1
|
Souder DC, McGregor ER, Clark JP, Rhoads TW, Porter TJ, Eliceiri KW, Moore DL, Puglielli L, Anderson RM. Neuron-specific isoform of PGC-1α regulates neuronal metabolism and brain aging. Nat Commun 2025; 16:2053. [PMID: 40021651 PMCID: PMC11871081 DOI: 10.1038/s41467-025-57363-y] [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: 05/14/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
The brain is a high-energy tissue, and although aging is associated with dysfunctional inflammatory and neuron-specific functional pathways, a direct connection to metabolism is not established. Here, we show that isoforms of mitochondrial regulator PGC-1α are driven from distinct brain cell-type specific promotors, repressed with aging, and integral in coordinating metabolism and growth signaling. Transcriptional and proteomic profiles of cortex from male adult, middle age, and advanced age mice reveal an aging metabolic signature linked to PGC-1α. In primary culture, a neuron-exclusive promoter produces the functionally dominant isoform of PGC-1α. Using growth repression as a challenge, we find that PGC-1α is regulated downstream of GSK3β independently across promoters. Broad cellular metabolic consequences of growth inhibition observed in vitro are mirrored in vivo, including activation of PGC-1α directed programs and suppression of aging pathways. These data place PGC-1α centrally in a growth and metabolism network directly relevant to brain aging.
Collapse
Affiliation(s)
- Dylan C Souder
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA
| | - Eric R McGregor
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA
| | - Josef P Clark
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA
| | - Timothy W Rhoads
- Department of Nutritional Sciences, University of Wisconsin Madison, Madison, WI, USA
| | - Tiaira J Porter
- Department of Neuroscience, University of Wisconsin Madison, Madison, WI, USA
| | - Kevin W Eliceiri
- Department of Medical Physics, University of Wisconsin Madison, Madison, WI, USA
| | - Darcie L Moore
- Department of Neuroscience, University of Wisconsin Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA
- GRECC William S, Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Rozalyn M Anderson
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA.
- GRECC William S, Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
2
|
Cencelli G, Pedini G, Ricci C, Rosina E, Cecchetti G, Gentile A, Aiello G, Pacini L, Garrone B, Ombrato R, Coletta I, Prati F, Milanese C, Bagni C. Early dysregulation of GSK3β impairs mitochondrial activity in Fragile X Syndrome. Neurobiol Dis 2024; 203:106726. [PMID: 39510449 DOI: 10.1016/j.nbd.2024.106726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/03/2024] [Accepted: 11/03/2024] [Indexed: 11/15/2024] Open
Abstract
The finely tuned regulation of mitochondria activity is essential for proper brain development. Fragile X Syndrome (FXS), the leading cause of inherited intellectual disability, is a neurodevelopmental disorder in which mitochondrial dysfunction has been increasingly implicated. This study investigates the role of Glycogen Synthase Kinase 3β (GSK3β) in FXS. Several studies have reported the dysregulation of GSK3β in FXS, and its role in mitochondrial function is also well established. However, the link between disrupted GSK3β activity and mitochondrial dysfunction in FXS remains unexplored. Utilizing Fmr1 knockout (KO) mice and human cell lines from individuals with FXS, we uncovered a developmental window where dysregulated GSK3β activity disrupts mitochondrial function. Notably, a partial inhibition of GSK3β activity in FXS fibroblasts from young individuals rescues the observed mitochondrial defects, suggesting that targeting GSK3β in the early stages may offer therapeutic benefits for this condition.
Collapse
Affiliation(s)
- Giulia Cencelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giorgia Pedini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carlotta Ricci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Eleonora Rosina
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giorgia Cecchetti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonietta Gentile
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giuseppe Aiello
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Laura Pacini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; Faculty of Medicine, UniCamillus, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | | | | | | | | | | | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
3
|
Ma X, Liu Y, Ding B, Lu S, Ni B, Chen Y, Yang L, Liu Y, Zhang Y, Wang Y, Yang Y, Liu X. Anthocyanins from blueberry ameliorated arsenic-induced memory impairment, oxidative stress, and mitochondrial-biosynthesis imbalance in rat hippocampal neurons. Cell Signal 2024; 119:111177. [PMID: 38621470 DOI: 10.1016/j.cellsig.2024.111177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/28/2024] [Accepted: 04/11/2024] [Indexed: 04/17/2024]
Abstract
In this study, blueberry anthocyanins extract (BAE) was used to investigate its protective effect on arsenic-induced rat hippocampal neurons damage. Arsenic exposure resulted in elevated levels of oxidative stress, decreased antioxidant capacity and increased apoptosis in rat hippocampal brain tissue and mitochondria. Immunohistochemical results showed that arsenic exposure also significantly decreased the expression of mitochondrial biosynthesis-related factors PGC-1α and TFAM. Treatment with BAE alleviated the decrease in antioxidant capacity, mitochondrial biogenesis related protein PGC-1α/NRF2/TFAM expression, and ATP production of arsenic induced hippocampal neurons in rats, and improved cognitive function in arsenic damaged rats. This study provides new insights into the detoxification effect of anthocyanins on the nervous system toxicity caused by metal exposure in the environment, indicating that anthocyanins may be a natural antioxidant against the nervous system toxicity caused by environmental metal exposure.
Collapse
Affiliation(s)
- Xinbo Ma
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Yang Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Bo Ding
- Nanning Center for Disease Control and Prevention, China
| | - Siqi Lu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Bangyao Ni
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Yuting Chen
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Liu Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Yanan Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Yuchen Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Yuxi Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Yanmei Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China.
| |
Collapse
|
4
|
Magdy O, Eshra M, Rashed L, Maher M, Hosny SA, ShamsEldeen AM. Amelioration of cisplatin-induced neurodegenerative changes in rats and restoration of mitochondrial biogenesis by 6-bromoindirubin-3'-oxime: The implication of the GSK-3β/PGC1-α axis. Tissue Cell 2024; 88:102393. [PMID: 38705086 DOI: 10.1016/j.tice.2024.102393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND The cognitive deficits observed after treatment with chemotherapeutic drugs are obvious clinical problems. For treating chemotherapy-induced cognitive deficits (CICD), the treatment modalities must target its underlying mechanisms. Specifically, cisplatin may activate glycogen synthase kinase-3β (GSK-3β), thereby enhancing neuronal apoptosis. 6-bromoindirubin-3'-oxime (6BIO) was not investigated previously in a model of CICD. Therefore, this investigation aimed to address the impacts of GSK3 inhibition on regulating cell signaling, which contributes to neurodegeneration and cognitive impairment. METHODS Thirty adult male Wistar rats were randomly allocated into control groups, while two experimental groups were exposed to repeated cisplatin injections (2 mg/kg intraperitoneally (ip), twice weekly, nine injections), termed chemobrain groups. The rats in the two experimental groups were equally divided into the chemobrain group (untreated) and the chemobrain-6BIO group (treated with 6BIO at a dose of 8.5 μg/kg ip every two days, started after the last dose of cisplatin and continued for two weeks). RESULTS Repeated exposure to cisplatin led to a marked decline in cognitive functions. GSK3 inhibition exerted neuroprotection by decreasing the expression of p-tau and amyloid β, thereby improving cognition. 6BIO, the GSK-3β inhibitor, restored mitochondrial biogenesis by augmenting the protein levels of PGC1-α and increasing the number of mitochondria in the cerebral cortex and hippocampus. CONCLUSION 6BIO provided neuroprotection and exhibited anti-apoptotic and anti-oxidative effects in a rat model of chemobrain.
