
Sex characteristics exhibit significant disparities in various human diseases, including prevalent cardiovascular diseases, cancers, metabolic disorders, autoimmune diseases, and neurodegenerative diseases. Risk profiles and pathological manifestations of these diseases exhibit notable variations between sexes. The underlying reasons for these sex disparities encompass multifactorial elements, such as physiology, genetics, and environment. Recent studies have shown that human body systems demonstrate sex-specific gene expression during critical developmental stages and gene editing processes. These genes, differentially expressed based on different sex, may be regulated by androgen or estrogen-responsive elements, thereby influencing the incidence and presentation of cardiovascular, oncological, metabolic, immune, and neurological diseases across sexes. However, despite the existence of sex differences in patients with human diseases, treatment guidelines predominantly rely on male data due to the underrepresentation of women in clinical trials. At present, there exists a substantial knowledge gap concerning sex-specific mechanisms and clinical treatments for diverse diseases. Therefore, this review aims to elucidate the advances of sex differences on human diseases by examining epidemiological factors, pathogenesis, and innovative progress of clinical treatments in accordance with the distinctive risk characteristics of each disease and provide a new theoretical and practical basis for further optimizing individualized treatment and improving patient prognosis.
Collapse
Affiliation(s)
- Yuncong Shi
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Jianshuai Ma
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Sijin Li
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Chao Liu
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Yuning Liu
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Jie Chen
- Department of Radiotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ningning Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shiming Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Hui Huang
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China.
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Chao YW, Tung YT, Yang SC, Shirakawa H, Su LH, Loe PY, Chiu WC. The Effects of Rice Bran on Neuroinflammation and Gut Microbiota in Ovariectomized Mice Fed a Drink with Fructose. Nutrients 2024; 16:2980. [PMID: 39275295 PMCID: PMC11397027 DOI: 10.3390/nu16172980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024] Open
Abstract
Rice bran, which is abundant in dietary fiber and phytochemicals, provides multiple health benefits. Nonetheless, its effects on neuroinflammation and gut microbiota in postmenopausal conditions are still not well understood. This study investigated the effects of rice bran and/or tea seed oil supplementation in d-galactose-injected ovariectomized (OVX) old mice fed a fructose drink. The combination of d-galactose injection, ovariectomy, and fructose drink administration creates a comprehensive model that simulates aging in females under multiple metabolic stressors, including oxidative stress, estrogen deficiency, and high-sugar diets, and allows the study of their combined impact on metabolic disorders and related diseases. Eight-week-old and 6-8-month-old female C57BL/6 mice were used. The mice were divided into six groups: a sham + young mice, a sham + old mice, an OVX + soybean oil, an OVX + soybean oil with rice bran, an OVX + tea seed oil (TO), and an OVX + TO with rice bran diet group. The OVX groups were subcutaneously injected with d-galactose (100 mg/kg/day) and received a 15% (v/v) fructose drink. The rice bran and tea seed oil supplementation formed 10% of the diet (w/w). The results showed that the rice bran with TO diet increased the number of short-chain fatty acid (SCFA)-producing Clostridia and reduced the number of endotoxin-producing Tannerellaceae, which mitigated imbalances in the gut-liver-brain axis. Rice bran supplementation reduced the relative weight of the liver, levels of hepatic triglycerides and total cholesterol; aspartate transaminase and alanine aminotransferase activity; brain levels of proinflammatory cytokines, including interleukin-1β and tumor necrosis factor-α; and plasma 8-hydroxy-2-deoxyguanosine. This study concludes that rice bran inhibits hepatic fat accumulation, which mitigates peripheral metaflammation and oxidative damage and reduces neuroinflammation in the brain.
Collapse
Affiliation(s)
- Yu-Wen Chao
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Tang Tung
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
- Advanced Plant and Food Crop Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Suh-Ching Yang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Hitoshi Shirakawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Li-Han Su
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Pei-Yu Loe
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Wan-Chun Chiu
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Department of Nutrition, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| |
Collapse
|
7
|
Cao Q, Zhang J, Hao X, Du S, Ao L, Zhu H, Huang W. The association between testosterone and serum soluble klotho in the females: evidence from the NHANES database. Front Endocrinol (Lausanne) 2024; 15:1335611. [PMID: 38818507 PMCID: PMC11137665 DOI: 10.3389/fendo.2024.1335611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/02/2024] [Indexed: 06/01/2024] Open
Abstract
Objective This research aimed to elucidate the relationship between testosterone levels and serum soluble klotho (S-klotho) concentrations in females aged 40-79 years using the National Health and Nutrition Examination Survey (NHANES) dataset. Design Associations between testosterone and S-klotho were assessed through multivariable linear regression methodologies, spanning nonadjusted, minimally adjusted, and fully adjusted models. Settings The investigation was conducted as a cross-sectional analysis utilizing the NHANES database. Participants From 20,146 NHANES participants between 2013 and 2016, 2,444 females met the stipulated inclusion and exclusion criteria. Results Free androgen index (FAI) showcased a negative correlation with S-klotho levels across all regression models (nonadjusted: β -7.08, 95% CI -13.39- -0.76; minimally adjusted: β -9.73, 95% CI -16.6- -2.84; fully adjusted: β -7.63, 95% CI -14.75-0.51). Conversely, total testosterone did not exhibit significant associations with S-klotho across the models. In the nonadjusted model, estradiol was positively associated with S-klotho concentrations (β 0.14, 95% CI 0.05-0.23), but this significance was not retained in subsequent regression models. Conclusion Findings suggest that in U.S. females aged 40-79 years, FAI negatively correlates with S-klotho concentrations, while there is the lack of significant associations for total testosterone and estradiol.
Collapse
Affiliation(s)
- Qi Cao
- Department of Reproductive Medical Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, Sichuan, China
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jiani Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, Sichuan, China
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaohu Hao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Siyu Du
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Lu Ao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huili Zhu
- Department of Reproductive Medical Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, Sichuan, China
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Wei Huang
- Department of Reproductive Medical Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, Sichuan, China
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Zhang Z, Liu X, Zhang S, Song Z, Lu K, Yang W. A review and analysis of key biomarkers in Alzheimer's disease. Front Neurosci 2024; 18:1358998. [PMID: 38445255 PMCID: PMC10912539 DOI: 10.3389/fnins.2024.1358998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects over 50 million elderly individuals worldwide. Although the pathogenesis of AD is not fully understood, based on current research, researchers are able to identify potential biomarker genes and proteins that may serve as effective targets against AD. This article aims to present a comprehensive overview of recent advances in AD biomarker identification, with highlights on the use of various algorithms, the exploration of relevant biological processes, and the investigation of shared biomarkers with co-occurring diseases. Additionally, this article includes a statistical analysis of key genes reported in the research literature, and identifies the intersection with AD-related gene sets from databases such as AlzGen, GeneCard, and DisGeNet. For these gene sets, besides enrichment analysis, protein-protein interaction (PPI) networks utilized to identify central genes among the overlapping genes. Enrichment analysis, protein interaction network analysis, and tissue-specific connectedness analysis based on GTEx database performed on multiple groups of overlapping genes. Our work has laid the foundation for a better understanding of the molecular mechanisms of AD and more accurate identification of key AD markers.
Collapse
Affiliation(s)
- Zhihao Zhang
- School of Computer Science and Technology, Xinjiang University, Ürümqi, China
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
| | - Xiangtao Liu
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
| | - Suixia Zhang
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Zhixin Song
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
| | - Ke Lu
- School of Computer Science and Technology, Xinjiang University, Ürümqi, China
| | - Wenzhong Yang
- School of Computer Science and Technology, Xinjiang University, Ürümqi, China
| |
Collapse
|
9
|
Lynch MA. A case for seeking sex-specific treatments in Alzheimer's disease. Front Aging Neurosci 2024; 16:1346621. [PMID: 38414633 PMCID: PMC10897030 DOI: 10.3389/fnagi.2024.1346621] [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: 11/29/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024] Open
Abstract
There is no satisfactory explanation for the sex-related differences in the incidence of many diseases and this is also true of Alzheimer's disease (AD), where females have a higher lifetime risk of developing the disease and make up about two thirds of the AD patient population. The importance of understanding the cause(s) that account for this disproportionate distribution cannot be overestimated, and is likely to be a significant factor in the search for therapeutic strategies that will combat the disease and, furthermore, potentially point to a sex-targeted approach to treatment. This review considers the literature in the context of what is known about the impact of sex on processes targeted by drugs that are in clinical trial for AD, and existing knowledge on differing responses of males and females to these drugs. Current knowledge strongly supports the view that trials should make assessing sex-related difference in responses a priority with a focus on exploring the sex-stratified treatments.
Collapse
|
10
|
Salinero AE, Venkataganesh H, Abi-Ghanem C, Riccio D, Kelly RD, Gannon OJ, Sura A, Brooks HL, Zuloaga KL. Effects of High Fat Diet on Metabolic Health Vary by Age of Menopause Onset. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576269. [PMID: 38293070 PMCID: PMC10827154 DOI: 10.1101/2024.01.18.576269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Menopause accelerates metabolic dysfunction, including (pre-)diabetes, obesity and visceral adiposity. However, the effects of endocrine vs. chronological aging in this progression are poorly understood. We hypothesize that menopause, especially in the context of middle-age, will exacerbate the metabolic effects of a high fat diet. Using young-adult and middle-aged C57BL/6J female mice, we modeled diet-induce obesity via chronic administration of high fat (HF) diet vs. control diet. We modeled peri-menopause/menopause via injections of 4-vinylcyclohexene diepoxide, which accelerates ovarian failure vs. vehicle. We performed glucose tolerance tests 2.5 and 7 months after diet onset, during the peri-menopausal and menopausal phases, respectively. Peri-menopause increased the severity of glucose intolerance and weight gain in middle-aged, HF-fed mice. Menopause increased weight gain in all mice regardless of age and diet, while chronological aging drove changes in adipose tissue distribution towards more visceral vs. subcutaneous adiposity. These data are in line with clinical data showing that post-menopausal women are more susceptible to metabolic dysfunction and suggest that greater chorological age exacerbates the effects of endocrine aging (menopause). This work highlights the importance of considering both chronological and endocrine aging in studies of metabolic health.
