1
|
Christensen A, McGill CJ, Qian W, Pike CJ. Effects of obesogenic diet and 17β-estradiol in female mice with APOE 3/3, 3/4, and 4/4 genotypes. Front Aging Neurosci 2024; 16:1415072. [PMID: 39347015 PMCID: PMC11427389 DOI: 10.3389/fnagi.2024.1415072] [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: 04/09/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
The main genetic risk factor for Alzheimer's disease (AD) is the apolipoprotein E ε4 allele (APOE4). AD risk associated with APOE4 disproportionately affects women. Furthermore, human and rodent studies indicate that the cognitive deficits associated with APOE4 are greater in females. One modifiable AD risk factor is obesity during middle age. Given that approximately two-thirds of US adults are overweight, it is important to understand how obesity affects AD risk, how it interacts with APOE4, and the extent to which its detrimental effects can be mitigated with therapeutics. One intervention study for women is estrogen-based hormone therapy, which can exert numerous health benefits when administered in early middle age. No experimental studies have examined the interactions among APOE4, obesity, and hormone therapy in aging females. To begin to explore these issues, we considered how obesity outcomes are affected by treatment with estradiol at the onset of middle age in female mice with human APOE3 and APOE4. Furthermore, to explore how gene dosage affects outcomes, we compared mice homozygous for APOE3 (3/3) and homozygous (4/4) or hemizygous (3/4) for APOE4. Mice were examined over a 4-month period that spans the transition into reproductive senescence, a normal age-related change that models many aspects of human perimenopause. Beginning at 5 months of age, mice were maintained on a control diet (10% fat) or high-fat diet (HFD; 60% fat). After 8 weeks, by which time obesity was present in all HFD groups, mice were implanted with an estradiol or vehicle capsule that was maintained for the final 8 weeks. Animals were assessed on a range of metabolic and neural measures. Overall, APOE4 was associated with poorer metabolic function and cognitive performance. However, an obesogenic diet induced relatively greater impairments in metabolic function and cognitive performance in APOE3/3 mice. Estradiol treatment improved metabolic and cognitive outcomes across all HFD groups, with APOE4/4 generally exhibiting the greatest benefit. APOE3/4 mice were intermediate to the homozygous genotypes on many measures but also exhibited unique profiles. Together, these findings highlight the importance of the APOE genotype as a modulator of the risks associated with obesity and the beneficial outcomes of estradiol.
Collapse
Affiliation(s)
| | | | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
2
|
Findley CA, McFadden S.A, Hill T, Peck MR, Quinn K, Hascup KN, Hascup ER. Sexual Dimorphism, Altered Hippocampal Glutamatergic Neurotransmission and Cognitive Impairment in APP Knock-In Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.05.570100. [PMID: 38106074 PMCID: PMC10723272 DOI: 10.1101/2023.12.05.570100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background It is well established that glutamatergic neurotransmission plays an essential role in learning and memory. Previous studies indicate that glutamate dynamics shift with Alzheimer's disease (AD) progression, contributing to negative cognitive outcomes. Objective In this study, we characterized hippocampal glutamatergic signaling with age and disease progression in a knock-in mouse model of AD (APPNL-F/NL-F). Methods At 2-4 and 18+ months old, male and female APPNL/NL, APPNL-F/NL-F, and C57BL/6 mice underwent cognitive assessment using Morris water maze (MWM) and Novel Object Recognition (NOR). Then, basal and 70 mM KCl stimulus-evoked glutamate release was measured in the dentate gyrus (DG), CA3, and CA1 regions of the hippocampus using a glutamate-selective microelectrode in anesthetized mice. Results Glutamate recordings support elevated stimulus-evoked glutamate release in the DG and CA3 of young APPNL-F/NL-F male mice that declined with age compared to age-matched control mice. Young female APPNL-F/NL-F mice exhibited increased glutamate clearance in the CA1 that slowed with age compared to age-matched control mice. Male and female APPNL-F/NL-F mice exhibited decreased CA1 basal glutamate levels, while males also showed depletion in the CA3. Cognitive assessment demonstrated impaired spatial cognition in aged male and female APPNL-F/NL-F mice, but only aged females displayed recognition memory deficits compared to age-matched control mice. Conclusions: These findings confirm a sex-dependent hyper-to-hypoactivation glutamatergic paradigm in APPNL-F/NL-F mice. Further, data illustrate a sexually dimorphic biological aging process resulting in a more severe cognitive phenotype for female APPNL-F/NL-F mice than their male counterparts. Research outcomes mirror that of human AD pathology and provide further evidence of divergent AD pathogenesis between sexes.