Collapse
Affiliation(s)
- Ola Magdy
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Mohammed Eshra
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Laila Rashed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | - Muhammed Maher
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Sara Adel Hosny
- Department of Histology, Faculty of Medicine, Cairo University, Egypt
| | | |
Collapse
|
5
|
Iqbal T, Nakagawa T. The therapeutic perspective of NAD + precursors in age-related diseases. Biochem Biophys Res Commun 2024; 702:149590. [PMID: 38340651 DOI: 10.1016/j.bbrc.2024.149590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is the fundamental molecule that performs numerous biological reactions and is crucial for maintaining cellular homeostasis. Studies have found that NAD+ decreases with age in certain tissues, and age-related NAD+ depletion affects physiological functions and contributes to various aging-related diseases. Supplementation of NAD+ precursor significantly elevates NAD+ levels in murine tissues, effectively mitigates metabolic syndrome, enhances cardiovascular health, protects against neurodegeneration, and boosts muscular strength. Despite the versatile therapeutic functions of NAD+ in animal studies, the efficacy of NAD+ precursors in clinical studies have been limited compared with that in the pre-clinical study. Clinical studies have demonstrated that NAD+ precursor treatment efficiently increases NAD+ levels in various tissues, though their clinical proficiency is insufficient to ameliorate the diseases. However, the latest studies regarding NAD+ precursors and their metabolism highlight the significant role of gut microbiota. The studies found that orally administered NAD+ intermediates interact with the gut microbiome. These findings provide compelling evidence for future trials to further explore the involvement of gut microbiota in NAD+ metabolism. Also, the reduced form of NAD+ precursor shows their potential to raise NAD+, though preclinical studies have yet to discover their efficacy. This review sheds light on NAD+ therapeutic efficiency in preclinical and clinical studies and the effect of the gut microbiota on NAD+ metabolism.
Collapse
Affiliation(s)
- Tooba Iqbal
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan; Research Center for Pre-Disease Science, University of Toyama, Toyama, Japan.
| |
Collapse
|
6
|
Souder DC, McGregor ER, Rhoads TW, Clark JP, Porter TJ, Eliceiri K, Moore DL, Puglielli L, Anderson RM. Mitochondrial regulator PGC-1a in neuronal metabolism and brain aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.559526. [PMID: 37808866 PMCID: PMC10557769 DOI: 10.1101/2023.09.29.559526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
The brain is a high energy tissue, and the cell types of which it is comprised are distinct in function and in metabolic requirements. The transcriptional co-activator PGC-1a is a master regulator of mitochondrial function and is highly expressed in the brain; however, its cell-type specific role in regulating metabolism has not been well established. Here, we show that PGC-1a is responsive to aging and that expression of the neuron specific PGC-1a isoform allows for specialization in metabolic adaptation. Transcriptional profiles of the cortex from male mice show an impact of age on immune, inflammatory, and neuronal functional pathways and a highly integrated metabolic response that is associated with decreased expression of PGC-1a. Proteomic analysis confirms age-related changes in metabolism and further shows changes in ribosomal and RNA splicing pathways. We show that neurons express a specialized PGC-1a isoform that becomes active during differentiation from stem cells and is further induced during the maturation of isolated neurons. Neuronal but not astrocyte PGC-1a responds robustly to inhibition of the growth sensitive kinase GSK3b, where the brain specific promoter driven dominant isoform is repressed. The GSK3b inhibitor lithium broadly reprograms metabolism and growth signaling, including significantly lower expression of mitochondrial and ribosomal pathway genes and suppression of growth signaling, which are linked to changes in mitochondrial function and neuronal outgrowth. In vivo, lithium treatment significantly changes the expression of genes involved in cortical growth, endocrine, and circadian pathways. These data place the GSK3b/PGC-1a axis centrally in a growth and metabolism network that is directly relevant to brain aging.
Collapse
Affiliation(s)
- Dylan C Souder
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI
| | - Eric R McGregor
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI
| | - Timothy W Rhoads
- Department of Nutritional Sciences, University of Wisconsin Madison, Madison, WI
| | - Josef P Clark
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI
| | - Tiaira J Porter
- Department of Neuroscience, University of Wisconsin Madison, Madison, WI
| | - Kevin Eliceiri
- Department of Medical Physics, University of Wisconsin Madison, Madison, WI
| | - Darcie L Moore
- Department of Neuroscience, University of Wisconsin Madison, Madison, WI
| | - Luigi Puglielli
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI
- GRECC William S, Middleton Memorial Veterans Hospital, Madison, WI
| | - Rozalyn M Anderson
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI
- GRECC William S, Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
7
|
Andrés-Benito P, Íñigo-Marco I, Brullas M, Carmona M, del Rio JA, Fernández-Irigoyen J, Santamaría E, Povedano M, Ferrer I. Proteostatic modulation in brain aging without associated Alzheimer's disease-and age-related neuropathological changes. Aging (Albany NY) 2023; 15:3295-3330. [PMID: 37179123 PMCID: PMC10449282 DOI: 10.18632/aging.204698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023]
Abstract
AIMS (Phospho)proteomics of old-aged subjects without cognitive or behavioral symptoms, and without AD-neuropathological changes and lacking any other neurodegenerative alteration will increase understanding about the physiological state of human brain aging without associate neurological deficits and neuropathological lesions. METHODS (Phospho)proteomics using conventional label-free- and SWATH-MS (Sequential window acquisition of all theoretical fragment ion spectra mass spectrometry) has been assessed in the frontal cortex (FC) of individuals without NFTs, senile plaques (SPs) and age-related co-morbidities classified by age (years) in four groups; group 1 (young, 30-44); group 2 (middle-aged: MA, 45-52); group 3 (early-elderly, 64-70); and group 4 (late-elderly, 75-85). RESULTS Protein levels and deregulated protein phosphorylation linked to similar biological terms/functions, but involving different individual proteins, are found in FC with age. The modified expression occurs in cytoskeleton proteins, membranes, synapses, vesicles, myelin, membrane transport and ion channels, DNA and RNA metabolism, ubiquitin-proteasome-system (UPS), kinases and phosphatases, fatty acid metabolism, and mitochondria. Dysregulated phosphoproteins are associated with the cytoskeleton, including microfilaments, actin-binding proteins, intermediate filaments of neurons and glial cells, and microtubules; membrane proteins, synapses, and dense core vesicles; kinases and phosphatases; proteins linked to DNA and RNA; members of the UPS; GTPase regulation; inflammation; and lipid metabolism. Noteworthy, protein levels of large clusters of hierarchically-related protein expression levels are stable until 70. However, protein levels of components of cell membranes, vesicles and synapses, RNA modulation, and cellular structures (including tau and tubulin filaments) are markedly altered from the age of 75. Similarly, marked modifications occur in the larger phosphoprotein clusters involving cytoskeleton and neuronal structures, membrane stabilization, and kinase regulation in the late elderly. CONCLUSIONS Present findings may increase understanding of human brain proteostasis modifications in the elderly in the subpopulation of individuals not having AD neuropathological change and any other neurodegenerative change in any telencephalon region.