Collapse
|
11
|
Liang G, Kow ASF, Yusof R, Tham CL, Ho YC, Lee MT. Menopause-Associated Depression: Impact of Oxidative Stress and Neuroinflammation on the Central Nervous System-A Review. Biomedicines 2024; 12:184. [PMID: 38255289 PMCID: PMC10813042 DOI: 10.3390/biomedicines12010184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Perimenopausal depression, occurring shortly before or after menopause, is characterized by symptoms such as emotional depression, anxiety, and stress, often accompanied by endocrine dysfunction, particularly hypogonadism and senescence. Current treatments for perimenopausal depression primarily provide symptomatic relief but often come with undesirable side effects. The development of agents targeting the specific pathologies of perimenopausal depression has been relatively slow. The erratic fluctuations in estrogen and progesterone levels during the perimenopausal stage expose women to the risk of developing perimenopausal-associated depression. These hormonal changes trigger the production of proinflammatory mediators and induce oxidative stress, leading to progressive neuronal damage. This review serves as a comprehensive overview of the underlying mechanisms contributing to perimenopausal depression. It aims to shed light on the complex relationship between perimenopausal hormones, neurotransmitters, brain-derived neurotrophic factors, chronic inflammation, oxidative stress, and perimenopausal depression. By summarizing the intricate interplay between hormonal fluctuations, neurotransmitter activity, brain-derived neurotrophic factors, chronic inflammation, oxidative stress, and perimenopausal depression, this review aims to stimulate further research in this field. The hope is that an increased understanding of these mechanisms will pave the way for the development of more effective therapeutic targets, ultimately reducing the risk of depression during the menopausal stage for the betterment of psychological wellbeing.
Collapse
Affiliation(s)
- Gengfan Liang
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | | | - Rohana Yusof
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Natural Medicines and Products Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Yu-Cheng Ho
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Ming Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
- Centre of Research for Mental Health and Well-Being, UCSI University, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
12
|
Nerattini M, Jett S, Andy C, Carlton C, Zarate C, Boneu C, Battista M, Pahlajani S, Loeb-Zeitlin S, Havryulik Y, Williams S, Christos P, Fink M, Brinton RD, Mosconi L. Systematic review and meta-analysis of the effects of menopause hormone therapy on risk of Alzheimer's disease and dementia. Front Aging Neurosci 2023; 15:1260427. [PMID: 37937120 PMCID: PMC10625913 DOI: 10.3389/fnagi.2023.1260427] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/25/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Despite a large preclinical literature demonstrating neuroprotective effects of estrogen, use of menopausal hormone therapy (HT) for Alzheimer's disease (AD) risk reduction has been controversial. Herein, we conducted a systematic review and meta-analysis of HT effects on AD and dementia risk. Methods Our systematic search yielded 6 RCT reports (21,065 treated and 20,997 placebo participants) and 45 observational reports (768,866 patient cases and 5.5 million controls). We used fixed and random effect meta-analysis to derive pooled relative risk (RR) and 95% confidence intervals (C.I.) from these studies. Results Randomized controlled trials conducted in postmenopausal women ages 65 and older show an increased risk of dementia with HT use compared with placebo [RR = 1.38, 95% C.I. 1.16-1.64, p < 0.001], driven by estrogen-plus-progestogen therapy (EPT) [RR = 1.64, 95% C.I. 1.20-2.25, p = 0.002] and no significant effects of estrogen-only therapy (ET) [RR = 1.19, 95% C.I. 0.92-1.54, p = 0.18]. Conversely, observational studies indicate a reduced risk of AD [RR = 0.78, 95% C.I. 0.64-0.95, p = 0.013] and all-cause dementia [RR = .81, 95% C.I. 0.70-0.94, p = 0.007] with HT use, with protective effects noted with ET [RR = 0.86, 95% C.I. 0.77-0.95, p = 0.002] but not with EPT [RR = 0.910, 95% C.I. 0.775-1.069, p = 0.251]. Stratified analysis of pooled estimates indicates a 32% reduced risk of dementia with midlife ET [RR = 0.685, 95% C.I. 0.513-0.915, p = 0.010] and non-significant reductions with midlife EPT [RR = 0.775, 95% C.I. 0.474-1.266, p = 0.309]. Late-life HT use was associated with increased risk, albeit not significant [EPT: RR = 1.323, 95% C.I. 0.979-1.789, p = 0.069; ET: RR = 1.066, 95% C.I. 0.996-1.140, p = 0.066]. Discussion These findings support renewed research interest in evaluating midlife estrogen therapy for AD risk reduction.
Collapse
Affiliation(s)
- Matilde Nerattini
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
- Department of Experimental and Clinical Biomedical Sciences, Nuclear Medicine Unit, University of Florence, Florence, Italy
| | - Steven Jett
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Caroline Carlton
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Camila Zarate
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Camila Boneu
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Michael Battista
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Susan Loeb-Zeitlin
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, United States
| | - Yelena Havryulik
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, United States
| | - Schantel Williams
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Paul Christos
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Matthew Fink
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Roberta Diaz Brinton
- Department of Neurology and Pharmacology, University of Arizona, Tucson, AZ, United States
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
- Department of Experimental and Clinical Biomedical Sciences, Nuclear Medicine Unit, University of Florence, Florence, Italy
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
13
|
Maturana W, Lobo I, Landeira-Fernandes J, Mograbi DC. Nondeclarative associative learning in Alzheimer's disease: An overview of eyeblink, fear, and other emotion-based conditioning. Physiol Behav 2023; 268:114250. [PMID: 37224936 DOI: 10.1016/j.physbeh.2023.114250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/04/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Alzheimer's disease is a neurodegenerative disorder that is characterized by progressive cognitive decline, particularly in declarative memory, and the presence of β-amyloid plaques, neurofibrillary tangles, and cortical atrophy (especially in the temporal lobe). Unlike the relationship between the temporal cortex and declarative memory, nondeclarative memories (e.g., motor, fear, and other emotion-based memories) involve distinct neural structures. The present review investigates nondeclarative associative learning ability in Alzheimer's disease. We discuss eyeblink conditioning, fear conditioning, and other emotion-based learning and present the functions and brain areas that are involved in each type of learning. Evidence suggests that nondeclarative learning is also affected by Alzheimer's disease, although some forms of learning may be relatively preserved. Details about each nondeclarative associative learning process and the implications of these findings are presented.
Collapse
|
14
|
Voskuhl R, Itoh Y. The X factor in neurodegeneration. J Exp Med 2022; 219:e20211488. [PMID: 36331399 PMCID: PMC9641640 DOI: 10.1084/jem.20211488] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/22/2022] [Accepted: 10/12/2022] [Indexed: 07/25/2023] Open
Abstract
Given the aging population, it is important to better understand neurodegeneration in aging healthy people and to address the increasing incidence of neurodegenerative diseases. It is imperative to apply novel strategies to identify neuroprotective therapeutics. The study of sex differences in neurodegeneration can reveal new candidate treatment targets tailored for women and men. Sex chromosome effects on neurodegeneration remain understudied and represent a promising frontier for discovery. Here, we will review sex differences in neurodegeneration, focusing on the study of sex chromosome effects in the context of declining levels of sex hormones during aging.
Collapse
Affiliation(s)
- Rhonda Voskuhl
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Yuichiro Itoh
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
15
|
Alruwais NM, Rusted JM, Tabet N, Dowell NG. Evidence of emerging BBB changes in mid-age apolipoprotein E epsilon-4 carriers. Brain Behav 2022; 12:e2806. [PMID: 36408825 PMCID: PMC9759141 DOI: 10.1002/brb3.2806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/23/2022] [Accepted: 10/08/2022] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Studies have recognized that the loss of the blood-brain barrier (BBB) integrity is a major structural biomarker where neurodegenerative disease potentially begins. Using a combination of high-quality neuroimaging techniques, we investigated potential subtle differences in BBB permeability in mid-age healthy people, comparing carriers of the apolipoprotein E epsilon-4 (APOEε4) genotype, the biggest risk factor for late onset, non-familial AD (LOAD) with APOEε3 carriers, the population norm. METHODS Forty-one cognitively healthy mid-age participants (42-59) were genotyped and pseudo-randomly selected to participate in the study by a third party. Blind to genotype, all participants had a structural brain scan acquisition including gadolinium-based dynamic contrast-enhanced magnetic resonance imaging acquired using a T1-weighted 3D vibe sequence. A B1 map and T1 map were acquired as part of the multi-parametric mapping acquisition. RESULTS Non-significant, but subtle differences in blood-brain barrier permeability were identified between healthy mid-age APOEε4 and APOEε3 carriers, matched on age, education, and gender. DISCUSSION This study demonstrated a tendency toward BBB permeability in APOEε4 participants emerging from mid-age, with quantitative differences observable on a number of the measures. While the differences did not reach a statistical significance, the results from this study hint at early changes in ε4 carrier BBB that may help identify at-risk populations and facilitate the development of early interventions to change the trajectory of decline.