Collapse
Affiliation(s)
- Caleigh A. Findley
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
- Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Samuel .A. McFadden
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
| | - Tiarra. Hill
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
| | - Mackenzie R. Peck
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
| | - Kathleen Quinn
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
| | - Kevin N. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
- Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
- Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
3
|
He W, Zhang S, Qi Z, Liu W. Unveiling the potential of estrogen: Exploring its role in neuropsychiatric disorders and exercise intervention. Pharmacol Res 2024; 204:107201. [PMID: 38704108 DOI: 10.1016/j.phrs.2024.107201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/01/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Neuropsychiatric disorders shorten human life spans through multiple ways and become major threats to human health. Exercise can regulate the estrogen signaling, which may be involved in depression, Alzheimer's disease (AD) and Parkinson's disease (PD), and other neuropsychiatric disorders as well in their sex differences. In nervous system, estrogen is an important regulator of cell development, synaptic development, and brain connectivity. Therefore, this review aimed to investigate the potential of estrogen system in the exercise intervention of neuropsychiatric disorders to better understand the exercise in neuropsychiatric disorders and its sex specific. Exercise can exert a protective effect in neuropsychiatric disorders through regulating the expression of estrogen and estrogen receptors, which are involved in neuroprotection, neurodevelopment, and neuronal glucose homeostasis. These processes are mediated by the downstream factors of estrogen signaling, including N-myc downstream regulatory gene 2 (Ndrg2), serotonin (5-HT), delta like canonical Notch ligand 1 (DLL1), NOD-like receptor thermal protein domain associated protein 3 (NLRP3), etc. In addition, exercise can act on the estrogen response element (ERE) fragment in the genes of estrogenic downstream factors like β-amyloid precursor protein cleavase 1 (BACE1). However, there are few studies on the relationship between exercise, the estrogen signaling pathway, and neuropsychiatric disorders. Hence, we review how the estrogen signaling mediates the mechanism of exercise intervention in neuropsychiatric disorders. We aim to provide a theoretical perspective for neuropsychiatric disorders affecting female health and provide theoretical support for the design of exercise prescriptions.
Collapse
Affiliation(s)
- Wenke He
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; College of Physical Education and Health, East China Normal University, Shanghai 200241,China
| | - Sen Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; College of Physical Education and Health, East China Normal University, Shanghai 200241,China
| | - Zhengtang Qi
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; College of Physical Education and Health, East China Normal University, Shanghai 200241,China.
| | - Weina Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; College of Physical Education and Health, East China Normal University, Shanghai 200241,China.
| |
Collapse
|
4
|
Baer SB, Dorn AD, Osborne DM. Sex differences in response to obesity and caloric restriction on cognition and hippocampal measures of autophagic-lysosomal transcripts and signaling pathways. BMC Neurosci 2024; 25:1. [PMID: 38166559 PMCID: PMC10759648 DOI: 10.1186/s12868-023-00840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 12/18/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Obesity rates in the U.S. continue to increase, with nearly 50% of the population being either obese or morbidly obese. Obesity, along with female sex, are leading risk factors for sporadic Alzheimer's Disease (AD) necessitating the need to better understand how these variables impact cellular function independent of age or genetic mutations. Animal and clinical studies both indicate that autophagy-lysosomal pathway (ALP) dysfunction is among the earliest known cellular systems to become perturbed in AD, preceding cognitive decline, yet little is known about how obesity and sex affects these cellular functions in the hippocampus, a brain region uniquely susceptible to the negative effects of obesity. We hypothesized that obesity would negatively affect key markers of ALP in the hippocampus, effects would vary based on sex, and that caloric restriction would counteract obesity effects. METHODS Female and male mice were placed on an obesogenic diet for 10 months, at which point half were switched to caloric restriction for three months, followed by cognitive testing in the Morris watermaze. Hippocampus was analyzed by western blot and qPCR. RESULTS Cognitive function in female mice responded differently to caloric restriction based on whether they were on a normal or obesogenic diet; male cognition was only mildly affected by caloric restriction and not obesity. Significant male-specific changes occurred in cellular markers of autophagy, including obesity increasing pAkt, Slc38a9, and Atg12, while caloric restriction reduced pRPS6 and increased Atg7. In contrast females experienced changes due to diet/caloric restriction predominately in lysosomal markers including increased TFE3, FLCN, FNIP2, and pAMPK. CONCLUSIONS Results support that hippocampal ALP is a target of obesity and that sex shapes molecular responses, while providing insight into how dietary manipulations affect learning and memory based on sex.