Collapse
Affiliation(s)
- Pol Andrés-Benito
- Neurologic Diseases and Neurogenetics Group - Bellvitge Institute for Biomedical Research (IDIBE LL), L’Hospitalet de Llobregat, Barcelona 08907, Spain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, L’Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Ignacio Íñigo-Marco
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), diSNA, Pamplona 31008, Spain
| | - Marta Brullas
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, L’Hospitalet de Llobregat, Barcelona 08907, Spain
- Neuropathology Group, Institute of Biomedical Research, IDIBELL, L’Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Margarita Carmona
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, L’Hospitalet de Llobregat, Barcelona 08907, Spain
- Neuropathology Group, Institute of Biomedical Research, IDIBELL, L’Hospitalet de Llobregat, Barcelona 08907, Spain
| | - José Antonio del Rio
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, L’Hospitalet de Llobregat, Barcelona 08907, Spain
- Molecular and Cellular Neurobiotechnology Group, Institute of Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology, Science Park Barcelona (PCB), Barcelona 08028, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08007, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), diSNA, Pamplona 31008, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), diSNA, Pamplona 31008, Spain
| | - Mónica Povedano
- Neurologic Diseases and Neurogenetics Group - Bellvitge Institute for Biomedical Research (IDIBE LL), L’Hospitalet de Llobregat, Barcelona 08907, Spain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, L’Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Isidro Ferrer
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, L’Hospitalet de Llobregat, Barcelona 08907, Spain
- Neuropathology Group, Institute of Biomedical Research, IDIBELL, L’Hospitalet de Llobregat, Barcelona 08907, Spain
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L’Hospitalet de Llobregat, Barcelona 08907, Spain
| |
Collapse
|
8
|
Wu Q, Yu M, Wang Z, Ai X, Liu Z, Zeng J, Li C, Yuan L, He J, Lin X, Wan W. Alternate-day fasting for the protection of cognitive impairment in c57BL/6J mice following whole-brain radiotherapy. Neurochem Int 2023; 162:105463. [PMID: 36513311 DOI: 10.1016/j.neuint.2022.105463] [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: 05/17/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
NLRP3 inflammasome activation is implicated in irradiation-induced cognitive dysfunction. Alternate-day fasting (ADF) has been demonstrated to improve neuroinflammation as a non-pharmacological intervention. However, the exact mechanism and the anti-inflammatory effect in irradiation-induced cognitive dysfunction still need further in-depth study. The present study examined the effects of eight-week ADF on the cognitive functions of mice as well as inflammasome-mediated hippocampal neuronal loss following irradiation in mouse models of irradiation-induced cognitive deficits using seven-week-old male C57BL/6J mice. The behavioral results of novel place recognition and object recognition tasks revealed that ADF ameliorated cognitive functions in irradiation-induced cognitive dysfunction mice. ADF inhibited the expression of components of the NLRP3 inflammasome (NLRP3, ASC, and Cl.caspase-1), the downstream inflammatory factor (IL-1β and IL-18), and apoptosis-related proteins (caspase-3) via western blotting. Furthermore, an increased number of neurons and activated astrocytes were observed in the hippocampus using immunohistochemistry and Sholl analysis, which was jointly confirmed by western blotting. According to our study, this is the first time we found that ADF improved cognitive dysfunction induced by irradiation, and the anti-inflammatory effect of ADF could be due to inhibition in NLRP3-mediated hippocampal neuronal loss by suppressing astrocyte activation.
Collapse
Affiliation(s)
- Qiong Wu
- Key Laboratory of Brain Science &Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, 571199, China
| | - Min Yu
- Department of Pharmacy, Chenzhou First People's Hospital, Chenzhou, 423001, China
| | - Zhen Wang
- Key Laboratory of Brain Science &Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, 571199, China; Department of Anatomy, Medical College of Hunan Vocational College of Environmental Biology, Hengyang, 421001, China
| | - Xiaohong Ai
- Department of Oncology and Radiotherapy, the First Affiliated Hospital of Nanhua University, Hengyang, 421001, China
| | - Zhenghai Liu
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang, 421001, China
| | - Jiayu Zeng
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang, 421001, China
| | - Cai Li
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang, 421001, China
| | - Lei Yuan
- Key Laboratory of Brain Science &Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, 571199, China
| | - Jie He
- Department of Pathology, Hainan Medical University, Haikou, 571199, China.
| | - Xinping Lin
- Yueyang Maternal and Child Health-care Hospital, Yueyang, 414021, China.
| | - Wei Wan
- Key Laboratory of Brain Science &Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
9
|
Habeos GI, Filippopoulou F, Habeos EE, Kalaitzopoulou E, Skipitari M, Papadea P, Lagoumintzis G, Niarchos A, Georgiou CD, Chartoumpekis DV. Maternal Calorie Restriction Induces a Transcriptional Cytoprotective Response in Embryonic Liver Partially Dependent on Nrf2. Antioxidants (Basel) 2022; 11:2274. [PMID: 36421460 PMCID: PMC9687455 DOI: 10.3390/antiox11112274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Calorie restriction is known to enhance Nrf2 signaling and longevity in adult mice, partially by reducing reactive oxygen species, but calorie restriction during pregnancy leads to intrauterine growth retardation. The latter is associated with fetal reprogramming leading to increased incidence of obesity, metabolic syndrome and diabetes in adult life. Transcription factor Nrf2 is a central regulator of the antioxidant response and its crosstalk with metabolic pathways is emerging. We hypothesized that the Nrf2 pathway is induced in embryos during calorie restriction in pregnant mothers. METHODS From gestational day 10 up to day 16, 50% of the necessary mouse diet was provided to Nrf2 heterozygous pregnant females with fathers being of the same genotype. Embryos were harvested at the end of gestational day 16 and fetal liver was used for qRT-PCR and assessment of oxidative stress (OS). RESULTS Intrauterine calorie restriction led to upregulation of mRNA expression of antioxidant genes (Nqo1, Gsta1, Gsta4) and of genes related to integrated stress response (Chac1, Ddit3) in WT embryos. The expression of a key gluconeogenic (G6pase) and two lipogenic genes (Acacb, Fasn) was repressed in calorie-restricted embryos. In Nrf2 knockout embryos, the induction of Nqo1 and Gsta1 genes was abrogated while that of Gsta4 was preserved, indicating an at least partially Nrf2-dependent induction of antioxidant genes after in utero calorie restriction. Measures of OS showed no difference (superoxide radical and malondialdehyde) or a small decrease (thiobarbituric reactive substances) in calorie-restricted WT embryos. CONCLUSIONS Calorie restriction during pregnancy elicits the transcriptional induction of cytoprotective/antioxidant genes in the fetal liver, which is at least partially Nrf2-dependent, with a physiological significance that warrants further investigation.
Collapse
Affiliation(s)
- George I. Habeos
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Fotini Filippopoulou
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Evagelia E. Habeos
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Electra Kalaitzopoulou
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Marianna Skipitari
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Polyxeni Papadea
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - George Lagoumintzis
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
- Department of Pharmacy, University of Patras, 26504 Patras, Greece
| | - Athanasios Niarchos
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Christos D. Georgiou
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Dionysios V. Chartoumpekis
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
10
|
Peluso A, Damgaard MV, Mori MAS, Treebak JT. Age-Dependent Decline of NAD +-Universal Truth or Confounded Consensus? Nutrients 2021; 14:nu14010101. [PMID: 35010977 PMCID: PMC8747183 DOI: 10.3390/nu14010101] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/16/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential molecule involved in various metabolic reactions, acting as an electron donor in the electron transport chain and as a co-factor for NAD+-dependent enzymes. In the early 2000s, reports that NAD+ declines with aging introduced the notion that NAD+ metabolism is globally and progressively impaired with age. Since then, NAD+ became an attractive target for potential pharmacological therapies aiming to increase NAD+ levels to promote vitality and protect against age-related diseases. This review summarizes and discusses a collection of studies that report the levels of NAD+ with aging in different species (i.e., yeast, C. elegans, rat, mouse, monkey, and human), to determine whether the notion that overall NAD+ levels decrease with aging stands true. We find that, despite systematic claims of overall changes in NAD+ levels with aging, the evidence to support such claims is very limited and often restricted to a single tissue or cell type. This is particularly true in humans, where the development of NAD+ levels during aging is still poorly characterized. There is a need for much larger, preferably longitudinal, studies to assess how NAD+ levels develop with aging in various tissues. This will strengthen our conclusions on NAD metabolism during aging and should provide a foundation for better pharmacological targeting of relevant tissues.
Collapse
Affiliation(s)
- Augusto Peluso
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark; (A.P.); (M.V.D.)
| | - Mads V. Damgaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark; (A.P.); (M.V.D.)
| | - Marcelo A. S. Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, São Paulo 13083-862, Brazil;
- Obesity and Comorbidities Research Center, University of Campinas, São Paulo 13083-862, Brazil
- Experimental Medicine Research Cluster, University of Campinas, São Paulo 13083-862, Brazil
| | - Jonas T. Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark; (A.P.); (M.V.D.)
