1
|
Khalil I, Sayad R, Kedwany AM, Sayed HH, Caprara ALF, Rissardo JP. Cardiovascular dysautonomia and cognitive impairment in Parkinson's disease (Review). MEDICINE INTERNATIONAL 2024; 4:70. [PMID: 39355336 PMCID: PMC11443310 DOI: 10.3892/mi.2024.194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024]
Abstract
Cognitive impairment is a prevalent non-motor symptom of Parkinson's disease (PD), which can result in significant disability and distress for patients and caregivers. There is a marked variation in the timing, characteristics and rate at which cognitive decline occurs in patients with PD. This decline can vary from normal cognition to mild cognitive impairment and dementia. Cognitive impairment is associated with several pathophysiological mechanisms, including the accumulation of β-amyloid and tau in the brain, oxidative stress and neuroinflammation. Cardiovascular autonomic dysfunctions are commonly observed in patients with PD. These dysfunctions play a role in the progression of cognitive impairment, the incidents of falls and even in mortality. The majority of symptoms of dysautonomia arise from changes in the peripheral autonomic nervous system, including both the sympathetic and parasympathetic nervous systems. Cardiovascular changes, including orthostatic hypotension, supine hypertension and abnormal nocturnal blood pressure (BP), can occur in both the early and advanced stages of PD. These changes tend to increase as the disease advances. The present review aimed to describe the cognitive changes in the setting of cardiovascular dysautonomia and to discuss strategies through which these changes can be modified and managed. It is a multifactorial process usually involving decreased blood flow to the brain, resulting in the development of cerebral ischemic lesions, an increased presence of abnormal white matter signals in the brain, and a potential influence on the process of neurodegeneration in PD. Another possible explanation is this association being independent observations of PD progression. Patients with clinical symptoms of dysautonomia should undergo 24-h ambulatory BP monitoring, as they are frequently subtle and underdiagnosed.
Collapse
Affiliation(s)
- Ibrahim Khalil
- Faculty of Medicine, Alexandria University, Alexandria 5372066, Egypt
| | - Reem Sayad
- Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | | | - Hager Hamdy Sayed
- Department of Nuclear Medicine, Assuit University, Assuit 71515, Egypt
| | | | | |
Collapse
|
2
|
Kipp BT, Nunes PT, Savage LM. Dysregulation of neurotrophin expression in prefrontal cortex and nucleus basalis magnocellularis during and after adolescent intermittent ethanol exposure. Alcohol 2024; 120:1-14. [PMID: 38897258 PMCID: PMC11390331 DOI: 10.1016/j.alcohol.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
A preclinical model of human adolescent binge drinking, adolescent intermittent ethanol exposure (AIE) recreates the heavy binge withdrawal consummatory patterns of adolescents and has identified the loss of basal forebrain cholinergic neurons as a pathological hallmark of this model. Cholinergic neurons of the nucleus basalis magnocellularis (NbM) that innervate the prefrontal cortex (PFC) are particularly vulnerable to alcohol related neurodegeneration. Target derived neurotrophins (nerve growth factor [NGF] and brain-derived neurotrophic factor [BDNF]) regulate cholinergic phenotype expression and survival. Evidence from other disease models implicates the role of immature neurotrophin, or proneurotrophins, activity at neurotrophic receptors in promoting cholinergic degeneration; however, it has yet to be explored in adolescent binge drinking. We sought to characterize the pro- and mature neurotrophin expression, alongside their cognate receptors and cholinergic markers in an AIE model. Male and female Sprague Dawley rats underwent 5 g/kg 20% EtOH or water gavage on two-day-on, two-day-off cycles from post-natal day 25-57. Rats were sacrificed 2 h, 24 h, or 3 weeks following the last gavage, and tissue were collected for protein measurement. Western blot analyses revealed that ethanol intoxication reduced the expression of BDNF and vesicular acetylcholine transporter (vAChT) in the PFC, while NGF was lower in the NbM of AIE treated animals. During acute alcohol withdrawal, proNGF in the PFC was increased while proBDNF decreased, and in the NbM proBDNF increased while NGF decreased. During AIE abstinence, the expression of neurotrophins, their receptors, and vAChT did not differ from controls in the PFC. In contrast, in the NbM the expression of both NGF and choline acetyltransferase (ChAT) were reduced long-term following AIE. Taken together these findings suggest that AIE alters the expression of proneurotrophins and neurotrophins during intoxication and withdrawal that favor prodegenerative mechanisms by increasing the expression of proNGF and proBDNF, while also reducing NGF and BDNF.