Collapse
Affiliation(s)
- Nourah M Alruwais
- Health science department, College of Applied Studies and Community Services, King Saud University, Riyadh, Saudi Arabia.,School of Psychology, University of Sussex, Brighton, UK
| | | | - Naji Tabet
- Brighton and Sussex Medical School (BSMS), Brighton, UK
| | | |
Collapse
|
16
|
Omer AB, Dalhat MH, Khan MK, Afzal O, Altamimi ASA, Alzarea SI, Almalki WH, Kazmi I. Butin Mitigates Memory Impairment in Streptozotocin-Induced Diabetic Rats by Inhibiting Oxidative Stress and Inflammatory Responses. Metabolites 2022; 12:1050. [PMID: 36355133 PMCID: PMC9694489 DOI: 10.3390/metabo12111050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/17/2022] [Accepted: 10/27/2022] [Indexed: 11/10/2023] Open
Abstract
It has been reported from the previous literature that butin restores mitochondrial dysfunction by modulation of oxidative stress and glutamate-induced neurotoxicity in mouse hippocampus HT22 cells. Butin also possesses an anti-Huntington's effect in rats. Considering the current background, this study was designed to evaluate the neuroprotective effect of butin against memory loss caused by streptozotocin (STZ). STZ (40 mg/kg) was intraperitoneally injected into rats. Three days later, diabetic rats were identified and included in the study. A total of 30 rats (12 nondiabetic and 18 diabetics) were grouped as Group A (control-non-diabetic rats) and Group B (STZ diabetic control) were treated with 1 mL of sodium CMC (0.5% w/v). Group C (STZ+ butin 25) were treated with butin 25 mg/kg. Group D (STZ+ butin 50) and Group E (butin per se) were administered with butin 50 mg/kg. Each therapy was administered orally once each day for 15-day. The Morris water maze and the Y-maze behavioural tests were run throughout the experimental programme. Animals were put to death on day 15 and their brains were removed for biochemical assays (CAT, SOD, GSH, MDA, nitrite, acetylcholinesterase (AchE), IL-1, and mitochondrial enzyme complexes). Rats with neurobehavioral impairments brought on by STZ have less spontaneous movement, learning capacity, and memory. Additionally, STZ decreased endogenous antioxidants and increased pro-inflammatory cytokines, nitrite, MDA, and AchE. Neurobehavioral deficits and metabolic markers were dramatically improved by butin.
Collapse
Affiliation(s)
- Asma B. Omer
- Department of Basic Health Sciences, Foundation Year for the Health Colleges, Princess Nourah bint Abdul Rahman University, Riyadh 11671, Saudi Arabia
| | - Mahmood Hassan Dalhat
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad Kaleem Khan
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Pharmacology, Dadasaheb Balpande College of Pharmacy, Nagpur 440037, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Abdulmalik S. A. Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
17
|
Branigan GL, Torrandell-Haro G, Vitali F, Brinton RD, Rodgers K. Age and sex differences on anti-hyperglycemic medication exposure and risk of newly diagnosed multiple sclerosis in propensity score matched type 2 diabetics. Heliyon 2022; 8:e11196. [PMID: 36325137 PMCID: PMC9618986 DOI: 10.1016/j.heliyon.2022.e11196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/30/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Background The association between exposure to anti-hyperglycemic medications (A-HgM) for Type 2 Diabetes Mellitus (T2D) treatment and Multiple Sclerosis (MS) in T2D patients is unclear. Methods This retrospective cohort analysis used the Mariner claims database. Patient records were surveyed for a diagnosis of MS starting 12 months after diagnosis of T2D. Patients were required to be actively enrolled in the Mariner claims records for six months prior and at least three years after the diagnosis of T2D without a history of previous neurodegenerative disease. Survival analysis was used to determine the association between A-HgM exposure and diagnosis of MS. A propensity score approach was used to minimize measured and unmeasured selection bias. The analyses were conducted between January 1st and April 28th, 2021. Findings In T2D patients younger than 45, A-HgM exposure was associated with a reduced risk of developing MS (RR: 0.22, 95%CI: 0.17-0.29, p-value <0.001). In contrast, A-HgM exposure in patients older than 45 was associated with an increased risk of MS with women exhibiting greater risk (RR: 1.53, 95%CI: 1.39-1.69, p < 0.001) than men (RR: 1.17, 95%CI: 1.01-1.37, p = 0 · 04). Patients who developed MS had a higher incidence of baseline comorbidities. Mean follow-up was 6.2 years with a standard deviation of 1.8 years. Interpretation In this study, A-HgM exposure in patients with T2D was associated with reduced risk of MS in patients younger than 45 whereas in patients older than 45, exposure to A-HgM was associated with an increased risk of newly diagnosed MS, particularly in women.
Collapse
Affiliation(s)
- Gregory L. Branigan
- Center for Innovation in Brain Science; University of Arizona, Tucson, Arizona, USA
- Department of Pharmacology; University of Arizona College of Medicine, Tucson, Arizona, USA
- MD-PhD Training Program; University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Georgina Torrandell-Haro
- Center for Innovation in Brain Science; University of Arizona, Tucson, Arizona, USA
- Department of Pharmacology; University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Francesca Vitali
- Center for Innovation in Brain Science; University of Arizona, Tucson, Arizona, USA
- Department of Neurology; University of Arizona College of Medicine, Tucson, Arizona, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, Arizona, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science; University of Arizona, Tucson, Arizona, USA
- Department of Pharmacology; University of Arizona College of Medicine, Tucson, Arizona, USA
- Department of Neurology; University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Kathleen Rodgers
- Center for Innovation in Brain Science; University of Arizona, Tucson, Arizona, USA
- Department of Pharmacology; University of Arizona College of Medicine, Tucson, Arizona, USA
| |
Collapse
|
18
|
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
|
19
|
McLean JW, Bhattrai A, Vitali F, Raikes AC, Wiegand JPL, Brinton RD. Contributions of sex and genotype to exploratory behavior differences in an aged humanized APOE mouse model of late-onset Alzheimer's disease. Learn Mem 2022; 29:321-331. [PMID: 36206387 PMCID: PMC9488030 DOI: 10.1101/lm.053588.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022]
Abstract
Age, genetics, and chromosomal sex have been identified as critical risk factors for late-onset Alzheimer's disease (LOAD). The predominant genetic risk factor for LOAD is the apolipoprotein E ε4 allele (APOE4), and the prevalence of LOAD is higher in females. However, the translational validity of APOE4 mouse models for AD-related cognitive impairment remains to be fully determined. The present study investigated the role of both sex and genotype on learning and memory in aged, humanized APOE knock-in mice. Aged (23.27 mo ± 1.21 mo; 39 male/37 female) APOE3/3, APOE3/4, and APOE4/4 mice performed a novel object recognition (NOR) assay. Task-related metrics were analyzed using two-way sex by genotype ANOVAs. Sex differences were more prominent relative to APOE genotype. Prior to NOR, female mice exhibited thigmotaxic center zone avoidance during the open field task relative to males, regardless of genotype. Within object familiarization and NOR tasks, females had greater object interaction and locomotion. Interestingly, only APOE4/4 females on average recognized the novel object. These results suggest that APOE4, although strongly related to LOAD pathogenesis, does not drive cognitive decline in the absence of other risk factors even in very aged mice. Chromosomal sex is a key driver of behavioral phenotypes and thus is a critical variable for translatability of interventions designed to preserve learning and memory in animal models of LOAD. Last, there was a very high degree of variability in behavioral performance across APOE genotypes. A cluster analysis of the behavioral data revealed a low-activity and a high-activity cluster. APOE4 carriers were overrepresented in the low-activity cluster, while male:female distributions did not differ. Collectively, the behavioral data indicate that chromosomal sex has the greatest impact on behavioral phenotype, and APOE4 carrier status may confer greater risk for cognitive decline in some animals.
Collapse
Affiliation(s)
- John W McLean
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona 85721, USA
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, Arizona 85721, USA
| | - Avnish Bhattrai
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona 85721, USA
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, USA
| | - Francesca Vitali
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona 85721, USA
| | - Adam C Raikes
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona 85721, USA
| | - Jean-Paul L Wiegand
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona 85721, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona 85721, USA
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, USA
| |
Collapse
|
20
|
Subramaniapillai S, Suri S, Barth C, Maximov II, Voldsbekk I, van der Meer D, Gurholt TP, Beck D, Draganski B, Andreassen OA, Ebmeier KP, Westlye LT, de Lange AG. Sex- and age-specific associations between cardiometabolic risk and white matter brain age in the UK Biobank cohort. Hum Brain Mapp 2022; 43:3759-3774. [PMID: 35460147 PMCID: PMC9294301 DOI: 10.1002/hbm.25882] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/24/2022] [Accepted: 04/05/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiometabolic risk (CMR) factors are associated with accelerated brain aging and increased risk for sex-dimorphic illnesses such as Alzheimer's disease (AD). Yet, it is unknown how CMRs interact with sex and apolipoprotein E-ϵ4 (APOE4), a known genetic risk factor for AD, to influence brain age across different life stages. Using age prediction based on multi-shell diffusion-weighted imaging data in 21,308 UK Biobank participants, we investigated whether associations between white matter Brain Age Gap (BAG) and body mass index (BMI), waist-to-hip ratio (WHR), body fat percentage (BF%), and APOE4 status varied (i) between males and females, (ii) according to age at menopause in females, and (iii) across different age groups in males and females. We report sex differences in associations between BAG and all three CMRs, with stronger positive associations among males compared to females. Independent of APOE4 status, higher BAG (older brain age relative to chronological age) was associated with greater BMI, WHR, and BF% in males, whereas in females, higher BAG was associated with greater WHR, but not BMI and BF%. These divergent associations were most prominent within the oldest group of females (66-81 years), where greater BF% was linked to lower BAG. Earlier menopause transition was associated with higher BAG, but no interactions were found with CMRs. In conclusion, the findings point to sex- and age-specific associations between CMRs and brain age. Incorporating sex as a factor of interest in studies addressing CMR may promote sex-specific precision medicine, consequently improving health care for both males and females.