- Correspondence: ; Tel.: +45-24805398
| |
Collapse
|
11
|
Souder DC, Dreischmeier IA, Smith AB, Wright S, Martin SA, Sagar MAK, Eliceiri KW, Salamat SM, Bendlin BB, Colman RJ, Beasley TM, Anderson RM. Rhesus monkeys as a translational model for late-onset Alzheimer's disease. Aging Cell 2021; 20:e13374. [PMID: 33951283 PMCID: PMC8208787 DOI: 10.1111/acel.13374] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/22/2021] [Accepted: 04/11/2021] [Indexed: 11/28/2022] Open
Abstract
Age is a major risk factor for late-onset Alzheimer's disease (AD) but seldom features in laboratory models of the disease. Furthermore, heterogeneity in size and density of AD plaques observed in individuals are not recapitulated in transgenic mouse models, presenting an incomplete picture. We show that the amyloid plaque microenvironment is not equivalent between rodent and primate species, and that differences in the impact of AD pathology on local metabolism and inflammation might explain established differences in neurodegeneration and functional decline. Using brain tissue from transgenic APP/PSEN1 mice, rhesus monkeys with age-related amyloid plaques, and human subjects with confirmed AD, we report altered energetics in the plaque microenvironment. Metabolic features included changes in mitochondrial distribution and enzymatic activity, and changes in redox cofactors NAD(P)H that were shared among species. A greater burden of lipofuscin was detected in the brains from monkeys and humans of advanced age compared to transgenic mice. Local inflammatory signatures indexed by astrogliosis and microglial activation were detected in each species; however, the inflamed zone was considerably larger for monkeys and humans. These data demonstrate the advantage of nonhuman primates in modeling the plaque microenvironment, and provide a new framework to investigate how AD pathology might contribute to functional loss.
Collapse
Affiliation(s)
- Dylan C. Souder
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
| | | | - Alex B. Smith
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
| | - Samantha Wright
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
| | - Stephen A. Martin
- Biology of Aging Laboratory Center for American Indian and Rural Health Equity Montana State University Bozeman MT USA
| | - Md Abdul Kader Sagar
- Department of Biomedical Engineering University of Wisconsin Madison Madison WI USA
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering University of Wisconsin Madison Madison WI USA
| | - Shahriar M. Salamat
- Department of Pathology Laboratory Medicine University of Wisconsin Madison Madison WI USA
- Neurological Surgery University of Wisconsin Madison Madison WI USA
| | | | - Ricki J. Colman
- Wisconsin National Primate Research Center University of Wisconsin Madison Madison WI USA
| | - T. Mark Beasley
- Department of Biostatistics University of Alabama Birmingham AL USA
- GRECC Birmingham/Atlanta Veterans Administration Hospital Birmingham AL USA
| | - Rozalyn M. Anderson
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
- GRECC William S. Middleton Memorial Veterans Hospital Madison WI USA
| |
Collapse
|
12
|
Orock A, Logan S, Deak F. Age-Related Cognitive Impairment: Role of Reduced Synaptobrevin-2 Levels in Deficits of Memory and Synaptic Plasticity. J Gerontol A Biol Sci Med Sci 2021; 75:1624-1632. [PMID: 30649208 DOI: 10.1093/gerona/glz013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Indexed: 01/02/2023] Open
Abstract
Cognitive impairment in the aging population is quickly becoming a health care priority, for which currently no disease-modifying treatment is available. Multiple domains of cognition decline with age even in the absence of neurodegenerative diseases. The cellular and molecular changes leading to cognitive decline with age remain elusive. Synaptobrevin-2 (Syb2), the major vesicular SNAP receptor protein, highly expressed in the cerebral cortex and hippocampus, is essential for synaptic transmission. We have analyzed Syb2 protein levels in mice and found a decrease with age. To investigate the functional consequences of lower Syb2 expression, we have used adult Syb2 heterozygous mice (Syb2+/-) with reduced Syb2 levels. This allowed us to mimic the age-related decrease of Syb2 in the brain in order to selectively test its effects on learning and memory. Our results show that Syb2+/- animals have impaired learning and memory skills and they perform worse with age in the radial arm water maze assay. Syb2+/- hippocampal neurons have reduced synaptic plasticity with reduced release probability and impaired long-term potentiation in the CA1 region. Syb2+/- neurons also have lower vesicular release rates when compared to WT controls. These results indicate that reduced Syb2 expression with age is sufficient to cause cognitive impairment.
Collapse
Affiliation(s)
- Albert Orock
- Oklahoma Center for Neuroscience, Reynolds Oklahoma Center on Aging, Oklahoma City.,Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, Oklahoma City.,Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Sreemathi Logan
- Oklahoma Center for Neuroscience, Reynolds Oklahoma Center on Aging, Oklahoma City.,Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, Oklahoma City.,Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Ferenc Deak
- Oklahoma Center for Neuroscience, Reynolds Oklahoma Center on Aging, Oklahoma City.,Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, Oklahoma City.,Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City.,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City.,Department of Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City
| |
Collapse
|
13
|
Mechanisms and Therapeutic Implications of GSK-3 in Treating Neurodegeneration. Cells 2021; 10:cells10020262. [PMID: 33572709 PMCID: PMC7911291 DOI: 10.3390/cells10020262] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders are spreading worldwide and are one of the greatest threats to public health. There is currently no adequate therapy for these disorders, and therefore there is an urgent need to accelerate the discovery and development of effective treatments. Although neurodegenerative disorders are broad ranging and highly complex, they may share overlapping mechanisms, and thus potentially manifest common targets for therapeutic interventions. Glycogen synthase kinase-3 (GSK-3) is now acknowledged to be a central player in regulating mood behavior, cognitive functions, and neuron viability. Indeed, many targets controlled by GSK-3 are critically involved in progressing neuron deterioration and disease pathogenesis. In this review, we focus on three pathways that represent prominent mechanisms linking GSK-3 with neurodegenerative disorders: cytoskeleton organization, the mammalian target of rapamycin (mTOR)/autophagy axis, and mitochondria. We also consider the challenges and opportunities in the development of GSK-3 inhibitors for treating neurodegeneration.
Collapse
|
14
|
Dias IR, Santos CDS, Magalhães CODE, de Oliveira LRS, Peixoto MFD, De Sousa RAL, Cassilhas RC. Does calorie restriction improve cognition? IBRO Rep 2020; 9:37-45. [PMID: 33336102 PMCID: PMC7733132 DOI: 10.1016/j.ibror.2020.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/29/2020] [Indexed: 01/14/2023] Open
Abstract
Calorie restriction (CR) has been considered the most effective non-pharmacological intervention to counteract aging-related diseases and improve longevity. This intervention has shown beneficial effects in the prevention and treatment of several chronic diseases and functional declines related to aging, such as Parkinson's, Alzheimer's, and neuroendocrine disorders. However, the effects of CR on cognition show controversial results since its effects vary according to intensity, duration, and the period of CR. This review focuses on the main studies published in the last ten years regarding the consequences of CR on cognition in different neurological diseases and conditions of experimental animals. Also, possible CR mimetics are discussed. These findings highlight the potential beneficial effects of CR of up to 40 % on cognition when started early in life in non human animals.