Collapse
Affiliation(s)
- Sadie B Baer
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR, USA
| | - Adrianah D Dorn
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR, USA
| | | |
Collapse
|
5
|
Balu D, Valencia-Olvera AC, Islam Z, Mielczarek C, Hansen A, Perez Ramos TM, York J, LaDu MJ, Tai LM. APOE genotype and sex modulate Alzheimer's disease pathology in aged EFAD transgenic mice. Front Aging Neurosci 2023; 15:1279343. [PMID: 38020764 PMCID: PMC10644540 DOI: 10.3389/fnagi.2023.1279343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Increasing evidence supports that age, APOE and sex interact to modulate Alzheimer's disease (AD) risk, however the underlying pathways are unclear. One way that AD risk factors may modulate cognition is by impacting amyloid beta (Aβ) accumulation as plaques, and/or neuroinflammation Therefore, the goal of the present study was to evaluate the extent to which age, APOE and sex modulate Aβ pathology, neuroinflammation and behavior in vivo. To achieve this goal, we utilized the EFAD mice, which express human APOE3 or APOE4 and have five familial AD mutations (FAD) that result in Aβ42 overproduction. We assessed Aβ levels, reactive glia and Morris water maze performance in 6-, 10-, 14-, and 18-month-old EFAD mice. Female APOE4 mice had the highest Aβ deposition, fibrillar amyloid deposits and neuroinflammation as well as earlier behavior deficits. Interestingly, we found that female APOE3 mice and male APOE4 mice had similar levels of pathology. Collectively our data support that the combination of APOE4 and female sex is the most detrimental combination for AD, and that at older ages, female sex may be equivalent to APOE4 genotype.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ana C. Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zarak Islam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Clare Mielczarek
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Allison Hansen
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Peoria, IL, United States
| | - Tamara M. Perez Ramos
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- School of Medicine, St. George’s University, St. George’s, Grenada
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
6
|
Walter KR, Ricketts DK, Presswood BH, Smith SM, Mooney SM. Prenatal alcohol exposure causes persistent microglial activation and age- and sex- specific effects on cognition and metabolic outcomes in an Alzheimer's Disease mouse model. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2023; 49:302-320. [PMID: 36194703 PMCID: PMC11040461 DOI: 10.1080/00952990.2022.2119571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/22/2022] [Accepted: 08/28/2022] [Indexed: 11/06/2022]
Abstract
Background: Prenatal alcohol exposure (PAE) causes behavioral deficits and increases risk of metabolic diseases. Alzheimer's Disease (AD) is a neurodegenerative disease that has a higher risk in adults with metabolic diseases. Both present with persistent neuroinflammation.Objectives: We tested whether PAE exacerbates AD-related cognitive decline in a mouse model (3xTg-AD; presenilin/amyloid precursor protein/tau), and assessed associations among cognition, metabolic impairment, and microglial reactivity.Methods: Alcohol-exposed (ALC) pregnant 3xTg-AD mice received 3 g/kg alcohol from embryonic day 8.5-17.5. We evaluated recognition memory and associative memory (fear conditioning) in 8-10 males and females per group at 3 months of age (3mo), 7mo, and 11mo, then assessed glucose tolerance, body composition, and hippocampal microglial activation at 12mo.Results: ALC females had higher body weights than controls from 5mo (p < .0001). Controls showed improved recognition memory at 11mo compared with 3mo (p = .007); this was not seen in ALC mice. Older animals froze more during fear conditioning than younger, and ALC mice were hyper-responsive to the fear-related cue (p = .017). Fasting blood glucose was lower in ALC males and higher in ALC females than controls. Positive associations occurred between glucose and fear-related context (p = .04) and adiposity and fear-related cue (p = .0002) in ALC animals. Hippocampal microglial activation was higher in ALC than controls (p < .0001); this trended to correlate with recognition memory.Conclusions: ALC animals showed age-related cognitive impairments that did not interact with AD risk but did correlate with metabolic dysfunction and somewhat with microglial activation. Thus, metabolic disorders may be a therapeutic target for people with FASDs.
