1
|
Fernandes AG, Poirier AC, Veilleux CC, Melin AD. Contributions and future potential of animal models for geroscience research on sensory systems. GeroScience 2025; 47:61-83. [PMID: 39312151 PMCID: PMC11872837 DOI: 10.1007/s11357-024-01327-5] [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: 04/15/2024] [Accepted: 08/23/2024] [Indexed: 03/04/2025] Open
Abstract
Sensory systems mediate our social interactions, food intake, livelihoods, and other essential daily functions. Age-related decline and disease in sensory systems pose a significant challenge to healthy aging. Research on sensory decline in humans is informative but can often be difficult, subject to sampling bias, and influenced by environmental variation. Study of animal models, including mice, rats, rabbits, pigs, cats, dogs, and non-human primates, plays a complementary role in biomedical research, offering advantages such as controlled conditions and shorter lifespans for longitudinal study. Various species offer different advantages and limitations but have provided key insights in geroscience research. Here we review research on age-related decline and disease in vision, hearing, olfaction, taste, and touch. For each sense, we provide an epidemiological overview of impairment in humans, describing the physiological processes and diseases for each sense. We then discuss contributions made by research on animal models and ideas for future research. We additionally highlight the need for integrative, multimodal research across the senses as well as across disciplines. Long-term studies spanning multiple generations, including on species with longer life spans, are also highly valuable. Overall, integrative studies of appropriate animal models have high translational potential for clinical applications, the development of novel diagnostics, therapies, and medical interventions and future research will continue to close gaps in these areas. Research on animal models to improve understanding of the biology of the aging senses and improve the healthspan and additional research on sensory systems hold special promise for new breakthroughs.
Collapse
Affiliation(s)
- Arthur G Fernandes
- Department of Anthropology and Archaeology, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 4N1, Canada.
| | - Alice C Poirier
- Department of Anthropology and Archaeology, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Carrie C Veilleux
- Department of Anthropology and Archaeology, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 4N1, Canada
- Department of Anatomy, Midwestern University, Glendale, AZ, USA
| | - Amanda D Melin
- Department of Anthropology and Archaeology, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 4N1, Canada.
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
2
|
Collinge CW, Razzoli M, Mansk R, McGonigle S, Lamming DW, Pacak CA, van der Pluijm I, Niedernhofer L, Bartolomucci A. The mouse Social Frailty Index (mSFI): a novel behavioral assessment for impaired social functioning in aging mice. GeroScience 2025; 47:85-107. [PMID: 38987495 PMCID: PMC11872866 DOI: 10.1007/s11357-024-01263-4] [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: 04/10/2024] [Accepted: 06/23/2024] [Indexed: 07/12/2024] Open
Abstract
Various approaches exist to quantify the aging process and estimate biological age on an individual level. Frailty indices based on an age-related accumulation of physical deficits have been developed for human use and translated into mouse models. However, declines observed in aging are not limited to physical functioning but also involve social capabilities. The concept of "social frailty" has been recently introduced into human literature, but no index of social frailty exists for laboratory mice yet. To fill this gap, we developed a mouse Social Frailty Index (mSFI) consisting of seven distinct assays designed to quantify social functioning which is relatively simple to execute and is minimally invasive. Application of the mSFI in group-housed male C57BL/6 mice demonstrated a progressively elevated levels of social frailty through the lifespan. Conversely, group-housed females C57BL/6 mice manifested social frailty only at a very old age. Female mice also showed significantly lower mSFI score from 10 months of age onward when compared to males. We also applied the mSFI in male C57BL/6 mice under chronic subordination stress and in chronic isolation, both of which induced larger increases in social frailty compared to age-matched group-housed males. Lastly, we show that the mSFI is enhanced in mouse models that show accelerated biological aging such as progeroid Ercc1-/Δ and Xpg-/- mice of both sexes compared to age matched littermate wild types. In summary, the mSFI represents a novel index to quantify trajectories of biological aging in mice and may help elucidate links between impaired social behavior and the aging process.
Collapse
Affiliation(s)
- Charles W Collinge
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Rachel Mansk
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Seth McGonigle
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Christina A Pacak
- Greg Marzolf Jr. Muscular Dystrophy Center & Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, and Department of Vascular Surgery, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Laura Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
3
|
Abu-Labdeh R, Omoluabi T, Yuan Q. Effects of Age and Atomoxetine on Olfactory Perception and Learning and Underlying Plasticity Mechanisms in Rats. Eur J Neurosci 2025; 61:e16649. [PMID: 39726209 DOI: 10.1111/ejn.16649] [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: 07/30/2024] [Revised: 11/20/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
The locus coeruleus (LC) plays a vital role in cognitive function through norepinephrine release. Impaired LC neuronal health and function is linked to cognitive decline during ageing and Alzheimer's disease. This study investigates age-related alterations in olfactory detection and discrimination learning, along with its reversal, in Long-Evans rats, and examines the effects of atomoxetine (ATM), a norepinephrine uptake inhibitor, on these processes. Adult (6-9 months) and aged (22-24 months) Long-Evans rats underwent odour detection threshold experiments with saline and two doses of ATM (0.3 and 1 mg/kg). Reward-based odour discrimination learning included simple, difficult and reversal learning tasks. LC neuron density, dopamine beta-hydroxylase and norepinephrine transporter expression in the piriform cortex (PC) and orbitofrontal cortex were measured. Reversal learning and olfactory threat extinction were used to measure behavioural flexibility. Immunohistochemistry and western blotting were used to analyse phosphorylated cAMP response element binding protein (pCREB) and cFos expression and ex vivo electrophysiology assessed long-term depression (LTD) in the PC. Whereas adult and aged cohorts showed similar odour detection and discrimination learning, fewer aged rats acquired reversal learning successfully. ATM improved reward-based odour discrimination in adults but hindered learning reversal. A delayed CREB phosphorylation in the posterior PC associated with atomoxetine administration possibly underlies learning enhancement. ATM resulted in less freezing behaviour in a threat conditioning and extinction paradigm at moderate, but not at higher doses. ATM administration ex vivo prevented PC LTD. These findings highlight the intricate effects of atomoxetine, influenced by target structures, and suggest potential interactions with other neurotransmitters. Our results contribute to understanding the impact of ageing and norepinephrine enhancers on cognitive processes.
Collapse
Affiliation(s)
- Ruhuf Abu-Labdeh
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Tamunotonye Omoluabi
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Qi Yuan
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
4
|
Jang MH, Song J. Adenosine and adenosine receptors in metabolic imbalance-related neurological issues. Biomed Pharmacother 2024; 177:116996. [PMID: 38897158 PMCID: PMC12021433 DOI: 10.1016/j.biopha.2024.116996] [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: 04/24/2024] [Revised: 06/08/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024] Open
Abstract
Metabolic syndromes (e.g., obesity) are characterized by insulin resistance, chronic inflammation, impaired glucose metabolism, and dyslipidemia. Recently, patients with metabolic syndromes have experienced not only metabolic problems but also neuropathological issues, including cognitive impairment. Several studies have reported blood-brain barrier (BBB) disruption and insulin resistance in the brain of patients with obesity and diabetes. Adenosine, a purine nucleoside, is known to regulate various cellular responses (e.g., the neuroinflammatory response) by binding with adenosine receptors in the central nervous system (CNS). Adenosine has four known receptors: A1R, A2AR, A2BR, and A3R. These receptors play distinct roles in various physiological and pathological processes in the brain, including endothelial cell homeostasis, insulin sensitivity, microglial activation, lipid metabolism, immune cell infiltration, and synaptic plasticity. Here, we review the recent findings on the role of adenosine receptor-mediated signaling in neuropathological issues related to metabolic imbalance. We highlight the importance of adenosine signaling in the development of therapeutic solutions for neuropathological issues in patients with metabolic syndromes.
Collapse
Affiliation(s)
- Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea.
| |
Collapse
|
5
|
Zhu Y, Hu CX, Liu X, Zhu RX, Wang BQ. Moderate coffee or tea consumption decreased the risk of cognitive disorders: an updated dose-response meta-analysis. Nutr Rev 2024; 82:738-748. [PMID: 37523229 DOI: 10.1093/nutrit/nuad089] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
CONTEXT Although several epidemiological studies have examined the association between coffee or tea intake and the risk of cognitive disorders, the results to date are inconsistent. OBJECTIVE An updated systematic review and dose-response meta-analysis was conducted to confirm the association between coffee, tea, and caffeine consumption and the risk of cognitive disorders. DATA SOURCES PubMed, Embase, and Web of Science were searched from inception to January 2022 for relevant studies, including dementia, Alzheimer disease (AD), and cognitive impairment or decline. DATA EXTRACTION Two reviewers independently performed data extraction and assessed the study quality. DATA ANALYSIS Restricted cubic splines were used to conduct the dose-response meta-analysis for coffee and tea intake. RESULTS Twenty-two prospective studies and 11 case-control studies involving 389 505 participants were eligible for this meta-analysis. Coffee and tea consumption was linked to a lower risk of cognitive disorders, with an overall relative risk (RR) of 0.73 (95% CI: 0.60-0.86) and 0.68 (95% CI: 0.56-0.80), respectively. The subgroup analysis revealed that ethnicity, sex, and outcomes had significant effects on this association. Protection was stronger for men than that for women in both coffee and tea consumption. A nonlinear relationship was found between coffee consumption and AD risk, and the strength of protection peaked at approximately 2.5 cups/day (RR: 0.74; 95% CI: 0.59-0.93). A linear relationship was found between tea consumption and cognitive disorders, and the risk decreased by 11% for every 1-cup/day increment. CONCLUSION This meta-analysis demonstrated that the consumption of 2.5 cups coffee/day minimizes the risk of AD, and 1 cup/day of tea intake leads to an 11% reduction in cognitive deficits. Effective interventions involving coffee and tea intake might prevent the occurrence of dementia.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Neurology, The First Hospital, China Medical University, Shenyang, China
| | - Chun-Xiang Hu
- Department of Neurology, The First Hospital, China Medical University, Shenyang, China
| | - Xu Liu
- Department of Neurology, The First Hospital, China Medical University, Shenyang, China
| | - Rui-Xia Zhu
- Department of Neurology, The First Hospital, China Medical University, Shenyang, China
| | - Ben-Qiao Wang
- Department of Neurology, The First Hospital, China Medical University, Shenyang, China
| |
Collapse
|
6
|
Cum M, Santiago Pérez JA, Wangia E, Lopez N, Wright ES, Iwata RL, Li A, Chambers AR, Padilla-Coreano N. A systematic review and meta-analysis of how social memory is studied. Sci Rep 2024; 14:2221. [PMID: 38278973 PMCID: PMC10817899 DOI: 10.1038/s41598-024-52277-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Social recognition is crucial for survival in social species, and necessary for group living, selective reproduction, pair bonding, and dominance hierarchies. Mice and rats are the most commonly used animal models in social memory research, however current paradigms do not account for the complex social dynamics they exhibit in the wild. To assess the range of social memories being studied, we conducted a systematic analysis of neuroscience articles testing the social memory of mice and rats published within the past two decades and analyzed their methods. Our results show that despite these rodent's rich social memory capabilities, the majority of social recognition papers explore short-term memories and short-term familiarity levels with minimal exposure between subject and familiar stimuli-a narrow type of social memory. We have identified several key areas currently understudied or underrepresented: kin relationships, mates, social ranks, sex variabilities, and the effects of aging. Additionally, reporting on social stimulus variables such as housing history, strain, and age, is limited, which may impede reproducibility. Overall, our data highlight large gaps in the diversity of social memories studied and the effects social variables have on social memory mechanisms.
