1
|
Mostafa F, Mantawy EM, Said RS, Azab SS, El-Demerdash E. Captopril attenuates oxidative stress and neuroinflammation implicated in cisplatin-induced cognitive deficits in rats. Psychopharmacology (Berl) 2025:10.1007/s00213-024-06706-6. [PMID: 39809925 DOI: 10.1007/s00213-024-06706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/18/2024] [Indexed: 01/16/2025]
Abstract
RATIONALE One of the most debilitating drawbacks of cisplatin chemotherapy is neurotoxicity which elicits memory impairment and cognitive dysfunction (chemobrain). This is primarily triggered by oxidative stress and inflammation. Captopril, an angiotensin-converting enzyme inhibitor, has been reported as a neuroprotective agent owing to its antioxidant and anti-inflammatory effects. OBJECTIVE We examined the possible neuroprotective effect of captopril against cisplatin-induced neurological and behavioral abnormalities in rats. METHODS Chemobrain was induced in rats by cisplatin (5 mg/kg, i.p.) on the 7th and 14th days of the study while captopril was administered orally (25 mg/kg) daily for three weeks. The effects of captopril were assessed by performing behavioral tests, histological examination, and evaluation of oxidative stress and inflammatory markers. RESULTS Cisplatin caused learning/memory dysfunction assessed by passive avoidance and Y-maze tests, decline in locomotion, and rotarod motor balance loss which were further verified by neurodegeneration observed in histological examination. Also, cisplatin aggravated oxidative stress by elevating lipid peroxidation (MDA) levels and diminishing catalase activity. Moreover, cisplatin upregulated the neuroinflammatory markers (TNF, IL-6, GFAP, and NF-κB). Captopril successfully ameliorated cisplatin damage on the levels of neurobehavioral and histopathological changes. Mechanistically, captopril significantly diminished MDA production and preserved catalase antioxidant activity. Captopril also counteracted neuroinflammation through inhibiting NF-κB and its downstream proinflammatory cytokines besides repressing astrocyte activity by reducing GFAP expression. CONCLUSION Our findings revealed that captopril could abrogate cisplatin neurotoxicity via reducing oxidative stress and neuroinflammation thus enhancing cognitive and behavioral performance. This could suggest the repurposing of captopril as a neuroprotective agent, especially in hypertensive cancer patients receiving cisplatin.
Collapse
Affiliation(s)
- Fatma Mostafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Riham S Said
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
2
|
Lana D, Ugolini F, Iovino L, Attorre S, Giovannini MG. Astrocytes phenomics as new druggable targets in healthy aging and Alzheimer's disease progression. Front Cell Neurosci 2025; 18:1512985. [PMID: 39835288 PMCID: PMC11743640 DOI: 10.3389/fncel.2024.1512985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
For over a century after their discovery astrocytes were regarded merely as cells located among other brain cells to hold and give support to neurons. Astrocytes activation, "astrocytosis" or A1 functional state, was considered a detrimental mechanism against neuronal survival. Recently, the scientific view on astrocytes has changed. Accumulating evidence indicate that astrocytes are not homogeneous, but rather encompass heterogeneous subpopulations of cells that differ from each other in terms of transcriptomics, molecular signature, function and response in physiological and pathological conditions. In this review, we report and discuss the recent literature on the phenomic differences of astrocytes in health and their modifications in disease conditions, focusing mainly on the hippocampus, a region involved in learning and memory encoding, in the age-related memory impairments, and in Alzheimer's disease (AD) dementia. The morphological and functional heterogeneity of astrocytes in different brain regions may be related to their different housekeeping functions. Astrocytes that express diverse transcriptomics and phenomics are present in strictly correlated brain regions and they are likely responsible for interactions essential for the formation of the specialized neural circuits that drive complex behaviors. In the contiguous and interconnected hippocampal areas CA1 and CA3, astrocytes show different, finely regulated, and region-specific heterogeneity. Heterogeneous astrocytes have specific activities in the healthy brain, and respond differently to physiological or pathological stimuli, such as inflammaging present in normal brain aging or beta-amyloid-dependent neuroinflammation typical of AD. To become reactive, astrocytes undergo transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. Alterations of astrocytes affect the neurovascular unit, the blood-brain barrier and reverberate to other brain cell populations, favoring or dysregulating their activities. It will be of great interest to understand whether the differential phenomics of astrocytes in health and disease can explain the diverse vulnerability of the hippocampal areas to aging or to different damaging insults, in order to find new astrocyte-targeted therapies that might prevent or treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Ludovica Iovino
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Selene Attorre
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
3
|
Dettori I, Bulli I, Venturini M, Magni G, Cherchi F, Rossi F, Lee H, Pedata F, Jacobson KA, Pugliese AM, Coppi E. MRS3997, a dual adenosine A 2A/A 2B receptor agonist, reduces brain ischemic damage and alleviates neuroinflammation in rats. Neuropharmacology 2025; 262:110214. [PMID: 39522676 PMCID: PMC11789432 DOI: 10.1016/j.neuropharm.2024.110214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The endogenous neuromodulator adenosine is massively released during hypoxic/ischemic insults and differentially modulates post-ischemic damage depending on the expression and recruitment of its four metabotropic receptor subtypes, namely A1, A2A, A2B and A3 receptors (A1Rs, A2ARs, A2BRs and A3Rs). We previously demonstrated, by using a model of transient middle cerebral artery occlusion (tMCAo) in rats, that selective activation of A2ARs, as well as A2BRs, ameliorates post-ischemic brain damage in contrast to neuroinflammation. In the present study, we investigated whether the multitarget nucleoside MRS3997, a full agonist at both A2ARs and A2BRs, would afford higher neuroprotection in post-ischemic damage. Chronic systemic treatment with MRS3997 reduced neurological deficit, body weight loss and infarct volume in the cortex and striatum measured 7 days after ischemia. The dual agonist counteracted neuronal loss, reduced myelin damage, and prevented morphological changes indicative of microglia and astrocyte activation. Finally, MRS3997 shifted plasma cytokine levels to an anti-inflammatory profile. These effects were preceded, at 2 days after the insult, by a reduced granulocyte infiltration in the ischemic cortex and, differently from what was observed with selective A2AR or A2BR agonism, also in striatum. In summary, we demonstrate here that MRS3997, systemically administered for 7 days after tMCAO, protects ischemic areas from neuronal and glial damage and inhibits neuroinflammation, therefore representing an attractive strategy to ameliorate post-stroke damage and neurological symptoms.
Collapse
Affiliation(s)
- Ilaria Dettori
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy.
| | - Irene Bulli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Martina Venturini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Giada Magni
- Institute of Applied Physics "Nello Carrara", National Research Council (IFAC-CNR), Sesto Fiorentino, Florence, Italy
| | - Federica Cherchi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Francesca Rossi
- Institute of Applied Physics "Nello Carrara", National Research Council (IFAC-CNR), Sesto Fiorentino, Florence, Italy
| | - Hobin Lee
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabe-tes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Felicita Pedata
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabe-tes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anna Maria Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy.
| | - Elisabetta Coppi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| |
Collapse
|
4
|
Afanvi HM, Esperance Broalet MY, Niemtiah O, James Y, Doukoure B, Tako NA, Metowogo K, Eklu-Gadegbeku K, Aklikokou K. Age-Related Cognitive and Volumetric Changes in the Brain of African Grasscutter ( Thryonomys swinderianus (Temminck, 1827)). Vet Med Int 2024; 2024:3349981. [PMID: 39712529 PMCID: PMC11661872 DOI: 10.1155/vmi/3349981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 12/24/2024] Open
Abstract
The African grasscutter (AGC) (Thryonomys swinderianus) is the second largest rodent in sub-Saharan Africa. It is bred for its organoleptic and culinary properties but also serves as a research model. The aim of this study was to investigate the relationship between age-related changes in brain weight, brain volume, and spatial and nonspatial memory performance in the AGC. A total of forty-two (42) captive-derived AGCs were divided into seven age groups: 6 neonates (6 days), 6 juveniles (1 month), 6 pubertals (3 months), 6 subadults (8 months), 6 young adults (2 years), 6 middle adults (4 years), and 6 old adults (5 years). The subjects were given a series of tests to assess their spatial memory (location test) and their nonspatial memory (object recognition test). Brain samples were then collected using basic neuroanatomical techniques. The weight and volume of the brain samples were determined and their encephalization quotient (EQ) was also calculated. The results showed that spatial and nonspatial memory in AGC develops into adulthood and then declines with age. Mean brain weight increased from neonates to mid-adulthood (5.20 ± 0.31 g-11.76 ± 0.23 g) and decreased in older AGC (11.75 ± 0.22 g). In contrast, the mean weight of the olfactory bulb (OB) increased from neonates to young adults (0.28 ± 0.02 g-0.80 ± 0.03 g) and the cerebellum increased from neonates to older (0.59 ± 0 0.01 g-1.86 ± 0.06 g). Finally, the EQ decreased with age (0.73 ± 0.05-0.29 ± 0.01). Mean brain volume increased with age from neonates to young adult (5 mL-11.25 mL). Conversely, the mean volume of the OB and cerebellum increases from neonates to older age (0.33 ± 0.03 mL-0.95 ± 0.04 mL). This study shows that spatial and nonspatial memory declines during the aging process in AGC. Neonates and juveniles have higher cognitive abilities than adults and older AGC. The weight of the brain, OB, and cerebellum increases from neonates to adult and decreases slightly from middle to old adults. However, the volume of the OB and cerebellum increases with age.
Collapse
Affiliation(s)
- Hounakey M. Afanvi
- Laboratory of Biology and Health, Neuroscience Unit, Felix Houphouët-Boigny University of Abidjan, BP V34, Abidjan 01, Ivory Coast
| | - Maman You Esperance Broalet
- Anatomy Laboratory, Medical Science Training and Research Unit, Alassane Ouattara University of Bouake, BP V 18 01, Bouake, Ivory Coast
| | - Ouattara Niemtiah
- Laboratory of Biology and Health, Neuroscience Unit, Felix Houphouët-Boigny University of Abidjan, BP V34, Abidjan 01, Ivory Coast
| | - Yaovi James
- Anatomy Laboratory, Faculty of Health Sciences, University of Lome, 01BP: 1515, Lome, Togo
| | - Brahima Doukoure
- Pathological Anatomy and Cytology Laboratory, Training and Research Unit-Medical Sciences, Felix Houphouët-Boigny University of Abidjan, BP V34, Abidjan 01, Ivory Coast
| | - Neme Antoine Tako
- Laboratory of Biology and Health, Neuroscience Unit, Felix Houphouët-Boigny University of Abidjan, BP V34, Abidjan 01, Ivory Coast
| | - Kossi Metowogo
- Physiology-Pharmacology Laboratory, Physiopathology Bioactive Substances and Safety Research Unit, University of Lome, 01BP: 1515, Lome, Togo
| | - Kwashi Eklu-Gadegbeku
- Physiology-Pharmacology Laboratory, Physiopathology Bioactive Substances and Safety Research Unit, University of Lome, 01BP: 1515, Lome, Togo
| | - Kodjo Aklikokou
- Physiology-Pharmacology Laboratory, Physiopathology Bioactive Substances and Safety Research Unit, University of Lome, 01BP: 1515, Lome, Togo
| |
Collapse
|
5
|
Nairuz T, Heo JC, Lee JH. Differential Glial Response and Neurodegenerative Patterns in CA1, CA3, and DG Hippocampal Regions of 5XFAD Mice. Int J Mol Sci 2024; 25:12156. [PMID: 39596222 PMCID: PMC11594373 DOI: 10.3390/ijms252212156] [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: 10/14/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
In this study, the distinct patterns of glial response and neurodegeneration within the CA1, CA3, and dentate gyrus (DG) regions of the hippocampus were examined in 5XFAD mice at 6 and 12 months of age. The primary feature of this transgenic mouse model is the rapid onset of amyloid pathology. We employed quantitative assessments via immunohistochemistry, incorporating double staining techniques, followed by observation with light microscopy and subsequent digital analysis of microscopic images. We identified significantly increased Aβ deposition in these three hippocampal regions at 6 and 12 months of transgenic mice. Moreover, the CA1 and CA3 regions showed higher vulnerability, with signs of reactive astrogliosis such as increased astrocyte density and elevated GFAP expression. Additionally, we observed a significant rise in microglia density, along with elevated inflammatory markers (TNFα) in these hippocampal regions. These findings highlight a non-uniform glial and neuronal response to Aβ plaque deposition within the hippocampal regions of 5xFAD mice, potentially contributing to the neurodegenerative and memory deficit characteristics of Alzheimer's disease in this model.