Collapse
Affiliation(s)
- Kathleen R. Walter
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis NC 28081, USA
| | - Dane K. Ricketts
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis NC 28081, USA
| | - Brandon H. Presswood
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis NC 28081, USA
| | - Susan M. Smith
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis NC 28081, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis NC 28081, USA
| | - Sandra M. Mooney
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis NC 28081, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis NC 28081, USA
| |
Collapse
|
7
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-specific effects of high-fat diet on rat brain glucose metabolism and early-onset dementia symptoms. Mech Ageing Dev 2023; 211:111795. [PMID: 36828273 DOI: 10.1016/j.mad.2023.111795] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/08/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023]
Abstract
Peripheral metabolic disturbances are associated with a variety of clinical health consequences and may contribute to the development of neurocognitive disorders. This study investigates whether long-term high-fat diet (HFD) consumption changes the brain glucose metabolism and impairs memory performance in a sex-dependent manner. Male and female rats, after weaning, were fed HFD or normal chow diet (NCD) for 16 weeks. Behavioral tests for spatial memory and an 18 F-FDG-PET scan were performed. Also, the expression of brain insulin resistance markers and Alzheimer's pathology-related genes was assessed by qPCR. The Morris water maze and Y-maze results showed, respectively, that memory retrieval and spatial working memory were impaired only in HFD male rats compared to NCD controls. In addition, measuring whole brain 18 F-FDG uptake indicated a significant reduction in glucose metabolism in male but not female HFD rats. Analysis of 15 genes related to glucose metabolism and Alzheimer's pathology, in the hippocampus, showed that expression of GLUT3, IRS2, and IDE is significantly reduced in HFD male rats. Our results suggest that sex affects the HFD-induced dysregulation of brain glucose metabolism and cognitive performance.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Coyle-Asbil B, Ogilvie LM, Simpson JA. Emerging roles for estrogen in regulating skeletal muscle physiology. Physiol Genomics 2023; 55:75-78. [PMID: 36622080 DOI: 10.1152/physiolgenomics.00158.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Bridget Coyle-Asbil
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada.,IMPART Investigator Team Canada, Saint John, New Brunswick, Canada
| | - Leslie M Ogilvie
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada.,IMPART Investigator Team Canada, Saint John, New Brunswick, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada.,IMPART Investigator Team Canada, Saint John, New Brunswick, Canada
| |
Collapse
|
9
|
Recent Advances in the Knowledge of the Mechanisms of Leptin Physiology and Actions in Neurological and Metabolic Pathologies. Int J Mol Sci 2023; 24:ijms24021422. [PMID: 36674935 PMCID: PMC9860943 DOI: 10.3390/ijms24021422] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
Excess body weight is frequently associated with low-grade inflammation. Evidence indicates a relationship between obesity and cancer, as well as with other diseases, such as diabetes and non-alcoholic fatty liver disease, in which inflammation and the actions of various adipokines play a role in the pathological mechanisms involved in these disorders. Leptin is mainly produced by adipose tissue in proportion to fat stores, but it is also synthesized in other organs, where leptin receptors are expressed. This hormone performs numerous actions in the brain, mainly related to the control of energy homeostasis. It is also involved in neurogenesis and neuroprotection, and central leptin resistance is related to some neurological disorders, e.g., Parkinson's and Alzheimer's diseases. In peripheral tissues, leptin is implicated in the regulation of metabolism, as well as of bone density and muscle mass. All these actions can be affected by changes in leptin levels and the mechanisms associated with resistance to this hormone. This review will present recent advances in the molecular mechanisms of leptin action and their underlying roles in pathological situations, which may be of interest for revealing new approaches for the treatment of diseases where the actions of this adipokine might be compromised.