Collapse
Affiliation(s)
- Meghan Cum
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | | | - Erika Wangia
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Naeliz Lopez
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Elizabeth S Wright
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Ryo L Iwata
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Albert Li
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Amelia R Chambers
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | | |
Collapse
|
7
|
Cum M, Pérez JS, Wangia E, Lopez N, Wright ES, Iwata RL, Li A, Chambers AR, Padilla-Coreano N. Mind the gap: A systematic review and meta-analysis of how social memory is studied. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572606. [PMID: 38187659 PMCID: PMC10769336 DOI: 10.1101/2023.12.20.572606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Social recognition is crucial for survival in social species, and necessary for group living, selective reproduction, pair bonding, and dominance hierarchies. Mice and rats are the most commonly used animal models in social memory research, however current paradigms do not account for the complex social dynamics they exhibit in the wild. To assess the range of social memories being studied, we conducted a systematic analysis of neuroscience articles testing the social memory of mice and rats published within the past two decades and analyzed their methods. Our results show that despite these rodent's rich social memory capabilities, the majority of social recognition papers explore short-term memories and short-term familiarity levels with minimal exposure between subject and familiar stimuli - a narrow type of social memory. We have identified several key areas currently understudied or underrepresented: kin relationships, mates, social ranks, sex variabilities, and the effects of aging. Additionally, reporting on social stimulus variables such as housing history, strain, and age, is limited, which may impede reproducibility. Overall, our data highlight large gaps in the diversity of social memories studied and the effects social variables have on social memory mechanisms.
Collapse
|
8
|
Ferroni NM, Chertoff MJ, Alberca CD, Berardino BG, Gianatiempo O, Brahamian M, Levi V, Urrutia L, Falasco G, Cánepa ET, Sonzogni SV. Oxidative stress associated with spatial memory impairment and social olfactory deterioration in female mice reveals premature aging aroused by perinatal protein malnutrition. Exp Neurol 2023; 368:114481. [PMID: 37463612 DOI: 10.1016/j.expneurol.2023.114481] [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: 02/21/2023] [Revised: 06/08/2023] [Accepted: 07/12/2023] [Indexed: 07/20/2023]
Abstract
Early-life adversity, like perinatal protein malnutrition, increases the vulnerability to develop long-term alterations in brain structures and function. This study aimed to determine whether perinatal protein malnutrition predisposes to premature aging in a murine model and to assess the cellular and molecular mechanisms involved. To this end, mouse dams were fed either with a normal (NP, casein 20%) or a low-protein diet (LP, casein 8%) during gestation and lactation. Female offspring were evaluated at 2, 7 and 12 months of age. Positron emission tomography analysis showed alterations in the hippocampal CA3 region and the accessory olfactory bulb of LP mice during aging. Protein malnutrition impaired spatial memory, coinciding with higher levels of reactive oxygen species in the hippocampus and sirt7 upregulation. Protein malnutrition also led to higher senescence-associated β-galactosidase activity and p21 expression. LP-12-month-old mice showed a higher number of newborn neurons that did not complete the maturation process. The social-odor discrimination in LP mice was impaired along life. In the olfactory bulb of LP mice, the senescence marker p21 was upregulated, coinciding with a downregulation of Sirt2 and Sirt7. Also, LP-12-month-old mice showed a downregulation of catalase and glutathione peroxidase, and LP-2-month-old mice showed a higher number of newborn neurons in the subventricular zone, which then returned to normal values. Our results show that perinatal protein malnutrition causes long-term impairment in cognitive and olfactory skills through an accelerated senescence phenotype accompanied by an increase in oxidative stress and altered sirtuin expression in the hippocampus and olfactory bulb.
Collapse
Affiliation(s)
- Nadina M Ferroni
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, C1428EGA Ciudad de Buenos Aires, Argentina
| | - Mariela J Chertoff
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, C1428EGA Ciudad de Buenos Aires, Argentina
| | - Carolina D Alberca
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, C1428EGA Ciudad de Buenos Aires, Argentina
| | - Bruno G Berardino
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, C1428EGA Ciudad de Buenos Aires, Argentina
| | - Octavio Gianatiempo
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, C1428EGA Ciudad de Buenos Aires, Argentina
| | - Martin Brahamian
- Bioterio central, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, C1428EGA Ciudad de Buenos Aires, Argentina
| | - Valeria Levi
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, C1428EGA Ciudad de Buenos Aires, Argentina
| | - Leandro Urrutia
- Centro de Imágenes Moleculares, Fleni, Belén de Escobar, B1625 Buenos Aires, Argentina
| | - Germán Falasco
- Centro de Imágenes Moleculares, Fleni, Belén de Escobar, B1625 Buenos Aires, Argentina
| | - Eduardo T Cánepa
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, C1428EGA Ciudad de Buenos Aires, Argentina
| | - Silvina V Sonzogni
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, C1428EGA Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
9
|
Cai C, Luo Q, Jia L, Xia Y, Lan X, Wei X, Shi S, Liu Y, Wang Y, Xiong Z, Shi R, Huang C, Chen Z. TRIM67 Implicates in Regulating the Homeostasis and Synaptic Development of Mitral Cells in the Olfactory Bulb. Int J Mol Sci 2023; 24:13439. [PMID: 37686246 PMCID: PMC10487898 DOI: 10.3390/ijms241713439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
In recent years, olfactory dysfunction has attracted increasingly more attention as a hallmark symptom of neurodegenerative diseases (ND). Deeply understanding the molecular basis underlying the development of the olfactory bulb (OB) will provide important insights for ND studies and treatments. Now, with a genetic knockout mouse model, we show that TRIM67, a new member of the tripartite motif (TRIM) protein family, plays an important role in regulating the proliferation and development of mitral cells in the OB. TRIM67 is abundantly expressed in the mitral cell layer of the OB. The genetic deletion of TRIM67 in mice leads to excessive proliferation of mitral cells in the OB and defects in its synaptic development, resulting in reduced olfactory function in mice. Finally, we show that TRIM67 may achieve its effect on mitral cells by regulating the Semaphorin 7A/Plexin C1 (Sema7A/PlxnC1) signaling pathway.
Collapse
Affiliation(s)
- Chunyu Cai
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
| | - Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu 611130, China
| | - Lanlan Jia
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu 611130, China
| | - Yu Xia
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
| | - Xinting Lan
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
| | - Xiaoli Wei
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
| | - Shuai Shi
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
| | - Yucong Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
| | - Yao Wang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
| | - Zongliang Xiong
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
| | - Riyi Shi
- Center for Paralysis Research, Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA;
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu 611130, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (C.C.); (Q.L.); (L.J.); (Y.X.); (X.L.); (X.W.); (S.S.); (Y.L.); (Y.W.); (Z.X.)
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu 611130, China
| |
Collapse
|
10
|
Cen XQ, Li P, Wang B, Chen X, Zhao Y, Yang N, Peng Y, Li CH, Ning YL, Zhou YG. Knockdown of adenosine A2A receptors in hippocampal neurons prevents post-TBI fear memory retrieval. Exp Neurol 2023; 364:114378. [PMID: 36907351 DOI: 10.1016/j.expneurol.2023.114378] [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: 12/08/2022] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
The formation of fear memory is crucial in emotional disorders such as PTSD and anxiety. Traumatic brain injury (TBI) can cause emotional disorders with dysregulated fear memory formation; however, their cross-interaction remains unclear and hurdled the treatment against TBI-related emotional disorders. While adenosine A2A receptor(A2AR) contributes to the physiological regulation of fear memory, this study aimed to evaluate the A2AR role and possible mechanisms in post-TBI fear memory formation using a craniocerebral trauma model, genetically modified A2AR mutant mice, and pharmacological A2AR agonist CGS21680 and antagonist ZM241385. Our finding showed (i) TBI enhanced mice freezing levels (fear memory) at seven days post-TBI; (ii) The A2AR agonist CGS21680 enhanced the post-TBI freezing levels; conversely, the A2AR antagonist ZM241385 reduced mice freezing level; further (iii) Genetic knockdown of neuronal A2AR in the hippocampal CA1, CA3, and DG regions reduced post-TBI freezing levels, while A2AR knockout in DG region yielded the most reduction in fear memory; finally, (iv) AAV-CaMKII-Cre virus-mediated DG deletion of A2AR on excitatory neurons led to a significant decreased freezing levels post-TBI. These findings indicate that brain trauma increases fear memory retrieval post-TBI, and A2AR on DG excitatory neurons plays a crucial role in this process. Importantly, inhibition of A2AR attenuates fear memory enhancement, which provides a new strategy to prevent fear memory formation/enhancement after TBI.
Collapse
Affiliation(s)
- Xiao-Qing Cen
- The College of Bioengineering, Chongqing University, No.174 Shazheng Street, Shapingba District, Chongqing 400044, China; Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Ping Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China; Institute of Brain and Intelligence, Army Medical University, Chongqing, China
| | - Bo Wang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Xing Chen
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Zhao
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Nan Yang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Peng
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Chang-Hong Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Ya-Lei Ning
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China; Institute of Brain and Intelligence, Army Medical University, Chongqing, China.
| | - Yuan-Guo Zhou
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China; Institute of Brain and Intelligence, Army Medical University, Chongqing, China.
| |
Collapse
|
11
|
Madeira D, Lopes CR, Simões AP, Canas PM, Cunha RA, Agostinho P. Astrocytic A 2A receptors silencing negatively impacts hippocampal synaptic plasticity and memory of adult mice. Glia 2023. [PMID: 37183905 DOI: 10.1002/glia.24384] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/16/2023]
Abstract
Astrocytes are wired to bidirectionally communicate with neurons namely with synapses, thus shaping synaptic plasticity, which in the hippocampus is considered to underlie learning and memory. Adenosine A2A receptors (A2A R) are a potential candidate to modulate this bidirectional communication, since A2A R regulate synaptic plasticity and memory and also control key astrocytic functions. Nonetheless, little is known about the role of astrocytic A2A R in synaptic plasticity and hippocampal-dependent memory. Here, we investigated the impact of genetic silencing astrocytic A2A R on hippocampal synaptic plasticity and memory of adult mice. The genetic A2A R silencing in astrocytes was accomplished by a bilateral injection into the CA1 hippocampal area of a viral construct (AAV5-GFAP-GFP-Cre) that inactivate A2A R expression in astrocytes of male adult mice carrying "floxed" A2A R gene, as confirmed by A2A R binding assays. Astrocytic A2A R silencing alters astrocytic morphology, typified by an increment of astrocytic arbor complexity, and led to deficits in spatial reference memory and compromised hippocampal synaptic plasticity, typified by a reduction of LTP magnitude and a shift of synaptic long-term depression (LTD) toward LTP. These data indicate that astrocytic A2A R control astrocytic morphology and influence hippocampal synaptic plasticity and memory of adult mice in a manner different from neuronal A2A R.
Collapse
Affiliation(s)
- Daniela Madeira
- Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Center for Neuroscience and Cell Biology- University of Coimbra (CNC- UC), Coimbra, Portugal
| | - Cátia R Lopes
- Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Center for Neuroscience and Cell Biology- University of Coimbra (CNC- UC), Coimbra, Portugal
| | - Ana P Simões
- Center for Neuroscience and Cell Biology- University of Coimbra (CNC- UC), Coimbra, Portugal
| | - Paula M Canas
- Center for Neuroscience and Cell Biology- University of Coimbra (CNC- UC), Coimbra, Portugal
| | - Rodrigo A Cunha
- Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Center for Neuroscience and Cell Biology- University of Coimbra (CNC- UC), Coimbra, Portugal
| | - Paula Agostinho
- Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Center for Neuroscience and Cell Biology- University of Coimbra (CNC- UC), Coimbra, Portugal
| |
Collapse
|
12
|
Korkutata M, Lazarus M. Adenosine A 2A receptors and sleep. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 170:155-178. [PMID: 37741690 DOI: 10.1016/bs.irn.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Adenosine, a known endogenous somnogen, induces sleep via A1 and A2A receptors. In this chapter, we review the current knowledge regarding the role of the adenosine A2A receptor and its agonists, antagonists, and allosteric modulators in sleep-wake regulation. Although many adenosine A2A receptor agonists, antagonists, and allosteric modulators have been identified, only a few have been tested to see if they can promote sleep or wakefulness. In addition, the growing popularity of natural sleep aids has led to an investigation of natural compounds that may improve sleep by activating the adenosine A2A receptor. Finally, we discuss the potential therapeutic advantage of allosteric modulators of adenosine A2A receptors over classic agonists and antagonists for treating sleep and neurologic disorders.