Collapse
Affiliation(s)
| | | | - Jong-Ha Lee
- Department of Biomedical Engineering, Keimyung University, Daegu 42601, Republic of Korea; (T.N.); (J.-C.H.)
| |
Collapse
|
6
|
Min-Kaung-Wint-Mon, Kida H, Kanehisa I, Kurose M, Ishikawa J, Sakimoto Y, Paw-Min-Thein-Oo, Kimura R, Mitsushima D. Adverse Effects of Aβ 1-42 Oligomers: Impaired Contextual Memory and Altered Intrinsic Properties of CA1 Pyramidal Neurons. Biomolecules 2024; 14:1425. [PMID: 39595601 PMCID: PMC11591707 DOI: 10.3390/biom14111425] [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: 09/26/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Aβ1-42 (amyloid beta) oligomers, the major neurotoxic culprits in Alzheimer's disease, initiate early pathophysiological events, including neuronal hyperactivity, that underlie aberrant network activity and cognitive impairment. Although several synaptotoxic effects have been extensively studied, neuronal hyperexcitability, which may also contribute to cognitive deficits, is not fully understood. Here, we found several adverse effects of in vivo injection of Aβ1-42 oligomers on contextual memory and intrinsic properties of CA1 pyramidal neurons. Male rats underwent behavioral and electrophysiological studies 1 week after microinjections into the dorsal CA1 region, followed by histological analysis. After 1 week, Aβ1-42 oligomers impaired contextual learning without affecting basic physiological functions and triggered training-induced neuronal excitability. Furthermore, riluzole, a persistent sodium current (INaP) blocker, dose-dependently reduced Aβ1-42 oligomer-induced hyperexcitability. Congo red staining, which detects insoluble amyloid deposits, further identified labeling of CA1 pyramidal neurons while immunohistochemistry with lecanemab, which detects soluble Aβ oligomers, revealed immunoreactivity of both pyramidal and non-pyramidal cells in the target area. Therefore, our study suggests that a single injection of Aβ1-42 oligomers resulted in contextual memory deficits along with concomitant neuronal hyperexcitability and amyloid deposition in the CA1 region after 1 week.
Collapse
Affiliation(s)
- Min-Kaung-Wint-Mon
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; (M.-K.-W.-M.); (H.K.); (I.K.); (M.K.); (J.I.); (Y.S.); (P.-M.-T.-O.)
| | - Hiroyuki Kida
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; (M.-K.-W.-M.); (H.K.); (I.K.); (M.K.); (J.I.); (Y.S.); (P.-M.-T.-O.)
| | - Itsuki Kanehisa
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; (M.-K.-W.-M.); (H.K.); (I.K.); (M.K.); (J.I.); (Y.S.); (P.-M.-T.-O.)
| | - Masahiko Kurose
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; (M.-K.-W.-M.); (H.K.); (I.K.); (M.K.); (J.I.); (Y.S.); (P.-M.-T.-O.)
| | - Junko Ishikawa
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; (M.-K.-W.-M.); (H.K.); (I.K.); (M.K.); (J.I.); (Y.S.); (P.-M.-T.-O.)
| | - Yuya Sakimoto
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; (M.-K.-W.-M.); (H.K.); (I.K.); (M.K.); (J.I.); (Y.S.); (P.-M.-T.-O.)
| | - Paw-Min-Thein-Oo
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; (M.-K.-W.-M.); (H.K.); (I.K.); (M.K.); (J.I.); (Y.S.); (P.-M.-T.-O.)
| | - Ryoichi Kimura
- Center for Liberal Arts and Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0884, Yamaguchi, Japan;
| | - Dai Mitsushima
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; (M.-K.-W.-M.); (H.K.); (I.K.); (M.K.); (J.I.); (Y.S.); (P.-M.-T.-O.)
- The Research Institute for Time Studies, Yamaguchi University, Yamaguchi 753-8511, Japan
| |
Collapse
|
7
|
Cooper JJM, Muthaiah R, Frost JR, Buck GT, Ravichandar R, Gadelkarim F, Raymond FD, Sim FJ. Clemastine Induces Oligodendrocyte Progenitor Pool Exhaustion and Senescence in the Context of Chronic Demyelination in a Rabbit Model. Ann Neurol 2024. [PMID: 39422297 DOI: 10.1002/ana.27098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 08/08/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES Clemastine has emerged as a promising therapy for the restoration of neurologic function in patients with multiple sclerosis (MS). However, clemastine and other agents with prodifferentiative effects on oligodendrocyte progenitor cells (OPCs) in rodent models have underperformed in clinical trials. We hypothesized that the preclinical studies showed more robust effects because of the abundance of OPCs in rodent models. To better examine the therapeutic potential of clemastine, we examined its effect on demyelinated white matter lesions in rabbits, which exhibit progenitor densities and limited remyelination more closely matching those found in tissues from patients with MS. METHODS We used lysolecithin to induce demyelination in white matter of New Zealand rabbits and then administered oral clemastine (10mg/kg/day) for various periods before assessing the OPC and oligodendrocyte (OL) populations in these lesions. RESULTS Daily administration of clemastine for the full study period (56 days) increased oligodendrogenesis in white matter lesions. However, shorter durations of treatment failed to increase overall OL density despite enhancing OPC-to-OL differentiation. This effect was due to exhaustion of the OPC pool, as the differentiating progenitors were not replaced because of reduced OPC proliferation. Notably, delayed administration of clemastine led to an accumulation of activated OPCs expressing markers of senescence. INTERPRETATION Although capable of driving OL differentiation, clemastine treatment in rabbits hampered progenitor pool replenishment, induced senescence, and promoted conversion of microglia/macrophages to a proinflammatory phenotype. Whether these effects would also occur in humans or with other prodifferentiative therapies should be studied further, but our data suggest the need to carefully consider progenitor dynamics in the treatment of MS. ANN NEUROL 2024.
Collapse
Affiliation(s)
- James J M Cooper
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Rupadevi Muthaiah
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Jon R Frost
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Gregory T Buck
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Roopa Ravichandar
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Farah Gadelkarim
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Faye D Raymond
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Fraser J Sim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
8
|
Lana D, Traini C, Bulli I, Sarti G, Magni G, Attorre S, Giovannini MG, Vannucchi MG. Chronic administration of prebiotics and probiotics ameliorates pathophysiological hallmarks of Alzheimer's disease in a APP/PS1 transgenic mouse model. Front Pharmacol 2024; 15:1451114. [PMID: 39166107 PMCID: PMC11333230 DOI: 10.3389/fphar.2024.1451114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
Introduction: The gut microbiota (MB), although one of the main producers of Aβ in the body, in physiological conditions contributes to the maintainance of a healthy brain. Dysbiosis, the dysbalance between Gram-negative and Gram-positive bacteria in the MB increases Aβ production, contributing to the accumulation of Aβ plaques in the brain, the main histopathological hallmark of Alzheimer's disease (AD). Administration of prebiotics and probiotics, maintaining or recovering gut-MB composition, could represent a nutraceutical strategy to prevent or reduce AD sympthomathology. Aim of this research was to evaluate whether treatment with pre- and probiotics could modify the histopathological signs of neurodegeneration in hippocampal CA1 and CA3 areas of a transgenic mouse model of AD (APP/PS1 mice). The hippocampus is one of the brain regions involved in AD. Methods: Tg mice and Wt littermates (Wt-T and Tg-T) were fed daily for 6 months from 2 months of age with a diet supplemented with prebiotics (a multi-extract of fibers and plant complexes, containing inulin/fruit-oligosaccharides) and probiotics (a 50%-50% mixture of Lactobacillus rhamnosus and Lactobacillus paracasei). Controls were Wt and Tg mice fed with a standard diet. Brain sections were immunostained for Aβ plaques, neurons, astrocytes, microglia, and inflammatory proteins that were evaluated qualitatively and quantitatively by immunofluorescence, confocal microscopy and digital imaging with ImageJ software. Results: Quantitative analyses demonstrated that: 1) The treatment with pre- and probiotics significantly decreased Aβ plaques in CA3, while in CA1 the reduction was not significant; 2) Neuronal damage in CA1 Stratum Pyramidalis was significantly prevented in Tg-T mice; no damage was found in CA3; 3) In both CA1 and CA3 the treatment significantly increased astrocytes density, and GFAP and IBA1 expression, especially around plaques; 4) Microglia reacted differently in CA1 and CA3: in CA3 of Tg-T mice there was a significant increase of CD68+ phagocytic microglia (ball-and-chain phenomic) and of CX3CR1 compared with CA1. Discussion: The higher microglia reactivity could be responsible for their more efficient scavenging activity towards Aβ plaques in CA3 in comparison to CA1. Treatment with pre- and probiotics, modifying many of the physiopathological hallmarks of AD, could be considered an effective nutraceutical strategy against AD symptomatology.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Chiara Traini
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Irene Bulli
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giorgia Sarti
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giada Magni
- Cnr — Istituto di Fisica Applicata “Nello Carrara”, Sesto Fiorentino, Italy
| | - Selene Attorre
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Giuliana Vannucchi
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
9
|
Man JHK, Breur M, van Gelder CAGH, Marcon G, Maderna E, Giaccone G, Altelaar M, van der Knaap MS, Bugiani M. Region-specific and age-related differences in astrocytes in the human brain. Neurobiol Aging 2024; 140:102-115. [PMID: 38763075 DOI: 10.1016/j.neurobiolaging.2024.02.016] [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: 08/07/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 05/21/2024]
Abstract
Astrocyte heterogeneity and its relation to aging in the normal human brain remain poorly understood. We here analyzed astrocytes in gray and white matter brain tissues obtained from donors ranging in age between the neonatal period to over 100 years. We show that astrocytes are differently distributed with higher density in the white matter. This regional difference in cellular density becomes less prominent with age. Additionally, we confirm the presence of morphologically distinct astrocytes, with gray matter astrocytes being morphologically more complex. Notably, gray matter astrocytes morphologically change with age, while white matter astrocytes remain relatively consistent in morphology. Using regional mass spectrometry-based proteomics, we did, however, identify astrocyte specific proteins with regional differences in abundance, reflecting variation in cellular density or expression level. Importantly, the expression of some astrocyte specific proteins region-dependently decreases with age. Taken together, we provide insights into region- and age-related differences in astrocytes in the human brain.
Collapse
Affiliation(s)
- Jodie H K Man
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Marjolein Breur
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Charlotte A G H van Gelder
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Netherlands Proteomics Center, Utrecht, the Netherlands
| | - Gabriella Marcon
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy; DAME, University of Udine, Udine, Italy
| | - Emanuela Maderna
- Division of Neurology 5 - Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giorgio Giaccone
- Division of Neurology 5 - Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Netherlands Proteomics Center, Utrecht, the Netherlands
| | - Marjo S van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam UMC, Amsterdam, the Netherlands.
| |
Collapse
|
10
|
Gáll Z, Csüdör Á, Sável IG, Kelemen K, Kolcsár M. Cholecalciferol Supplementation Impacts Behavior and Hippocampal Neuroglial Reorganization in Vitamin D-Deficient Rats. Nutrients 2024; 16:2326. [PMID: 39064769 PMCID: PMC11279879 DOI: 10.3390/nu16142326] [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: 07/02/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Vitamin D deficiency (VDD) is widespread around the world and has been extensively documented to affect various health conditions, including the cognitive functioning of the brain. Serum 25-hydroxylated forms of vitamin D are traditionally used to determine vitamin D status. However, there is now evidence that cholecalciferol activation can occur and be controlled by locally expressed enzymes in the brain. This study aimed to investigate the effects of cholecalciferol supplementation on cognitive function in rats who underwent transient VDD in adulthood. Thirty-six adult Wistar rats were administered paricalcitol (seven doses of 32 ng injected every other day) along with a "vitamin D-free" diet to induce VDD, which was confirmed using a LC-MS/MS serum analysis of the cholecalciferol and 25-hydroxyvitamin D3 levels. Treatment was performed by including 1000 IU/kg and 10,000 IU/kg cholecalciferol in the diet. Cognitive performance was evaluated using the novel object recognition (NOR), Morris water maze (MWM), and radial arm maze (RAM) tests. An immunohistochemical analysis of the brain regions involved in learning and memory was performed by quantifying the neurons, astrocytes, and microglia labelled with anti-neuronal nuclei (NeuN), glial fibrillary acidic protein (GFAP), and ionized calcium-binding adaptor molecule 1 (Iba-1) antibodies, respectively. The vitamin D deficient group showed the lowest performance in both the MWM and RAM tests. In contrast, the cholecalciferol-treated groups exhibited a faster learning curve. However, no difference was detected between the groups in the NOR test. On the other hand, differences in the cellular organization of the hippocampus and amygdala were observed between the groups. Cholecalciferol supplementation decreased the density of the Iba-1- and GFAP-labeled cells in the hilus and cornu Ammonis 3 (CA3) regions of the hippocampus and in the amygdala. These results support vitamin D's substantial role in learning and memory. They also highlight that subtle changes of cognitive function induced by transient VDD could be reversed by cholecalciferol supplementation. Further studies are needed to better understand VDD and cholecalciferol's effects on the brain structure and function.
Collapse
Affiliation(s)
- Zsolt Gáll
- Department of Pharmacology and Clinical Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| | - Ágnes Csüdör
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| | - István-Gábor Sável
- Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| | - Krisztina Kelemen
- Department of Physiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| | - Melinda Kolcsár
- Department of Pharmacology and Clinical Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| |
Collapse
|
11
|
Santana NNM, Silva EHA, Santos SFD, Bezerra LLF, da Silva MMO, Cavalcante JS, Fiuza FP, Morais PLADG, Engelberth RC. Neuronal Stability, Volumetric Changes, and Decrease in GFAP Expression of Marmoset (Callithrix jacchus) Subcortical Visual Nuclei During Aging. J Comp Neurol 2024; 532:e25649. [PMID: 38967410 DOI: 10.1002/cne.25649] [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: 12/14/2023] [Revised: 05/06/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024]
Abstract
The physiological aging process is well known for functional decline in visual abilities. Among the components of the visual system, the dorsal lateral geniculate nucleus (DLG) and superior colliculus (SC) provide a good model for aging investigations, as these structures constitute the main visual pathways for retinal inputs reaching the visual cortex. However, there are limited data available on quantitative morphological and neurochemical aspects in DLG and SC across lifespan. Here, we used optical density to determine immunoexpression of glial fibrillary acidic protein (GFAP) and design-based stereological probes to estimate the neuronal number, total volume, and layer volume of the DLG and SC in marmosets (Callithrix jacchus), ranging from 36 to 143 months of age. Our results revealed an age-related increase in total volume and layer volume of the DLG, with an overall stability in SC volume. Furthermore, a stable neuronal number was demonstrated in DLG and superficial layers of SC (SCv). A decrease in GFAP immunoexpression was observed in both visual centers. The results indicate region-specific variability in volumetric parameter, possibly attributed to structural plastic events in response to inflammation and compensatory mechanisms at the cellular and subcellular level. Additionally, the DLG and SCv seem to be less vulnerable to aging effects in terms of neuronal number. The neuropeptidergic data suggest that reduced GFAP expression may reflect morphological atrophy in the astroglial cells. This study contributes to updating the current understanding of aging effects in the visual system and stablishes a crucial foundation for future research on visual perception throughout the aging process.