Collapse
Affiliation(s)
- Isabella Rocha Dias
- Neuroplasticity and Exercise Study Group (Grupo de Estudos em Neuroplasticidade e Exercício - GENE), UFVJM, Diamantina, MG, Brazil.,Multicenter Post Graduation Program in Physiological Sciences (PMPGCF), UFVJM, Brazilian Society of Physiology, Diamantina, MG, Brazil
| | - Carina de Sousa Santos
- Multicenter Post Graduation Program in Physiological Sciences (PMPGCF), UFVJM, Brazilian Society of Physiology, Diamantina, MG, Brazil
| | - Caíque Olegário Diniz E Magalhães
- Neuroplasticity and Exercise Study Group (Grupo de Estudos em Neuroplasticidade e Exercício - GENE), UFVJM, Diamantina, MG, Brazil.,Multicenter Post Graduation Program in Physiological Sciences (PMPGCF), UFVJM, Brazilian Society of Physiology, Diamantina, MG, Brazil
| | - Lucas Renan Sena de Oliveira
- Neuroplasticity and Exercise Study Group (Grupo de Estudos em Neuroplasticidade e Exercício - GENE), UFVJM, Diamantina, MG, Brazil.,Multicenter Post Graduation Program in Physiological Sciences (PMPGCF), UFVJM, Brazilian Society of Physiology, Diamantina, MG, Brazil
| | - Marco Fabrício Dias Peixoto
- Department of Physical Education, Federal University of the Valleys of Jequitinhonha and Mucuri (UFVJM), Diamantina, MG, Brazil.,Neuroplasticity and Exercise Study Group (Grupo de Estudos em Neuroplasticidade e Exercício - GENE), UFVJM, Diamantina, MG, Brazil.,Multicenter Post Graduation Program in Physiological Sciences (PMPGCF), UFVJM, Brazilian Society of Physiology, Diamantina, MG, Brazil.,Post Graduation Program in Health Science (PPGCS), UFVJM, Diamantina, MG, Brazil
| | - Ricardo Augusto Leoni De Sousa
- Neuroplasticity and Exercise Study Group (Grupo de Estudos em Neuroplasticidade e Exercício - GENE), UFVJM, Diamantina, MG, Brazil.,Multicenter Post Graduation Program in Physiological Sciences (PMPGCF), UFVJM, Brazilian Society of Physiology, Diamantina, MG, Brazil
| | - Ricardo Cardoso Cassilhas
- Department of Physical Education, Federal University of the Valleys of Jequitinhonha and Mucuri (UFVJM), Diamantina, MG, Brazil.,Neuroplasticity and Exercise Study Group (Grupo de Estudos em Neuroplasticidade e Exercício - GENE), UFVJM, Diamantina, MG, Brazil.,Multicenter Post Graduation Program in Physiological Sciences (PMPGCF), UFVJM, Brazilian Society of Physiology, Diamantina, MG, Brazil.,Post Graduation Program in Health Science (PPGCS), UFVJM, Diamantina, MG, Brazil
| |
Collapse
|
15
|
GSK3β Regulates Brain Energy Metabolism. Cell Rep 2019; 23:1922-1931.e4. [PMID: 29768193 PMCID: PMC6082412 DOI: 10.1016/j.celrep.2018.04.045] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 01/08/2018] [Accepted: 04/11/2018] [Indexed: 01/01/2023] Open
Abstract
GSK3β is a serine threonine kinase implicated in the progression of Alzheimer’s disease. Although the role of GSK3β in growth and pathology has been extensively studied, little is known about the metabolic consequences of GSK3β manipulation, particularly in the brain. Here, we show that GSK3β regulates mitochondrial energy metabolism in human H4 neuroglioma cells and rat PC12-derived neuronal cells and that inhibition of GSK3β in mice in vivo alters metabolism in the hippocampus in a region-specific manner. We demonstrate that GSK3β inhibition increases mitochondrial respiration and membrane potential and alters NAD(P)H metabolism. These metabolic effects are associated with increased PGC-1α protein stabilization, enhanced nuclear localization, and increased transcriptional co-activation. In mice treated with the GSK3β inhibitor lithium carbonate, changes in hippocampal energy metabolism are linked to increased PGC-1α. These data highlight a metabolic role for brain GSK3β and suggest that the GSK3β/PGC-1α axis may be important in neuronal metabolic integrity.
Collapse
|
16
|
Lin X, Chen Y, Zhang P, Chen G, Zhou Y, Yu X. The potential mechanism of postoperative cognitive dysfunction in older people. Exp Gerontol 2019; 130:110791. [PMID: 31765741 DOI: 10.1016/j.exger.2019.110791] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/18/2019] [Accepted: 11/21/2019] [Indexed: 02/08/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a common disorder following surgery, which seriously threatens the quality of patients' life, especially the older people. Accumulating attention has been paid to POCD worldwide in pace with the popularization of anesthesia/surgery. The development of medical humanities and rehabilitation medicine sets higher demands on accurate diagnosis and safe treatment system of POCD. Although the research on POCD is in full swing, underlying pathogenesis is still inconclusive due to these conflicting results and controversial evidence. Generally, POCD is closely related to neuropsychiatric diseases such as dementia, depression and Alzheimer's disease in molecular pathways. Researchers have come up with various hypotheses to reveal the mechanisms of POCD, including neuroinflammation, oxidative stress, autophagy disorder, impaired synaptic function, lacking neurotrophic support, etc. Recent work focused on molecular mechanism of POCD in older people has been thoroughly reviewed and summed up here, concerning the changes of peripheral circulation, pathological pathways of central nervous system (CNS), the microbiota-gut-brain axis and the related brain regions. Accordingly, this article provides a better perspective to understand the development situation of POCD in older people, which is conductive to uncover the pathological mechanism and exploit reasonable treatment strategy of POCD.
Collapse
Affiliation(s)
- Xianyi Lin
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Piao Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China.
| | - Youfa Zhou
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Xin Yu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
17
|
Foster TC. Senescent neurophysiology: Ca 2+ signaling from the membrane to the nucleus. Neurobiol Learn Mem 2019; 164:107064. [PMID: 31394200 DOI: 10.1016/j.nlm.2019.107064] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/29/2019] [Accepted: 08/03/2019] [Indexed: 12/16/2022]
Abstract
The current review provides a historical perspective on the evolution of hypothesized mechanisms for senescent neurophysiology, focused on the CA1 region of the hippocampus, and the relationship of senescent neurophysiology to impaired hippocampal-dependent memory. Senescent neurophysiology involves processes linked to calcium (Ca2+) signaling including an increase in the Ca2+-dependent afterhyperpolarization (AHP), decreasing pyramidal cell excitability, hyporesponsiveness of N-methyl-D-aspartate (NMDA) receptor function, and a shift in Ca2+-dependent synaptic plasticity. Dysregulation of intracellular Ca2+ and downstream signaling of kinase and phosphatase activity lies at the core of senescent neurophysiology. Ca2+-dysregulation involves a decrease in Ca2+ influx through NMDA receptors and an increase release of Ca2+ from internal Ca2+ stores. Recent work has identified changes in redox signaling, arising in middle-age, as an initiating factor for senescent neurophysiology. The shift in redox state links processes of aging, oxidative stress and inflammation, with functional changes in mechanisms required for episodic memory. The link between age-related changes in Ca2+ signaling, epigenetics and gene expression is an exciting area of research. Pharmacological and behavioral intervention, initiated in middle-age, can promote memory function by initiating transcription of neuroprotective genes and rejuvenating neurophysiology. However, with more advanced age, or under conditions of neurodegenerative disease, epigenetic changes may weaken the link between environmental influences and transcription, decreasing resilience of memory function.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience and Genetics and Genomics Program, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
18
|
An expanding GSK3 network: implications for aging research. GeroScience 2019; 41:369-382. [PMID: 31313216 DOI: 10.1007/s11357-019-00085-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/02/2019] [Indexed: 10/26/2022] Open
Abstract
The last few decades of longevity research have been very exciting. We now know that longevity and healthspan can be manipulated across species, from unicellular eukaryotes to nonhuman primates, and that while aging itself is inevitable, how we age is malleable. Numerous dietary, genetic, and pharmacological studies now point to links between metabolism and growth regulation as a central aspect in determining longevity and, perhaps more importantly, health with advancing age. Here, we focus on a relatively new player in aging studies GSK3, glycogen synthase kinase, a key factor in growth and metabolism whose name fails to convey the extensive breadth of its role in cellular adaptation. First, we provide a brief overview of GSK3, touching on those aspects that are likely relevant to aging. Then, we outline the role of GSK3 in cellular functions including growth signaling, cell fate, and metabolism. Next, we describe evidence demonstrating a direct role for GSK3 in a range of age-related diseases, despite the fact that they differ considerably in their etiology and pathology. Finally, we discuss the role that GSK3 may play in normative aging and how GSK3 might be a suitable target to oppose age-related disease vulnerability.