Collapse
Affiliation(s)
- Sivaniya Subramaniapillai
- LREN, Centre for Research in Neurosciences, Department of Clinical NeurosciencesLausanne University Hospital (CHUV) and University of LausanneLausanneSwitzerland
- Department of Psychology, Faculty of ScienceMcGill UniversityMontrealQuebecCanada
- Department of PsychologyUniversity of OsloOsloNorway
| | - Sana Suri
- Department of PsychiatryUniversity of OxfordOxfordUK
- Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
| | - Claudia Barth
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- Department of Psychiatric ResearchDiakonhjemmet HospitalOsloNorway
| | - Ivan I. Maximov
- Department of PsychologyUniversity of OsloOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- Department of Health and FunctioningWestern Norway University of Applied SciencesBergenNorway
| | - Irene Voldsbekk
- Department of PsychologyUniversity of OsloOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
| | - Dennis van der Meer
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- School of Mental Health and Neuroscience, Faculty of Health Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Tiril P. Gurholt
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
| | - Dani Beck
- Department of PsychologyUniversity of OsloOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- Department of Psychiatric ResearchDiakonhjemmet HospitalOsloNorway
| | - Bogdan Draganski
- LREN, Centre for Research in Neurosciences, Department of Clinical NeurosciencesLausanne University Hospital (CHUV) and University of LausanneLausanneSwitzerland
- Department of NeurologyMax Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
| | | | - Lars T. Westlye
- Department of PsychologyUniversity of OsloOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
| | - Ann‐Marie G. de Lange
- LREN, Centre for Research in Neurosciences, Department of Clinical NeurosciencesLausanne University Hospital (CHUV) and University of LausanneLausanneSwitzerland
- Department of PsychologyUniversity of OsloOsloNorway
- Department of PsychiatryUniversity of OxfordOxfordUK
| |
Collapse
|
21
|
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
|
22
|
Lin Y, Qiu T, Wei G, Que Y, Wang W, Kong Y, Xie T, Chen X. Role of Histone Post-Translational Modifications in Inflammatory Diseases. Front Immunol 2022; 13:852272. [PMID: 35280995 PMCID: PMC8908311 DOI: 10.3389/fimmu.2022.852272] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Inflammation is a defensive reaction for external stimuli to the human body and generally accompanied by immune responses, which is associated with multiple diseases such as atherosclerosis, type 2 diabetes, Alzheimer’s disease, psoriasis, asthma, chronic lung diseases, inflammatory bowel disease, and multiple virus-associated diseases. Epigenetic mechanisms have been demonstrated to play a key role in the regulation of inflammation. Common epigenetic regulations are DNA methylation, histone modifications, and non-coding RNA expression; among these, histone modifications embrace various post-modifications including acetylation, methylation, phosphorylation, ubiquitination, and ADP ribosylation. This review focuses on the significant role of histone modifications in the progression of inflammatory diseases, providing the potential target for clinical therapy of inflammation-associated diseases.
Collapse
Affiliation(s)
- Yingying Lin
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Ting Qiu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Guifeng Wei
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yueyue Que
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Wenxin Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yichao Kong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
23
|
Jett S, Malviya N, Schelbaum E, Jang G, Jahan E, Clancy K, Hristov H, Pahlajani S, Niotis K, Loeb-Zeitlin S, Havryliuk Y, Isaacson R, Brinton RD, Mosconi L. Endogenous and Exogenous Estrogen Exposures: How Women's Reproductive Health Can Drive Brain Aging and Inform Alzheimer's Prevention. Front Aging Neurosci 2022; 14:831807. [PMID: 35356299 PMCID: PMC8959926 DOI: 10.3389/fnagi.2022.831807] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/07/2022] [Indexed: 01/14/2023] Open
Abstract
After advanced age, female sex is the major risk factor for late-onset Alzheimer's disease (AD), the most common cause of dementia affecting over 24 million people worldwide. The prevalence of AD is higher in women than in men, with postmenopausal women accounting for over 60% of all those affected. While most research has focused on gender-combined risk, emerging data indicate sex and gender differences in AD pathophysiology, onset, and progression, which may help account for the higher prevalence in women. Notably, AD-related brain changes develop during a 10-20 year prodromal phase originating in midlife, thus proximate with the hormonal transitions of endocrine aging characteristic of the menopause transition in women. Preclinical evidence for neuroprotective effects of gonadal sex steroid hormones, especially 17β-estradiol, strongly argue for associations between female fertility, reproductive history, and AD risk. The level of gonadal hormones to which the female brain is exposed changes considerably across the lifespan, with relevance to AD risk. However, the neurobiological consequences of hormonal fluctuations, as well as that of hormone therapies, are yet to be fully understood. Epidemiological studies have yielded contrasting results of protective, deleterious and null effects of estrogen exposure on dementia risk. In contrast, brain imaging studies provide encouraging evidence for positive associations between greater cumulative lifetime estrogen exposure and lower AD risk in women, whereas estrogen deprivation is associated with negative consequences on brain structure, function, and biochemistry. Herein, we review the existing literature and evaluate the strength of observed associations between female-specific reproductive health factors and AD risk in women, with a focus on the role of endogenous and exogenous estrogen exposures as a key underlying mechanism. Chief among these variables are reproductive lifespan, menopause status, type of menopause (spontaneous vs. induced), number of pregnancies, and exposure to hormonal therapy, including hormonal contraceptives, hormonal therapy for menopause, and anti-estrogen treatment. As aging is the greatest risk factor for AD followed by female sex, understanding sex-specific biological pathways through which reproductive history modulates brain aging is crucial to inform preventative and therapeutic strategies for AD.
Collapse
Affiliation(s)
- Steven Jett
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Niharika Malviya
- 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
| | - Eva Jahan
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Katherine Clancy
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Hollie Hristov
- Department of Neurology, 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
| | - Kellyann Niotis
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Susan Loeb-Zeitlin
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, United States
| | - Yelena Havryliuk
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, United States
| | - Richard Isaacson
- Department of Neurology, 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
|
24
|
Mishra A, Wang Y, Yin F, Vitali F, Rodgers KE, Soto M, Mosconi L, Wang T, Brinton RD. A tale of two systems: Lessons learned from female mid-life aging with implications for Alzheimer's prevention & treatment. Ageing Res Rev 2022; 74:101542. [PMID: 34929348 PMCID: PMC8884386 DOI: 10.1016/j.arr.2021.101542] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 12/05/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
Neurological aging is frequently viewed as a linear process of decline, whereas in reality, it is a dynamic non-linear process. The dynamic nature of neurological aging is exemplified during midlife in the female brain. To investigate fundamental mechanisms of midlife aging that underlie risk for development of Alzheimer's disease (AD) in late life, we investigated the brain at greatest risk for the disease, the aging female brain. Outcomes of our research indicate that mid-life aging in the female is characterized by the emergence of three phases: early chronological (pre-menopause), endocrinological (peri-menopause) and late chronological (post-menopause) aging. The endocrinological aging program is sandwiched between early and late chronological aging. Throughout the three stages of midlife aging, two systems of biology, metabolic and immune, are tightly integrated through a network of signaling cascades. The network of signaling between these two systems of biology underlie an orchestrated sequence of adaptative starvation responses that shift the brain from near exclusive dependence on a single fuel, glucose, to utilization of an auxiliary fuel derived from lipids, ketone bodies. The dismantling of the estrogen control of glucose metabolism during mid-life aging is a critical contributor to the shift in fuel systems and emergence of dynamic neuroimmune phenotype. The shift in fuel reliance, puts the largest reservoir of local fatty acids, white matter, at risk for catabolism as a source of lipids to generate ketone bodies through astrocytic beta oxidation. APOE4 genotype accelerates the tipping point for emergence of the bioenergetic crisis. While outcomes derived from research conducted in the female brain are not directly translatable to the male brain, the questions addressed in a female centric program of research are directly applicable to investigation of the male brain. Like females, males with AD exhibit deficits in the bioenergetic system of the brain, activation of the immune system and hallmark Alzheimer's pathologies. The drivers and trajectory of mechanisms underlying neurodegeneration in the male brain will undoubtedly share common aspects with the female in addition to factors unique to the male. Preclinical and clinical evidence indicate that midlife endocrine aging can also be a transitional bridge to autoimmune disorders. Collectively, the data indicate that endocrinological aging is a critical period "tipping point" in midlife which can initiate emergence of the prodromal stage of late-onset-Alzheimer's disease. Interventions that target both immune and metabolic shifts that occur during midlife aging have the potential to alter the trajectory of Alzheimer's risk in late life. Further, to achieve precision medicine for AD, chromosomal sex is a critical variable to consider along with APOE genotype, other genetic risk factors and stage of disease.
Collapse
Affiliation(s)
- Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Fei Yin
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Francesca Vitali
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Kathleen E Rodgers
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Maira Soto
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Tian Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Roberta D Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA.
| |
Collapse
|
25
|
Reddy V, McCarthy M, Raval AP. Xenoestrogens impact brain estrogen receptor signaling during the female lifespan: A precursor to neurological disease? Neurobiol Dis 2021; 163:105596. [PMID: 34942334 DOI: 10.1016/j.nbd.2021.105596] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/08/2021] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
Xenoestrogens, foreign synthetic chemicals mimicking estrogens, are lurking in our surroundings. Climate change may alter their toxicity and bioavailability. Since xenoestrogens have extremely high lipid solubility and are structurally similar to natural endogenous estrogens, they can bind to estrogen receptors (ERs) -alpha (ER-α) and -beta (ER-β). Scientific evidence accumulated over the past decades have suggested that natural 17β-estradiol (E2; a potent estrogen), via activation of its receptors, plays a pivotal role in regulation of brain development, differentiation, metabolism, synaptic plasticity, neuroprotection, cognition, anxiety, body temperature, feeding and sexual behavior. In the brain, ER-β is predominantly expressed in the various regions, including cerebral cortex and hippocampus, that have been shown to play a key role in cognition. Therefore, disturbances in function of ER-β mediated E2 signaling by xenoestrogens can lead to deleterious effects that potentiate a variety of neurological diseases starting from prenatal to post-menopause in women. The goal of this review is to identify the possible neurological effects of xenoestrogens that can alter estrogen receptor-mediated signaling in the brain during different stages of the female lifespan.