Collapse
Affiliation(s)
- Nelyane N M Santana
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Eryck H A Silva
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Sâmarah F Dos Santos
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Lyzandro L F Bezerra
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Maria M O da Silva
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Jeferson S Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Felipe P Fiuza
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Paulo L A de G Morais
- Laboratory of Experimental Neurology, College of the Health Sciences, University of the State of Rio Grande do Norte, Mossoró, Brazil
| | - Rovena Clara Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
12
|
Bencze N, Scheich B, Szőke É, Wilhelm I, Körmöndi S, Botz B, Helyes Z. Osteosarcoma-Induced Pain Is Mediated by Glial Cell Activation in the Spinal Dorsal Horn, but Not Capsaicin-Sensitive Nociceptive Neurons: A Complex Functional and Morphological Characterization in Mice. Cancers (Basel) 2024; 16:1788. [PMID: 38791867 PMCID: PMC11120600 DOI: 10.3390/cancers16101788] [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: 02/15/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024] Open
Abstract
Bone cancer and its related chronic pain are huge clinical problems since the available drugs are often ineffective or cannot be used long term due to a broad range of side effects. The mechanisms, mediators and targets need to be identified to determine potential novel therapies. Here, we characterize a mouse bone cancer model induced by intratibial injection of K7M2 osteosarcoma cells using an integrative approach and investigate the role of capsaicin-sensitive peptidergic sensory nerves. The mechanical pain threshold was assessed by dynamic plantar aesthesiometry, limb loading by dynamic weight bearing, spontaneous pain-related behaviors via observation, knee diameter with a digital caliper, and structural changes by micro-CT and glia cell activation by immunohistochemistry in BALB/c mice of both sexes. Capsaicin-sensitive peptidergic sensory neurons were defunctionalized by systemic pretreatment with a high dose of the transient receptor potential vanilloid 1 (TRPV1) agonist resiniferatoxin (RTX). During the 14- and 28-day experiments, weight bearing on the affected limb and the paw mechanonociceptive thresholds significantly decreased, demonstrating secondary mechanical hyperalgesia. Signs of spontaneous pain and osteoplastic bone remodeling were detected both in male and female mice without any sex differences. Microglia activation was shown by the increased ionized calcium-binding adapter molecule 1 (Iba1) immunopositivity on day 14 and astrocyte activation by the enhanced glial fibrillary acidic protein (GFAP)-positive cell density on day 28 in the ipsilateral spinal dorsal horn. Interestingly, defunctionalization of the capsaicin-sensitive afferents representing approximately 2/3 of the nociceptive fibers did not alter any functional parameters. Here, we provide the first complex functional and morphological characterization of the K7M2 mouse osteosarcoma model. Bone-cancer-related chronic pain and hyperalgesia are likely to be mediated by central sensitization involving neuroinflammation via glial cell activation in the spinal dorsal horn, but not the capsaicin-sensitive sensory neuronal system.
Collapse
Affiliation(s)
- Noémi Bencze
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
| | - Bálint Scheich
- Department of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary;
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN-PTE), 7624 Pécs, Hungary
| | - Imola Wilhelm
- Institute of Biophysics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary;
| | - Sándor Körmöndi
- Department of Traumatology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Bálint Botz
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- Department of Medical Imaging, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN-PTE), 7624 Pécs, Hungary
- PharmInVivo Ltd., Szondy György Str. 10, 7629 Pécs, Hungary
| |
Collapse
|
13
|
Horn MD, Forest SC, Saied AA, MacLean AG. Astrocyte expression of aging-associated markers positively correlates with neurodegeneration in the frontal lobe of the rhesus macaque brain. Front Aging Neurosci 2024; 16:1368517. [PMID: 38577492 PMCID: PMC10993697 DOI: 10.3389/fnagi.2024.1368517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/06/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction As the population over the age of 65 increases, rates of neurodegenerative disorders and dementias will rise - necessitating further research into the cellular and molecular mechanisms that contribute to brain aging. With the critical importance of astrocytes to neuronal health and functioning, we hypothesized that alterations in astrocyte expression of aging-associated markers p16INK4a (p16) and sirtuin 1 (SIRT1) with age would correlate with increased rates of neurodegeneration, as measured by FluoroJade C (FJC) staining. Methods To test this hypothesis, 19 rhesus macaques at the Tulane National Primate Research Center were selected based on the following criteria: archival FFPE CNS tissue available to use, no noted neuropathology, and an age range of 5-30 years. Tissues were cut at 5 μm and stained for GFAP, p16, SIRT1, and FJC, followed by whole-slide imaging and HALO® image analysis for percentage of marker-positive cells and relative intensity of each stain. Results We found the percentage of p16+ cells increases with age in total cells and astrocytes of the frontal (p = 0.0021, p = 0.0012 respectively) and temporal (p = 0.0226, p = 0.0203 respectively) lobes, as well as the relative intensity of p16 staining (frontal lobe: p = 0.0060; temporal lobe: p = 0.0269). For SIRT1, we found no correlation with age except for an increase in the relative intensity of SIRT1 in the temporal lobe (p = 0.0033). There was an increase in neurodegeneration, as measured by the percentage of FJC+ cells in the frontal lobe with age (p = 0.0057), as well as in the relative intensity of FJC staining in the frontal (p = 0.0030) and parietal (p = 0.0481) lobes. Importantly, increased p16 and SIRT1 expression in astrocytes correlated with increasing neurodegeneration in the frontal lobe (p = 0.0009, p = 0.0095 respectively). Discussion Together, these data suggest that age-associated alterations in astrocytes contribute to neurodegeneration and provide a target for mechanistic studies in the future.
Collapse
Affiliation(s)
- Miranda D Horn
- Brain Institute, Tulane University, New Orleans, LA, United States
| | | | - Ahmad A Saied
- Tulane National Primate Research Center, Covington, LA, United States
| | - Andrew G MacLean
- Brain Institute, Tulane University, New Orleans, LA, United States
- Tulane National Primate Research Center, Covington, LA, United States
- Tulane Center for Aging, New Orleans, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
14
|
Marques KL, Rodrigues V, Balduci CTN, Montes GC, Barradas PC, Cunha-Rodrigues MC. Emerging therapeutic strategies in hypoxic-ischemic encephalopathy: a focus on cognitive outcomes. Front Pharmacol 2024; 15:1347529. [PMID: 38469401 PMCID: PMC10925695 DOI: 10.3389/fphar.2024.1347529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/30/2024] [Indexed: 03/13/2024] Open
Abstract
Perinatal hypoxia-ischemia represents a significant risk to CNS development, leading to high mortality rates, diverse damages, and persistent neurological deficits. Despite advances in neonatal medicine in recent decades, the incidence of HIE remains substantial. Motor deficits can manifest early, while cognitive impairments may be diagnosed later, emphasizing the need for extended follow-up. This review aims to explore potential candidates for therapeutic interventions for hypoxic-ischemic encephalopathy (HIE), with a focus on cognitive deficits. We searched randomized clinical trials (RCT) that tested drug treatments for HIE and evaluated cognitive outcomes. The results included studies on erythropoietin, melatonin, magnesium sulfate, topiramate, and a combination of vitamin C and ibuprofen. Although there are several indications of the efficacy of these drugs among animal models, considering neuroprotective properties, the RCTs failed to provide complete effectiveness in the context of cognitive impairments derived from HIE. More robust RCTs are still needed to advance our knowledge and to establish standardized treatments for HIE.
Collapse
Affiliation(s)
- Kethely L. Marques
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor Rodrigues
- Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cassiana T. N. Balduci
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Rare Diseases Sales Force, Daiichi Sankyo Brazil, São Paulo, Brazil
| | - Guilherme C. Montes
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Penha C. Barradas
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marta C. Cunha-Rodrigues
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
June A, Matysik W, Marlicz M, Zucker E, Wagley PK, Kuan CY, Burnsed J. Acute seizure activity in neonatal inflammation-sensitized hypoxia-ischemia in mice. PLoS One 2024; 19:e0295860. [PMID: 38206902 PMCID: PMC10783742 DOI: 10.1371/journal.pone.0295860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/30/2023] [Indexed: 01/13/2024] Open
Abstract
OBJECTIVE To examine acute seizure activity and neuronal damage in a neonatal mouse model of inflammation-sensitized hypoxic-ischemic (IS-HI) brain injury utilizing continuous electroencephalography (cEEG) and neurohistology. METHODS Neonatal mice were exposed to either IS-HI with Escherichia coli lipopolysaccharide (LPS) or HI alone on postnatal (p) day 10 using unilateral carotid artery ligation followed by global hypoxia (n = 10 [5 female, 5 male] for IS-HI, n = 12 [5 female, 7 male] for HI alone). Video cEEG was recorded for the duration of the experiment and analyzed for acute seizure activity and behavior. Brain tissue was stained and scored based on the degree of neuronal injury in the hippocampus, cortex, and thalamus. RESULTS There was no significant difference in acute seizure activity among mice exposed to IS-HI compared to HI with regards to seizure duration (mean = 63 ± 6 seconds for HI vs mean 62 ± 5 seconds for IS-HI, p = 0.57) nor EEG background activity. Mice exposed to IS-HI had significantly more severe neural tissue damage at p30 as measured by neuropathologic scores (mean = 8 ± 1 vs 23 ± 3, p < 0.0001). INTERPRETATION In a neonatal mouse model of IS-HI, there was no significant difference in acute seizure activity among mice exposed to IS-HI compared to HI. Mice exposed to IS-HI did show more severe neuropathologic damage at a later age, which may indicate the presence of chronic inflammatory mechanisms of brain injury distinct from acute seizure activity.
Collapse
Affiliation(s)
- Angelina June
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Weronika Matysik
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Maria Marlicz
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Emily Zucker
- College of Arts and Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Pravin K. Wagley
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Chia-Yi Kuan
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jennifer Burnsed
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Neurology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
16
|
Karperien AL, Jelinek HF. Morphology and Fractal-Based Classifications of Neurons and Microglia in Two and Three Dimensions. ADVANCES IN NEUROBIOLOGY 2024; 36:149-172. [PMID: 38468031 DOI: 10.1007/978-3-031-47606-8_7] [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: 03/13/2024]
Abstract
Microglia and neurons live physically intertwined, intimately related structurally and functionally in a dynamic relationship in which microglia change continuously over a much shorter timescale than do neurons. Although microglia may unwind and depart from the neurons they attend under certain circumstances, in general, together both contribute to the fractal topology of the brain that defines its computational capabilities. Both neuronal and microglial morphologies are well-described using fractal analysis complementary to more traditional measures. For neurons, the fractal dimension has proved valuable for classifying dendritic branching and other neuronal features relevant to pathology and development. For microglia, fractal geometry has substantially contributed to classifying functional categories, where, in general, the more pathological the biological status, the lower the fractal dimension for individual cells, with some exceptions, including hyper-ramification. This chapter provides a review of the intimate relationships between neurons and microglia, by introducing 2D and 3D fractal analysis methodology and its applications in neuron-microglia function in health and disease.
Collapse
Affiliation(s)
- Audrey L Karperien
- School of Community Health, Charles Sturt University, Albury, NSW, Australia
| | - Herbert F Jelinek
- Department of Medical Sciences and Biotechnology Center, Khalifa University, Abu Dhabi, UAE
| |
Collapse
|
17
|
Landucci E, Ribaudo G, Anyanwu M, Oselladore E, Giannangeli M, Mazzantini C, Lana D, Giovannini MG, Memo M, Pellegrini-Giampietro DE, Gianoncelli A. Virtual Screening-Accelerated Discovery of a Phosphodiesterase 9 Inhibitor with Neuroprotective Effects in the Kainate Toxicity In Vitro Model. ACS Chem Neurosci 2023; 14:3826-3838. [PMID: 37726213 PMCID: PMC10587872 DOI: 10.1021/acschemneuro.3c00431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023] Open
Abstract
In the central nervous system, some specific phosphodiesterase (PDE) isoforms modulate pathways involved in neuronal plasticity. Accumulating evidence suggests that PDE9 may be a promising therapeutic target for neurodegenerative diseases. In the current study, computational techniques were used to identify a nature-inspired PDE9 inhibitor bearing the scaffold of an isoflavone, starting from a database of synthetic small molecules using a ligand-based approach. Furthermore, docking studies supported by molecular dynamics investigations allowed us to evaluate the features of the ligand-target complex. In vitro assays confirmed the computational results, showing that the selected compound inhibits the enzyme in the nanomolar range. Additionally, we evaluated the expression of gene and protein levels of PDE9 in organotypic hippocampal slices, observing an increase following exposure to kainate (KA). Importantly, the PDE9 inhibitor reduced CA3 damage induced by KA in a dose-dependent manner in organotypic hippocampal slices. Taken together, these observations strongly support the potential of the identified nature-inspired PDE9 inhibitor and suggest that such a molecule could represent a promising lead compound to develop novel therapeutic tools against neurological diseases..