Collapse
|
19
|
Orefice NS, Souchet B, Braudeau J, Alves S, Piguet F, Collaud F, Ronzitti G, Tada S, Hantraye P, Mingozzi F, Ducongé F, Cartier N. Real-Time Monitoring of Exosome Enveloped-AAV Spreading by Endomicroscopy Approach: A New Tool for Gene Delivery in the Brain. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:237-251. [PMID: 31440523 PMCID: PMC6699252 DOI: 10.1016/j.omtm.2019.06.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 06/21/2019] [Indexed: 12/15/2022]
Abstract
Exosomes represent a strategy for optimizing the adeno-associated virus (AAV) toward the development of novel therapeutic options for neurodegenerative disorders. However, in vivo spreading of exosomes and AAVs after intracerebral administration is poorly understood. This study provides an assessment and comparison of the spreading into the brain of exosome-enveloped AAVs (exo-AAVs) or unassociated AAVs (std-AAVs) through in vivo optical imaging techniques like probe-based confocal laser endomicroscopy (pCLE) and ex vivo fluorescence microscopy. The std-AAV serotypes (AAV6 and AAV9) encoding the GFP were enveloped in exosomes and injected into the ipsilateral hippocampus. At 3 months post-injection, pCLE detected enhanced GFP expression of both exo-AAV serotypes in contralateral hemispheres compared to std-AAVs. Although sparse GFP-positive astrocytes were observed using exo-AAVs, our results show that the enhancement of the transgene expression resulting from exo-AAVs was largely restricted to neurons and oligodendrocytes. Our results suggest (1) the possibility of combining gene therapy with an endoscopic approach to enable tracking of exo-AAV spread, and (2) exo-AAVs allow for widespread, long-term gene expression in the CNS, supporting the use of exo-AAVs as an efficient gene delivery tool.
Collapse
Affiliation(s)
- Nicola Salvatore Orefice
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay 94100, France.,CEA, Fundamental Research Division (DRF), Institut of Biology Francois Jacob, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses 92265, France
| | - Benoît Souchet
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay 94100, France.,CEA, Fundamental Research Division (DRF), Institut of Biology Francois Jacob, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses 92265, France
| | - Jérôme Braudeau
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay 94100, France.,CEA, Fundamental Research Division (DRF), Institut of Biology Francois Jacob, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses 92265, France
| | - Sandro Alves
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay 94100, France.,CEA, Fundamental Research Division (DRF), Institut of Biology Francois Jacob, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses 92265, France
| | - Françoise Piguet
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay 94100, France.,CEA, Fundamental Research Division (DRF), Institut of Biology Francois Jacob, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses 92265, France
| | - Fanny Collaud
- INTEGRARE, Genethon, INSERM, Université Evry, Université Paris-Saclay, Evry 91002, France
| | - Giuseppe Ronzitti
- INTEGRARE, Genethon, INSERM, Université Evry, Université Paris-Saclay, Evry 91002, France
| | - Satoru Tada
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay 94100, France.,CEA, Fundamental Research Division (DRF), Institut of Biology Francois Jacob, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses 92265, France
| | - Philippe Hantraye
- CEA, Fundamental Research Division (DRF), Institut of Biology Francois Jacob, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses 92265, France.,Neurodegenerative Diseases Laboratory, CNRS Laboratory of Neurodegenerative Diseases (UMR9199), Fontenay-aux-Roses 92265, France
| | - Federico Mingozzi
- INTEGRARE, Genethon, INSERM, Université Evry, Université Paris-Saclay, Evry 91002, France
| | - Frédéric Ducongé
- CEA, Fundamental Research Division (DRF), Institut of Biology Francois Jacob, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses 92265, France.,Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, Fontenay-aux-Roses 92265, France
| | - Nathalie Cartier
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay 94100, France.,CEA, Fundamental Research Division (DRF), Institut of Biology Francois Jacob, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses 92265, France
| |
Collapse
|
20
|
Effects of methyl-beta-cyclodextrin on blood-brain barrier permeability in angiotensin II-induced hypertensive rats. Brain Res 2019; 1715:148-155. [DOI: 10.1016/j.brainres.2019.03.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/01/2019] [Accepted: 03/22/2019] [Indexed: 01/06/2023]
|
21
|
Wahl D, Solon-Biet SM, Cogger VC, Fontana L, Simpson SJ, Le Couteur DG, Ribeiro RV. Aging, lifestyle and dementia. Neurobiol Dis 2019; 130:104481. [PMID: 31136814 DOI: 10.1016/j.nbd.2019.104481] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Aging is the greatest risk factor for most diseases including cancer, cardiovascular disorders, and neurodegenerative disease. There is emerging evidence that interventions that improve metabolic health with aging may also be effective for brain health. The most robust interventions are non-pharmacological and include limiting calorie or protein intake, increasing aerobic exercise, or environmental enrichment. In humans, dietary patterns including the Mediterranean, Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) and Okinawan diets are associated with improved age-related health and may reduce neurodegenerative disease including dementia. Rapamycin, metformin and resveratrol act on nutrient sensing pathways that improve cardiometabolic health and decrease the risk for age-associated disease. There is some evidence that they may reduce the risk for dementia in rodents. There is a growing recognition that improving metabolic function may be an effective way to optimize brain health during aging.
Collapse
Affiliation(s)
- Devin Wahl
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia.
| | - Samantha M Solon-Biet
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia
| | - Luigi Fontana
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| | - David G Le Couteur
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia
| | - Rosilene V Ribeiro
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| |
Collapse
|
22
|
Dong Y, Digman MA, Brewer GJ. Age- and AD-related redox state of NADH in subcellular compartments by fluorescence lifetime imaging microscopy. GeroScience 2019; 41:51-67. [PMID: 30729413 DOI: 10.1007/s11357-019-00052-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/17/2019] [Indexed: 02/06/2023] Open
Abstract
Nicotinamide adenine dinucleotide (reduced form: NADH) serves as a vital redox-energy currency for reduction-oxidation homeostasis and fulfilling energetic demands. While NADH exists as free and bound forms, only free NADH is utilized for complex I to power oxidative phosphorylation, especially important in neurons. Here, we studied how much free NADH remains available for energy production in mitochondria of old living neurons. We hypothesize that free NADH in neurons from old mice is lower than the levels in young mice and even lower in neurons from the 3xTg-AD Alzheimer's disease (AD) mouse model. To assess free NADH, we used lifetime imaging of NADH autofluorescence with 2-photon excitation to be able to resolve the pool of NADH in mitochondria, cytoplasm, and nuclei. Primary neurons from old mice were characterized by a lower free/bound NADH ratio than young neurons from both non-transgenic (NTg) and more so in 3xTg-AD mice. Mitochondrial compartments maintained 26 to 41% more reducing NADH redox state than cytoplasm for each age, genotype, and sex. Aging diminished the mitochondrial free NADH concentration in NTg neurons by 43% and in 3xTg-AD by 50%. The lower free NADH with age suggests a decline in capacity to regenerate free NADH for energetic supply to power oxidative phosphorylation which further worsens in AD. Applying this non-invasive approach, we showed the most explicit measures yet of bioenergetic deficits in free NADH with aging at the subcellular level in live neurons from in-bred mice and an AD model.