Collapse
Affiliation(s)
- Varun Reddy
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Micheline McCarthy
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
26
|
Beckman D, Morrison JH. Towards developing a rhesus monkey model of early Alzheimer's disease focusing on women's health. Am J Primatol 2021; 83:e23289. [PMID: 34056733 DOI: 10.1002/ajp.23289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/11/2021] [Accepted: 05/16/2021] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of elderly dementia, affecting nearly 50 million people worldwide, with two-thirds of the cases in the USA in women. Despite considerable investment, this prevalence is expected to increase further in the coming decades, based on the projected demographics of the population. Currently, most of the preclinical AD studies rely on transgenic mice carrying mutations associated with the early onset familiar form of AD, although the vast majority of cases are sporadic. A prevailing current hypothesis is that the cascade of events leading to AD starts with the accumulation of small soluble oligomers of the Aβ peptide (AβOs) that target and disrupt synapses. Taking advantage of the high translational power of rhesus monkeys due to their physiological and genetic similarities to humans, we recently developed a female rhesus monkey model of early AD pathogenesis based on exogenous administration AβOs. Here we review and discuss how soluble oligomers of Aβ can target vulnerable spines in the neocortex and hippocampus of female middle-aged monkeys and induce neuroinflammatory responses, similar to what is known to occur in the human brain. Developing a rhesus monkey model of early AD focusing on women's health is critical for the understanding of how hormonal changes during menopause transition affect brain health and ultimately may contribute to AD neurodegeneration.
Collapse
Affiliation(s)
- Danielle Beckman
- California National Primate Research Center, UC Davis, Davis, California, USA
| | - John H Morrison
- California National Primate Research Center, UC Davis, Davis, California, USA
- Department of Neurology, School of Medicine, UC Davis, Davis, California, USA
| |
Collapse
|
27
|
Li S, Qu L, Wang X, Kong L. Novel insights into RIPK1 as a promising target for future Alzheimer's disease treatment. Pharmacol Ther 2021; 231:107979. [PMID: 34480965 DOI: 10.1016/j.pharmthera.2021.107979] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/30/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is an intractable neurodegenerative disease showing a clinical manifestation with memory loss, cognitive impairment and behavioral dysfunction. The predominant pathological characteristics of AD include neuronal loss, β-amyloid (Aβ) deposition and hyperphosphorylated Tau induced neurofibrillary tangles (NFTs), while considerable studies proved these could be triggered by neuronal death and neuroinflammation. Receptor-interacting protein kinase 1 (RIPK1) is a serine/threonine kinase existed at the cross-point of cell death and inflammatory signaling pathways. Emerging investigations have shed light on RIPK1 for its potential role in AD progression. The present review makes a bird's eye view on the functions of RIPK1 and mainly focus on the underlying linkages between RIPK1 and AD from comprehensive aspects including neuronal death, Aβ and Tau, inflammasome activation, BBB rupture, AMPK/mTOR, mitochondrial dysfunction and O-glcNAcylation. Moreover, the discovery of RIPK1 inhibitors, ongoing clinical trials along with future RIPK1-targeted therapeutics are also reviewed.
Collapse
Affiliation(s)
- Shang Li
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lailiang Qu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiaobing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
28
|
Identification of the Hub Genes in Alzheimer's Disease. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6329041. [PMID: 34326892 PMCID: PMC8302378 DOI: 10.1155/2021/6329041] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/03/2021] [Indexed: 12/29/2022]
Abstract
Purpose Alzheimer's disease (AD) is considered to be the most common neurodegenerative disease and also one of the major fatal diseases affecting the elderly, thus bringing a huge burden to society. Therefore, identifying AD-related hub genes is extremely important for developing novel strategies against AD. Materials and Methods Here, we extracted the gene expression profile GSE63061 from the National Center for Biotechnology Information (NCBI) GEO database. Once the unverified gene chip was removed, we standardized the microarray data after quality control. We utilized the Limma software package to screen the differentially expressed genes (DEGs). We conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of DEGs. Subsequently, we constructed a protein-protein interaction (PPI) network using the STRING database. Result We screened 2169 DEGs, comprising 1313 DEGs with upregulation and 856 DEGs with downregulation. Functional enrichment analysis showed that the response of immune, the degranulation of neutrophils, lysosome, and the differentiation of osteoclast were greatly enriched in DEGs with upregulation; peptide biosynthetic process, translation, ribosome, and oxidative phosphorylation were dramatically enriched in DEGs with downregulation. 379 nodes and 1149 PPI edges were demonstrated in the PPI network constructed by upregulated DEGs; 202 nodes and 1963 PPI edges were shown in the PPI network constructed by downregulated DEGs. Four hub genes, including GAPDH, RHOA, RPS29, and RPS27A, were identified to be the newly produced candidates involved in AD pathology. Conclusion GAPDH, RHOA, RPS29, and RPS27A are expected to be key candidates for AD progression. The results of this study can provide comprehensive insight into understanding AD's pathogenesis and potential new therapeutic targets.
Collapse
|
29
|
Bakeberg MC, Gorecki AM, Kenna JE, Jefferson A, Byrnes M, Ghosh S, Horne MK, McGregor S, Stell R, Walters S, Mastaglia FL, Anderton RS. Elevated HDL Levels Linked to Poorer Cognitive Ability in Females With Parkinson's Disease. Front Aging Neurosci 2021; 13:656623. [PMID: 34177552 PMCID: PMC8226251 DOI: 10.3389/fnagi.2021.656623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Cholesterol levels have been associated with age-related cognitive decline, however, such an association has not been comprehensively explored in people with Parkinson's disease (PD). To address this uncertainty, the current cross-sectional study examined the cholesterol profile and cognitive performance in a cohort of PD patients. Methods Cognitive function was evaluated using two validated assessments (ACE-R and SCOPA-COG) in 182 people with PD from the Australian Parkinson's Disease Registry. Total cholesterol (TC), high-density lipoprotein (HDL), low-density lipoprotein (LDL), and Triglyceride (TRG) levels were examined within this cohort. The influence of individual lipid subfractions on domain-specific cognitive performance was investigated using covariate-adjusted generalised linear models. Results Females with PD exhibited significantly higher lipid subfraction levels (TC, HDL, and LDL) when compared to male counterparts. While accounting for covariates, HDL levels were strongly associated with poorer performance across multiple cognitive domains in females but not males. Conversely, TC and LDL levels were not associated with cognitive status in people with PD. Conclusion Higher serum HDL associates with poorer cognitive function in females with PD and presents a sex-specific biomarker for cognitive impairment in PD.
Collapse
Affiliation(s)
- Megan C Bakeberg
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Anastazja M Gorecki
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,School of Biological Sciences, The University of Western Australia, Perth, WA, Australia
| | - Jade E Kenna
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Alexa Jefferson
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Michelle Byrnes
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Soumya Ghosh
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Malcolm K Horne
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.,Centre for Clinical Neurosciences and Neurological Research, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia
| | - Sarah McGregor
- Centre for Clinical Neurosciences and Neurological Research, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia
| | - Rick Stell
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Sue Walters
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Frank L Mastaglia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Ryan S Anderton
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia.,School of Health Sciences, Institute for Health Research, The University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
30
|
Toro CA, Hansen J, Siddiq MM, Johnson K, Zhao W, Azulai D, Das DK, Bauman W, Sebra R, Cai D, Iyengar R, Cardozo CP. The Human ApoE4 Variant Reduces Functional Recovery and Neuronal Sprouting After Incomplete Spinal Cord Injury in Male Mice. Front Cell Neurosci 2021; 15:626192. [PMID: 33679326 PMCID: PMC7930340 DOI: 10.3389/fncel.2021.626192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/01/2021] [Indexed: 01/01/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating form of neurotrauma. Patients who carry one or two apolipoprotein E (ApoE)4 alleles show worse functional outcomes and longer hospital stays after SCI, but the cellular and molecular underpinnings for this genetic link remain poorly understood. Thus, there is a great need to generate animal models to accurately replicate the genetic determinants of outcomes after SCI to spur development of treatments that improve physical function. Here, we examined outcomes after a moderate contusion SCI of transgenic mice expressing human ApoE3 or ApoE4. ApoE4 mice have worse locomotor function and coordination after SCI. Histological examination revealed greater glial staining in ApoE4 mice after SCI associated with reduced levels of neuronal sprouting markers. Bulk RNA sequencing revealed that subcellular processes (SCPs), such as extracellular matrix organization and inflammatory responses, were highly ranked among upregulated genes at 7 days after SCI in ApoE4 variants. Conversely, SCPs related to neuronal action potential and neuron projection development were increased in ApoE3 mice at 21 days. In summary, our results reveal a clinically relevant SCI mouse model that recapitulates the influence of ApoE genotypes on post SCI function in individuals who carry these alleles and suggest that the mechanisms underlying worse recovery for ApoE4 animals involve glial activation and loss of sprouting and synaptic activity.
Collapse
Affiliation(s)
- Carlos A Toro
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Spinal Cord Injuries and Disorders System of Care, United States Department of Veterans Affairs, New York, NY, United States
| | - Jens Hansen
- Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mustafa M Siddiq
- Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kaitlin Johnson
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States.,Spinal Cord Injuries and Disorders System of Care, United States Department of Veterans Affairs, New York, NY, United States
| | - Wei Zhao
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Spinal Cord Injuries and Disorders System of Care, United States Department of Veterans Affairs, New York, NY, United States
| | - Daniella Azulai
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States.,Spinal Cord Injuries and Disorders System of Care, United States Department of Veterans Affairs, New York, NY, United States
| | - Dibash K Das
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Spinal Cord Injuries and Disorders System of Care, United States Department of Veterans Affairs, New York, NY, United States
| | - William Bauman
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States.,Spinal Cord Injuries and Disorders System of Care, United States Department of Veterans Affairs, New York, NY, United States
| | - Robert Sebra
- Department of Genetics and Genomic Studies, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Dongming Cai
- Department of Neurology, James J. Peters VA Medical Center, New York, NY, United States.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States.,Department of Rehabilitative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
31
|
The Cholinesterase Inhibitory Properties of Stephaniae Tetrandrae Radix. Molecules 2020; 25:molecules25245914. [PMID: 33327436 PMCID: PMC7764916 DOI: 10.3390/molecules25245914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 11/18/2022] Open
Abstract
Stephaniae tetrandrae radix (STR) is a commonly used traditional Chinese medicine in alleviating edema by inducing diuresis. In the clinic, STR extracts or its components are widely used in the treatment of edema, dysuria, and rheumatism for the regulation of water metabolism. Furthermore, STR has been used in treating emotional problems for years by combining with other Chinese herbs. However, the material basis and mechanism of STR on the nervous system have not been revealed. Here, the main components of STR extracts with different extracting solvents were identified, including three major alkaloids, i.e., cyclanoline, fangchinoline, and tetrandrine. The cholinesterase inhibitory activity of STR extracts and its alkaloids was determined using the Ellman assay. Both cyclanoline and fangchinoline showed acetylcholinesterase (AChE) inhibitory activity, demonstrating noncompetitive enzyme inhibition. In contrast, tetrandrine did not show enzymatic inhibition. The synergism of STR alkaloids with huperzine A or donepezil was calculated by the median-effect principle. The drug combination of fangchinoline–huperzine A or donepezil synergistically inhibited AChE, having a combination index (CI) < 1 at Fa = 0.5. Furthermore, the molecular docking results showed that fangchinoline bound with AChE residues in the peripheral anionic site, and cyclanoline bound with AChE residues in the peripheral anionic site, anionic site, and catalytic site. In parallel, cyclanoline bound with butyrylcholinesterase (BChE) residues in the anionic site, catalytic site, and aromatic site. The results support that fangchinoline and cyclanoline, alkaloids derived from STR, could account for the anti-AChE function of STR. Thus, STR extract or its alkaloids may potentially be developed as a therapeutic strategy for Alzheimer’s patients.