Collapse
Affiliation(s)
- Mustafa Korkutata
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS) and Institute of Medicine, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
13
|
Chen JF, Choi DS, Cunha RA. Striatopallidal adenosine A 2A receptor modulation of goal-directed behavior: Homeostatic control with cognitive flexibility. Neuropharmacology 2023; 226:109421. [PMID: 36634866 PMCID: PMC10132052 DOI: 10.1016/j.neuropharm.2023.109421] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/30/2022] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
Dysfunction of goal-directed behaviors under stressful or pathological conditions results in impaired decision-making and loss of flexibility of thoughts and behaviors, which underlie behavioral deficits ranging from depression, obsessive-compulsive disorders and drug addiction. Tackling the neuromodulators fine-tuning this core behavioral element may facilitate the development of effective strategies to control these deficits present in multiple psychiatric disorders. The current investigation of goal-directed behaviors has concentrated on dopamine and glutamate signaling in the corticostriatal pathway. In accordance with the beneficial effects of caffeine intake on mood and cognitive dysfunction, we now propose that caffeine's main site of action - adenosine A2A receptors (A2AR) - represent a novel target to homeostatically control goal-directed behavior and cognitive flexibility. A2AR are abundantly expressed in striatopallidal neurons and colocalize and interact with dopamine D2, NMDA and metabotropic glutamate 5 receptors to integrate dopamine and glutamate signaling. Specifically, striatopallidal A2AR (i) exert an overall "break" control of a variety of cognitive processes, making A2AR antagonists a novel strategy for improving goal-directed behavior; (ii) confer homeostatic control of goal-directed behavior by acting at multiple sites with often opposite effects, to enhance cognitive flexibility; (iii) integrate dopamine and adenosine signaling through multimeric A2AR-D2R heterocomplexes allowing a temporally precise fine-tuning in response to local signaling changes. As the U.S. Food and Drug Administration recently approved the A2AR antagonist Nourianz® (istradefylline) to treat Parkinson's disease, striatal A2AR-mediated control of goal-directed behavior may offer a new and real opportunity for improving deficits of goal-directed behavior and enhance cognitive flexibility under various neuropsychiatric conditions. This article is part of the Special Issue on "Purinergic Signaling: 50 years".
Collapse
Affiliation(s)
- Jiang-Fan Chen
- Molecular Neuropharmacology Laboratory, Wenzhou Medical University, Wenzhou, China; Department of Neurology, School of Medicine, Boston University, Boston, MA, USA.
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; FMUC-Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
14
|
Hoerner F, Lawrenz A, Oerke AK, Müller DWH, Azogu-Sepe I, Roller M, Damerau K, Preisfeld A. Long-Term Olfactory Memory in African Elephants. Animals (Basel) 2023; 13:ani13040679. [PMID: 36830466 PMCID: PMC9951961 DOI: 10.3390/ani13040679] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
African elephants are capable of discriminating scents up to a single changed molecule and show the largest reported repertoire of olfactory receptor genes. Olfaction plays an important role in family bonding. However, to the best of our knowledge, no empirical data exist on their ability to remember familiar scents long-term. In an ethological experiment, two mother-daughter pairs were presented with feces of absent kin, absent non-kin, and present non-kin. Video recordings showed reactions of elephants recognizing kin after long-term separation but only minor reactions to non-kin. Results give the empirical implication that elephants have an olfactory memory longer than 1 year and up to 12 years and can distinguish between kin and non-kin just by scent. These findings confirm the significance of scent for family bonds in African elephants.
Collapse
Affiliation(s)
- Franziska Hoerner
- Department of Zoology, University of Wuppertal, 42119 Wuppertal, Germany
- Correspondence:
| | | | - Ann-Kathrin Oerke
- Endocrinology Laboratory, German Primate Centre, 37077 Goettingen, Germany
| | | | - Idu Azogu-Sepe
- Serengeti-Park Department of Research, Serengeti-Park Hodenhagen, 29693 Hodenhagen, Germany
| | | | - Karsten Damerau
- Department of Ecology, Europa-Universität Flensburg, 24943 Flensburg, Germany
| | - Angelika Preisfeld
- Department of Zoology, University of Wuppertal, 42119 Wuppertal, Germany
| |
Collapse
|
15
|
Lin YS, Weibel J, Landolt HP, Santini F, Slawik H, Borgwardt S, Cajochen C, Reichert CF. Brain activity during a working memory task after daily caffeine intake and caffeine withdrawal: a randomized double-blind placebo-controlled trial. Sci Rep 2023; 13:1002. [PMID: 36653409 PMCID: PMC9849460 DOI: 10.1038/s41598-022-26808-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
Acute caffeine intake has been found to increase working memory (WM)-related brain activity in healthy adults without improving behavioral performances. The impact of daily caffeine intake-a ritual shared by 80% of the population worldwide-and of its discontinuation on working memory and its neural correlates remained unknown. In this double-blind, randomized, crossover study, we examined working memory functions in 20 young healthy non-smokers (age: 26.4 ± 4.0 years; body mass index: 22.7 ± 1.4 kg/m2; and habitual caffeine intake: 474.1 ± 107.5 mg/day) in a 10-day caffeine (150 mg × 3 times/day), a 10-day placebo (3 times/day), and a withdrawal condition (9-day caffeine followed by 1-day placebo). Throughout the 10th day of each condition, participants performed four times a working memory task (N-Back, comprising 3- and 0-back), and task-related blood-oxygen-level-dependent (BOLD) activity was measured in the last session with functional magnetic resonance imaging. Compared to placebo, participants showed a higher error rate and a longer reaction time in 3- against 0-back trials in the caffeine condition; also, in the withdrawal condition we observed a higher error rate compared to placebo. However, task-related BOLD activity, i.e., an increased attention network and decreased default mode network activity in 3- versus 0-back, did not show significant differences among three conditions. Interestingly, irrespective of 3- or 0-back, BOLD activity was reduced in the right hippocampus in the caffeine condition compared to placebo. Adding to the earlier evidence showing increasing cerebral metabolic demands for WM function after acute caffeine intake, our data suggest that such demands might be impeded over daily intake and therefore result in a worse performance. Finally, the reduced hippocampal activity may reflect caffeine-associated hippocampal grey matter plasticity reported in the previous analysis. The findings of this study reveal an adapted neurocognitive response to daily caffeine exposure and highlight the importance of classifying impacts of caffeine on clinical and healthy populations.
Collapse
Affiliation(s)
- Yu-Shiuan Lin
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland. .,Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland. .,Neuropsychiatry and Brain Imaging, Psychiatric Hospital of the University of Basel, Basel, Switzerland.
| | - Janine Weibel
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland.,Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Sleep and Health Zurich, University Center of Competence, University of Zurich, Zurich, Switzerland
| | - Francesco Santini
- Division of Radiological Physics, Department of Radiology, University Hospital Basel, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Helen Slawik
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland.,Clinical Sleep Laboratory, Psychiatric Hospital of the University of Basel, Basel, Switzerland
| | - Stefan Borgwardt
- Neuropsychiatry and Brain Imaging, Psychiatric Hospital of the University of Basel, Basel, Switzerland
| | - Christian Cajochen
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland. .,Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland.
| | - Carolin Franziska Reichert
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland.,Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
| |
Collapse
|
16
|
Golub EM, Conner B, Edwards M, Gilllis L, Lacreuse A. Potential trade-off between olfactory and visual discrimination learning in common marmosets (Callithrix jacchus): Implications for the assessment of age-related cognitive decline. Am J Primatol 2022; 84:e23427. [PMID: 35942572 PMCID: PMC9444974 DOI: 10.1002/ajp.23427] [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: 02/07/2022] [Revised: 06/17/2022] [Accepted: 07/23/2022] [Indexed: 11/09/2022]
Abstract
Olfactory dysfunction has been identified as an early biomarker for dementia risk but has rarely been assessed in nonhuman primate models of human aging. To better characterize common marmosets as such models, we assessed olfactory discrimination performance in a sample of 10 animals (5 females), aged 2.5-8.9 years old. The monkeys were proficient in the discrimination and reversal of visual stimuli but naïve to odor stimuli. For olfactory discrimination, the monkeys performed a series of six discriminations of increasing difficulty between two odor stimuli. We found no evidence for an age-related decline as both young and older individuals were able to perform the discriminations in roughly the same number of trials. In addition, the older monkeys had faster responses than the younger animals. However, we noted that when adjusted for age, the speed of acquisition of the first discrimination in the olfactory modality was inversely correlated to the speed of acquisition of their first discrimination of two visual stimuli months earlier. These results suggest that marmosets may compensate for sensory deficits in one modality with higher sensory performance in another. These data have broad implications for the assessment of age-related cognitive decline and the categorization of animals as impaired or nonimpaired.
Collapse
Affiliation(s)
| | - Bryce Conner
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst MA
| | - Mélise Edwards
- Neuroscience and Behavior Program, University of Massachusetts Amherst MA
| | - Lacey Gilllis
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst MA
| | - Agnès Lacreuse
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst MA
- Neuroscience and Behavior Program, University of Massachusetts Amherst MA
| |
Collapse
|
17
|
Caffeine-Induced Acute and Delayed Responses in Cerebral Metabolism of Control and Schizophrenia-Like Wisket Rats. Int J Mol Sci 2022; 23:ijms23158186. [PMID: 35897774 PMCID: PMC9331118 DOI: 10.3390/ijms23158186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 12/04/2022] Open
Abstract
Recently, morphological impairments have been detected in the brain of a triple-hit rat schizophrenia model (Wisket), and delayed depressive effects of caffeine treatment in both control and Wisket animals have also been shown. The aims of this study were to determine the basal and caffeine-induced acute (30 min) and delayed (24 h) changes in the cerebral 18fluorodeoxyglucose (18F-FDG) uptake by positron emission tomography (PET) in control and Wisket rats. No significant differences were identified in the basal whole-brain metabolism between the two groups, and the metabolism was not modified acutely by a single intraperitoneal caffeine (20 mg/kg) injection in either group. However, one day after caffeine administration, significantly enhanced 18F-FDG uptake was detected in the whole brain and the investigated areas (hippocampus, striatum, thalamus, and hypothalamus) in the control group. Although the Wisket animals showed only moderate enhancements in the 18F-FDG uptake, significantly lower brain metabolism was observed in this group than in the caffeine-treated control group. This study highlights that the basal brain metabolism of Wisket animals was similar to control rats, and that was not influenced acutely by single caffeine treatment at the whole-brain level. Nevertheless, the distinct delayed responsiveness to this psychostimulant in Wisket model rats suggests impaired control of the cerebral metabolism.
Collapse
|
18
|
Pathophysiological Role and Medicinal Chemistry of A 2A Adenosine Receptor Antagonists in Alzheimer's Disease. Molecules 2022; 27:molecules27092680. [PMID: 35566035 PMCID: PMC9102440 DOI: 10.3390/molecules27092680] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 12/20/2022] Open
Abstract
The A2A adenosine receptor is a protein belonging to a family of four GPCR adenosine receptors. It is involved in the regulation of several pathophysiological conditions in both the central nervous system and periphery. In the brain, its localization at pre- and postsynaptic level in striatum, cortex, hippocampus and its effects on glutamate release, microglia and astrocyte activation account for a crucial role in neurodegenerative diseases, including Alzheimer’s disease (AD). This ailment is considered the main form of dementia and is expected to exponentially increase in coming years. The pathological tracts of AD include amyloid peptide-β extracellular accumulation and tau hyperphosphorylation, causing neuronal cell death, cognitive deficit, and memory loss. Interestingly, in vitro and in vivo studies have demonstrated that A2A adenosine receptor antagonists may counteract each of these clinical signs, representing an important new strategy to fight a disease for which unfortunately only symptomatic drugs are available. This review offers a brief overview of the biological effects mediated by A2A adenosine receptors in AD animal and human studies and reports the state of the art of A2A adenosine receptor antagonists currently in clinical trials. As an original approach, it focuses on the crucial role of pharmacokinetics and ability to pass the blood–brain barrier in the discovery of new agents for treating CNS disorders. Considering that A2A receptor antagonist istradefylline is already commercially available for Parkinson’s disease treatment, if the proof of concept of these ligands in AD is confirmed and reinforced, it will be easier to offer a new hope for AD patients.