Collapse
|
10
|
Gee LMV, Barron-Millar B, Leslie J, Richardson C, Zaki MYW, Luli S, Burgoyne RA, Cameron RIT, Smith GR, Brain JG, Innes B, Jopson L, Dyson JK, McKay KRC, Pechlivanis A, Holmes E, Berlinguer-Palmini R, Victorelli S, Mells GF, Sandford RN, Palmer J, Kirby JA, Kiourtis C, Mokochinski J, Hall Z, Bird TG, Borthwick LA, Morris CM, Hanson PS, Jurk D, Stoll EA, LeBeau FEN, Jones DEJ, Oakley F. Anti-Cholestatic Therapy with Obeticholic Acid Improves Short-Term Memory in Bile Duct-Ligated Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:11-26. [PMID: 36243043 DOI: 10.1016/j.ajpath.2022.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/03/2022] [Accepted: 09/28/2022] [Indexed: 12/12/2022]
Abstract
Patients with cholestatic liver disease, including those with primary biliary cholangitis, can experience symptoms of impaired cognition or brain fog. This phenomenon remains unexplained and is currently untreatable. Bile duct ligation (BDL) is an established rodent model of cholestasis. In addition to liver changes, BDL animals develop cognitive symptoms early in the disease process (before development of cirrhosis and/or liver failure). The cellular mechanisms underpinning these cognitive symptoms are poorly understood. Herein, the study explored the neurocognitive symptom manifestations, and tested potential therapies, in BDL mice, and used human neuronal cell cultures to explore translatability to humans. BDL animals exhibited short-term memory loss and showed reduced astrocyte coverage of the blood-brain barrier, destabilized hippocampal network activity, and neuronal senescence. Ursodeoxycholic acid (first-line therapy for most human cholestatic diseases) did not reverse symptomatic or mechanistic aspects. In contrast, obeticholic acid (OCA), a farnesoid X receptor agonist and second-line anti-cholestatic agent, normalized memory function, suppressed blood-brain barrier changes, prevented hippocampal network deficits, and reversed neuronal senescence. Co-culture of human neuronal cells with either BDL or human cholestatic patient serum induced cellular senescence and increased mitochondrial respiration, changes that were limited again by OCA. These findings provide new insights into the mechanism of cognitive symptoms in BDL animals, suggesting that OCA therapy or farnesoid X receptor agonism could be used to limit cholestasis-induced neuronal senescence.
Collapse
Affiliation(s)
- Lucy M V Gee
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ben Barron-Millar
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Claire Richardson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Marco Y W Zaki
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Biochemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Saimir Luli
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rachel A Burgoyne
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rainie I T Cameron
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Graham R Smith
- Bioinformatics Support Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John G Brain
- Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Barbara Innes
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Laura Jopson
- Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Jessica K Dyson
- Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Katherine R C McKay
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alexandros Pechlivanis
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Elaine Holmes
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | - Stella Victorelli
- Department of Physiology and Biomedical Engineering, Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - George F Mells
- Academic Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Richard N Sandford
- Academic Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Jeremy Palmer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John A Kirby
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Joao Mokochinski
- MRC London Institute of Medical Sciences, London, United Kingdom
| | - Zoe Hall
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Thomas G Bird
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom; MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Lee A Borthwick
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher M Morris
- Medical Toxicology Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Peter S Hanson
- Medical Toxicology Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Diana Jurk
- Department of Physiology and Biomedical Engineering, Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | | | - Fiona E N LeBeau
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David E J Jones
- Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
11
|
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
|
12
|
Deng C, Chen H, Meng Z, Meng S. Roles of traditional chinese medicine regulating neuroendocrinology on AD treatment. Front Endocrinol (Lausanne) 2022; 13:955618. [PMID: 36213283 PMCID: PMC9533021 DOI: 10.3389/fendo.2022.955618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/01/2022] [Indexed: 11/20/2022] Open
Abstract
The incidence of sporadic Alzheimer's disease (AD) is increasing in recent years. Studies have shown that in addition to some genetic abnormalities, the majority of AD patients has a history of long-term exposure to risk factors. Neuroendocrine related risk factors have been proved to be strongly associated with AD. Long-term hormone disorder can have a direct detrimental effect on the brain by producing an AD-like pathology and result in cognitive decline by impairing neuronal metabolism, plasticity and survival. Traditional Chinese Medicine(TCM) may regulate the complex process of endocrine disorders, and improve metabolic abnormalities, as well as the resulting neuroinflammation and oxidative damage through a variety of pathways. TCM has unique therapeutic advantages in treating early intervention of AD-related neuroendocrine disorders and preventing cognitive decline. This paper reviewed the relationship between neuroendocrine and AD as well as the related TCM treatment and its mechanism. The advantages of TCM intervention on endocrine disorders and some pending problems was also discussed, and new insights for TCM treatment of dementia in the future was provided.