Collapse
|
32
|
Mishra A, Shang Y, Wang Y, Bacon ER, Yin F, Brinton RD. Dynamic Neuroimmune Profile during Mid-life Aging in the Female Brain and Implications for Alzheimer Risk. iScience 2020; 23:101829. [PMID: 33319170 PMCID: PMC7724165 DOI: 10.1016/j.isci.2020.101829] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/13/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022] Open
Abstract
Aging and endocrine transition states can significantly impact inflammation across organ systems. Neuroinflammation is well documented in Alzheimer disease (AD). Herein, we investigated neuroinflammation that emerges during mid-life aging, chronological and endocrinological, in the female brain as an early initiating mechanism driving AD risk later in life. Analyses were conducted in a translational rodent model of mid-life chronological and endocrinological aging followed by validation in transcriptomic profiles from women versus age-matched men. In the translational model, the neuroinflammatory profile of mid-life aging in females was endocrine and chronological state specific, dynamic, anatomically distributed, and persistent. Microarray dataset analyses of aging human hippocampus indicated a sex difference in neuroinflammatory profile in which women exhibited a profile comparable to the pattern discovered in our translational rodent model, whereas age-matched men exhibited a profile consistent with low neuroimmune activation. Translationally, these findings have implications for therapeutic interventions during mid-life to decrease late-onset AD risk.
Collapse
Affiliation(s)
- Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Yuan Shang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Eliza R Bacon
- Department of Medical Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Fei Yin
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Roberta D Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| |
Collapse
|
33
|
Tang MHE, Blair JPM, Bager CL, Bay-Jensen AC, Henriksen K, Christiansen C, Karsdal MA. Matrix metalloproteinase-degraded type I collagen is associated with APOE/TOMM40 variants and preclinical dementia. NEUROLOGY-GENETICS 2020; 6:e508. [PMID: 33134509 PMCID: PMC7577557 DOI: 10.1212/nxg.0000000000000508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/28/2020] [Indexed: 01/06/2023]
Abstract
Objective Dysregulation of type I collagen metabolism has a great impact on human health. We have previously seen that matrix metalloproteinase–degraded type I collagen (C1M) is associated with early death and age-related pathologies. To dissect the biological impact of type I collagen dysregulation, we have performed a genome-wide screening of the genetic factors related to type I collagen turnover. Methods Patient registry data and genotypes have been collected for a total of 4,981 Danish postmenopausal women. Genome-wide association with serum levels of C1M was assessed and phenotype-genotype association analysis performed. Results Twenty-two genome-wide significant variants associated with C1M were identified in the APOE-C1/TOMM40 gene cluster. The APOE-C1/TOMM40 gene cluster is associated with hyperlipidemia and cognitive disorders, and we further found that C1M levels correlated with tau degradation markers and were decreased in women with preclinical cognitive impairment. Conclusions Our study provides elements for better understanding the role of the collagen metabolism in the onset of cognitive impairment.
Collapse
Affiliation(s)
- Man-Hung Eric Tang
- ProScion (M.-H.E.T., J.P.M.B., C.L.B.), Herlev; Faculty of Health and Medical Sciences (J.P.M.B.), University of Copenhagen; ImmunoScience (A.-C.B.-J., C.C., M.A.K.), and Endocrinology (K.H.), Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - Joseph P M Blair
- ProScion (M.-H.E.T., J.P.M.B., C.L.B.), Herlev; Faculty of Health and Medical Sciences (J.P.M.B.), University of Copenhagen; ImmunoScience (A.-C.B.-J., C.C., M.A.K.), and Endocrinology (K.H.), Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - Cecilie Liv Bager
- ProScion (M.-H.E.T., J.P.M.B., C.L.B.), Herlev; Faculty of Health and Medical Sciences (J.P.M.B.), University of Copenhagen; ImmunoScience (A.-C.B.-J., C.C., M.A.K.), and Endocrinology (K.H.), Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - Anne-Christine Bay-Jensen
- ProScion (M.-H.E.T., J.P.M.B., C.L.B.), Herlev; Faculty of Health and Medical Sciences (J.P.M.B.), University of Copenhagen; ImmunoScience (A.-C.B.-J., C.C., M.A.K.), and Endocrinology (K.H.), Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - Kim Henriksen
- ProScion (M.-H.E.T., J.P.M.B., C.L.B.), Herlev; Faculty of Health and Medical Sciences (J.P.M.B.), University of Copenhagen; ImmunoScience (A.-C.B.-J., C.C., M.A.K.), and Endocrinology (K.H.), Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - Claus Christiansen
- ProScion (M.-H.E.T., J.P.M.B., C.L.B.), Herlev; Faculty of Health and Medical Sciences (J.P.M.B.), University of Copenhagen; ImmunoScience (A.-C.B.-J., C.C., M.A.K.), and Endocrinology (K.H.), Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - Morten Asser Karsdal
- ProScion (M.-H.E.T., J.P.M.B., C.L.B.), Herlev; Faculty of Health and Medical Sciences (J.P.M.B.), University of Copenhagen; ImmunoScience (A.-C.B.-J., C.C., M.A.K.), and Endocrinology (K.H.), Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| |
Collapse
|
34
|
de Lange AG, Barth C, Kaufmann T, Anatürk M, Suri S, Ebmeier KP, Westlye LT. The maternal brain: Region-specific patterns of brain aging are traceable decades after childbirth. Hum Brain Mapp 2020; 41:4718-4729. [PMID: 32767637 PMCID: PMC7555081 DOI: 10.1002/hbm.25152] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/30/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
Pregnancy involves maternal brain adaptations, but little is known about how parity influences women's brain aging trajectories later in life. In this study, we replicated previous findings showing less apparent brain aging in women with a history of childbirths, and identified regional brain aging patterns linked to parity in 19,787 middle- and older-aged women. Using novel applications of brain-age prediction methods, we found that a higher number of previous childbirths were linked to less apparent brain aging in striatal and limbic regions. The strongest effect was found in the accumbens-a key region in the mesolimbic reward system, which plays an important role in maternal behavior. While only prospective longitudinal studies would be conclusive, our findings indicate that subcortical brain modulations during pregnancy and postpartum may be traceable decades after childbirth.
Collapse
Affiliation(s)
- Ann‐Marie G. de Lange
- Department of PsychiatryUniversity of OxfordOxfordUK
- Department of PsychologyUniversity of OsloOsloNorway
- NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Claudia Barth
- NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Tobias Kaufmann
- NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Melis Anatürk
- Department of PsychiatryUniversity of OxfordOxfordUK
- Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
| | - Sana Suri
- Department of PsychiatryUniversity of OxfordOxfordUK
- Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
| | | | - Lars T. Westlye
- Department of PsychologyUniversity of OsloOsloNorway
- NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and AddictionOslo University HospitalOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
| |
Collapse
|
35
|
McCarthy M, Raval AP. The peri-menopause in a woman's life: a systemic inflammatory phase that enables later neurodegenerative disease. J Neuroinflammation 2020; 17:317. [PMID: 33097048 PMCID: PMC7585188 DOI: 10.1186/s12974-020-01998-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023] Open
Abstract
The peri-menopause or menopausal transition—the time period that surrounds the final years of a woman’s reproductive life—is associated with profound reproductive and hormonal changes in a woman’s body and exponentially increases a woman’s risk of cerebral ischemia and Alzheimer’s disease. Although our understanding of the exact timeline or definition of peri-menopause is limited, it is clear that there are two stages to the peri-menopause. These are the early menopausal transition, where menstrual cycles are mostly regular, with relatively few interruptions, and the late transition, where amenorrhea becomes more prolonged and lasts for at least 60 days, up to the final menstrual period. Emerging evidence is showing that peri-menopause is pro-inflammatory and disrupts estrogen-regulated neurological systems. Estrogen is a master regulator that functions through a network of estrogen receptors subtypes alpha (ER-α) and beta (ER-β). Estrogen receptor-beta has been shown to regulate a key component of the innate immune response known as the inflammasome, and it also is involved in regulation of neuronal mitochondrial function. This review will present an overview of the menopausal transition as an inflammatory event, with associated systemic and central nervous system inflammation, plus regulation of the innate immune response by ER-β-mediated mechanisms.