Collapse
|
19
|
Siracusa ER, Higham JP, Snyder-Mackler N, Brent LJN. Social ageing: exploring the drivers of late-life changes in social behaviour in mammals. Biol Lett 2022; 18:20210643. [PMID: 35232274 PMCID: PMC8889194 DOI: 10.1098/rsbl.2021.0643] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Social interactions help group-living organisms cope with socio-environmental challenges and are central to survival and reproductive success. Recent research has shown that social behaviour and relationships can change across the lifespan, a phenomenon referred to as 'social ageing'. Given the importance of social integration for health and well-being, age-dependent changes in social behaviour can modulate how fitness changes with age and may be an important source of unexplained variation in individual patterns of senescence. However, integrating social behaviour into ageing research requires a deeper understanding of the causes and consequences of age-based changes in social behaviour. Here, we provide an overview of the drivers of late-life changes in sociality. We suggest that explanations for social ageing can be categorized into three groups: changes in sociality that (a) occur as a result of senescence; (b) result from adaptations to ameliorate the negative effects of senescence; and/or (c) result from positive effects of age and demographic changes. Quantifying the relative contribution of these processes to late-life changes in sociality will allow us to move towards a more holistic understanding of how and why these patterns emerge and will provide important insights into the potential for social ageing to delay or accelerate other patterns of senescence.
Collapse
Affiliation(s)
- Erin R Siracusa
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter, UK
| | - James P Higham
- Department of Anthropology, New York University, New York, NY, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA.,School of Life Sciences, Arizona State University, Tempe, AZ, USA.,School for Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
| | - Lauren J N Brent
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter, UK
| |
Collapse
|
20
|
Xu Y, Ning Y, Zhao Y, Peng Y, Luo F, Zhou Y, Li P. Caffeine Functions by Inhibiting Dorsal and Ventral Hippocampal Adenosine 2A Receptors to Modulate Memory and Anxiety, Respectively. Front Pharmacol 2022; 13:807330. [PMID: 35185566 PMCID: PMC8847668 DOI: 10.3389/fphar.2022.807330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/11/2022] [Indexed: 11/16/2022] Open
Abstract
As a nonspecific antagonist of the adenosine A2A receptor (A2AR), caffeine enhances learning and improves memory impairment. Simultaneously, the consumption of caffeine correlates with a feeling of anxiety. The hippocampus is functionally differentiated along its dorsal/ventral axis and plays a crucial role both in memory and anxiety. Whether caffeine exerts its regulation by inhibiting A2ARs in different subregions of the hippocampus is still unknown. In the present study, we found that after chronic intake of drinking water containing caffeine (1 g/L, 3 weeks), mice exhibited aggravated anxiety-like behavior and enhanced memory function. Tissue-specific, functional disruption of dorsal hippocampal A2ARs by the CRE-LoxP system prevented the memory-enhancing effect of caffeine, while selective disruption of ventral hippocampal A2ARs blocked the impact of caffeine on anxiety. These results, together with the enhanced memory of dorsal hippocampus A2AR knockout mice and greater anxiety-like behavior of ventral hippocampus A2AR knockout mice without caffeine, indicates a dissociation between the roles of ventral and dorsal hippocampal A2A receptors in caffeine’s effects on anxiety-like and memory-related behavioral measures, respectively. Furthermore, optogenetic activation of dorsal or ventral hippocampal A2ARs reversed the behavioral alterations caused by drinking caffeine, leading to impaired memory or decreased anxiety-like behaviors, respectively. Taken together, our findings suggest that the memory- and anxiety-enhancing effects of caffeine are related to the differential effects of inhibiting A2ARs in the dorsal and ventral hippocampus, respectively.
Collapse
Affiliation(s)
- Yawei Xu
- State Key Laboratory of Trauma, Department of Army Occupational Disease, The Molecular Biology Center, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yalei Ning
- State Key Laboratory of Trauma, Department of Army Occupational Disease, The Molecular Biology Center, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan Zhao
- State Key Laboratory of Trauma, Department of Army Occupational Disease, The Molecular Biology Center, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan Peng
- State Key Laboratory of Trauma, Department of Army Occupational Disease, The Molecular Biology Center, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fen Luo
- State Key Laboratory of Trauma, Department of Army Occupational Disease, The Molecular Biology Center, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuanguo Zhou
- State Key Laboratory of Trauma, Department of Army Occupational Disease, The Molecular Biology Center, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ping Li
- State Key Laboratory of Trauma, Department of Army Occupational Disease, The Molecular Biology Center, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
21
|
Gardener SL, Rainey-Smith SR, Villemagne VL, Fripp J, Doré V, Bourgeat P, Taddei K, Fowler C, Masters CL, Maruff P, Rowe CC, Ames D, Martins RN. Higher Coffee Consumption Is Associated With Slower Cognitive Decline and Less Cerebral Aβ-Amyloid Accumulation Over 126 Months: Data From the Australian Imaging, Biomarkers, and Lifestyle Study. Front Aging Neurosci 2021; 13:744872. [PMID: 34867277 PMCID: PMC8641656 DOI: 10.3389/fnagi.2021.744872] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/23/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Worldwide, coffee is one of the most popular beverages consumed. Several studies have suggested a protective role of coffee, including reduced risk of Alzheimer's disease (AD). However, there is limited longitudinal data from cohorts of older adults reporting associations of coffee intake with cognitive decline, in distinct domains, and investigating the neuropathological mechanisms underpinning any such associations. Methods: The aim of the current study was to investigate the relationship between self-reported habitual coffee intake, and cognitive decline assessed using a comprehensive neuropsychological battery in 227 cognitively normal older adults from the Australian Imaging, Biomarkers, and Lifestyle (AIBL) study, over 126 months. In a subset of individuals, we also investigated the relationship between habitual coffee intake and cerebral Aβ-amyloid accumulation (n = 60) and brain volumes (n = 51) over 126 months. Results: Higher baseline coffee consumption was associated with slower cognitive decline in executive function, attention, and the AIBL Preclinical AD Cognitive Composite (PACC; shown reliably to measure the first signs of cognitive decline in at-risk cognitively normal populations), and lower likelihood of transitioning to mild cognitive impairment or AD status, over 126 months. Higher baseline coffee consumption was also associated with slower Aβ-amyloid accumulation over 126 months, and lower risk of progressing to "moderate," "high," or "very high" Aβ-amyloid burden status over the same time-period. There were no associations between coffee intake and atrophy in total gray matter, white matter, or hippocampal volume. Discussion: Our results further support the hypothesis that coffee intake may be a protective factor against AD, with increased coffee consumption potentially reducing cognitive decline by slowing cerebral Aβ-amyloid accumulation, and thus attenuating the associated neurotoxicity from Aβ-amyloid-mediated oxidative stress and inflammatory processes. Further investigation is required to evaluate whether coffee intake could be incorporated as a modifiable lifestyle factor aimed at delaying AD onset.
Collapse
Affiliation(s)
- Samantha L Gardener
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Perth, WA, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Perth, WA, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,School of Psychological Science, University of Western Australia, Perth, WA, Australia
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jurgen Fripp
- CSIRO Health and Biosecurity/Australian e-Health Research Centre, Herston, QLD, Australia
| | - Vincent Doré
- CSIRO Health and Biosecurity/Australian e-Health Research Centre, Herston, QLD, Australia.,Department of Molecular Imaging and Therapy, Centre for PET, Austin Health, Heidelberg, VIC, Australia
| | - Pierrick Bourgeat
- CSIRO Health and Biosecurity/Australian e-Health Research Centre, Herston, QLD, Australia
| | - Kevin Taddei
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Perth, WA, Australia
| | - Christopher Fowler
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Maruff
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.,Cogstate Ltd., Melbourne, VIC, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Centre for PET, Austin Health, Heidelberg, VIC, Australia.,The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - David Ames
- National Ageing Research Institute, Parkville, VIC, Australia.,Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, VIC, Australia
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Perth, WA, Australia.,Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | | |
Collapse
|
22
|
Khani E, Khiali S, Beheshtirouy S, Entezari-Maleki T. Potential pharmacologic treatments for COVID-19 smell and taste loss: A comprehensive review. Eur J Pharmacol 2021; 912:174582. [PMID: 34678243 PMCID: PMC8524700 DOI: 10.1016/j.ejphar.2021.174582] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/08/2021] [Accepted: 10/18/2021] [Indexed: 12/30/2022]
Abstract
The acute loss of taste and smell following COVID-19 are hallmark symptoms that affect 20-85% of patients. However, the pathophysiology and potential treatments of COVID-19 smell and taste loss are not fully understood. We searched the literature to review the potential pathologic pathways and treatment options for COVID-19 smell and taste loss. The interaction of novel coronavirus with ACE-2 receptors expressed on sustentacular cells and taste buds results in direct damage to the olfactory and gustatory systems. Also, the invasion of the virus to the olfactory neurons and consequent local inflammation are other proposed mechanisms. Therefore, COVID-19 patients with smell or taste loss may benefit from neuroprotective, anti-inflammatory, or depolarizing agents. Based on the current evidence, phosphodiesterase inhibitors, insulin, and corticosteroids can be promising for the management of COVID-19 smell and taste loss. This review provided crucial information for treating COVID-19-related smell and/or taste loss, urging to perform large clinical trials to find optimum treatment options.
Collapse
Affiliation(s)
- Elnaz Khani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajad Khiali
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samineh Beheshtirouy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Entezari-Maleki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran,Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Corresponding author. Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, P.O. Box: 51664-14766 12, Iran
| |
Collapse
|
23
|
Been LE, Sheppard PAS, Galea LAM, Glasper ER. Hormones and neuroplasticity: A lifetime of adaptive responses. Neurosci Biobehav Rev 2021; 132:679-690. [PMID: 34808191 DOI: 10.1016/j.neubiorev.2021.11.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022]
Abstract
Major life transitions often co-occur with significant fluctuations in hormones that modulate the central nervous system. These hormones enact neuroplastic mechanisms that prepare an organism to respond to novel environmental conditions and/or previously unencountered cognitive, emotional, and/or behavioral demands. In this review, we will explore several examples of how hormones mediate neuroplastic changes in order to produce adaptive responses, particularly during transitions in life stages. First, we will explore hormonal influences on social recognition in both males and females as they transition to sexual maturity. Next, we will probe the role of hormones in mediating the transitions to motherhood and fatherhood, respectively. Finally, we will survey the long-term impact of reproductive experience on neuroplasticity in females, including potential protective effects and risk factors associated with reproductive experience in mid-life and beyond. Ultimately, a more complete understanding of how hormones influence neuroplasticity throughout the lifespan, beyond development, is necessary for understanding how individuals respond to life changes in adaptive ways.