Collapse
Affiliation(s)
- Elisa Landucci
- Department
of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze 50139, Italy
| | - Giovanni Ribaudo
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Margrate Anyanwu
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Erika Oselladore
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Matteo Giannangeli
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Costanza Mazzantini
- Department
of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze 50139, Italy
| | - Daniele Lana
- Department
of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze 50139, Italy
| | - Maria Grazia Giovannini
- Department
of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze 50139, Italy
| | - Maurizio Memo
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | | | - Alessandra Gianoncelli
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| |
Collapse
|
18
|
Lana D, Branca JJV, Delfino G, Giovannini MG, Casamenti F, Nardiello P, Bucciantini M, Stefani M, Zach P, Zecchi-Orlandini S, Nosi D. Morphofunctional Investigation in a Transgenic Mouse Model of Alzheimer's Disease: Non-Reactive Astrocytes Are Involved in Aβ Load and Reactive Astrocytes in Plaque Build-Up. Cells 2023; 12:2258. [PMID: 37759482 PMCID: PMC10526848 DOI: 10.3390/cells12182258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The term neuroinflammation defines the reactions of astrocytes and microglia to alterations in homeostasis in the diseased central nervous system (CNS), the exacerbation of which contributes to the neurodegenerative effects of Alzheimer's disease (AD). Local environmental conditions, such as the presence of proinflammatory molecules, mechanical properties of the extracellular matrix (ECM), and local cell-cell interactions, are determinants of glial cell phenotypes. In AD, the load of the cytotoxic/proinflammatory amyloid β (Aβ) peptide is a microenvironmental component increasingly growing in the CNS, imposing time-evolving challenges on resident cells. This study aimed to investigate the temporal and spatial variations of the effects produced by this process on astrocytes and microglia, either directly or by interfering in their interactions. Ex vivo confocal analyses of hippocampal sections from the mouse model TgCRND8 at different ages have shown that overproduction of Aβ peptide induced early and time-persistent disassembly of functional astroglial syncytium and promoted a senile phenotype of reactive microglia, hindering Aβ clearance. In the late stages of the disease, these patterns were altered in the presence of Aβ-plaques, surrounded by typically reactive astrocytes and microglia. Morphofunctional characterization of peri-plaque gliosis revealed a direct contribution of astrocytes in plaque buildup that might result in shielding Aβ-peptide cytotoxicity and, as a side effect, in exacerbating neuroinflammation.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, University of Florence, 50134 Florence, Italy; (D.L.); (M.G.G.)
| | - Jacopo Junio Valerio Branca
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (J.J.V.B.); (S.Z.-O.)
| | - Giovanni Delfino
- Department of Biology, University of Florence, 50121 Florence, Italy;
| | - Maria Grazia Giovannini
- Department of Health Sciences, University of Florence, 50134 Florence, Italy; (D.L.); (M.G.G.)
| | - Fiorella Casamenti
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50134 Florence, Italy;
| | - Pamela Nardiello
- General Laboratory, Careggi University Hospital, 50134 Florence, Italy;
| | - Monica Bucciantini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (M.B.); (M.S.)
| | - Massimo Stefani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (M.B.); (M.S.)
| | - Petr Zach
- Department of Anatomy, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
| | - Sandra Zecchi-Orlandini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (J.J.V.B.); (S.Z.-O.)
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (J.J.V.B.); (S.Z.-O.)
- DMSC Imaging Platform, 50134 Florence, Italy
| |
Collapse
|
19
|
Lana D, Magni G, Landucci E, Wenk GL, Pellegrini-Giampietro DE, Giovannini MG. Phenomic Microglia Diversity as a Druggable Target in the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13668. [PMID: 37761971 PMCID: PMC10531074 DOI: 10.3390/ijms241813668] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Phenomics, the complexity of microglia phenotypes and their related functions compels the continuous study of microglia in disease animal models to find druggable targets for neurodegenerative disorders. Activation of microglia was long considered detrimental for neuron survival, but more recently it has become apparent that the real scenario of microglia morphofunctional diversity is far more complex. In this review, we discuss the recent literature on the alterations in microglia phenomics in the hippocampus of animal models of normal brain aging, acute neuroinflammation, ischemia, and neurodegenerative disorders, such as AD. Microglia undergo phenomic changes consisting of transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. The classical subdivision of microglia into M1 and M2, two different, all-or-nothing states is too simplistic, and does not correspond to the variety of phenotypes recently discovered in the brain. We will discuss the phenomic modifications of microglia focusing not only on the differences in microglia reactivity in the diverse models of neurodegenerative disorders, but also among different areas of the brain. For instance, in contiguous and highly interconnected regions of the rat hippocampus, microglia show a differential, finely regulated, and region-specific reactivity, demonstrating that microglia responses are not uniform, but vary significantly from area to area in response to insults. It is of great interest to verify whether the differences in microglia reactivity may explain the differential susceptibility of different brain areas to insults, and particularly the higher sensitivity of CA1 pyramidal neurons to inflammatory stimuli. Understanding the spatiotemporal heterogeneity of microglia phenomics in health and disease is of paramount importance to find new druggable targets for the development of novel microglia-targeted therapies in different CNS disorders. This will allow interventions in three different ways: (i) by suppressing the pro-inflammatory properties of microglia to limit the deleterious effect of their activation; (ii) by modulating microglia phenotypic change to favor anti-inflammatory properties; (iii) by influencing microglia priming early in the disease process.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Florence, Italy;
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Gary L. Wenk
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA;
| | - Domenico Edoardo Pellegrini-Giampietro
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| |
Collapse
|
20
|
Zhou H, Wang J, Wen T. The molecular neural mechanism underlying the acceleration of brain aging due to Dcf1 deficiency. Mol Cell Neurosci 2023; 126:103884. [PMID: 37506857 DOI: 10.1016/j.mcn.2023.103884] [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: 05/01/2023] [Revised: 07/11/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023] Open
Abstract
Owing to the continuous increase in human life expectancy, the management of aging-related diseases has become an urgent issue. The brain dominates the central nervous system; therefore, brain aging is a key area of aging-related research. We previously uncovered that dendritic cell factor 1 (Dcf1) maintains the stemness of neural stem cells and its expression in Drosophila can prolong lifespan, suggesting an association between Dcf1 and aging; however, the specific underlying neural mechanism remains unclear. In the present study, we show for the first time that hippocampal neurogenesis is decreased in aged Dcf1-/- mice, which leads to a decrease in the number of brain neurons and an increased number of senescent cells. Moreover, astrocytes proliferate abnormally and express elevated mRNA levels of aging-related factors, in addition to displaying increased activation of Akt and Foxo3a. Finally, behavioral tests confirm that aged Dcf1-/- mice exhibit a significant decline in cognitive abilities related to learning and memory. In conclusion, we reveal a novel mechanism underlying brain aging triggered by Dcf1 deficiency at the molecular, cellular, tissue, and behavioral levels, providing a new perspective for the exploration of brain aging.
Collapse
Affiliation(s)
- Haicong Zhou
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China; Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University Shanghai, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University Shanghai, China
| | - Tieqiao Wen
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University Shanghai, China.
| |
Collapse
|
21
|
Csemer A, Kovács A, Maamrah B, Pocsai K, Korpás K, Klekner Á, Szücs P, Nánási PP, Pál B. Astrocyte- and NMDA receptor-dependent slow inward currents differently contribute to synaptic plasticity in an age-dependent manner in mouse and human neocortex. Aging Cell 2023; 22:e13939. [PMID: 37489544 PMCID: PMC10497838 DOI: 10.1111/acel.13939] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/26/2023] Open
Abstract
Slow inward currents (SICs) are known as excitatory events of neurons elicited by astrocytic glutamate via activation of extrasynaptic NMDA receptors. By using slice electrophysiology, we tried to provide evidence that SICs can elicit synaptic plasticity. Age dependence of SICs and their impact on synaptic plasticity was also investigated in both on murine and human cortical slices. It was found that SICs can induce a moderate synaptic plasticity, with features similar to spike timing-dependent plasticity. Overall SIC activity showed a clear decline with aging in humans and completely disappeared above a cutoff age. In conclusion, while SICs contribute to a form of astrocyte-dependent synaptic plasticity both in mice and humans, this plasticity is differentially affected by aging. Thus, SICs are likely to play an important role in age-dependent physiological and pathological alterations of synaptic plasticity.
Collapse
Affiliation(s)
- Andrea Csemer
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- Doctoral School of Molecular MedicineUniversity of DebrecenDebrecenHungary
| | - Adrienn Kovács
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Baneen Maamrah
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- Doctoral School of Molecular MedicineUniversity of DebrecenDebrecenHungary
| | - Krisztina Pocsai
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Kristóf Korpás
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Álmos Klekner
- Department of Neurosurgery, Clinical CentreUniversity of DebrecenDebrecenHungary
| | - Péter Szücs
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Péter P. Nánási
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- Department of Dental Physiology and Pharmacology, Faculty of DentistryUniversity of DebrecenDebrecenHungary
| | - Balázs Pál
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- Doctoral School of Molecular MedicineUniversity of DebrecenDebrecenHungary
| |
Collapse
|
22
|
de Deus JL, Amorim MR, da Silva Junior RMP, Jesus AA, de Barcellos Filho PCG, Cárnio EC, Cunha AOS, Leão RM, Branco LG. Inhaled molecular hydrogen reduces hippocampal neuroinflammation, glial reactivity and ameliorates memory impairment during systemic inflammation. Brain Behav Immun Health 2023; 31:100654. [PMID: 37449286 PMCID: PMC10336161 DOI: 10.1016/j.bbih.2023.100654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Sepsis is associated with numerous physiological and biochemical abnormalities that result in a life-threatening condition. The involvement of the Central Nervous System (CNS) during sepsis has received considerable attention, especially the hippocampus which plays a key role in the learning and memory processes. The increased interest in this limbic region during systemic inflammation (SI) is related to the number of sepsis survivor patients who have cognitive impairments. A single injection of lipopolysaccharide (LPS)-induced systemic inflammation is the most commonly used murine endotoxemia model because it replicates several pathophysiological changes observed in severe sepsis. Molecular hydrogen (H2) has been used as an anti-inflammatory therapeutic strategy to prevent neuroinflammation. However, the mechanisms by which inhaled H2 mitigate memory loss during SI remains unknown. To understand how H2 acts in the hippocampus, the current study focused on specific mechanisms that may be involved in reducing neuroinflammation in rats during SI. We hypothesized that inhaled H2 decreases LPS-induced hippocampal pro-inflammatory cytokines surges and this effect is associated with reduced memory loss. Using different and integrative approaches, i.e., from hippocampal cells electrophysiology to animal behavior, we report that inhaled H2 decreased LPS-induced peripheral and hippocampal inflammation, decreased microglial and astrocytic activation, lessen memory loss without affecting long-term potentiation (LTP). To our knowledge, this is the first evidence showing that inhaled H2 reduces hippocampal microglial and glial cells inflammation, which may be associated with a reduced memory impairment induced by SI.
Collapse
Affiliation(s)
- Júnia Lara de Deus
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Mateus Ramos Amorim
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Aline Alves Jesus
- Department of Physiology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | | | - Evelin Capellari Cárnio
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | - Ricardo Maurício Leão
- Department of Physiology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Luiz G.S. Branco
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
23
|
Mitroshina EV, Krivonosov MI, Pakhomov AM, Yarullina LE, Gavrish MS, Mishchenko TA, Yarkov RS, Vedunova MV. Unravelling the Collective Calcium Dynamics of Physiologically Aged Astrocytes under a Hypoxic State In Vitro. Int J Mol Sci 2023; 24:12286. [PMID: 37569663 PMCID: PMC10419080 DOI: 10.3390/ijms241512286] [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: 06/28/2023] [Revised: 07/24/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Astrocytes serve many functions in the brain related to maintaining nerve tissue homeostasis and regulating neuronal function, including synaptic transmission. It is assumed that astrocytes are crucial players in determining the physiological or pathological outcome of the brain aging process and the development of neurodegenerative diseases. Therefore, studies on the peculiarities of astrocyte physiology and interastrocytic signaling during aging are of utmost importance. Calcium waves are one of the main mechanisms of signal transmission between astrocytes, and in the present study we investigated the features of calcium dynamics in primary cultures of murine cortical astrocytes in physiological aging and hypoxia modeling in vitro. Specifically, we focused on the assessment of calcium network dynamics and the restructuring of the functional network architecture in primary astrocytic cultures. Calcium imaging was performed on days 21 ("young" astrocyte group) and 150 ("old" astrocyte group) of cultures' development in vitro. While the number of active cells and frequency of calcium events were decreased, we observed a reduced degree of correlation in calcium dynamics between neighboring cells, which was accompanied by a reduced number of functionally connected cells with fewer and slower signaling events. At the same time, an increase in the mRNA expression of anti-apoptotic factor Bcl-2 and connexin 43 was observed in "old" astrocytic cultures, which can be considered as a compensatory response of cells with a decreased level of intercellular communication. A hypoxic episode aggravates the depression of the connectivity of calcium dynamics of "young" astrocytes rather than that of "old" ones.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Mikhail I. Krivonosov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Alexander M. Pakhomov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
- Federal Research Center Institute of Applied Physics of the Russian Academy of Sciences (IAP RAS), 603950 Nizhny Novgorod, Russia
| | - Laysan E. Yarullina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Maria S. Gavrish
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Roman S. Yarkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| |
Collapse
|
24
|
Razee A, Banerjee S, Hong J, Magaki S, Fishbein G, Ajijola OA, Umar S. Thoracic Spinal Cord Neuroinflammation as a Novel Therapeutic Target in Pulmonary Hypertension. Hypertension 2023; 80:1297-1310. [PMID: 37092338 PMCID: PMC10192067 DOI: 10.1161/hypertensionaha.122.20782] [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: 12/09/2022] [Accepted: 04/03/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) is associated with aberrant sympathoexcitation leading to right ventricular failure (RVF), arrhythmias, and death. Microglial activation and neuroinflammation have been implicated in sympathoexcitation in experimental PH. We recently reported the first evidence of thoracic spinal cord (TSC) neuroinflammation in PH rats. Here, we hypothesize that PH is associated with increased cardiopulmonary afferent signaling leading to TSC-specific neuroinflammation and sympathoexcitation. Furthermore, inhibition of TSC neuroinflammation rescues experimental PH and RVF. METHODS We performed transcriptomic analysis and its validation on the TSC of monocrotaline (n=8) and Sugen hypoxia (n=8) rat models of severe PH-RVF. A group of monocrotaline rats received either daily intrathecal microglial activation inhibitor minocycline (200 μg/kg per day, n=5) or PBS (n=5) from day 14 through 28. Echocardiography and right ventricle-catheterization were performed terminally. Real-time quantitative reverse transcription PCR, immunolocalization, microglia+astrocyte quantification, and terminal deoxynucleotidyl transferase dUTP nick end labeling were assessed. Plasma catecholamines were measured by ELISA. Human spinal cord autopsy samples (Control n=3; pulmonary arterial hypertension n=3) were assessed to validate preclinical findings. RESULTS Increased cardiopulmonary afferent signaling was demonstrated in preclinical and clinical PH. Our findings delineated common dysregulated genes and pathways highlighting neuroinflammation and apoptosis in the remodeled TSC and highlighted increased sympathoexcitation in both rat models. Moreover, we validated significantly increased microglial and astrocytic activation and CX3CL1 expression in TSC of human pulmonary arterial hypertension. Finally, amelioration of TSC neuroinflammation by minocycline in monocrotaline rats inhibited microglial activation, decreased proinflammatory cytokines, sympathetic nervous system activation and significantly attenuated PH and RVF. CONCLUSIONS Targeting neuroinflammation and associated molecular pathways and genes in the TSC may yield novel therapeutic strategies for PH and RVF.