Collapse
Affiliation(s)
- Chujun Deng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Huize Chen
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Zeyu Meng
- The Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shengxi Meng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
13
|
Gannon OJ, Robison LS, Salinero AE, Abi-Ghanem C, Mansour FM, Kelly RD, Tyagi A, Brawley RR, Ogg JD, Zuloaga KL. High-fat diet exacerbates cognitive decline in mouse models of Alzheimer's disease and mixed dementia in a sex-dependent manner. J Neuroinflammation 2022; 19:110. [PMID: 35568928 PMCID: PMC9107741 DOI: 10.1186/s12974-022-02466-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 04/21/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Approximately 70% of Alzheimer's disease (AD) patients have co-morbid vascular contributions to cognitive impairment and dementia (VCID); this highly prevalent overlap of dementia subtypes is known as mixed dementia (MxD). AD is more prevalent in women, while VCID is slightly more prevalent in men. Sex differences in risk factors may contribute to sex differences in dementia subtypes. Unlike metabolically healthy women, diabetic women are more likely to develop VCID than diabetic men. Prediabetes is 3× more prevalent than diabetes and is linked to earlier onset of dementia in women, but not men. How prediabetes influences underlying pathology and cognitive outcomes across different dementia subtypes is unknown. To fill this gap in knowledge, we investigated the impact of diet-induced prediabetes and biological sex on cognitive function and neuropathology in mouse models of AD and MxD. METHODS Male and female 3xTg-AD mice received a sham (AD model) or unilateral common carotid artery occlusion surgery to induce chronic cerebral hypoperfusion (MxD model). Mice were fed a control or high fat (HF; 60% fat) diet from 3 to 7 months of age. In both sexes, HF diet elicited a prediabetic phenotype (impaired glucose tolerance) and weight gain. RESULTS In females, but not males, metabolic consequences of a HF diet were more severe in AD or MxD mice compared to WT. In both sexes, HF-fed AD or MxD mice displayed deficits in spatial memory in the Morris water maze (MWM). In females, but not males, HF-fed AD and MxD mice also displayed impaired spatial learning in the MWM. In females, but not males, AD or MxD caused deficits in activities of daily living, regardless of diet. Astrogliosis was more severe in AD and MxD females compared to males. Further, AD/MxD females had more amyloid beta plaques and hippocampal levels of insoluble amyloid beta 40 and 42 than AD/MxD males. In females, but not males, more severe glucose intolerance (prediabetes) was correlated with increased hippocampal microgliosis. CONCLUSIONS High-fat diet had a wider array of metabolic, cognitive, and neuropathological consequences in AD and MxD females compared to males. These findings shed light on potential underlying mechanisms by which prediabetes may lead to earlier dementia onset in women.
Collapse
Affiliation(s)
- Olivia J. Gannon
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Lisa S. Robison
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA ,grid.261241.20000 0001 2168 8324Department of Psychology & Neuroscience, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL 33314 USA ,grid.264307.40000 0000 9688 1551Department of Psychology, Stetson University, 421 N Woodland Blvd, DeLand, FL 32723 USA
| | - Abigail E. Salinero
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Charly Abi-Ghanem
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Febronia M. Mansour
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Richard D. Kelly
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Alvira Tyagi
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Rebekah R. Brawley
- grid.264307.40000 0000 9688 1551Department of Psychology, Stetson University, 421 N Woodland Blvd, DeLand, FL 32723 USA
| | - Jordan D. Ogg
- grid.264307.40000 0000 9688 1551Department of Psychology, Stetson University, 421 N Woodland Blvd, DeLand, FL 32723 USA
| | - Kristen L. Zuloaga
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| |
Collapse
|
14
|
Rivera DS, Lindsay CB, Oliva CA, Bozinovic F, Inestrosa NC. A Multivariate Assessment of Age-Related Cognitive Impairment in Octodon degus. Front Integr Neurosci 2021; 15:719076. [PMID: 34526882 PMCID: PMC8437396 DOI: 10.3389/fnint.2021.719076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/23/2021] [Indexed: 01/27/2023] Open
Abstract
Aging is a progressive functional decline characterized by a gradual deterioration in physiological function and behavior. The most important age-related change in cognitive function is decline in cognitive performance (i.e., the processing or transformation of information to make decisions that includes speed of processing, working memory, and learning). The purpose of this study is to outline the changes in age-related cognitive performance (i.e., short-term recognition memory and long-term learning and memory) in long-lived Octodon degus. The strong similarity between degus and humans in social, metabolic, biochemical, and cognitive aspects makes it a unique animal model for exploring the mechanisms underlying the behavioral and cognitive deficits related to natural aging. In this study, we examined young adult female degus (12- and 24-months-old) and aged female degus (38-, 56-, and 75-months-old) that were exposed to a battery of cognitive-behavioral tests. Multivariate analyses of data from the Social Interaction test or Novel Object/Local Recognition (to measure short-term recognition memory), and the Barnes maze test (to measure long-term learning and memory) revealed a consistent pattern. Young animals formed a separate group of aged degus for both short- and long-term memories. The association between the first component of the principal component analysis (PCA) from short-term memory with the first component of the PCA from long-term memory showed a significant negative correlation. This suggests age-dependent differences in both memories, with the aged degus having higher values of long-term memory ability but poor short-term recognition memory, whereas in the young degus an opposite pattern was found. Approximately 5% of the young and 80% of the aged degus showed an impaired short-term recognition memory; whereas for long-term memory about 32% of the young degus and 57% of the aged degus showed decreased performance on the Barnes maze test. Throughout this study, we outlined age-dependent cognitive performance decline during natural aging in degus. Moreover, we also demonstrated that the use of a multivariate approach let us explore and visualize complex behavioral variables, and identified specific behavioral patterns that allowed us to make powerful conclusions that will facilitate further the study on the biology of aging. In addition, this study could help predict the onset of the aging process based on behavioral performance.