Collapse
Affiliation(s)
- Laura E Been
- Department of Psychology, Haverford College, 370 Lancaster Avenue, Haverford, PA, 19041, USA.
| | - Paul A S Sheppard
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, 1151 Richmond St, London, Ontario, N6A 5B7, Canada.
| | - Liisa A M Galea
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada.
| | - Erica R Glasper
- Department of Psychology, University of Maryland, College Park, MD, 20742 USA.
| |
Collapse
|
24
|
Zhang RC, Madan CR. How does caffeine influence memory? Drug, experimental, and demographic factors. Neurosci Biobehav Rev 2021; 131:525-538. [PMID: 34563564 DOI: 10.1016/j.neubiorev.2021.09.033] [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: 05/19/2021] [Revised: 09/10/2021] [Accepted: 09/17/2021] [Indexed: 11/29/2022]
Abstract
Caffeine is a widely used nootropic drug, but its effects on memory in healthy participants have not been sufficiently evaluated. Here we review evidence of the effects of caffeine on different types of memory, and the associated drug, experimental, and demographical factors. There is limited evidence that caffeine affects performance in memory tasks beyond improved reaction times. For drug factors, a dose-response relationship may exist but findings are inconsistent. Moreover, there is evidence that the source of caffeine can modulate its effects on memory. For experimental factors, past studies often lacked a baseline control for diet and sleep and none discussed the possible reversal of withdrawal effect due to pre-experimental fasting. For demographic factors, caffeine may interact with sex and age, and the direction of the effect may depend on the dose, individual tolerance, and metabolism at baseline. Future studies should incorporate these considerations, as well as providing continued evidence on the effect of caffeine in visuospatial, prospective, and implicit memory measures.
Collapse
Affiliation(s)
- Ruo-Chong Zhang
- School of Psychology, University of Nottingham, Nottingham, UK.
| | | |
Collapse
|
25
|
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
|
26
|
Zhou X, Zhang L. The Neuroprotective Effects of Moderate and Regular Caffeine Consumption in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5568011. [PMID: 34447487 PMCID: PMC8384510 DOI: 10.1155/2021/5568011] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/27/2021] [Accepted: 07/19/2021] [Indexed: 12/25/2022]
Abstract
The increasing numbers of elderly Alzheimer's disease (AD) patients because of a steady increase in the average lifespan and aging society attract great scientific concerns, while there were fewer effective treatments on AD progression due to unclear exact causes and pathogenesis of AD. Moderate (200-500 mg/d) and regular caffeine consumption from coffee and tea are considered to alleviate the risk of AD and have therapeutic potential. This paper reviewed epidemiological studies about the relationship of caffeine intake from coffee or/and tea with the risk of AD and summarized the caffeine-related AD therapies based on experimental models. And further well-designed and well-conducted studies are suggested to investigate the optimal dosages, frequencies, and durations of caffeine consumption to slow down AD progression and treat AD.
Collapse
Affiliation(s)
- Xiangyu Zhou
- School of Food Science and Nutrition, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK
| | - Lin Zhang
- The Key Laboratory for Special Medical Food Process in Hunan Province, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| |
Collapse
|
27
|
Adenosine A 2A receptor neurons in the olfactory bulb mediate odor-guided behaviors in mice. Brain Res 2021; 1768:147590. [PMID: 34310936 DOI: 10.1016/j.brainres.2021.147590] [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: 04/20/2021] [Revised: 07/09/2021] [Accepted: 07/21/2021] [Indexed: 11/21/2022]
Abstract
Depression, rapid eye movement (REM) sleep behavior disorder, and altered olfaction are often present in Parkinson's disease. Our previous studies demonstrated the role of the olfactory bulb (OB) in causing REM sleep disturbances in depression. Furthermore, adenosine A2A receptors (A2AR) which are richly expressed in the OB, play an important role in the regulation of REM sleep. Caffeine, an adenosine A1 receptors and A2AR antagonist, and other A2AR antagonists were reported to improve olfactory function and restore age-related olfactory deficits. Therefore, we hypothesized that the A2AR neurons in the OB may regulate olfaction or odor-guided behaviors in mice. In the present study, we employed chemogenetics to specifically activate or inhibit neuronal activity. Then, buried food test and olfactory habituation/dishabituation test were performed to measure the changes in the mice's olfactory ability. We demonstrated that activation of OB neurons or OB A2AR neurons shortened the latency of buried food test and enhanced olfactory habituation to the same odors and dishabituation to different odors; inhibition of these neurons showed the opposite effects. Photostimulation of ChR2-expressing OB A2AR neuron terminals evoked inward current in the olfactory tubercle (OT) and the piriform cortex (Pir), which was blocked by glutamate receptor antagonists 2-amino-5-phosphonopentanoic acid and 6-cyano-7nitroquinoxaline-2,3-dione. Collectively, these results suggest that the OB mediates olfaction via A2AR neurons in mice. Moreover, the excitatory glutamatergic release from OB neurons to the OT and the Pir were found responsible for the olfaction-mediated effects of OB A2AR neurons.
Collapse
|
28
|
Caffeine - treat or trigger? Disparate behavioral and long-term dopaminergic changes in control and schizophrenia-like Wisket rats. Physiol Behav 2021; 236:113410. [PMID: 33819453 DOI: 10.1016/j.physbeh.2021.113410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/12/2021] [Accepted: 03/26/2021] [Indexed: 11/20/2022]
Abstract
The influence of caffeine on behavioral functions in both healthy and schizophrenic subjects is controversial. Here we aimed to reveal the effects of repeated caffeine pre- and post-training treatments on motor and exploratory activities and cognitive functions in a reward-based test (Ambitus) along with a brain region-specific dopamine D2 receptor profile in control and schizophrenia-like WISKET model rats. In the control animals, pre-treatment caused temporary enhancement in motor activity, while permanent improvement in learning function was detected in the WISKET animals. Post-treatment produced significant impairments in both groups. Caffeine caused short-lasting hyperactivity followed by a rebound in the inactive phase determined in undisturbed circumstance. Caffeine treatment substantially enhanced the dopamine D2 receptor mediated G-protein activation in the prefrontal cortex and olfactory bulb of both groups, while it increased in the dorsal striatum and cerebral cortex only in the WISKET animals. Caffeine enhanced the maximal binding capacity in the hippocampus and cerebral cortex of WISKET animals, but it decreased in the prefrontal cortex of the control animals. Regarding the dopamine D2 receptor mRNA expression, caffeine treatment caused significant enhancement in the prefrontal cortex of WISKET animals, while it increased the hippocampal dopamine D2 receptor protein amount in both groups. This study highlights the disparate effects of caffeine pre- versus post-training treatments on behavioral parameters in both control and schizophrenia-like animals and the prolonged changes in the dopaminergic system. It is supposed that the delayed depressive effects of caffeine might be compensated by frequent coffee intake, as observed in schizophrenic patients.
Collapse
|
29
|
Rojic-Becker D, Portero-Tresserra M, Martí-Nicolovius M, Vale-Martínez A, Guillazo-Blanch G. Effects of caloric restriction on monoaminergic neurotransmission, peripheral hormones, and olfactory memory in aged rats. Behav Brain Res 2021; 409:113328. [PMID: 33930470 DOI: 10.1016/j.bbr.2021.113328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/30/2021] [Accepted: 04/25/2021] [Indexed: 01/02/2023]
Abstract
Aging is associated with a reduced ability to identify and discriminate scents, and olfactory dysfunction has been linked to preclinical stages of neurodegenerative diseases in humans. Moreover, emerging evidence suggests that smell-driven behaviors are regulated by hormones like insulin or leptin, and by metabolic parameters like glucose, which in turn may influence monoaminergic neurotransmission in brain areas related to cognition. Several studies have suggested that dietary interventions like caloric restriction (CR) can mitigate the age-induced decline in memory by modifying metabolic parameters and brain monoaminergic levels. The present study explored the effects of CR on age-dependent olfactory memory deficits, as well as their relationship with peripheral leptin, insulin and glucose levels, and brain monoamines. To this end, aged rats (24-months-old) fed on a CR diet or with ad libitum access to food, and adult rats (3-4 months), were trained in an odor discrimination task (ODT). The peripheral plasma levels of insulin, leptin, and glucose, and of monoamines and metabolites/precursors in brain areas related to olfactory learning and memory processes, such as the striatum and frontal cortex (FC), were determined. The data obtained indicated that CR attenuated the age-dependent decline in olfactory sensitivity in old animals fed ad libitum, which was correlated with the performance in ODT retention trial, as well as with leptin plasma levels. CR enhanced dopamine levels in the striatum, while it attenuated the age-related decline in serotonin levels in the striatum and FC. Such findings support a positive effect of CR on age-dependent olfactory sensitivity decline and dysfunctions in brain monoamine levels.
Collapse
Affiliation(s)
- Divka Rojic-Becker
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Portero-Tresserra
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Margarita Martí-Nicolovius
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Vale-Martínez
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Guillazo-Blanch
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
30
|
Olopade FE, Femi-Akinlosotu OM, Adekanmbi AJ, Ighogboja OO, Shokunbi MT. Chronic Caffeine Ingestion Improves Motor Function and Increases Dendritic Length and Arborization in the Motor Cortex, Striatum, and Cerebellum. J Caffeine Adenosine Res 2021. [DOI: 10.1089/caff.2020.0017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
| | | | | | | | - Matthew T. Shokunbi
- Department of Anatomy and University of Ibadan, Ibadan, Nigeria
- Department of Surgery, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
31
|
Impaired olfactory neurogenesis affects the performance of olfactory-guided behavior in aged female opossums. Sci Rep 2021; 11:4418. [PMID: 33627729 PMCID: PMC7904797 DOI: 10.1038/s41598-021-83834-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/01/2021] [Indexed: 12/23/2022] Open
Abstract
Increasing evidence has indicated that adult neurogenesis contributes to brain plasticity, although function of new neurons is still under debate. In opossums, we performed an olfactory-guided behavior task and examined the association between olfactory discrimination-guided behavior and adult neurogenesis in the olfactory bulb (OB). We found that young and aged opossums of either sex learned to find food buried in litter using olfactory cues. However, aged females required more time to find food compared to aged males and young opossums of both sexes. The levels of doublecortin, that is used as a marker for immature neurons, were the lowest in the OB of aged female opossums. Another protein, HuD that is associated with learning and memory, was detected in all layers of the OB, except the granule cell layer, where a high density of DCX cells was detected. The level of HuD was higher in aged opossums compared to young opossums. This indicates that HuD is involved in plasticity and negatively regulates olfactory perception. The majority of 2-year-old female opossums are in the post-reproductive age but males of this age are still sexually active. We suggest that in aged female opossums neural plasticity induced by adult neurogenesis decreases due to their hormonal decline.
Collapse
|
32
|
Lin YS, Weibel J, Landolt HP, Santini F, Meyer M, Brunmair J, Meier-Menches SM, Gerner C, Borgwardt S, Cajochen C, Reichert C. Daily Caffeine Intake Induces Concentration-Dependent Medial Temporal Plasticity in Humans: A Multimodal Double-Blind Randomized Controlled Trial. Cereb Cortex 2021; 31:3096-3106. [PMID: 33585896 DOI: 10.1093/cercor/bhab005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/25/2020] [Accepted: 12/27/2020] [Indexed: 12/18/2022] Open
Abstract
Caffeine is commonly used to combat high sleep pressure on a daily basis. However, interference with sleep-wake regulation could disturb neural homeostasis and insufficient sleep could lead to alterations in human gray matter. Hence, in this double-blind, randomized, cross-over study, we examined the impact of 10-day caffeine (3 × 150 mg/day) on human gray matter volumes (GMVs) and cerebral blood flow (CBF) by fMRI MP-RAGE and arterial spin-labeling sequences in 20 habitual caffeine consumers, compared with 10-day placebo (3 × 150 mg/day). Sleep pressure was quantified by electroencephalographic slow-wave activity (SWA) in the previous nighttime sleep. Nonparametric voxel-based analyses revealed a significant reduction in GMV in the medial temporal lobe (mTL) after 10 days of caffeine intake compared with 10 days of placebo, voxel-wisely adjusted for CBF considering the decreased perfusion after caffeine intake compared with placebo. Larger GMV reductions were associated with higher individual concentrations of caffeine and paraxanthine. Sleep SWA was, however, neither different between conditions nor associated with caffeine-induced GMV reductions. Therefore, the data do not suggest a link between sleep depth during daily caffeine intake and changes in brain morphology. In conclusion, daily caffeine intake might induce neural plasticity in the mTL depending on individual metabolic processes.