Collapse
Affiliation(s)
- Micheline McCarthy
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA. .,Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
36
|
de Lange AMG, Barth C, Kaufmann T, Maximov II, van der Meer D, Agartz I, Westlye LT. Women's brain aging: Effects of sex-hormone exposure, pregnancies, and genetic risk for Alzheimer's disease. Hum Brain Mapp 2020; 41:5141-5150. [PMID: 32856754 PMCID: PMC7670641 DOI: 10.1002/hbm.25180] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/17/2020] [Accepted: 08/04/2020] [Indexed: 12/27/2022] Open
Abstract
Sex hormones such as estrogen fluctuate across the female lifespan, with high levels during reproductive years and natural decline during the transition to menopause. Women's exposure to estrogen may influence their heightened risk of Alzheimer's disease (AD) relative to men, but little is known about how it affects normal brain aging. Recent findings from the UK Biobank demonstrate less apparent brain aging in women with a history of multiple childbirths, highlighting a potential link between sex-hormone exposure and brain aging. We investigated endogenous and exogenous sex-hormone exposure, genetic risk for AD, and neuroimaging-derived biomarkers for brain aging in 16,854 middle to older-aged women. The results showed that as opposed to parity, higher cumulative sex-hormone exposure was associated with more evident brain aging, indicating that i) high levels of cumulative exposure to sex-hormones may have adverse effects on the brain, and ii) beneficial effects of pregnancies on the female brain are not solely attributable to modulations in sex-hormone exposure. In addition, for women using hormonal replacement therapy (HRT), starting treatment earlier was associated with less evident brain aging, but only in women with a genetic risk for AD. Genetic factors may thus contribute to how timing of HRT initiation influences women's brain aging trajectories.
Collapse
Affiliation(s)
- Ann-Marie G de Lange
- Department of Psychology, University of Oslo, Oslo, Norway.,NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.,Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Claudia Barth
- NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Tobias Kaufmann
- NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ivan I Maximov
- Department of Psychology, University of Oslo, Oslo, Norway.,NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Dennis van der Meer
- NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.,School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Ingrid Agartz
- NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.,Department of Psychiatry, Diakonhjemmet Hospital, Oslo, Norway.,Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institute, Stockholm, Sweden.,KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Lars T Westlye
- Department of Psychology, University of Oslo, Oslo, Norway.,NORMENT, Institute of Clinical Medicine, University of Oslo, & Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.,KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| |
Collapse
|
37
|
Barth C, de Lange AMG. Towards an understanding of women's brain aging: the immunology of pregnancy and menopause. Front Neuroendocrinol 2020; 58:100850. [PMID: 32504632 DOI: 10.1016/j.yfrne.2020.100850] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/23/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
Abstract
Women are at significantly greater risk of developing Alzheimer's disease and show higher prevalence of autoimmune conditions relative to men. Women's brain health is historically understudied, and little is therefore known about the mechanisms underlying epidemiological sex differences in neurodegenerative diseases, and how female-specific factors may influence women's brain health across the lifespan. In this review, we summarize recent studies on the immunology of pregnancy and menopause, emphasizing that these major immunoendocrine transition phases may play a critical part in women's brain aging trajectories.
Collapse
Affiliation(s)
- Claudia Barth
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Ann-Marie G de Lange
- Department of Psychology, University of Oslo, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK.
| |
Collapse
|
38
|
Vegeto E, Villa A, Della Torre S, Crippa V, Rusmini P, Cristofani R, Galbiati M, Maggi A, Poletti A. The Role of Sex and Sex Hormones in Neurodegenerative Diseases. Endocr Rev 2020; 41:5572525. [PMID: 31544208 PMCID: PMC7156855 DOI: 10.1210/endrev/bnz005] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDs) are a wide class of disorders of the central nervous system (CNS) with unknown etiology. Several factors were hypothesized to be involved in the pathogenesis of these diseases, including genetic and environmental factors. Many of these diseases show a sex prevalence and sex steroids were shown to have a role in the progression of specific forms of neurodegeneration. Estrogens were reported to be neuroprotective through their action on cognate nuclear and membrane receptors, while adverse effects of male hormones have been described on neuronal cells, although some data also suggest neuroprotective activities. The response of the CNS to sex steroids is a complex and integrated process that depends on (i) the type and amount of the cognate steroid receptor and (ii) the target cell type-either neurons, glia, or microglia. Moreover, the levels of sex steroids in the CNS fluctuate due to gonadal activities and to local metabolism and synthesis. Importantly, biochemical processes involved in the pathogenesis of NDs are increasingly being recognized as different between the two sexes and as influenced by sex steroids. The aim of this review is to present current state-of-the-art understanding on the potential role of sex steroids and their receptors on the onset and progression of major neurodegenerative disorders, namely, Alzheimer's disease, Parkinson's diseases, amyotrophic lateral sclerosis, and the peculiar motoneuron disease spinal and bulbar muscular atrophy, in which hormonal therapy is potentially useful as disease modifier.
Collapse
Affiliation(s)
- Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze della Salute (DiSS), Università degli Studi di Milano, Italy
| | - Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Valeria Crippa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Paola Rusmini
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Riccardo Cristofani
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Mariarita Galbiati
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| |
Collapse
|
39
|
Shi XL, Yan N, Cui YJ, Liu ZP. A Unique GSK-3β inhibitor B10 Has a Direct Effect on Aβ, Targets Tau and Metal Dyshomeostasis, and Promotes Neuronal Neurite Outgrowth. Cells 2020; 9:cells9030649. [PMID: 32155989 PMCID: PMC7140427 DOI: 10.3390/cells9030649] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/02/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
Due to the complicated pathogenesis of Alzheimer's disease (AD), the development of multitargeted agents to simultaneously interfere with multiple pathological processes of AD is a potential choice. Glycogen synthase kinase-3β (GSK-3β) plays a vital role in the AD pathological process. In this study, we discovered a novel 1H-pyrrolo[2,3-b]pyridine derivative B10 as a GSK-3β inhibitor that features with a quinolin-8-ol moiety to target the metal dyshomeostasis of AD. B10 potently inhibited GSK-3β with an IC50 of 66 ± 2.5 nM. At the concentration of 20 μM, B10 increased β-catenin abundance (β-catenin/GAPDH: 0.83 ± 0.086 vs. 0.30 ± 0.016), phosphorylated GSK-3β at Ser9 (p-GSK-3β/GAPDH: 0.53 ± 0.045 vs. 0.35 ± 0.012), and decreased the phosphorylated tau level (p-tau/GAPDH: 0.33 ± 0.065 vs. 0.83 ± 0.061) in SH-SY5Y cells. Unlike other GSK-3β inhibitors, B10 had a direct effect on Aβ by inhibiting Aβ1-42 aggregation and promoting the Aβ1-42 aggregate disassociation. It selectively chelated with Cu2+, Zn2+, Fe3+, and Al3+, and targeted AD metal dyshomeostasis. Moreover, B10 effectively increased the mRNA expression of the recognized neurogenesis markers, GAP43, N-myc, and MAP-2, and promoted the differentiated neuronal neurite outgrowth, possibly through the GSK-3β and β-catenin signal pathways. Therefore, B10 is a potent and unique GSK-3β inhibitor that has a direct on Aβ and serves as a multifunctional anti-AD agent for further investigations.
Collapse
|
40
|
Wang Y, Mishra A, Brinton RD. Transitions in metabolic and immune systems from pre-menopause to post-menopause: implications for age-associated neurodegenerative diseases. F1000Res 2020; 9. [PMID: 32047612 PMCID: PMC6993821 DOI: 10.12688/f1000research.21599.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
The brain undergoes two aging programs: chronological and endocrinological. This is particularly evident in the female brain, which undergoes programs of aging associated with reproductive competency. Comprehensive understanding of the dynamic metabolic and neuroinflammatory aging process in the female brain can illuminate windows of opportunities to promote healthy brain aging. Bioenergetic crisis and chronic low-grade inflammation are hallmarks of brain aging and menopause and have been implicated as a unifying factor causally connecting genetic risk factors for Alzheimer's disease and other neurodegenerative diseases. In this review, we discuss metabolic phenotypes of pre-menopausal, peri-menopausal, and post-menopausal aging and their consequent impact on the neuroinflammatory profile during each transition state. A critical aspect of the aging process is the dynamic metabolic neuro-inflammatory profiles that emerge during chronological and endocrinological aging. These dynamic systems of biology are relevant to multiple age-associated neurodegenerative diseases and provide a therapeutic framework for prevention and delay of neurodegenerative diseases of aging. While these findings are based on investigations of the female brain, they have a broader fundamental systems of biology strategy for investigating the aging male brain. Molecular characterization of alterations in fuel utilization and neuroinflammatory mechanisms during these neuro-endocrine transition states can inform therapeutic strategies to mitigate the risk of Alzheimer's disease in women. We further discuss a precision hormone replacement therapy approach to target symptom profiles during endocrine and chronological aging to reduce risk for age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
41
|
Shang Y, Mishra A, Wang T, Wang Y, Desai M, Chen S, Mao Z, Do L, Bernstein AS, Trouard TP, Brinton RD. Evidence in support of chromosomal sex influencing plasma based metabolome vs APOE genotype influencing brain metabolome profile in humanized APOE male and female mice. PLoS One 2020; 15:e0225392. [PMID: 31917799 PMCID: PMC6952084 DOI: 10.1371/journal.pone.0225392] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/29/2019] [Indexed: 01/18/2023] Open
Abstract
Late onset Alzheimer’s disease (LOAD) is a progressive neurodegenerative disease with four well-established risk factors: age, APOE4 genotype, female chromosomal sex, and maternal history of AD. Each risk factor impacts multiple systems, making LOAD a complex systems biology challenge. To investigate interactions between LOAD risk factors, we performed multiple scale analyses, including metabolomics, transcriptomics, brain magnetic resonance imaging (MRI), and beta-amyloid assessment, in 16 months old male and female mice with humanized human APOE3 (hAPOE3) or APOE4 (hAPOE4) genes. Metabolomic analyses indicated a sex difference in plasma profile whereas APOE genotype determined brain metabolic profile. Consistent with the brain metabolome, gene and pathway-based RNA-Seq analyses of the hippocampus indicated increased expression of fatty acid/lipid metabolism related genes and pathways in both hAPOE4 males and females. Further, female transcription of fatty acid and amino acids pathways were significantly different from males. MRI based imaging analyses indicated that in multiple white matter tracts, hAPOE4 males and females exhibited lower fractional anisotropy than their hAPOE3 counterparts, suggesting a lower level of white matter integrity in hAPOE4 mice. Consistent with the brain metabolomic and transcriptomic profile of hAPOE4 carriers, beta-amyloid generation was detectable in 16-month-old male and female brains. These data provide therapeutic targets based on chromosomal sex and APOE genotype. Collectively, these data provide a framework for developing precision medicine interventions during the prodromal phase of LOAD, when the potential to reverse, prevent and delay LOAD progression is greatest.