Collapse
Affiliation(s)
- Asif Razee
- Department of Anesthesiology and Perioperative Medicine Division of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Somanshu Banerjee
- Department of Anesthesiology and Perioperative Medicine Division of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jason Hong
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Los Angeles, CA, USA
| | - Shino Magaki
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Greg Fishbein
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, Los Angeles, CA, USA
| | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine Division of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
25
|
Ajenikoko MK, Ajagbe AO, Onigbinde OA, Okesina AA, Tijani AA. Review of Alzheimer's disease drugs and their relationship with neuron-glia interaction. IBRO Neurosci Rep 2023; 14:64-76. [PMID: 36593897 PMCID: PMC9803919 DOI: 10.1016/j.ibneur.2022.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Because Alzheimer's disease has no known treatment, sufferers and their caregivers must concentrate on symptom management. Astrocytes and microglia are now known to play distinct physiological roles in synaptic function, the blood-brain barrier, and neurovascular coupling. Consequently, the search for drugs that can slow the degenerative process in dementia sufferers continues because existing drugs are designed to alleviate the symptoms of Alzheimer's disease. Drugs that address pathological changes without interfering with the normal function of glia, such as eliminating amyloid-beta deposits, are prospective treatments for neuroinflammatory illnesses. Because neuron-astrocytes-microglia interactions are so complex, developing effective, preventive, and therapeutic medications for AD will necessitate novel methodologies and strategic targets. This review focused on existing medications used in treating AD amongst which include Donepezil, Choline Alphoscerate, Galantamine, Dextromethorphan, palmitoylethanolamide, citalopram, resveratrol, and solanezumab. This review summarizes the effects of these drugs on neurons, astrocytes, and microglia interactions based on their pharmacokinetic properties, mechanism of action, dosing, and clinical presentations.
Collapse
Affiliation(s)
- Michael Kunle Ajenikoko
- Department of Anatomy, Faculty of Biomedical Sciences, Kampala International University, Western Campus, Ishaka, Uganda
| | - Abayomi Oyeyemi Ajagbe
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, P.M.B. 900001 Abuja, Nigeria
| | - Oluwanisola Akanji Onigbinde
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, P.M.B. 900001 Abuja, Nigeria
| | - Akeem Ayodeji Okesina
- Department of Clinical Medicine and Community Health, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Ahmad Adekilekun Tijani
- Department of Anatomy, Faculty of Basic Medical Sciences, Modibbo Adama University, Yola, Nigeria
| |
Collapse
|
26
|
Suthard RL, Jellinger AL, Surets M, Shpokayte M, Pyo AY, Buzharsky MD, Senne RA, Dorst K, Leblanc H, Ramirez S. Chronic Gq activation of ventral hippocampal neurons and astrocytes differentially affects memory and behavior. Neurobiol Aging 2023; 125:9-31. [PMID: 36801699 DOI: 10.1016/j.neurobiolaging.2023.01.007] [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: 09/15/2022] [Revised: 12/20/2022] [Accepted: 01/13/2023] [Indexed: 02/01/2023]
Abstract
Network dysfunction is implicated in numerous diseases and psychiatric disorders, and the hippocampus serves as a common origin for these abnormalities. To test the hypothesis that chronic modulation of neurons and astrocytes induces impairments in cognition, we activated the hM3D(Gq) pathway in CaMKII+ neurons or GFAP+ astrocytes within the ventral hippocampus across 3, 6, and 9 months. CaMKII-hM3Dq activation impaired fear extinction at 3 months and acquisition at 9 months. Both CaMKII-hM3Dq manipulation and aging had differential effects on anxiety and social interaction. GFAP-hM3Dq activation impacted fear memory at 6 and 9 months. GFAP-hM3Dq activation impacted anxiety in the open field only at the earliest time point. CaMKII-hM3Dq activation modified the number of microglia, while GFAP-hM3Dq activation impacted microglial morphological characteristics, but neither affected these measures in astrocytes. Overall, our study elucidates how distinct cell types can modify behavior through network dysfunction, while adding a more direct role for glia in modulating behavior.
Collapse
Affiliation(s)
- Rebecca L Suthard
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Alexandra L Jellinger
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Michelle Surets
- Undergraduate Program in Neuroscience, Boston University, Boston, MA, USA
| | - Monika Shpokayte
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Angela Y Pyo
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA, USA
| | | | - Ryan A Senne
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Kaitlyn Dorst
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Heloise Leblanc
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Steve Ramirez
- Department of Biomedical Engineering, Boston University, Boston, MA, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA, USA.
| |
Collapse
|
27
|
Cunha Feio Leal MD, Amaral Junior FLD, Silva Arruda BFD, Kurosawa JAA, Vieira AA, Maia JCC, Scalfoni VVB, Silveira Junior AMD, Feijó MO, Albuquerque FBAD, Marta MHM, Normando MPN, Silva AGOCD, Trindade FCPD, Siqueira Mendes FDCCD, Sosthenes MCK. The Masticatory Activity Interference in Quantitative Estimation of CA1, CA3 and Dentate Gyrus Hippocampal Astrocytes of Aged Murine Models and under Environmental Stimulation. Int J Mol Sci 2023; 24:ijms24076529. [PMID: 37047502 PMCID: PMC10095286 DOI: 10.3390/ijms24076529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/22/2023] [Accepted: 03/01/2023] [Indexed: 04/03/2023] Open
Abstract
Studies indicating the influence of masticatory dysfunction, due to a soft diet or lack of molars, on impairing spatial memory and learning have led to research about neuronal connections between areas and cell populations possibly affected. In this sense, with scarce detailed data on the subfields of hippocampus in dementia neurodegeneration, there is no information about astrocytic responses in its different layers. Thus, considering this context, the present study evaluated the effects of deprivation and rehabilitation of masticatory activity, aging, and environmental enrichment on the stereological quantification of hippocampal astrocytes from layers CA1, CA3, and DG. For this purpose, we examined mature (6-month-old; 6M), and aged (18-month-old; 18M) mice, subjected to distinct masticatory regimens and environments. Three different regimens of masticatory activity were applied: continuous normal mastication with hard pellets (HD); normal mastication followed by deprived mastication with equal periods of pellets followed by soft powder (HD/SD); or rehabilitated masticatory activity with equal periods of HD, followed by powder, followed by pellets (HD/SD/HD). Under each specific regimen, half of the animals were raised in standard cages (impoverished environment (IE)) and the other half in enriched cages (enriched environment (EE)), mimicking sedentary or active lifestyles. Microscopic stereological, systematic, and random sampling approaches with an optical dissector of GFAP-immunolabeled astrocytes were done, allowing for an astrocyte numerical estimate. Stratum moleculare and hilus, from the dentate gyrus (DG) and Strata Lacunosum-Moleculare, Oriens, and Radiatum, similarly to the dentate gyrus, showed no significant change in any of the investigated variables (age, diet, or environment) in these layers. However, in Stratum radiatum, it was possible to observe significant differences associated with diet regimens and age. Therefore, diet-related differences were found when the HD 18M IE group was compared to the HD/SD/HD 18-month-old group in the same environment (IE) (p = 0.007). In the present study, we present modulatory factors (masticatory function, environmental enrichment, and aging) for the differentiated quantitative laminar response in the hippocampal regions, suggesting other studies to read the plasticity and responsiveness of astrocytes, including the molecular background.
Collapse
Affiliation(s)
- Marília da Cunha Feio Leal
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Fabio Leite do Amaral Junior
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Bernardo Freire da Silva Arruda
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | | | - Amanda Almeida Vieira
- Curso de Medicina, Centro Universitário do Estado do Pará, Belém 66613-903, PA, Brazil
| | | | | | - Antonio Morais da Silveira Junior
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Matheus Oliveira Feijó
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Fernanda Beatriz Araújo de Albuquerque
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | | | | | - Alana Gabriele Oliveira Cabeça da Silva
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Fernanda Catharina Pires da Trindade
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Fabíola de Carvalho Chaves de Siqueira Mendes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
- Curso de Medicina, Centro Universitário do Estado do Pará, Belém 66613-903, PA, Brazil
| | - Marcia Consentino Kronka Sosthenes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| |
Collapse
|
28
|
Villablanca C, Vidal R, Gonzalez-Billault C. Are cytoskeleton changes observed in astrocytes functionally linked to aging? Brain Res Bull 2023; 196:59-67. [PMID: 36935053 DOI: 10.1016/j.brainresbull.2023.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/22/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
Astrocytes are active participants in the performance of the Central Nervous System (CNS) in both health and disease. During aging, astrocytes are susceptible to reactive astrogliosis, a molecular state characterized by functional changes in response to pathological situations, and cellular senescence, characterized by loss of cell division, apoptosis resistance, and gain of proinflammatory functions. This results in two different states of astrocytes, which can produce proinflammatory phenotypes with harmful consequences in chronic conditions. Reactive astrocytes and senescent astrocytes share morpho-functional features that are dependent on the organization of the cytoskeleton. However, such changes in the cytoskeleton have yet to receive the necessary attention to explain their role in the alterations of astrocytes that are associated with aging and pathologies. In this review, we summarize all the available findings that connect changes in the cytoskeleton of the astrocytes with aging. In addition, we discuss future avenues that we believe will guide such a novel topic.
Collapse
Affiliation(s)
- Cristopher Villablanca
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - René Vidal
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Christian Gonzalez-Billault
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Institute for Nutrition and Food Technologies, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
29
|
Chen Z, Jin J, Lu Q. STAMP2 alleviates microglial neurotoxicity by inhibiting LPS-induced NF-κB activation. Mol Cell Toxicol 2023. [DOI: 10.1007/s13273-023-00346-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
30
|
Bondi H, Chiazza F, Masante I, Bortolotto V, Canonico PL, Grilli M. Heterogenous response to aging of astrocytes in murine Substantia Nigra pars compacta and pars reticulata. Neurobiol Aging 2023; 123:23-34. [PMID: 36630756 DOI: 10.1016/j.neurobiolaging.2022.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
Currently, little is known about the impact of aging on astrocytes in substantia nigra pars compacta (SNpc), where dopaminergic neurons degenerate both in physiological aging and in Parkinson's disease, an age-related neurodegenerative disorder. We performed a morphometric analysis of GFAP+ astrocytes in SNpc and, for comparison, in the pars reticulata (SNpr) of young (4-6 months), middle-aged (14-17 months) and old (20-24 months) C57BL/6J male mice. We demonstrated an age-dependent increase of structural complexity only in astrocytes localized in SNpc, and not in SNpr. Astrocytic structural remodelling was not accompanied by changes in GFAP expression, while GFAP increased in SNpr of old compared to young mice. In parallel, transcript levels of selected astrocyte-enriched genes were evaluated. With aging, decreased GLT1 expression occurred only in SNpc, while xCT transcript increased both in SNpc and SNpr, suggesting a potential loss of homeostatic control of extracellular glutamate only in the subregion where age-dependent neurodegeneration occurs. Altogether, our results support an heterogenous morphological and biomolecular response to aging of GFAP+ astrocytes in SNpc and SNpr.