Collapse
Affiliation(s)
- Yu-Shiuan Lin
- Centre for Chronobiology, University Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland.,Transfaculty Research Platform, Molecular and Cognitive Neurosciences, University of Basel, 4055 Basel, Switzerland.,Neuropsychiatry and Brain Imaging, University Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland
| | - Janine Weibel
- Centre for Chronobiology, University Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland.,Transfaculty Research Platform, Molecular and Cognitive Neurosciences, University of Basel, 4055 Basel, Switzerland
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland.,Sleep & Health Zurich, University Center of Competence, University of Zurich, 8091 Zurich, Switzerland
| | - Francesco Santini
- Department of Radiology, Division of Radiological Physics, University Hospital Basel, 4031 Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, 4123 Allschwil, Switzerland
| | - Martin Meyer
- Centre for Chronobiology, University Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland.,Transfaculty Research Platform, Molecular and Cognitive Neurosciences, University of Basel, 4055 Basel, Switzerland.,Clinical Sleep Laboratory, University Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland
| | - Julia Brunmair
- Department of Analytical Chemistry, University of Vienna, 1090 Vienna A, Austria
| | | | - Christopher Gerner
- Department of Analytical Chemistry, University of Vienna, 1090 Vienna A, Austria.,Joint Metabolome Facility, University of Vienna and Medical University of Vienna, 1090 Vienna A, Austria
| | - Stefan Borgwardt
- Neuropsychiatry and Brain Imaging, University Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland
| | - Christian Cajochen
- Centre for Chronobiology, University Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland.,Transfaculty Research Platform, Molecular and Cognitive Neurosciences, University of Basel, 4055 Basel, Switzerland
| | - Carolin Reichert
- Centre for Chronobiology, University Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland.,Transfaculty Research Platform, Molecular and Cognitive Neurosciences, University of Basel, 4055 Basel, Switzerland
| |
Collapse
|
33
|
Yang Q, Li B, Chen S, Tang J, Li Y, Li Y, Zhang S, Shi C, Zhang Y, Mou M, Xue W, Zhu F. MMEASE: Online meta-analysis of metabolomic data by enhanced metabolite annotation, marker selection and enrichment analysis. J Proteomics 2021; 232:104023. [PMID: 33130111 DOI: 10.1016/j.jprot.2020.104023] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 10/12/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022]
Abstract
Large-scale and long-term metabolomic studies have attracted widespread attention in the biomedical studies yet remain challenging despite recent technique progresses. In particular, the ineffective way of experiment integration and limited capacity in metabolite annotation are known issues. Herein, we constructed an online tool MMEASE enabling the integration of multiple analytical experiments with an enhanced metabolite annotation and enrichment analysis (https://idrblab.org/mmease/). MMEASE was unique in capable of (1) integrating multiple analytical blocks; (2) providing enriched annotation for >330 thousands of metabolites; (3) conducting enrichment analysis using various categories/sub-categories. All in all, MMEASE aimed at supplying a comprehensive service for large-scale and long-term metabolomics, which might provide valuable guidance to current biomedical studies. SIGNIFICANCE: To facilitate the studies of large-scale and long-term metabolomic analysis, MMEASE was developed to (1) achieve the online integration of multiple datasets from different analytical experiments, (2) provide the most diverse strategies for marker discovery, enabling performance assessment and (3) significantly amplify metabolite annotation and subsequent enrichment analysis. MMEASE aimed at supplying a comprehensive service for long-term and large-scale metabolomics, which might provide valuable guidance to current biomedical studies.
Collapse
Affiliation(s)
- Qingxia Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing, Chongqing 401331, China
| | - Sijie Chen
- School of Pharmaceutical Sciences, School of Big Data and Software Engineering, Chongqing University, Chongqing, Chongqing 401331, China
| | - Jing Tang
- Department of Bioinformatics, Chongqing Medical University, Chongqing, Chongqing 400016, China
| | - Yinghong Li
- School of Pharmaceutical Sciences, School of Big Data and Software Engineering, Chongqing University, Chongqing, Chongqing 401331, China
| | - Yi Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Song Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Cheng Shi
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ying Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, School of Big Data and Software Engineering, Chongqing University, Chongqing, Chongqing 401331, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; School of Pharmaceutical Sciences, School of Big Data and Software Engineering, Chongqing University, Chongqing, Chongqing 401331, China.
| |
Collapse
|
34
|
Effects of sub-chronic caffeine ingestion on memory and the hippocampal Akt, GSK-3β and ERK signaling in mice. Brain Res Bull 2021; 170:137-145. [PMID: 33556562 DOI: 10.1016/j.brainresbull.2021.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/22/2020] [Accepted: 02/03/2021] [Indexed: 12/20/2022]
Abstract
Caffeine, one of the most widely consumed psychoactive substance in the world, has been shown to affect mood, memory, alertness, and cognitive performance. This study aimed to assess the effect of sub-chronic oral gavage of caffeine on memory and the phosphorylation levels of hippocampal Akt (protein kinase B), GSK-3β (Glycogen Synthase Kinase-3beta) and ERK (extracellular signal-regulated kinase) in mice. Adult male NMRI mice were administered with caffeine at the doses of 0.25, 0.5, 0.75 and 1.5 mg/kg/oral gavage for 10 days before behavioral assessments. Upon completion of the behavioral tasks, the hippocampi were isolated for western blot analysis to detect the phosphorylated and total levels of Akt, GSK-3β and ERK proteins. The results showed that sub-chronic caffeine ingestion at the dose of 0.5 mg/kg improves memory in mice both in passive avoidance and novel object recognition tasks. Furthermore, this memory enhancing dose of caffeine elevated the ratios of phosphorylated to total contents of hippocampal Akt, GSK-3β and ERK. This study suggests that sub-chronic low dose of caffeine improves memory and increases the phosphorylation of hippocampal Akt, GSK-3β and ERK proteins.
Collapse
|
35
|
Jenner P, Mori A, Kanda T. Can adenosine A2A receptor antagonists be used to treat cognitive impairment, depression or excessive sleepiness in Parkinson's disease? Parkinsonism Relat Disord 2020; 80 Suppl 1:S28-S36. [DOI: 10.1016/j.parkreldis.2020.09.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/15/2020] [Indexed: 01/29/2023]
|
36
|
Tang D, Yan R, Sun Y, Kai G, Chen K, Li J. Material basis, effect, and mechanism of ethanol extract of Pinellia ternata tubers on oxidative stress-induced cell senescence. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 77:153275. [PMID: 32659678 DOI: 10.1016/j.phymed.2020.153275] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/25/2020] [Accepted: 06/29/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND The tuber of Pinellia ternata has been used for a thousand years in China. P. ternata possessed the activities of anti-emetic, sedative-hypnotic, anti-cancer, anti-asthmatic, anti-tussive, and anti-inflammatory. It is the representative of expectorant medicines in Traditional Chinese Medicine (TCM). Phlegm is the pathological product and a new pathogenic factor of the metabolite, which is analogous to the damage of oxidative stress. PURPOSE The objectives of the study were to investigate the protective activity and mechanism of ethanol extract of P. ternata tubers (PTE) and its main constituents on oxidative stress-induced cell senescence. METHODS H2O2 and AAPH were used to establish cellular senescence models. The anti-aging effects of PTE and its components were evaluated by SA-β-gal staining, flow cytometry, scanning electron microscope (SEM), and multiple microplate reader, the molecular mechanisms of them were investigated by qRT-PCR and Western Blot. RESULTS We found PTE exhibited the apparent effect on cell senescence, evidenced by inhibiting senescence β-Galactosidase (SA-β-gal) expression, lipofuscin accumulation, cell cycle arrest at the G2/M phase, oxidative damage and apoptosis, and increasing telomerase activity. Their mechanisms were related to increase expressions of SIRT1, forkhead box 3a (Foxo3a), Bcl-2, active regulator of SIRT1, RPS19BP1 (AROS), and Hu antigen R (HuR), but decrease Bax, p53 and deleted in breast cancer 1 (DBC1) levels. Furthermore, adenosine, and succinic acid, as the critical substances in PTE, could also inhibit SA-β-gal expression and cell cycle arrest, down-regulate the expression of Bax, and up-regulate Bcl-2, SirT1, and Foxo3a. CONCLUSIONS We have demonstrated that PTE slows down oxidative stress-induced cell senescence, and adenosine and succinic acid are the key active components.
Collapse
Affiliation(s)
- Ding Tang
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Renyi Yan
- Tianjin Ubasio Biotechnology Group Co., Ltd., Tianjin 300457, China
| | - Yuan Sun
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
| | - Guoyin Kai
- College of pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Keli Chen
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
| | - Juan Li
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China.
| |
Collapse
|
37
|
Aging Alters Olfactory Bulb Network Oscillations and Connectivity: Relevance for Aging-Related Neurodegeneration Studies. Neural Plast 2020; 2020:1703969. [PMID: 32774353 PMCID: PMC7396091 DOI: 10.1155/2020/1703969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/27/2020] [Accepted: 02/12/2020] [Indexed: 11/18/2022] Open
Abstract
The aging process eventually cause a breakdown in critical synaptic plasticity and connectivity leading to deficits in memory function. The olfactory bulb (OB) and the hippocampus, both regions of the brain considered critical for the processing of odors and spatial memory, are commonly affected by aging. Using an aged wild-type C57B/6 mouse model, we sought to define the effects of aging on hippocampal plasticity and the integrity of cortical circuits. Specifically, we measured the long-term potentiation of high-frequency stimulation (HFS-LTP) at the Shaffer-Collateral CA1 pyramidal synapses. Next, local field potential (LFP) spectra, phase-amplitude theta-gamma coupling (PAC), and connectivity through coherence were assessed in the olfactory bulb, frontal and entorhinal cortices, CA1, and amygdala circuits. The OB of aged mice showed a significant increase in the number of histone H2AX-positive neurons, a marker of DNA damage. While the input-output relationship measure of basal synaptic activity was found not to differ between young and aged mice, a pronounced decline in the slope of field excitatory postsynaptic potential (fEPSP) and the population spike amplitude (PSA) were found in aged mice. Furthermore, aging was accompanied by deficits in gamma network oscillations, a shift to slow oscillations, reduced coherence and theta-gamma PAC in the OB circuit. Thus, while the basal synaptic activity was unaltered in older mice, impairment in hippocampal synaptic transmission was observed only in response to HFS. However, age-dependent alterations in neural network appeared spontaneously in the OB circuit, suggesting the neurophysiological basis of synaptic deficits underlying olfactory processing. Taken together, the results highlight the sensitivity and therefore potential use of LFP quantitative network oscillations and connectivity at the OB level as objective electrophysiological markers that will help reveal specific dysfunctional circuits in aging-related neurodegeneration studies.
Collapse
|
38
|
Temido-Ferreira M, Ferreira DG, Batalha VL, Marques-Morgado I, Coelho JE, Pereira P, Gomes R, Pinto A, Carvalho S, Canas PM, Cuvelier L, Buée-Scherrer V, Faivre E, Baqi Y, Müller CE, Pimentel J, Schiffmann SN, Buée L, Bader M, Outeiro TF, Blum D, Cunha RA, Marie H, Pousinha PA, Lopes LV. Age-related shift in LTD is dependent on neuronal adenosine A 2A receptors interplay with mGluR5 and NMDA receptors. Mol Psychiatry 2020; 25:1876-1900. [PMID: 29950682 PMCID: PMC7387321 DOI: 10.1038/s41380-018-0110-9] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 05/02/2018] [Accepted: 05/14/2018] [Indexed: 01/31/2023]
Abstract
Synaptic dysfunction plays a central role in Alzheimer's disease (AD), since it drives the cognitive decline. An association between a polymorphism of the adenosine A2A receptor (A2AR) encoding gene-ADORA2A, and hippocampal volume in AD patients was recently described. In this study, we explore the synaptic function of A2AR in age-related conditions. We report, for the first time, a significant overexpression of A2AR in hippocampal neurons of aged humans, which is aggravated in AD patients. A similar profile of A2AR overexpression in rats was sufficient to drive age-like memory impairments in young animals and to uncover a hippocampal LTD-to-LTP shift. This was accompanied by increased NMDA receptor gating, dependent on mGluR5 and linked to enhanced Ca2+ influx. We confirmed the same plasticity shift in memory-impaired aged rats and APP/PS1 mice modeling AD, which was rescued upon A2AR blockade. This A2AR/mGluR5/NMDAR interaction might prove a suitable alternative for regulating aberrant mGluR5/NMDAR signaling in AD without disrupting their constitutive activity.