Collapse
Affiliation(s)
- Daniela S Rivera
- GEMA Center for Genomics, Ecology and Environment, Facultad de Estudios Interdisciplinarios, Universidad Mayor, Santiago, Chile
| | - Carolina B Lindsay
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina A Oliva
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Bozinovic
- Center for Applied Ecology and Sustainability (CAPES), Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
15
|
Yuan X, Chen R, Zhang Y, Lin X, Yang X. Sexual dimorphism of gut microbiota at different pubertal status. Microb Cell Fact 2020; 19:152. [PMID: 32723385 PMCID: PMC7390191 DOI: 10.1186/s12934-020-01412-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
Background Accumulating evidence infer that gut microbiome-host relations are key mediators or modulators driving the observed sexual dimorphism in some disease onset and progression. To date, the sex-differences of gut microbiota at different pubertal status have not been reported. Objective To determine the characteristics of gut microbiota of both genders at different pubertal status. Methods Gut microbiota was analyzed in 89 Chinese participants aged 5–15 years. Participants were divided into pre-puberty and puberty groups for both male and female. The composition of gut microbiota was investigated by 16S rRNA-based metagenomics. Ecological representations of microbial communities were computed. The prediction of metagenomic functional content from 16S rRNA gene surveys was conducted. Results There were 49 males (9.76 ± 2.15 years) and 40 females (9.74 ± 1.63 years); 21 males and 26 females were at puberty. At genus level, Alistipes, Megamonas, Oscillospira and Parabacteroides were more prevalent in girls than in boys (p < 0.05). There were no significantly differences of alpha-diversity between genders, which was independent of pubertal status. The beta-diversity was significantly different in pubertal subjects between genders. Using statistical analyses, we assigned genera Dorea, Megamonas, Bilophila, Parabacteroides and Phascolarctobacterium as microbial markers for pubertal subjects. The predicted metabolic profiles differ in both pubertal and pre-pubertal groups between genders. Conclusion This cross-sectional study revealed that sex differences in the gut microbiota composition and predicted metabolic profiles exist before puberty, which become more significant at puberty. The identification of novel puberty bacterial markers may disclose a potential effects of gender-related microbiota profiles on puberty onset.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Endocrinology, Fuzhou Children's Hospital of Fujian Medical University, NO. 145, 817 Middle Road, Fuzhou, 350005, China
| | - Ruimin Chen
- Department of Endocrinology, Fuzhou Children's Hospital of Fujian Medical University, NO. 145, 817 Middle Road, Fuzhou, 350005, China.
| | - Ying Zhang
- Department of Endocrinology, Fuzhou Children's Hospital of Fujian Medical University, NO. 145, 817 Middle Road, Fuzhou, 350005, China
| | - Xiangquan Lin
- Department of Endocrinology, Fuzhou Children's Hospital of Fujian Medical University, NO. 145, 817 Middle Road, Fuzhou, 350005, China
| | - Xiaohong Yang
- Department of Endocrinology, Fuzhou Children's Hospital of Fujian Medical University, NO. 145, 817 Middle Road, Fuzhou, 350005, China
| |
Collapse
|