Collapse
Affiliation(s)
- Yuan Shang
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona, United States of America
| | - Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona, United States of America
| | - Tian Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona, United States of America
| | - Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona, United States of America
| | - Maunil Desai
- School of Pharmacy, University of Southern California, Los Angeles, California, United States of America
| | - Shuhua Chen
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona, United States of America
| | - Zisu Mao
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona, United States of America
| | - Loi Do
- Biomedical Engineering, University of Arizona, Tucson, Arizona, United States of America
| | - Adam S. Bernstein
- College of Medicine, University of Arizona, Tucson, Arizona, United States of America
| | - Theodore P. Trouard
- Biomedical Engineering, University of Arizona, Tucson, Arizona, United States of America
| | - Roberta D. Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
42
|
|
43
|
Toro CA, Das DK, Cai D, Cardozo CP. Elucidating the Role of Apolipoprotein E Isoforms in Spinal Cord Injury-Associated Neuropathology. J Neurotrauma 2019; 36:3317-3322. [PMID: 31218915 DOI: 10.1089/neu.2018.6334] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating, life-altering, neurological event that affects ∼300,000 individuals in the United States. Currently, there are no effective treatments to reverse the neurological impairments caused by the lesion. Until a cure is available, there is an urgent need for strategies that can either spare injured neurons or promote neuroplasticity and functional recovery. Genetic links to outcomes after SCI may provide insights into the pathological mechanisms, and possible new avenues for drug development. In the present review, we discuss the current knowledge linking apolipoprotein E genotypes with better or worse functional outcomes after an SCI, and the possible molecular mechanisms that may contribute to this association.
Collapse
Affiliation(s)
- Carlos A Toro
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Bronx, New York
| | - Dibash K Das
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Bronx, New York
| | - Dongming Cai
- Neurology Service, James J. Peters VA, Bronx, New York
- Department of Neurology, Icahn School of Medicine at Mount Sinai, Bronx, New York
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Bronx, New York
- Department of Rehabilitative Medicine, Icahn School of Medicine at Mount Sinai, Bronx, New York
| |
Collapse
|
44
|
Short H. 'Unheard and undertreated': Menopausal mental health (September editorial). Post Reprod Health 2019; 25:118-119. [PMID: 31630614 DOI: 10.1177/2053369119877086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
|
45
|
Johnson DB, Manouchehri A, Haugh AM, Quach HT, Balko JM, Lebrun-Vignes B, Mammen A, Moslehi JJ, Salem JE. Neurologic toxicity associated with immune checkpoint inhibitors: a pharmacovigilance study. J Immunother Cancer 2019; 7:134. [PMID: 31118078 PMCID: PMC6530194 DOI: 10.1186/s40425-019-0617-x] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) produce durable antitumor responses but provoke autoimmune toxicities, including uncommon but potentially devastating neurologic toxicities. The clinical features, including the spectrum, timing, and outcomes, of ICI-induced neurologic toxicities are not well characterized. METHODS We performed disproportionality analysis using Vigibase, the World Health Organization pharmacovigilance database, comparing neurologic adverse event (AE) reporting in patients receiving ICIs vs. the full database. Neurologic AEs were classified by group queries using Medical Dictionary for Regulatory Activities, between database inception to September 28, 2018. Associations between ICIs and neurologic AEs were assessed using reporting odds ratios (ROR) and information component (IC). IC compares observed and expected values to find associations between drugs and AEs using disproportionate Bayesian reporting; IC025 (lower end of the IC 95% credibility interval) > 0 is considered statistically significant. RESULTS Among the full database, 18,518,994 AEs were reported, including 48,653 with ICIs. ICIs were associated with higher incidence of myasthenia gravis (0.47% of ICI reports vs. 0.04% of the full database, ROR 16.5 [95% CI 14.5-18.9]; IC025 3.31), encephalitis (0.51% vs. 0.05%, ROR 10.4 [95% CI 9.2-11.8]; IC025 3.15), peripheral neuropathy (1.16% vs. 0.67%, IC025 0.68), and meningitis (0.15% vs. 0.06%, ROR 3.1 [95% CI 2.5-3.9]; IC025 1.01). Myasthenia gravis and encephalitis were associated with anti-PD-1 whereas other neurologic AEs were associated with anti-CTLA-4. Myasthenia gravis was characterized by high fatality rates (~ 20%), early onset (median 29 days), and frequent concurrent myocarditis and myositis; whereas other neurologic AEs had lower fatality rates (6-12%), later onset (median 61-80 days), and were non-overlapping. CONCLUSIONS ICIs produce a spectrum of distinct classes of neurologic AEs that can cause significant morbidity and mortality and tend to occur early and with class-specific associations.
Collapse
Affiliation(s)
- Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.
| | - Ali Manouchehri
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA
| | - Alexandra M Haugh
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA
| | - Henry T Quach
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA
| | - Benedicte Lebrun-Vignes
- Department of Pharmacology, Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Regional Pharmacovigilance Centre, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrew Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Javid J Moslehi
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA
| | - Joe-Elie Salem
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA
- Department of Pharmacology, Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Regional Pharmacovigilance Centre, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
46
|
Luo H, Xiang Y, Qu X, Liu H, Liu C, Li G, Han L, Qin X. Apelin-13 Suppresses Neuroinflammation Against Cognitive Deficit in a Streptozotocin-Induced Rat Model of Alzheimer's Disease Through Activation of BDNF-TrkB Signaling Pathway. Front Pharmacol 2019; 10:395. [PMID: 31040784 PMCID: PMC6477031 DOI: 10.3389/fphar.2019.00395] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/29/2019] [Indexed: 01/14/2023] Open
Abstract
Alzheimer’s disease (AD), a progressive neurodegenerative disease characterized by impairments of cognitive function as a result of synaptic deficits and neuronal loss, is associated with inflammation. Apelin-13, a predominant neuropeptide with inhibiting effect on inflammation, has beneficial effects on cognition memory and neuronal damage. However, whether apelin-13 can protect neurons to ameliorate cognitive deficits in AD by inhibiting the inflammatory response remains largely unknown. To test this hypothesis, rats were intracerebroventricularly (ICV) injected with streptozotocin (3 mg/kg) alone or in combination with apelin-13 (2 μg). And tyrosine receptor kinase B (TrkB) blocker K252a (200 nM) was administrated 10 min before apelin injection. Furthermore, cognitive performance was assessed by new object recognition (NOR) and Y-maze tests. Protein expression of apelin, APJ, microglial marker (IBA1), astroglia marker (GFAP), interleukin 1 beta (IL-1β), tumor necrosis factor-α (TNF-α), synaptophysin (SYP), brain-derived neurotrophic factor (BDNF), TrkB, phospho-TrkB (p-TrkB) in the hippocampus were examined by western blotting or immunohistochemistry. And the gene expression of IBA1, GFAP, IL-1β, TNF-α, and SYP were detected by real-time quantitative polymerase chain reaction (PCR). Inflammatory disorder in the hippocampus was tested by hematoxylin and eosin (H&E) staining. The enzyme-linked immunosorbent assay (ELISA) was used to study the expression level of acetylcholine. And the activity of acetylcholinesterase was detected by Acetylcholinesterase Assay Kit. We observed that apelin/APJ signaling was downregulated in the hippocampus of rats administrated with STZ. Apelin-13 was found to significantly ameliorate STZ-induced AD-like phenotypes including congnitive deficit, cholinergic disfunction and the damage of neuron and synaptic plasticity. Moreover, apelin-13 inhibited microglia and astrocyte activation, reduced IL-1β and TNF-α expression and hippocampal BDNF/TrkB expression deficit in AD rats. Finally, apelin-13-mediated effects were blocked by TrkB receptor antagonist K252a. These results suggest that apelin-13 upregulates BDNF/TrkB pathway against cognitive deficit in a STZ-induced rat model of sporadic AD by attenuating inflammation.
Collapse
Affiliation(s)
- Huaiqing Luo
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Chi Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Guangyi Li
- Department of Anatomy, Histology and Embryology, Changsha Medical University, Changsha, China
| | - Li Han
- Department of Physiology, School of Basic Medical Science, Changsha Medical University, Changsha, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
| |
Collapse
|
47
|
Man MQ, Yang B, Elias PM. Benefits of Hesperidin for Cutaneous Functions. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:2676307. [PMID: 31061668 PMCID: PMC6466919 DOI: 10.1155/2019/2676307] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022]
Abstract
Hesperidin is a bioflavonoid, with high concentration in citrus fruits. In addition to its well-known benefits for cardiovascular function, type II diabetes, and anti-inflammation, recent studies have demonstrated multiple benefits of hesperidin for cutaneous functions, including wound healing, UV protection, anti-inflammation, antimicrobial, antiskin cancer, and skin lightening. In addition, hesperidin enhances epidermal permeability barrier homeostasis in both normal young and aged skin. The mechanisms by which hesperidin benefits cutaneous functions are attributable to its antioxidant properties, inhibition of MAPK-dependent signaling pathways, and stimulation of epidermal proliferation, differentiation, and lipid production. Because of its low cost, wide availability, and superior safety, hesperidin could prove useful for the management of a variety of cutaneous conditions.
Collapse
Affiliation(s)
- Mao-Qiang Man
- Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
- Department of Dermatology, University of California San Francisco and Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Peter M. Elias
- Department of Dermatology, University of California San Francisco and Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| |
Collapse
|