Collapse
Grants
- FCT - Fundação para a Ciência e Tecnologia
- Région Hauts de France (PARTNAIRR COGNADORA), ANR (ADORATAU and SPREADTAU), LECMA/Alzheimer Forschung Initiative, Programmes d’Investissements d’Avenir LabEx (excellence laboratory) DISTALZ (Development of Innovative Strategies for a Transdisciplinary approach to ALZheimer’s disease), France Alzheimer/Fondation de France, the FHU VasCog research network (Lille, France), Fondation pour la Recherche Médicale, Fondation Plan Alzheimer, INSERM, CNRS, Université Lille 2, Lille Métropole Communauté Urbaine, FEDER, DN2M, LICEND and CoEN.
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Goettingen
- ATIP/AVENIR program (Centre National de la Recherche Scientifique - CNRS)
- ATIP/AVENIR program (Centre National de la Recherche Scientifique - CNRS), by the Foundation Plan Alzheimer (Senior Innovative Grant 2010)
Collapse
Affiliation(s)
- Mariana Temido-Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Diana G Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal
- MedInUP-Center for Drug Discovery and Innovative Medicines, University of Porto, 4200-450, Porto, Portugal
| | - Vânia L Batalha
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Inês Marques-Morgado
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Joana E Coelho
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Pedro Pereira
- Laboratory of Neuropathology, Department of Neurosciences, Hospital de Santa Maria, CHLN, EPE, 1649-035, Lisbon, Portugal
| | - Rui Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
- Faculdade de Ciências da Universidade de Lisboa, 1749-016, Lisbon, Portugal
| | - Andreia Pinto
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Sara Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Paula M Canas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Laetitia Cuvelier
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), 1070, Brussels, Belgium
| | - Valerie Buée-Scherrer
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172 JPArc, "Alzheimer & Tauopathie", LabEx DISTALZ, Lille, France
| | - Emilie Faivre
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172 JPArc, "Alzheimer & Tauopathie", LabEx DISTALZ, Lille, France
| | - Younis Baqi
- PharmaCenter Bonn, Pharmazeutische Chemie I, Pharmazeutisches Institut, University of Bonn, Bonn, Germany
- Department of Chemistry, Faculty of Science, Sultan Qaboos University, PO Box 36, Postal Code 123, Muscat, Oman
| | - Christa E Müller
- PharmaCenter Bonn, Pharmazeutische Chemie I, Pharmazeutisches Institut, University of Bonn, Bonn, Germany
| | - José Pimentel
- Laboratory of Neuropathology, Department of Neurosciences, Hospital de Santa Maria, CHLN, EPE, 1649-035, Lisbon, Portugal
| | - Serge N Schiffmann
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), 1070, Brussels, Belgium
| | - Luc Buée
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172 JPArc, "Alzheimer & Tauopathie", LabEx DISTALZ, Lille, France
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), 13125, Berlin, Germany
- Charité-University Medicine, 10117, Berlin, Germany
- Institute of Biology, University of Lübeck, 23652, Lübeck, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
- Max Planck Institute for Experimental Medicine, 37075, Göttingen, Germany
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082, Lisbon, Portugal
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, United Kingdom
| | - David Blum
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172 JPArc, "Alzheimer & Tauopathie", LabEx DISTALZ, Lille, France
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Hélène Marie
- Université Côte d'Azur, CNRS UMR7276, IPMC, 06560, Valbonne, France
| | - Paula A Pousinha
- Université Côte d'Azur, CNRS UMR7276, IPMC, 06560, Valbonne, France
| | - Luísa V Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal.
| |
Collapse
|
39
|
Caffeine Consumption plus Physical Exercise Improves Behavioral Impairments and Stimulates Neuroplasticity in Spontaneously Hypertensive Rats (SHR): an Animal Model of Attention Deficit Hyperactivity Disorder. Mol Neurobiol 2020; 57:3902-3919. [PMID: 32621279 DOI: 10.1007/s12035-020-02002-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a prevalent and disabling disorder, mainly characterized by hyperactivity, inattention, and impulsivity, but also by olfactory and memory impairments that frequently persist throughout lifetime. The pathophysiology of ADHD is complex, involving several brain regions and neural pathways including alterations in adenosine neuromodulation. The administration of caffeine (a non-selective adenosine receptor antagonist) and physical exercise have been independently pointed as effective approaches for the management of ADHD symptoms. Here, we evaluated the effects of caffeine consumption (0.3 mg/mL in drinking water) plus physical exercise in running wheels during 6 weeks-starting during either adolescence (30 days old) or adulthood (4-5 months old)-on behavioral performance (including olfactory discrimination, open field, object recognition, and water maze tests) on the brain levels of monoamines (by high-performance liquid chromatography), on proteins related to synaptic plasticity and on brain-derived neurotrophic factor signaling (by Western blot analysis) in spontaneously hypertensive rats (SHRs), a validated animal model of ADHD. SHRs displayed persistent impairments of olfactory and short-term recognition memory from adolescence to adulthood, which were accompanied by lower levels of synaptosomal-associated protein 25 (SNAP-25) in the prefrontal cortex and hippocampus. The association of caffeine plus physical exercise during adolescence or adulthood restored the olfactory discrimination ability and, in an independent manner, improved short-term recognition memory of SHRs. These benefits were not associated to alterations in locomotor activity or in the hypertensive phenotype. The association of caffeine consumption plus physical exercise during adolescence increased the levels of SNAP-25, syntaxin, and serotonin in the hippocampus and prefrontal cortex, and striatal dopamine levels in SHRs. These results provide new evidence of the potential of caffeine and physical exercise, starting at adolescence or adult life, to improve behavioral impairments and stimulate neuroplasticity in ADHD.
Collapse
|
40
|
Petković B, Kesić S, Pešić V. Critical View on the Usage of Ribavirin in Already Existing Psychostimulant-Use Disorder. Curr Pharm Des 2020; 26:466-484. [PMID: 31939725 PMCID: PMC8383468 DOI: 10.2174/1381612826666200115094642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022]
Abstract
Substance-use disorder represents a frequently hidden non-communicable chronic disease. Patients with intravenous drug addiction are at high risk of direct exposure to a variety of viral infections and are considered to be the largest subpopulation infected with the hepatitis C virus. Ribavirin is a synthetic nucleoside analog that has been used as an integral component of hepatitis C therapy. However, ribavirin medication is quite often associated with pronounced psychiatric adverse effects. It is not well understood to what extent ribavirin per se contributes to changes in drug-related neurobehavioral disturbances, especially in the case of psychostimulant drugs, such as amphetamine. It is now well-known that repeated amphetamine usage produces psychosis in humans and behavioral sensitization in animals. On the other hand, ribavirin has an affinity for adenosine A1 receptors that antagonistically modulate the activity of dopamine D1 receptors, which play a critical role in the development of behavioral sensitization. This review will focus on the current knowledge of neurochemical/ neurobiological changes that exist in the psychostimulant drug-addicted brain itself and the antipsychotic-like efficiency of adenosine agonists. Particular attention will be paid to the potential side effects of ribavirin therapy, and the opportunities and challenges related to its application in already existing psychostimulant-use disorder.
Collapse
Affiliation(s)
- Branka Petković
- Address correspondence to this author at the Department of Neurophysiology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana Blvd. 142, 11060, Belgrade, Serbia; Tel: +381-11-20-78-300; Fax: +381-11-27-61-433; E-mail:
| | | | | |
Collapse
|
41
|
The effects of caffeine on olfactory function and mood: an exploratory study. Psychopharmacology (Berl) 2020; 237:3511-3517. [PMID: 33123819 PMCID: PMC7683449 DOI: 10.1007/s00213-020-05695-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/23/2020] [Indexed: 10/28/2022]
Abstract
Caffeine has been demonstrated to enhance olfactory function in rodents, but to date, the sparse research in humans has not shown any equivalent effects. However, due to the methodological nature of those human studies, a number of questions remain unanswered, which the present study aimed to investigate. Using a double-blind experimental design, participants (n = 40) completed baseline mood measures, standardised threshold and identification tests and were then randomly allocated to receive a capsule containing either 100 mg of caffeine or placebo, followed by the same olfactory tests and mood measures. Results revealed that despite a trend toward elevated arousal following caffeine for habitual caffeine consumers, there were no changes in odour function. In contrast, for non-caffeine consumers, caffeine acted to enhance odour (threshold) sensitivity but reduce odour identification. Overall, these findings demonstrate a complex profile of effects of caffeine on odour function and, given the evidence from the wider caffeine literature, it is proposed that the effects of caffeine might be limited to older populations.
Collapse
|
42
|
Potential new therapies against a toxic relationship: neuroinflammation and Parkinson’s disease. Behav Pharmacol 2019; 30:676-688. [DOI: 10.1097/fbp.0000000000000512] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
43
|
Perkins AE, Varlinskaya EI, Deak T. From adolescence to late aging: A comprehensive review of social behavior, alcohol, and neuroinflammation across the lifespan. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:231-303. [PMID: 31733665 DOI: 10.1016/bs.irn.2019.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The passage of time dictates the pace at which humans and other organisms age but falls short of providing a complete portrait of how environmental, lifestyle and underlying biological processes contribute to senescence. Two fundamental features of the human experience that change dramatically across the lifespan include social interactions and, for many, patterns of alcohol consumption. Rodent models show great utility for understanding complex interactions among aging, social behavior and alcohol use and abuse, yet little is known about the neural changes in late aging that contribute to the natural decline in social behavior. Here, we posit that aging-related neuroinflammation contributes to the insipid loss of social motivation across the lifespan, an effect that is exacerbated by patterns of repeated alcohol consumption observed in many individuals. We provide a comprehensive review of (i) neural substrates crucial for the expression of social behavior under non-pathological conditions; (ii) unique developmental/lifespan vulnerabilities that may contribute to the divergent effects of low-and high-dose alcohol exposure; and (iii) aging-associated changes in neuroinflammation that may sit at the intersection between social processes and alcohol exposure. In doing so, we provide an overview of correspondence between lifespan/developmental periods between common rodent models and humans, give careful consideration to model systems used to aptly probe social behavior, identify points of coherence between human and animal models, and point toward a multitude of unresolved issues that should be addressed in future studies. Together, the combination of low-dose and high-dose alcohol effects serve to disrupt the normal development and maintenance of social relationships, which are critical for both healthy aging and quality of life across the lifespan. Thus, a more complete understanding of neural systems-including neuroinflammatory processes-which contribute to alcohol-induced changes in social behavior will provide novel opportunities and targets for promoting healthy aging.