Collapse
Affiliation(s)
- Heather Bondi
- Laboratory of Neuroplasticity, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Fausto Chiazza
- Laboratory of Neuroplasticity, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Irene Masante
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Valeria Bortolotto
- Laboratory of Neuroplasticity, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Mariagrazia Grilli
- Laboratory of Neuroplasticity, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy.
| |
Collapse
|
31
|
Graciani AL, Gutierre MU, Coppi AA, Arida RM, Gutierre RC. MYELIN, AGING, AND PHYSICAL EXERCISE. Neurobiol Aging 2023; 127:70-81. [PMID: 37116408 DOI: 10.1016/j.neurobiolaging.2023.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023]
Abstract
Myelin sheath is a structure in neurons fabricated by oligodendrocytes and Schwann cells responsible for increasing the efficiency of neural synapsis, impulse transmission, and providing metabolic support to the axon. They present morpho-functional changes during health aging as deformities of the sheath and its fragmentation, causing an increased load on microglial phagocytosis, with Alzheimer's disease aggravating. Physical exercise has been studied as a possible protective agent for the nervous system, offering benefits to neuroplasticity. In this regard, studies in animal models for Alzheimer's and depression reported the efficiency of physical exercise in protecting against myelin degeneration. A reduction of myelin damage during aging has also been observed in healthy humans. Physical activity promotes oligodendrocyte proliferation and myelin preservation during old age, although some controversies remain. In this review, we will address how effective physical exercise can be as a protective agent of the myelin sheath against the effects of aging in physiological and pathological conditions.
Collapse
|
32
|
Rawji KS, Neumann B, Franklin RJM. Glial aging and its impact on central nervous system myelin regeneration. Ann N Y Acad Sci 2023; 1519:34-45. [PMID: 36398864 DOI: 10.1111/nyas.14933] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aging is a major risk factor for several neurodegenerative diseases and is associated with cognitive decline. In addition to affecting neuronal function, the aging process significantly affects the functional phenotype of the glial cell compartment, comprising oligodendrocyte lineage cells, astrocytes, and microglia. These changes result in a more inflammatory microenvironment, resulting in a condition that is favorable for neuron and synapse loss. In addition to facilitating neurodegeneration, the aging glial cell population has negative implications for central nervous system remyelination, a regenerative process that is of particular importance to the chronic demyelinating disease multiple sclerosis. This review will discuss the changes that occur with aging in the three main glial populations and provide an overview of the studies documenting the impact these changes have on remyelination.
Collapse
Affiliation(s)
- Khalil S Rawji
- Altos Labs, Cambridge Institute of Science, Cambridge, UK
| | - Björn Neumann
- Altos Labs, Cambridge Institute of Science, Cambridge, UK
| | | |
Collapse
|
33
|
Gudkov SV, Burmistrov DE, Kondakova EV, Sarimov RM, Yarkov RS, Franceschi C, Vedunova MV. An emerging role of astrocytes in aging/neuroinflammation and gut-brain axis with consequences on sleep and sleep disorders. Ageing Res Rev 2023; 83:101775. [PMID: 36334910 DOI: 10.1016/j.arr.2022.101775] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/05/2022] [Accepted: 10/30/2022] [Indexed: 11/18/2022]
Abstract
Understanding the role of astrocytes in the central nervous system has changed dramatically over the last decade. The accumulating findings indicate that glial cells are involved not only in the maintenance of metabolic and ionic homeostasis and in the implementation of trophic functions but also in cognitive functions and information processing in the brain. Currently, there are some controversies regarding the role of astrocytes in complex processes such as aging of the nervous system and the pathogenesis of age-related neurodegenerative diseases. Many findings confirm the important functional role of astrocytes in age-related brain changes, including sleep disturbance and the development of neurodegenerative diseases and particularly Alzheimer's disease. Until recent years, neurobiological research has focused mainly on neuron-glial interactions, in which individual astrocytes locally modulate neuronal activity and communication between neurons. The review considers the role of astrocytes in the physiology of sleep and as an important "player" in the development of neurodegenerative diseases. In addition, the features of the astrocytic network reorganization during aging are discussed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Dmitriy E Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Elena V Kondakova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Ruslan M Sarimov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Roman S Yarkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Maria V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| |
Collapse
|
34
|
Verkhratsky A, Semyanov A. Astrocytes in Ageing. Subcell Biochem 2023; 103:253-277. [PMID: 37120471 DOI: 10.1007/978-3-031-26576-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Ageing is associated with a morphological and functional decline of astrocytes with a prevalence of morphological atrophy and loss of function. In particular, ageing is manifested by the shrinkage of astrocytic processes: branches and leaflets, which decreases synaptic coverage. Astrocytic dystrophy affects multiple functions astrocytes play in the brain active milieu. In particular, and in combination with an age-dependent decline in the expression of glutamate transporters, astrocytic atrophy translates into deficient glutamate clearance and K+ buffering. Decreased astrocyte presence may contribute to age-dependent remodelling of brain extracellular space, hence affecting extrasynaptic signalling. Old astrocytes lose endfeet polarisation of AQP4 water channels, thus limiting the operation of the glymphatic system. In ageing, astrocytes down-regulate their antioxidant capacity leading to decreased neuroprotection. All these changes may contribute to an age-dependent cognitive decline.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
- Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain.
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Physiology, Jiaxing University College of Medicine, Jiaxing, Zhejiang Pro, China
| |
Collapse
|
35
|
Cvetanovic M, Gray M. Contribution of Glial Cells to Polyglutamine Diseases: Observations from Patients and Mouse Models. Neurotherapeutics 2023; 20:48-66. [PMID: 37020152 PMCID: PMC10119372 DOI: 10.1007/s13311-023-01357-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 04/07/2023] Open
Abstract
Neurodegenerative diseases are broadly characterized neuropathologically by the degeneration of vulnerable neuronal cell types in a specific brain region. The degeneration of specific cell types has informed on the various phenotypes/clinical presentations in someone suffering from these diseases. Prominent neurodegeneration of specific neurons is seen in polyglutamine expansion diseases including Huntington's disease (HD) and spinocerebellar ataxias (SCA). The clinical manifestations observed in these diseases could be as varied as the abnormalities in motor function observed in those who have Huntington's disease (HD) as demonstrated by a chorea with substantial degeneration of striatal medium spiny neurons (MSNs) or those with various forms of spinocerebellar ataxia (SCA) with an ataxic motor presentation primarily due to degeneration of cerebellar Purkinje cells. Due to the very significant nature of the degeneration of MSNs in HD and Purkinje cells in SCAs, much of the research has centered around understanding the cell autonomous mechanisms dysregulated in these neuronal cell types. However, an increasing number of studies have revealed that dysfunction in non-neuronal glial cell types contributes to the pathogenesis of these diseases. Here we explore these non-neuronal glial cell types with a focus on how each may contribute to the pathogenesis of HD and SCA and the tools used to evaluate glial cells in the context of these diseases. Understanding the regulation of supportive and harmful phenotypes of glia in disease could lead to development of novel glia-focused neurotherapeutics.
Collapse
Affiliation(s)
- Marija Cvetanovic
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, Minneapolis, USA
| | - Michelle Gray
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
36
|
Sharma S, Borski C, Hanson J, Garcia MA, Link CD, Hoeffer C, Chatterjee A, Nagpal P. Identifying an Optimal Neuroinflammation Treatment Using a Nanoligomer Discovery Engine. ACS Chem Neurosci 2022; 13:3247-3256. [PMID: 36410860 DOI: 10.1021/acschemneuro.2c00365] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Acute activation of innate immune response in the brain, or neuroinflammation, protects this vital organ from a range of external pathogens and promotes healing after traumatic brain injury. However, chronic neuroinflammation leading to the activation of immune cells like microglia and astrocytes causes damage to the nervous tissue, and it is causally linked to a range of neurodegenerative diseases such as Alzheimer's diseases (AD), Multiple Sclerosis (MS), Parkinson's disease (PD), and many others. While neuroinflammation is a key target for a range of neuropathological diseases, there is a lack of effective countermeasures to tackle it, and existing experimental therapies require fairly invasive intracerebral and intrathecal delivery due to difficulty associated with the therapeutic crossover between the blood-brain barrier, making such treatments impractical to treat neuroinflammation long-term. Here, we present the development of an optimal neurotherapeutic using our Nanoligomer Discovery Engine, by screening downregulation of several proinflammatory cytokines (e.g., Interleukin-1β or IL-1β, tumor necrosis factor-alpha or TNF-α, TNF receptor 1 or TNFR1, Interleukin 6 or IL-6), inflammasomes (e.g., NLRP1), key transcription factors (e.g., nuclear factor kappa-B or NF-κβ) and their combinations, as upstream regulators and canonical pathway targets, to identify and validate the best-in-class treatment. Using our high-throughput drug discovery, target validation, and lead molecule identification via a bioinformatics and artificial intelligence-based ranking method to design sequence-specific peptide molecules to up- or downregulate gene expression of the targeted gene at will, we used our discovery engine to perturb and identify most effective upstream regulators and canonical pathways for therapeutic intervention to reverse neuroinflammation. The lead neurotherapeutic was a combination of Nanoligomers targeted to NF-κβ (SB.201.17D.8_NF-κβ1) and TNFR1 (SB.201.18D.6_TNFR1), which were identified using in vitro cell-based screening in donor-derived human astrocytes and further validated in vivo using a mouse model of lipopolysaccharide (LPS)-induced neuroinflammation. The combination treatment SB_NI_111 was delivered without any special formulation using a simple intraperitoneal injection of low dose (5 mg/kg) and was found to significantly suppress the expression of LPS-induced neuroinflammation in mouse hippocampus. These results point to the broader applicability of this approach towards the development of therapies for chronic neuroinflammation-linked neurodegenerative diseases, sleep countermeasures, and others, and the potential for further investigation of the lead neurotherapeutic molecule as reversible gene therapy.
Collapse
Affiliation(s)
- Sadhana Sharma
- Sachi Bioworks, Colorado Technology Center, 685 S Arthur AvenueLouisville, Colorado 80027, United States
| | - Curtis Borski
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States.,Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Jessica Hanson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States.,Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Micklaus A Garcia
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Christopher D Link
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Charles Hoeffer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States.,Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Anushree Chatterjee
- Sachi Bioworks, Colorado Technology Center, 685 S Arthur AvenueLouisville, Colorado 80027, United States
| | - Prashant Nagpal
- Sachi Bioworks, Colorado Technology Center, 685 S Arthur AvenueLouisville, Colorado 80027, United States
| |
Collapse
|
37
|
Lin N, Jin JW, Lai ZM, Zhang DF, Chen Y, Guo HG, Liu JL. Mdivi-1 improves postoperative neurocognitive disorders in aged rats undergoing splenectomy by inhibiting dynamin-related protein-1. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1338. [PMID: 36660632 PMCID: PMC9843339 DOI: 10.21037/atm-22-5496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/05/2022] [Indexed: 12/28/2022]
Abstract
Background The regulatory role of mitochondria in the inflammatory response of the nervous system postoperatively remains unclear. This study explored the relationship between mitochondria and postoperative neurocognitive dysfunction (PNCD) by regulating mitochondrial function in aged rats undergoing splenectomy. Methods A total of 120 aged rats were randomly divided into five groups (n=24) as follows: Control group (not subjected to any form of treatment), Sham group (subjected only to sham-splenectomized operation after anesthesia), Splenectomy group (only underwent splenectomy after anesthesia), Synonyms Mitochondrial division inhibitor 1 (Mdivi-1) group [treated with Mdivi-1, a dynamin-relatedprotein 1 (Drp1) inhibitor], and Dimethyl Sulfoxide (DMSO) group (treated with DMSO, a solvent). Inflammatory markers, namely interleukin-1β (IL-1β) and tumor necrosis factor α (TNF-α), were measured in the plasma and brains of the rats. Cognitive function was assessed using the Morris water maze test. Results During the perioperative period, the physiological parameters did not differ among the five groups (P>0.05). The results of the Morris water maze experiments showed that the memory of the rats was significantly impaired after splenectomy, which was alleviated by Mdivi-1 administration (P=0.04). Postoperatively, the proinflammatory cytokine levels in the serum and hippocampus tissue were upregulated, while Mdivi-1 administration reduced this increase. The electron microscopy and hematoxylin-eosin (HE) staining results indicated that the structure of neurons and mitochondria was minimally impaired in the Mdivi-1 group. Conclusions Aged rats that underwent splenectomy exhibited significant postoperative cognitive impairments. The selective inhibitor of Drp1, Mdivi-1, exerted protective effects against PNCD by ameliorating mitochondrial dysfunction and reducing the inflammatory response.