Collapse
Affiliation(s)
- Yue Dong
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Michelle A Digman
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.,Laboratory of Fluorescence Dynamics, Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Gregory J Brewer
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA. .,MIND Institute, Center for Neurobiology of Learning and Memory, University of California, Irvine, CA, USA.
| |
Collapse
|
23
|
Metzler-Baddeley C, Mole JP, Sims R, Fasano F, Evans J, Jones DK, Aggleton JP, Baddeley RJ. Fornix white matter glia damage causes hippocampal gray matter damage during age-dependent limbic decline. Sci Rep 2019; 9:1060. [PMID: 30705365 PMCID: PMC6355929 DOI: 10.1038/s41598-018-37658-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
Aging leads to gray and white matter decline but their causation remains unclear. We explored two classes of models of age and dementia risk related brain changes. The first class of models emphasises the importance of gray matter: age and risk-related processes cause neurodegeneration and this causes damage in associated white matter tracts. The second class of models reverses the direction of causation: aging and risk factors cause white matter damage and this leads to gray matter damage. We compared these models with linear mediation analysis and quantitative MRI indices (from diffusion, quantitative magnetization transfer and relaxometry imaging) of tissue properties in two limbic structures implicated in age-related memory decline: the hippocampus and the fornix in 166 asymptomatic individuals (aged 38–71 years). Aging was associated with apparent glia but not neurite density damage in the fornix and the hippocampus. Mediation analysis supported white matter damage causing gray matter decline; controlling for fornix glia damage, the correlations between age and hippocampal damage disappear, but not vice versa. Fornix and hippocampal differences were both associated with reductions in episodic memory performance. These results suggest that fornix white matter glia damage may cause hippocampal gray matter damage during age-dependent limbic decline.
Collapse
Affiliation(s)
- Claudia Metzler-Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK.
| | - Jilu P Mole
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Rebecca Sims
- Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Fabrizio Fasano
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK.,Siemens Healthcare, Head Office, Sir William Siemens Square, Surrey, GU16 8QD, UK
| | - John Evans
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Derek K Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK.,School of Psychology, Faculty of Health Sciences, Australian Catholic University, Melbourne, Victoria, 3065, Australia
| | - John P Aggleton
- School of Psychology, Cardiff University, Tower Building, 70 Park Place, Cardiff, CF10 3AT, UK
| | - Roland J Baddeley
- Experimental Psychology, University of Bristol, 12a Priory Road, Bristol, BS8 1TU, UK
| |
Collapse
|
24
|
Booker SA, Campbell GR, Mysiak KS, Brophy PJ, Kind PC, Mahad DJ, Wyllie DJA. Loss of protohaem IX farnesyltransferase in mature dentate granule cells impairs short-term facilitation at mossy fibre to CA3 pyramidal cell synapses. J Physiol 2017; 595:2147-2160. [PMID: 28083896 PMCID: PMC5350446 DOI: 10.1113/jp273581] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/06/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Neurodegenerative disorders can exhibit dysfunctional mitochondrial respiratory chain complex IV activity. Conditional deletion of cytochrome c oxidase, the terminal enzyme in the respiratory electron transport chain of mitochondria, from hippocampal dentate granule cells in mice does not affect low-frequency dentate to CA3 glutamatergic synaptic transmission. High-frequency dentate to CA3 glutamatergic synaptic transmission and feedforward inhibition are significantly attenuated in cytochrome c oxidase-deficient mice. Intact presynaptic mitochondrial function is critical for the short-term dynamics of mossy fibre to CA3 synaptic function. ABSTRACT Neurodegenerative disorders are characterized by peripheral and central symptoms including cognitive impairments which have been associated with reduced mitochondrial function, in particular mitochondrial respiratory chain complex IV or cytochrome c oxidase activity. In the present study we conditionally removed a key component of complex IV, protohaem IX farnesyltransferase encoded by the COX10 gene, in granule cells of the adult dentate gyrus. Utilizing whole-cell patch-clamp recordings from morphologically identified CA3 pyramidal cells from control and complex IV-deficient mice, we found that reduced mitochondrial function did not result in overt deficits in basal glutamatergic synaptic transmission at the mossy-fibre synapse because the amplitude, input-output relationship and 50 ms paired-pulse facilitation were unchanged following COX10 removal from dentate granule cells. However, trains of stimuli given at high frequency (> 20 Hz) resulted in dramatic reductions in short-term facilitation and, at the highest frequencies (> 50 Hz), also reduced paired-pulse facilitation, suggesting a requirement for adequate mitochondrial function to maintain glutamate release during physiologically relevant activity patterns. Interestingly, local inhibition was reduced, suggesting the effect observed was not restricted to synapses with CA3 pyramidal cells via large mossy-fibre boutons, but rather to all synapses formed by dentate granule cells. Therefore, presynaptic mitochondrial function is critical for the short-term dynamics of synapse function, which may contribute to the cognitive deficits observed in pathological mitochondrial dysfunction.
Collapse
Affiliation(s)
- Sam A Booker
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Graham R Campbell
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Karolina S Mysiak
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Peter J Brophy
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Peter C Kind
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
| | - Don J Mahad
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - David J A Wyllie
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
| |
Collapse
|
25
|
Horowitz AM, Villeda SA. Therapeutic potential of systemic brain rejuvenation strategies for neurodegenerative disease. F1000Res 2017; 6:1291. [PMID: 28815019 PMCID: PMC5539850 DOI: 10.12688/f1000research.11437.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2017] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases are a devastating group of conditions that cause progressive loss of neuronal integrity, affecting cognitive and motor functioning in an ever-increasing number of older individuals. Attempts to slow neurodegenerative disease advancement have met with little success in the clinic; however, a new therapeutic approach may stem from classic interventions, such as caloric restriction, exercise, and parabiosis. For decades, researchers have reported that these systemic-level manipulations can promote major functional changes that extend organismal lifespan and healthspan. Only recently, however, have the functional effects of these interventions on the brain begun to be appreciated at a molecular and cellular level. The potential to counteract the effects of aging in the brain, in effect rejuvenating the aged brain, could offer broad therapeutic potential to combat dementia-related neurodegenerative disease in the elderly. In particular, results from heterochronic parabiosis and young plasma administration studies indicate that pro-aging and rejuvenating factors exist in the circulation that can independently promote or reverse age-related phenotypes. The recent demonstration that human umbilical cord blood similarly functions to rejuvenate the aged brain further advances this work to clinical translation. In this review, we focus on these blood-based rejuvenation strategies and their capacity to delay age-related molecular and functional decline in the aging brain. We discuss new findings that extend the beneficial effects of young blood to neurodegenerative disease models. Lastly, we explore the translational potential of blood-based interventions, highlighting current clinical trials aimed at addressing therapeutic applications for the treatment of dementia-related neurodegenerative disease in humans.
Collapse
Affiliation(s)
- Alana M. Horowitz
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, California, 94143, USA
- Department of Anatomy, University of California San Francisco, San Francisco, California, 94143, USA
| | - Saul A. Villeda
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, California, 94143, USA
- Department of Anatomy, University of California San Francisco, San Francisco, California, 94143, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California, 94143, USA
| |
Collapse
|
26
|
Colman RJ. Non-human primates as a model for aging. Biochim Biophys Acta Mol Basis Dis 2017; 1864:2733-2741. [PMID: 28729086 DOI: 10.1016/j.bbadis.2017.07.008] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/28/2017] [Accepted: 07/08/2017] [Indexed: 02/07/2023]
Abstract
There has been, and continues to be, a dramatic shift in the human population towards older ages necessitating biomedical research aimed at better understanding the basic biology of aging and age-related diseases and facilitating new and improved therapeutic options. As it is not practical to perform the breadth of this research in humans, animal models are necessary to recapitulate the complexity of the aging environment. The mouse model is most frequently chosen for these endeavors, however, they are frequently not the most appropriate model. Non-human primates, on the other hand, are more closely related to humans and recapitulate the human aging process and development of age-related diseases. Extensive aging research has been performed in the well-characterized rhesus macaque aging model. More recently, the common marmoset, a small non-human primate with a shorter lifespan, has been explored as a potential aging model. This model holds particular promise as an aging disease model in part due to the successful creation of transgenic marmosets. Limitations to the use of non-human primates in aging research exist but can be mitigated somewhat by the existence of available resources supported by the National Institutes of Health. This article is part of a Special Issue entitled: Animal models of aging - edited by "Houtkooper Riekelt".