Collapse
Affiliation(s)
- Amy E Perkins
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Elena I Varlinskaya
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States.
| |
Collapse
|
44
|
Han YY, Chen ZH, Shang YJ, Yan WW, Wu BY, Li CH. Cordycepin improves behavioral-LTP and dendritic structure in hippocampal CA1 area of rats. J Neurochem 2019; 151:79-90. [PMID: 31314908 DOI: 10.1111/jnc.14826] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022]
Abstract
Cordycepin, an adenosine analog, has been reported to improve cognitive function, but which seems to be inconsistent with the reports showing that cordycepin inhibited long-term potentiation (LTP). Behavioral-LTP is usually used to study long-term synaptic plasticity induced by learning tasks in freely moving animals. In order to investigate simultaneously the effects of cordycepin on LTP and behavior in rats, we applied the model of behavioral-LTP induced by Y-maze learning task through recording population spikes in hippocampal CA1 region. Golgi staining and Sholl analysis were employed to assess the morphological structure of dendrites in pyramidal cells of hippocampal CA1 area, and western blotting was used to examine the level of adenosine A1 receptors and A2A receptors (A2AR). We found that cordycepin significantly improved behavioral-LTP magnitude, accompanied by increases in the total length of dendrites, the number of intersections and spine density but did not affect Y-maze learning task. Furthermore, cordycepin obviously reduced A2AR level without altering adenosine A1 receptors level; and the agonist of A2AR (CGS 21680) rather than antagonist (SCH 58261) could reverse the potentiation of behavioral-LTP induced by cordycepin. These results suggested that cordycepin improved behavioral-LTP and morphological structure of dendrite in hippocampal CA1 but did not contribute to the improvement of learning and memory. And cordycepin improved behavioral-LTP may be through reducing the level of A2AR in hippocampus. Collectively, the effects of cordycepin on cognitive function and LTP were complex and involved multiple mechanisms.
Collapse
Affiliation(s)
- Yuan-Yuan Han
- School of Life Science, South China Normal University, Guangzhou, China
| | - Zhao-Hui Chen
- School of Life Science, South China Normal University, Guangzhou, China
| | - Ying-Jie Shang
- School of Life Science, South China Normal University, Guangzhou, China
| | - Wen-Wen Yan
- School of Life Science, South China Normal University, Guangzhou, China
| | - Bao-Yan Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Chu-Hua Li
- School of Life Science, South China Normal University, Guangzhou, China
| |
Collapse
|
45
|
Samal J, Rebelo AL, Pandit A. A window into the brain: Tools to assess pre-clinical efficacy of biomaterials-based therapies on central nervous system disorders. Adv Drug Deliv Rev 2019; 148:68-145. [PMID: 30710594 DOI: 10.1016/j.addr.2019.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/04/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
Therapeutic conveyance into the brain is a cardinal requirement for treatment of diverse central nervous system (CNS) disorders and associated pathophysiology. Effectual shielding of the brain by the blood-brain barrier (BBB) sieves out major proportion of therapeutics with the exception of small lipophilic molecules. Various nano-delivery systems (NDS) provide an effective solution around this obstacle owing to their small size and targeting properties. To date, these systems have been used for several pre-clinical disease models including glioma, neurodegenerative diseases and psychotic disorders. An efficacy screen for these systems involves a test battery designed to probe into the multiple facets of therapeutic delivery. Despite their wide application in redressing various disease targets, the efficacy evaluation strategies for all can be broadly grouped into four modalities, namely: histological, bio-imaging, molecular and behavioural. This review presents a comprehensive insight into all of these modalities along with their strengths and weaknesses as well as perspectives on an ideal design for a panel of tests to screen brain nano-delivery systems.
Collapse
Affiliation(s)
- Juhi Samal
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Ana Lucia Rebelo
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
46
|
Lopes JP, Pliássova A, Cunha RA. The physiological effects of caffeine on synaptic transmission and plasticity in the mouse hippocampus selectively depend on adenosine A 1 and A 2A receptors. Biochem Pharmacol 2019; 166:313-321. [PMID: 31199895 DOI: 10.1016/j.bcp.2019.06.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/07/2019] [Indexed: 12/25/2022]
Abstract
Caffeine is the most consumed psychoactive drug worldwide and its intake in moderate amounts prevents neurodegenerative disorders. However, the molecular targets of caffeine to modulate activity in brain circuits are ill-defined. By electrophysiologically recording synaptic transmission and plasticity in Schaffer fibers-CA1 pyramid synapses of mouse hippocampal slices, we characterized the impact of caffeine using a concentration reached in the brain parenchyma upon moderate caffeine consumption. Caffeine (50 µM) facilitated synaptic transmission by 40%, while decreasing paired-pulse facilitation, and also decreased by 35% the amplitude of long-term potentiation (LTP). Clearance of extracellular adenosine with adenosine deaminase (2 U/mL) blunted all the effects of caffeine on synaptic transmission and plasticity. The A1R antagonist DPCPX (100 nM) only eliminated caffeine-induced facilitation of synaptic transmission while not affecting caffeine-induced depression of LTP; conversely, the genetic (using A2AR knockout mice) or the pharmacological blockade (with SCH58261, 50 nM) of A2AR eliminated the effect of caffeine on LTP while not affecting caffeine-induced facilitation of synaptic transmission. Finally, blockade of GABAA or of ryanodine receptors with bicuculline (10 μM) or dantrolene (10 μM), respectively, did not affect the ability of caffeine to alter synaptic transmission or plasticity. These results show that the effects of caffeine on synaptic transmission and plasticity in the hippocampus are selectively mediated by antagonizing adenosine receptors, where A1R are responsible for the impact of caffeine on synaptic transmission and A2AR regulate the impact of caffeine on LTP.
Collapse
Affiliation(s)
- João P Lopes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal.
| | - Anna Pliássova
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
47
|
Sampaio TB, Soares de Souza B, Roversi K, Schuh T, Poli A, Takahashi RN, Prediger RD. Temporal development of behavioral impairments in rats following locus coeruleus lesion induced by 6-hydroxydopamine: Involvement of β 3-adrenergic receptors. Neuropharmacology 2019; 151:98-111. [PMID: 30959019 DOI: 10.1016/j.neuropharm.2019.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 01/17/2023]
Abstract
Noradrenergic degeneration in the locus coeruleus (LC) seems a convergent neuropathological marker of different neurodegenerative diseases. Herein, we investigated the temporal development of apoptotic signaling activation in the LC, noradrenergic dysfunction and behavioral impairments in rats following the noradrenergic lesion of the LC. For this purpose, the dopamine reuptake inhibitor nomifensine was administered 1 h before the stereotaxic bilateral injections of 6-hydroxydopamine (6-OHDA; 5, 10 or 20 μg/hem) into the LC. The behavioral and neurochemical analyses were performed at 7, 21 and 42 days after 6-OHDA injections. All doses of 6-OHDA induced neuronal death in LC, but only the highest dose (20 μg/hem) disrupted the motor function. 6-OHDA (5 μg/hem) injection induced short-term memory deficits in all periods, olfactory discrimination and long-term memory impairments at 7 days, and depressive-like behaviors at 21 and 42 days after injection. Moreover, 6-OHDA infusion increased Bax/Bcl2 ratio and caspase 3 levels, and decreased the dopamine β-hydroxylase immunocontent in the LC. Noradrenergic neurotransmission dysfunction was observed in the LC, olfactory bulb, prefrontal cortex, hippocampus and striatum. The intranasal (i.n.) noradrenaline (NA) infusion restored the impairments in the olfactory discrimination, short-term memory and depressive-like behavior of 6-OHDA-lesioned rats. In addition, these effects were blocked by the prior i.n. infusion of the β3-adrenergic receptor antagonist SR59230A. These findings indicate that the 6-OHDA injection into the LC induced the apoptosis signaling activation, noradrenergic neurotransmission dysfunction and behavioral impairments that were restored via β3-adrenergic receptors activation mediated by the i.n. NA administration.
Collapse
Affiliation(s)
- Tuane Bazanella Sampaio
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Bruna Soares de Souza
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Katiane Roversi
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Tayná Schuh
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Anicleto Poli
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Reinaldo Naoto Takahashi
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Rui Daniel Prediger
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil.
| |
Collapse
|
48
|
Boyer F, Jaouen F, Ibrahim EC, Gascon E. Deficits in Social Behavior Precede Cognitive Decline in Middle-Aged Mice. Front Behav Neurosci 2019; 13:55. [PMID: 30971905 PMCID: PMC6445840 DOI: 10.3389/fnbeh.2019.00055] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/06/2019] [Indexed: 11/13/2022] Open
Abstract
An extensive literature details deterioration of multiple brain functions, especially memory and learning, during aging in humans and in rodents. In contrast, the decline of social functions is less well understood. It is presently not clear whether age-dependent deficits observed in social behavior mainly reflect the disruption of social networks activity or are simply secondary to a more general impairment of cognitive and executive functions in older individuals. To address this issue, we carried out a battery of behavioral tasks exploring different brain functions in young (3 months) and middle-aged wild-type mice (9 months). Consistent with previous reports, our results show no obvious differences between these two groups in most of the domains investigated including learning and memory. Surprisingly, in social tasks, middle-aged animals showed significantly reduced levels of interactions when exposed to a new juvenile mouse. In the absence of overt cognitive decline, our findings suggest that social impairments may precede the disruption of other brain functions and argue for a selective vulnerability of social circuits during aging.
Collapse
Affiliation(s)
- Flora Boyer
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), Marseille, France
| | - Florence Jaouen
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), Marseille, France
| | - El Chérif Ibrahim
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), Marseille, France
| | - Eduardo Gascon
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), Marseille, France
| |
Collapse
|
49
|
Han K, Lee J, Choi BY, Jeong H, Cho JH, Kim JK. Does Improved Attention Induced by Caffeine Intake Affect Olfactory Function? Clin Exp Otorhinolaryngol 2019; 13:52-57. [PMID: 30813710 PMCID: PMC7010495 DOI: 10.21053/ceo.2018.01424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/20/2018] [Indexed: 11/27/2022] Open
Abstract
Objectives Past several studies have proven that caffeine facilitates attentional enhancement by acting as an adenosine antagonist once it is absorbed by the body, resulting in improved psycho-behavioral function. Modern clinical olfactory function tests are usually assessed by psychophysical tests but due to a paucity of data, the influence of enhanced attention by caffeine on olfactory function still remains unclear. The objective of this study was to compare results of cognitive function (attention) and olfactory function before and after caffeine administration in order to analyze effects of caffeine on olfactory function in normosmic subjects. Methods This study enrolled 49 participants of Konkuk University Hospital with a mean age of 27.7 years who had patent olfactory clefts and no olfactory dysfunction from May 2015 to February 2016. Subjects were restrained from caffeine 10 hours before the test. On day 1, participant’s subjective olfactory function was evaluated before and after uptake of either caffeinated or decaffeinated coffee using visual analog scale (VAS) score, minimum cross-sectional area (MCA) measured by acoustic rhinometry, and the Korean version of Sniffin’ Stick II (KVSS II). Evaluation of participant’s attentional degree was measured by d2 test. On day 2, the same procedure was carried out with counterpart substance. The type of coffee initially administrated was randomly selected. Results After administration, caffeinated coffee resulted in significant attentional enhancement than decaffeinated coffee. Results of d2 test showed statistically significant differences in the parameters of total number of errors and omission errors. In both the caffeinated and decaffeinated groups, the patients showed slight increase in VAS score and nasal cavity area; however, the difference was not statistically significant. Also, caffeinated coffee intake compared to decaffeinated coffee intake showed no significant relevance to olfactory function. Conclusion Caffeine may significantly improve attentional congnitive function, while not have acute effects on olfactory function.
Collapse
Affiliation(s)
- Kyujin Han
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University School of Medicine, Seoul, Korea
| | - Jiyeon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University School of Medicine, Seoul, Korea
| | - Bo Yoon Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University School of Medicine, Seoul, Korea
| | - Hamin Jeong
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University School of Medicine, Seoul, Korea
| | - Jae Hoon Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University School of Medicine, Seoul, Korea
| | - Jin Kook Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
50
|
Caffeine improves contrast sensitivity of freely moving rats. Physiol Behav 2019; 199:111-117. [DOI: 10.1016/j.physbeh.2018.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 11/06/2018] [Accepted: 11/12/2018] [Indexed: 11/22/2022]
|