Collapse
Affiliation(s)
- Nan Lin
- Department of Anesthesiology, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Jian-Wen Jin
- Department of Clinical Medicine, Fujian Health College, Fuzhou, China
| | - Zhong-Meng Lai
- Department of Anesthesiology, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Dan-Feng Zhang
- Department of Anesthesiology, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Ye Chen
- Department of Anesthesiology, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Hong-Gang Guo
- Hangzhou Medical College, Zhejiang Provincial Key Laboratory of laboratory Animal and Safety Research, Hangzhou, China
| | - Jun-Le Liu
- Department of Anesthesiology, Union Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
38
|
Cannabidiol inhibits microglia activation and mitigates neuronal damage induced by kainate in an in-vitro seizure model. Neurobiol Dis 2022; 174:105895. [DOI: 10.1016/j.nbd.2022.105895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/24/2022] Open
|
39
|
Patel SV, DeCarlo CM, Book SA, Schormans AL, Whitehead SN, Allman BL, Hayes SH. Noise exposure in early adulthood causes age-dependent and brain region-specific impairments in cognitive function. Front Neurosci 2022; 16:1001686. [PMID: 36312027 PMCID: PMC9606802 DOI: 10.3389/fnins.2022.1001686] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
Hearing loss is a chronic health condition that affects millions of people worldwide. In addition to age-related hearing impairment, excessive noise exposure is a leading cause of hearing loss. Beyond the devastating effects of hearing impairment itself, epidemiological studies have identified hearing loss as a major risk factor for age-related cognitive decline, including dementia. At present, we currently lack a full understanding of the brain regions and underlying molecular changes that are responsible for mediating the link between hearing loss and cognitive impairment across aging. In the present study, we exposed 6-month-old rats to an occupational-like noise (100 dB SPL, 4 h/day × 30 days) or sham exposure and investigated both hippocampal-dependent (i.e., spatial learning and memory, assessed using the Morris water maze) and striatal-dependent (i.e., visuomotor associative learning, assessed using an operant-conditioning task) cognitive function across aging at 7, 10, and 13 months of age. We also investigated brain region-specific changes in microglial expression following noise/sham exposure in order to assess the potential contribution of this cell type to noise-induced cognitive impairments. Consistent with human studies, the occupational-like noise exposure resulted in high-frequency hearing loss, evidenced by a significant increase in hearing thresholds at 20 kHz. Ultimately, our results suggest that not all higher-level cognitive tasks or their associated brain regions appear to be equally susceptible to noise-induced deficits during aging, as the occupational-like noise exposure caused an age-dependent deficit in spatial but not visuomotor associative learning, as well as altered microglial expression in the hippocampus but not the striatum. Interestingly, we found no significant relationships between spatial learning ability and the level of hearing loss or altered microglial density in the hippocampus following noise exposure, suggesting that other changes in the brain likely contribute to hippocampal-dependent cognitive dysfunction following noise exposure. Lastly, we found that a subset of younger animals also showed noise-induced deficits in spatial learning; findings which suggest that noise exposure may represent an increased risk for cognitive impairment in vulnerable subjects. Overall, our findings highlight that even a mild occupational-like noise exposure earlier in adulthood can have long lasting implications for cognitive function later in life.
Collapse
|
40
|
Lana D, Landucci E, Mazzantini C, Magni G, Pellegrini-Giampietro DE, Giovannini MG. The Protective Effect of CBD in a Model of In Vitro Ischemia May Be Mediated by Agonism on TRPV2 Channel and Microglia Activation. Int J Mol Sci 2022; 23:12144. [PMID: 36292998 PMCID: PMC9603301 DOI: 10.3390/ijms232012144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 09/21/2023] Open
Abstract
Cannabinoids, used for centuries for recreational and medical purposes, have potential therapeutic value in stroke treatment. Cannabidiol (CBD), a non-psychoactive compound and partial agonist of TRPV2 channels, is efficacious in many neurological disorders. We investigated the effects of CBD or Δ9-tetrahydrocannabinol (THC) in rat organotypic hippocampal slices exposed to oxygen-glucose deprivation (OGD), an in vitro model of ischemia. Neuronal TRPV2 expression decreased after OGD, but it increased in activated, phagocytic microglia. CBD increased TRPV2 expression, decreased microglia phagocytosis, and increased rod microglia after OGD. THC had effects contrary to those of CBD. Our results show that cannabinoids have different effects in ischemia. CBD showed neuroprotective effects, mediated, at least in part, by TRPV2 channels, since the TRPV2 antagonist tranilast blocked them, while THC worsened the neurodegeneration caused by ischemia. In conclusion, our results suggest that different cannabinoid molecules play different roles in the mechanisms of post-ischemic neuronal death. These different effects of cannabinoid observed in our experiments caution against the indiscriminate use of cannabis or cannabinoid preparations for recreational or therapeutic use. It was observed that the positive effects of CBD may be counteracted by the negative effects caused by high levels of THC.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Costanza Mazzantini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Sesto Fiorentino, Italy
| | | | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| |
Collapse
|
41
|
Bychkov R, Juhaszova M, Calvo-Rubio Barrera M, Donald LAH, Coletta C, Shumaker C, Moorman K, Sirenko ST, Maltsev AV, Sollott SJ, Lakatta EG. The Heart's Pacemaker Mimics Brain Cytoarchitecture and Function: Novel Interstitial Cells Expose Complexity of the SAN. JACC Clin Electrophysiol 2022; 8:1191-1215. [PMID: 36182566 DOI: 10.1016/j.jacep.2022.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/27/2022] [Accepted: 07/01/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND The sinoatrial node (SAN) of the heart produces rhythmic action potentials, generated via calcium signaling within and among pacemaker cells. Our previous work has described the SAN as composed of a hyperpolarization-activated cyclic nucleotide-gated potassium channel 4 (HCN4)-expressing pacemaker cell meshwork, which merges with a network of connexin 43+/F-actin+ cells. It is also known that sympathetic and parasympathetic innervation create an autonomic plexus in the SAN that modulates heart rate and rhythm. However, the anatomical details of the interaction of this plexus with the pacemaker cell meshwork have yet to be described. OBJECTIVES This study sought to describe the 3-dimensional cytoarchitecture of the mouse SAN, including autonomic innervation, peripheral glial cells, and pacemaker cells. METHODS The cytoarchitecture of SAN whole-mount preparations was examined by three-dimensional confocal laser-scanning microscopy of triple immunolabeled with combinations of antibodies for HCN4, S100 calcium-binding protein B (S100B), glial fibrillary acidic protein (GFAP), choline acetyltransferase, or vesicular acetylcholine transporter, and tyrosine hydroxylase, and transmission electron microscopy. RESULTS The SAN exhibited heterogeneous autonomic innervation, which was accompanied by a web of peripheral glial cells and a novel S100B+/GFAP- interstitial cell population, with a unique morphology and a distinct distribution pattern, creating complex interactions with other cell types in the node, particularly with HCN4-expressing cells. Transmission electron microscopy identified a similar population of interstitial cells as telocytes, which appeared to secrete vesicles toward pacemaker cells. Application of S100B to SAN preparations desynchronized Ca2+ signaling in HCN4-expressing cells and increased variability in SAN impulse rate and rhythm. CONCLUSIONS The autonomic plexus, peripheral glial cell web, and a novel S100B+/GFAP- interstitial cell type embedded within the HCN4+ cell meshwork increase the structural and functional complexity of the SAN and provide a new regulatory pathway of rhythmogenesis.
Collapse
Affiliation(s)
- Rostislav Bychkov
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Magdalena Juhaszova
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Miguel Calvo-Rubio Barrera
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Lorenzo A H Donald
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Christopher Coletta
- Laboratory of Genetics and Genomics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Chad Shumaker
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Kayla Moorman
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Syevda Tagirova Sirenko
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Alexander V Maltsev
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Steven J Sollott
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.
| |
Collapse
|
42
|
Anticonvulsant Action and Long-Term Effects of Chronic Cannabidiol Treatment in the Rat Pentylenetetrazole-Kindling Model of Epilepsy. Biomedicines 2022; 10:biomedicines10081811. [PMID: 36009358 PMCID: PMC9405483 DOI: 10.3390/biomedicines10081811] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Cannabidiol (CBD) showed anticonvulsant action in several preclinical models and is currently approved by regulatory agencies to treat childhood epilepsy syndromes. However, CBD treatment has limited benefits, and its long-term effects on cognition are not fully understood yet. This study aimed to examine the impact of long-term CBD treatment in the pentylenetetrazole (PTZ)-kindling model of epilepsy. Adult male Wistar rats (N = 24) received PTZ (35 mg/kg intraperitoneally) every other day until two consecutive generalized seizures occurred. CBD (60 mg/kg body weight) was administered daily by the oral route until the kindled state was achieved (n = 12). To confirm that the formulation and administration techniques were not of concern, liquid chromatography–mass spectrometry was performed to test the brain penetration of the CBD formula. As a result of CBD treatment, a lower mortality rate and significantly prolonged generalized seizure latency (925.3 ± 120.0 vs. 550.1 ± 69.62 s) were observed, while the frequency and duration of generalized seizures were not influenced. The CBD-treated group showed a significant decrease in vertical exploration in the open field test and a significant decrease in the discrimination index in the novel object recognition (NOR) test (−0.01 ± 0.17 vs. 0.57 ± 0.15, p = 0.04). The observed behavioral characteristics may be connected to the decreased thickness of the stratum pyramidale or the decreased astrogliosis observed in the hippocampus. In conclusion, CBD treatment did not prevent kindling, nor did it affect seizure frequency or duration. However, it did increase the latency to the first seizure and decreased the prolonged status epilepticus-related mortality in PTZ-kindled rats. The cognitive impairment observed in the NOR test may be related to the high dose used in this study, which may warrant further investigation.
Collapse
|
43
|
Coker CR, White M, Singal A, Bingaman SS, Paul A, Arnold AC, Silberman Y. Minocycline Reduces Hypothalamic Microglia Activation and Improves Metabolic Dysfunction in High Fat Diet-Induced Obese Mice. Front Physiol 2022; 13:933706. [PMID: 35784876 PMCID: PMC9244633 DOI: 10.3389/fphys.2022.933706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/27/2022] [Indexed: 01/27/2023] Open
Abstract
Obesity is associated with insulin resistance, glucose intolerance, inflammation, and altered neuronal activity in brain regions controlling metabolic functions including food intake, energy expenditure, and glucose homeostasis, such as the hypothalamus. In this study, we tested the hypothesis that inhibiting inflammation with minocycline could reduce adverse metabolic consequences associated with high-fat diet (HFD)-induced obesity in mice and sought to determine if metabolic improvements were associated with reduced hypothalamic microglia activity. Male C57Bl/6J mice were placed on 60% HFD for 12 weeks, with minocycline (40 mg/kg, p.o.) or normal tap water given during the last 6 weeks of diet. Age-matched mice maintained on control diet were used as an additional comparator group. Metabolic function was assessed during the last week of treatment. Ramified (resting) and non-ramified (active) microglia were quantified in the hypothalamus following immunohistochemical staining of ionized calcium-binding adaptor 1 (Iba-1) and further assessed by RNAseq. In HFD fed mice, minocycline attenuated body mass and adiposity without altering food intake suggesting enhanced energy expenditure. Minocycline also attenuated hyperinsulinemia and improved insulin sensitivity in HFD mice. Increased microglial activation and autophagy gene network changes were observed in the paraventricular nucleus (PVN) of the hypothalamus of HFD mice, which was prevented by minocycline treatment. Contrary to PVN findings, there were no significant effects of either HFD or minocycline on microglia activation in the hypothalamic arcuate nucleus or central amygdala. Together, these findings suggest that minocycline improves HFD-induced weight gain and insulin resistance in part by reducing inflammatory processes in the PVN, a key hypothalamic region regulating metabolic function.
Collapse
Affiliation(s)
- Caitlin R. Coker
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University School of Medicine, Washington, DC, United States
| | - Melissa White
- Department of Comparative Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Aneesh Singal
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Sarah S. Bingaman
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Anirban Paul
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Amy C. Arnold
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Yuval Silberman
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
44
|
Lee RHC, Wu CYC, Citadin CT, Couto E Silva A, Possoit HE, Clemons GA, Acosta CH, de la Llama VA, Neumann JT, Lin HW. Activation of Neuropeptide Y2 Receptor Can Inhibit Global Cerebral Ischemia-Induced Brain Injury. Neuromolecular Med 2022; 24:97-112. [PMID: 34019239 PMCID: PMC8606017 DOI: 10.1007/s12017-021-08665-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022]
Abstract
Cardiopulmonary arrest (CA) can greatly impact a patient's life, causing long-term disability and death. Although multi-faceted treatment strategies against CA have improved survival rates, the prognosis of CA remains poor. We previously reported asphyxial cardiac arrest (ACA) can cause excessive activation of the sympathetic nervous system (SNS) in the brain, which contributes to cerebral blood flow (CBF) derangements such as hypoperfusion and, consequently, neurological deficits. Here, we report excessive activation of the SNS can cause enhanced neuropeptide Y levels. In fact, mRNA and protein levels of neuropeptide Y (NPY, a 36-amino acid neuropeptide) in the hippocampus were elevated after ACA-induced SNS activation, resulting in a reduced blood supply to the brain. Post-treatment with peptide YY3-36 (PYY3-36), a pre-synaptic NPY2 receptor agonist, after ACA inhibited NPY release and restored brain circulation. Moreover, PYY3-36 decreased neuroinflammatory cytokines, alleviated mitochondrial dysfunction, and improved neuronal survival and neurological outcomes. Overall, NPY is detrimental during/after ACA, but attenuation of NPY release via PYY3-36 affords neuroprotection. The consequences of PYY3-36 inhibit ACA-induced 1) hypoperfusion, 2) neuroinflammation, 3) mitochondrial dysfunction, 4) neuronal cell death, and 5) neurological deficits. The present study provides novel insights to further our understanding of NPY's role in ischemic brain injury.
Collapse
Affiliation(s)
- Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Cristiane T Citadin
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Alexandre Couto E Silva
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Harlee E Possoit
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Garrett A Clemons
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Christina H Acosta
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Victoria A de la Llama
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Jake T Neumann
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA.