Collapse
Affiliation(s)
- Ricki J Colman
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Wisconsin Institutes for Medical Research, 1111 Highland Avenue, Madison, WI 53705, USA; Wisconsin National Primate Research Center, University of Wisconsin, 1220 Capitol Court, Madison, WI 53715, USA.
| |
Collapse
|
27
|
Miller KN, Burhans MS, Clark JP, Howell PR, Polewski MA, DeMuth TM, Eliceiri KW, Lindstrom MJ, Ntambi JM, Anderson RM. Aging and caloric restriction impact adipose tissue, adiponectin, and circulating lipids. Aging Cell 2017; 16:497-507. [PMID: 28156058 PMCID: PMC5418198 DOI: 10.1111/acel.12575] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2016] [Indexed: 11/26/2022] Open
Abstract
Adipose tissue expansion has been associated with system-wide metabolic dysfunction and increased vulnerability to diabetes, cancer, and cardiovascular disease. A reduction in adiposity is a hallmark of caloric restriction (CR), an intervention that extends longevity and delays the onset of these same age-related conditions. Despite these parallels, the role of adipose tissue in coordinating the metabolism of aging is poorly defined. Here, we show that adipose tissue metabolism and secretory profiles change with age and are responsive to CR. We conducted a cross-sectional study of CR in adult, late-middle-aged, and advanced-aged mice. Adiposity and the relationship between adiposity and circulating levels of the adipose-derived peptide hormone adiponectin were age-sensitive. CR impacted adiposity but only levels of the high molecular weight isoform of adiponectin responded to CR. Activators of metabolism including PGC-1a, SIRT1, and NAMPT were differentially expressed with CR in adipose tissues. Although age had a significant impact on NAD metabolism, as detected by biochemical assay and multiphoton imaging, the impact of CR was subtle and related to differences in reliance on oxidative metabolism. The impact of age on circulating lipids was limited to composition of circulating phospholipids. In contrast, the impact of CR was detected in all lipid classes regardless of age, suggesting a profound difference in lipid metabolism. These data demonstrate that aspects of adipose tissue metabolism are life phase specific and that CR is associated with a distinct metabolic state, suggesting that adipose tissue signaling presents a suitable target for interventions to delay aging.
Collapse
Affiliation(s)
- Karl N. Miller
- Division of GeriatricsDepartment of MedicineSMPHMadisonWI53706USA
- Department of Nutritional SciencesUniversity of Wisconsin MadisonMadisonWI53706USA
| | - Maggie S. Burhans
- Division of GeriatricsDepartment of MedicineSMPHMadisonWI53706USA
- Present address: Fred Hutchinson Cancer CenterSeattleWAUSA
| | - Josef P. Clark
- Division of GeriatricsDepartment of MedicineSMPHMadisonWI53706USA
| | - Porsha R. Howell
- Division of GeriatricsDepartment of MedicineSMPHMadisonWI53706USA
| | | | - Tyler M. DeMuth
- Division of GeriatricsDepartment of MedicineSMPHMadisonWI53706USA
| | - Kevin W. Eliceiri
- Laboratory for Optical and Computational InstrumentationUniversity of Wisconsin MadisonMadisonWI53706USA
| | - Mary J. Lindstrom
- Department of Biostatistics and Medical InformaticsUniversity of WisconsinMadisonWI53705USA
| | - James M. Ntambi
- Department of Nutritional SciencesUniversity of Wisconsin MadisonMadisonWI53706USA
- Department of BiochemistryUniversity of WisconsinMadisonWI53706USA
| | - Rozalyn M. Anderson
- Division of GeriatricsDepartment of MedicineSMPHMadisonWI53706USA
- Laboratory for Optical and Computational InstrumentationUniversity of Wisconsin MadisonMadisonWI53706USA
- GRECCWilliam S. Middleton Memorial Veterans HospitalMadisonWI53705USA
| |
Collapse
|
28
|
Niraula A, Sheridan JF, Godbout JP. Microglia Priming with Aging and Stress. Neuropsychopharmacology 2017; 42:318-333. [PMID: 27604565 PMCID: PMC5143497 DOI: 10.1038/npp.2016.185] [Citation(s) in RCA: 283] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/22/2016] [Accepted: 08/25/2016] [Indexed: 01/08/2023]
Abstract
The population of aged individuals is increasing worldwide and this has significant health and socio-economic implications. Clinical and experimental studies on aging have discovered myriad changes in the brain, including reduced neurogenesis, increased synaptic aberrations, higher metabolic stress, and augmented inflammation. In rodent models of aging, these alterations are associated with cognitive decline, neurobehavioral deficits, and increased reactivity to immune challenges. In rodents, caloric restriction and young blood-induced revitalization reverses the behavioral effects of aging. The increased inflammation in the aged brain is attributed, in part, to the resident population of microglia. For example, microglia of the aged brain are marked by dystrophic morphology, elevated expression of inflammatory markers, and diminished expression of neuroprotective factors. Importantly, the heightened inflammatory profile of microglia in aging is associated with a 'sensitized' or 'primed' phenotype. Mounting evidence points to a causal link between the primed profile of the aged brain and vulnerability to secondary insults, including infections and psychological stress. Conversely, psychological stress may also induce aging-like sensitization of microglia and increase reactivity to secondary challenges. This review delves into the characteristics of neuroinflammatory signaling and microglial sensitization in aging, its implications in psychological stress, and interventions that reverse aging-associated deficits.
Collapse
Affiliation(s)
- Anzela Niraula
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - John F Sheridan
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Division of Biosciences, The Ohio State University, College of Dentistry, Columbus, OH, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, 231 IBMR Bld, 460 Medical Center Drive Columbus, OH 43210, USA, Tel: +614 293 3456, Fax: +614 366 2097, E-mail:
| |
Collapse
|
29
|
Gouw AM, Efe G, Barakat R, Preecha A, Mehdizadeh M, Garan SA, Brooks GA. Roles of estrogen receptor-alpha in mediating life span: the hypothalamic deregulation hypothesis. Physiol Genomics 2016; 49:88-95. [PMID: 28011880 DOI: 10.1152/physiolgenomics.00073.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In several species caloric restriction (CR) extends life span. In this paper we integrate data from studies on CR and other sources to articulate the hypothalamic deregulation hypothesis by which estrogen receptor-alpha (ER-α) signaling in the hypothalamus and limbic system affects life span under the stress of CR in mammals. ER-α is one of two principal estrogen-binding receptors differentially expressed in the amygdala, hippocampus, and several key hypothalamic nuclei: the arcuate nucleus (ARN), preoptic area (POA), ventromedial nucleus (VMN), antero ventral periventricular nucleus (AVPV), paraventricular nucleus (PVN), supraoptic nucleus (SON), and suprachiasmatic nucleus (SCN). Estradiol signaling via ER-α is essential in basal level functioning of reproductive cycle, sexually receptive behaviors, physiological stress responses, as well as sleep cycle, and other nonsexual behaviors. When an organism is placed under long-term CR, which introduces an external stress to this ER-α signaling, the reduction of ER-α expression is attenuated over time in the hypothalamus. This review paper seeks to characterize the downstream effects of ER-α in the hypothalamus and limbic system that affect normal endocrine functioning.
Collapse
Affiliation(s)
- Arvin M Gouw
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and.,Department of Integrative Biology, University of California at Berkeley, Berkeley, California
| | - Gizem Efe
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - Rita Barakat
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - Andrew Preecha
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - Morvarid Mehdizadeh
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - Steven A Garan
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - George A Brooks
- Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and .,Department of Integrative Biology, University of California at Berkeley, Berkeley, California
| |
Collapse
|