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA.
| |
Collapse
|
45
|
Indari O, Jakhmola S, Pathak DK, Tanwar M, Kandpal M, Mishra A, Kumar R, Jha HC. Comparative Account of Biomolecular Changes Post Epstein Barr Virus Infection of the Neuronal and Glial Cells Using Raman Microspectroscopy. ACS Chem Neurosci 2022; 13:1627-1637. [PMID: 35561419 DOI: 10.1021/acschemneuro.2c00081] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Raman microspectroscopy is a vibrational spectroscopy technique used for investigating molecular fingerprints of a wide range of liquid or solid samples. The technique can be efficiently utilized to understand the virus-mediated cellular changes and could provide valuable insights into specific biomolecular alterations. The Epstein Barr virus (EBV) has been associated with various types of cancers as well as neurodegenerative diseases. However, EBV-mediated neurological ailments are yet underexplored in terms of biomolecular changes in neuronal and glial cells (astrocytes and microglia). In continuation of our earlier exploration of EBV-influenced glial cells, we tried to decipher biomolecular changes in EBV-infected neuronal cells using Raman microspectroscopy. Additionally, we compared the consecutive biomolecular changes observed in neuronal cells with both the glial cells. We observed that EBV infection gets differentially regulated in the neuronal cells, astrocytes, and microglia. The viral entry and initiation of infection-mediated cellular modulation could start as soon as 2 h post infection but may regulate a distinct biomolecular milieu in different time intervals. Similar to the early timespan, the 24-36 h interval could also be important for EBV to manipulate neuronal as well as glial cells as depicted from elevated biomolecular activities. At these time intervals, some common biomolecules such as proline, glucose, lactic acid, nucleotides, or cholesterol were observed in the cells. However, at these time intervals, some distinct biomolecules were also observed in each cell, such as collagen, lipid, and protein stretches in the neuronal nucleus (2-4 h); tyrosine and RNA in the astrocyte nucleus (2-4 h nucleus); and fatty acids in the microglia nucleus (24-36 h). The observed biomolecular entities could ultimately play pivotal roles in the viral usurpation of cells. We also provided insights into whether these biomolecular changes can be correlated to each other and mediate virus-associated manifestations which can be linked to neurological complications. Our study aids in the understanding of EBV-mediated biomolecular changes in the various compartments of the central nervous system.
Collapse
Affiliation(s)
- Omkar Indari
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Shweta Jakhmola
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Devesh K. Pathak
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology Indore, Simrol, Indore 453552, India
- Department of Chemical Engineering, University of Seoul, Seoul 02504, Republic of Korea
| | - Manushree Tanwar
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Meenakshi Kandpal
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur 342011, India
| | - Rajesh Kumar
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| |
Collapse
|
46
|
Liao HY, Ran R, Da CM, Wang ZQ, Zhou KS, Zhang HH. Ski Regulates the Inflammatory Response of Reactive Astrocytes Induced by Oxygen Glucose Deprivation/Reoxygenation (OGD/R) Through the NF-κB Pathway. Neuroscience 2022; 490:250-263. [PMID: 35339646 DOI: 10.1016/j.neuroscience.2022.02.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/07/2022] [Accepted: 02/14/2022] [Indexed: 10/18/2022]
Abstract
Spinal cord injury (SCI) is a common disease of the nervous system, including primary and secondary injuries. Neuronal inflammation after SCI is the most important pathological process of SCI and a chemical barrier to nerve function recovery after injury. Ski, an evolutionarily conserved functional transcriptional regulator protein, is upregulated in reactive astrocytes after SCI and regulates the biological characteristics of astrocytes. However, its role in the glial inflammatory response triggered by reactive astrocytes after spinal cord ischemia and its exact mechanism remains unclear. This study investigated the role and mechanism of Ski in the inflammatory response triggered by reactive astrocytes induced by oxygen and sugar deprivation/reoxygenation (OGD/R) model in vitro. In the ODG/R model, Ski expression was upregulated. In contrast, Ski upregulation was accompanied by increased levels of iNOS, IL-1β, IL-6, TNF-α, and other inflammation-related factors. These results indicated that the inflammatory response triggered by astrocytes was significantly enhanced in OGD/R-stimulated astrocytes. Astrocytes were transfected with Ski specific siRNA to knock out Ski and subsequently attenuate OGD-induced astrocyte-triggered inflammation. Our results also suggest that Ski downregulation downregulates the expression of iNOS, IL-1β, IL-6, and TNF-α in OGD/R-induced reactive astrocytes by inhibiting the activity of the NF-κB signaling pathway. In conclusion, downregulation of Ski can effectively inhibit glial inflammation in SCI by inhibiting the activity of the NF-κB pathway. These findings suggest that Ski is a promising therapeutic target for inflammatory responses after SCI.In conclusion, Ski downregulation can effectively inhibit glial inflammation in SCI by inhibiting the activity of the NF-κB pathway. These findings suggest that Ski might serve as a promising target for the treatment of inflammatory responses after SCI.
Collapse
Affiliation(s)
- Hai-Yang Liao
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Rui Ran
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Chao-Ming Da
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China; Gansu Provincial Maternal and Child Health Hospital, 143 Qilihe North Street, Lanzhou 730050, PR China
| | - Zhi-Qiang Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Kai-Sheng Zhou
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China.
| |
Collapse
|
47
|
Vitamin D3 suppresses astrocyte activation and ameliorates coal dust-induced mood disorders in mice. J Affect Disord 2022; 303:138-147. [PMID: 35157949 DOI: 10.1016/j.jad.2022.02.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Pneumoconiosis patients exhibit significantly more anxiety and depression than healthy individuals. However, the mechanism of coal dust-induced anxiety and depression remains unclear. METHODS A pneumoconiosis mouse model with anxiety- and depression-like behaviors were established after 28 days of exposure to coal dust. Vitamin D3 treatment (1200 IU/kg/week) was administered intraperitoneally for 3 months starting from the first coal exposure. Tail suspension test (TST), open field test (OFT), and elevated plus-maze (EPM) test were used to assess anxiety- and depression-like behaviors. Theserum concentration of 25(OH)D3 and fibrillary acid protein (GFAP) expression were determined. In addition, the morphology and distribution of GFAP and neurogenic differentiation factor1 expression (NeuroD1) in different cerebral hippocampus were observed. RESULTS In coal dust-exposed mice, immobility time decreased in OFT and increased in TST,and the frequency of entering the open arm decreased in the EPM compared with the control mice. Coal dust increased hippocampal GFAP expression and astrocyte activation and reduced neurogenic differentiation factor1 expression (NeuroD1). In addition, Vitamin D3 significantly alleviated anxiety- and depressive-like behaviors in TST and EPM test, decreased GFAP expression level, modified hippocampal astrocyte activation pattern, and advanced brain-derived neurotrophic factor (BDNF) distribution and expression in CA1 and CA3 of the hippocampus. CONCLUSIONS Taken together, our results suggest that, by inhibiting the over-activation of astrocytes and increasing BDNF and neuron protection, vitamin D treatment ameliorates coal-dust-induced depressive and anxiety-like behavior, which is the first evidence that vitamin D may be a new approach for treating mood disorders caused by particulate matter.
Collapse
|
48
|
Mills WA, Woo AM, Jiang S, Martin J, Surendran D, Bergstresser M, Kimbrough IF, Eyo UB, Sofroniew MV, Sontheimer H. Astrocyte plasticity in mice ensures continued endfoot coverage of cerebral blood vessels following injury and declines with age. Nat Commun 2022; 13:1794. [PMID: 35379828 PMCID: PMC8980042 DOI: 10.1038/s41467-022-29475-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 03/11/2022] [Indexed: 01/30/2023] Open
Abstract
Astrocytes extend endfeet that enwrap the vasculature, and disruptions to this association which may occur in disease coincide with breaches in blood-brain barrier (BBB) integrity. Here we investigate if focal ablation of astrocytes is sufficient to disrupt the BBB in mice. Targeted two-photon chemical apoptotic ablation of astrocytes induced a plasticity response whereby surrounding astrocytes extended processes to cover vascular vacancies. In young animals, replacement processes occur in advance of endfoot retraction, but this is delayed in aged animals. Stimulation of replacement astrocytes results in constriction of pre-capillary arterioles, suggesting that replacement astrocytes are functional. Pharmacological inhibition of pSTAT3, as well as astrocyte specific deletion of pSTAT3, reduces astrocyte replacement post-ablation, without perturbations to BBB integrity. Similar endfoot replacement occurs following astrocyte cell death due to reperfusion in a stroke model. Together, these studies uncover the ability of astrocytes to maintain cerebrovascular coverage via substitution from nearby cells.
Collapse
Affiliation(s)
- William A. Mills
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XRobert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.438526.e0000 0001 0694 4940Graduate Program in Translational Biology, Medicine, & Health, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - AnnaLin M. Woo
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Shan Jiang
- grid.168010.e0000000419368956Department of Material Science and Engineering, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Joelle Martin
- grid.438526.e0000 0001 0694 4940Graduate Program in Translational Biology, Medicine, & Health, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Dayana Surendran
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Matthew Bergstresser
- grid.438526.e0000 0001 0694 4940School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Ian F. Kimbrough
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Ukpong B. Eyo
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XRobert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Michael V. Sofroniew
- grid.19006.3e0000 0000 9632 6718Department of Neurobiology, University of California, Los Angeles, CA USA
| | - Harald Sontheimer
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA
| |
Collapse
|
49
|
Bouvier DS, Fixemer S, Heurtaux T, Jeannelle F, Frauenknecht KBM, Mittelbronn M. The Multifaceted Neurotoxicity of Astrocytes in Ageing and Age-Related Neurodegenerative Diseases: A Translational Perspective. Front Physiol 2022; 13:814889. [PMID: 35370777 PMCID: PMC8969602 DOI: 10.3389/fphys.2022.814889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
In a healthy physiological context, astrocytes are multitasking cells contributing to central nervous system (CNS) homeostasis, defense, and immunity. In cell culture or rodent models of age-related neurodegenerative diseases (NDDs), such as Alzheimer’s disease (AD) and Parkinson’s disease (PD), numerous studies have shown that astrocytes can adopt neurotoxic phenotypes that could enhance disease progression. Chronic inflammatory responses, oxidative stress, unbalanced phagocytosis, or alteration of their core physiological roles are the main manifestations of their detrimental states. However, if astrocytes are directly involved in brain deterioration by exerting neurotoxic functions in patients with NDDs is still controversial. The large spectrum of NDDs, with often overlapping pathologies, and the technical challenges associated with the study of human brain samples complexify the analysis of astrocyte involvement in specific neurodegenerative cascades. With this review, we aim to provide a translational overview about the multi-facets of astrocyte neurotoxicity ranging from in vitro findings over mouse and human cell-based studies to rodent NDDs research and finally evidence from patient-related research. We also discuss the role of ageing in astrocytes encompassing changes in physiology and response to pathologic stimuli and how this may prime detrimental responses in NDDs. To conclude, we discuss how potentially therapeutic strategies could be adopted to alleviate or reverse astrocytic toxicity and their potential to impact neurodegeneration and dementia progression in patients.
Collapse
Affiliation(s)
- David S. Bouvier
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Systems Biomedicine (LCSB), University of Luxembourg (UL), Belvaux, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- *Correspondence: David S. Bouvier,
| | - Sonja Fixemer
- Luxembourg Center of Systems Biomedicine (LCSB), University of Luxembourg (UL), Belvaux, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
| | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Systems Biology Group, Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
| | - Félicia Jeannelle
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
| | - Katrin B. M. Frauenknecht
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Institute of Neuropathology, Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michel Mittelbronn
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Systems Biomedicine (LCSB), University of Luxembourg (UL), Belvaux, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Department of Cancer Research (DOCR), Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
- Faculty of Science, Technology, and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Michel Mittelbronn,
| |
Collapse
|
50
|
Archie SR, Sharma S, Burks E, Abbruscato T. Biological determinants impact the neurovascular toxicity of nicotine and tobacco smoke: A pharmacokinetic and pharmacodynamics perspective. Neurotoxicology 2022; 89:140-160. [PMID: 35150755 PMCID: PMC8958572 DOI: 10.1016/j.neuro.2022.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/30/2022] [Accepted: 02/05/2022] [Indexed: 01/01/2023]
Abstract
Accumulating evidence suggests that the detrimental effect of nicotine and tobacco smoke on the central nervous system (CNS) is caused by the neurotoxic role of nicotine on blood-brain barrier (BBB) permeability, nicotinic acetylcholine receptor expression, and the dopaminergic system. The ultimate consequence of these nicotine associated neurotoxicities can lead to cerebrovascular dysfunction, altered behavioral outcomes (hyperactivity and cognitive dysfunction) as well as future drug abuse and addiction. The severity of these detrimental effects can be associated with several biological determinants. Sex and age are two important biological determinants which can affect the pharmacokinetics and pharmacodynamics of several systemically available substances, including nicotine. With regard to sex, the availability of gonadal hormone is impacted by the pregnancy status and menstrual cycle resulting in altered metabolism rate of nicotine. Additionally, the observed lower smoking cessation rate in females compared to males is a consequence of differential effects of sex on pharmacokinetics and pharmacodynamics of nicotine. Similarly, age-dependent alterations in the pharmacokinetics and pharmacodynamics of nicotine have also been observed. One such example is related to severe vulnerability of adolescence towards addiction and long-term behavioral changes which may continue through adulthood. Considering the possible neurotoxic effects of nicotine on the central nervous system and the deterministic role of sex as well as age on these neurotoxic effects of smoking, it has become important to consider sex and age to study nicotine induced neurotoxicity and development of treatment strategies for combating possible harmful effects of nicotine. In the future, understanding the role of sex and age on the neurotoxic actions of nicotine can facilitate the individualization and optimization of treatment(s) to mitigate nicotine induced neurotoxicity as well as smoking cessation therapy. Unfortunately, however, no such comprehensive study is available which has considered both the sex- and age-dependent neurotoxicity of nicotine, as of today. Hence, the overreaching goal of this review article is to analyze and summarize the impact of sex and age on pharmacokinetics and pharmacodynamics of nicotine and possible neurotoxic consequences associated with nicotine in order to emphasize the importance of including these biological factors for such studies.
Collapse
Affiliation(s)
- Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Elizabeth Burks
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Thomas Abbruscato
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA.
| |
Collapse
|