Collapse
Affiliation(s)
- Brian T Kipp
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - Polliana T Nunes
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - Lisa M Savage
- Department of Psychology, Binghamton University of the State University of New York, New York, USA.
| |
Collapse
|
3
|
Jardine KH, Minard EP, Wideman CE, Edwards H, Abouelnaga KH, Messer WS, Winters BD. M1 muscarinic receptor activation reverses age-related memory updating impairment in mice. Neurobiol Aging 2024; 145:65-75. [PMID: 39489146 DOI: 10.1016/j.neurobiolaging.2024.10.007] [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: 04/15/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Previously consolidated memories can become temporarily labile upon reactivation. Reactivation-based memory updating is chiefly studied in young subjects, so we aimed to assess this process across the lifespan. To do this, we developed a behavioural paradigm wherein a reactivated object memory is updated with contextual information; 3-month-old and 6-month-old male C57BL/6 mice displayed object memory updating, but 12-month-old mice did not. We found that M1 muscarinic acetylcholine receptor signaling during reactivation was necessary for object memory updating in the young mice. Next, we targeted this mechanism in an attempt to facilitate object memory updating in aging mice. Remarkably, systemic pharmacological M1 receptor activation reversed the age-related deficit. Quantification of cholinergic system markers within perirhinal cortex revealed subtle cellular changes that may contribute to differential performance across age groups. These findings suggest that natural cholinergic change across the lifespan contributes to inflexible memory in the aging brain.
Collapse
Affiliation(s)
- Kristen H Jardine
- Department of Psychology and Collaborative Neuroscience Program, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| | - Emily P Minard
- Department of Psychology and Collaborative Neuroscience Program, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Cassidy E Wideman
- Department of Psychology and Collaborative Neuroscience Program, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Haley Edwards
- Department of Psychology and Collaborative Neuroscience Program, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Karim H Abouelnaga
- Department of Psychology and Collaborative Neuroscience Program, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - William S Messer
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, 2801 Bancroft Street, Toledo, OH 43606, United States
| | - Boyer D Winters
- Department of Psychology and Collaborative Neuroscience Program, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
4
|
Fecik MJ, Nunes PT, Vetreno RP, Savage LM. Voluntary wheel running exercise rescues behaviorally-evoked acetylcholine efflux in the medial prefrontal cortex and epigenetic changes in ChAT genes following adolescent intermittent ethanol exposure. PLoS One 2024; 19:e0311405. [PMID: 39436939 PMCID: PMC11495633 DOI: 10.1371/journal.pone.0311405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
Adolescent intermittent ethanol (AIE) exposure, which models heavy binge ethanol intake in adolescence, leads to a variety of deficits that persist into adulthood-including suppression of the cholinergic neuron phenotype within the basal forebrain. This is accompanied by a reduction in acetylcholine (ACh) tone in the medial prefrontal cortex (mPFC). Voluntary wheel running exercise (VEx) has been shown to rescue AIE-induced suppression of the cholinergic phenotype. Therefore, the goal of the current study is to determine if VEx will also rescue ACh efflux in the mPFC during spontaneous alternation, attention set shifting performance, and epigenetic silencing of the cholinergic phenotype following AIE. Male and female rats were subjected to 16 intragastric gavages of 20% ethanol or tap water on a two-day on/two-day off schedule from postnatal day (PD) 25-54, before being assigned to either VEx or stationary control groups. In Experiment 1, rats were tested on a four-arm spontaneous alternation maze with concurrent in vivo microdialysis for ACh in the mPFC. An operant attention set-shifting task was used to measure changes in cognitive and behavioral flexibility. In Experiment 2, a ChIP analysis of choline acetyltransferase (ChAT) genes was performed on basal forebrain tissue. It was found that VEx increased ACh efflux in the mPFC in both AIE and control male and female rats, as well as rescued the AIE-induced epigenetic methylation changes selectively at the Chat promoter CpG island across sexes. Overall, these data support the restorative effects of exercise on damage to the cholinergic projections to the mPFC and demonstrate the plasticity of cholinergic system for recovery after alcohol induced brain damage.
Collapse
Affiliation(s)
- Matthew J. Fecik
- Department of Psychology, Behavioral Neuroscience Area, Binghamton University-State University of New York, Binghamton, NY, United States of America
| | - Polliana T. Nunes
- Department of Psychology, Behavioral Neuroscience Area, Binghamton University-State University of New York, Binghamton, NY, United States of America
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Lisa M. Savage
- Department of Psychology, Behavioral Neuroscience Area, Binghamton University-State University of New York, Binghamton, NY, United States of America
| |
Collapse
|
5
|
Song Y, Gordon PC, Roy O, Metsomaa J, Belardinelli P, Rostami M, Ziemann U. Involvement of muscarinic acetylcholine receptor-mediated cholinergic neurotransmission in TMS-EEG responses. Prog Neuropsychopharmacol Biol Psychiatry 2024; 136:111167. [PMID: 39383933 DOI: 10.1016/j.pnpbp.2024.111167] [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: 06/24/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
The combination of transcranial magnetic stimulation and electroencephalography (TMS-EEG) is emerging as a valuable tool for investigating brain functions in health and disease. However, the detailed neural mechanisms underlying TMS-EEG responses, including TMS-evoked EEG potentials (TEPs) and TMS-induced EEG oscillations (TIOs), remain largely unknown. Combining TMS-EEG with pharmacological interventions provides a unique opportunity to elucidate the roles of specific receptor-mediated neurotransmissions in these responses. Here, we investigated the involvement of muscarinic acetylcholine receptor (mAChR)-mediated cholinergic neurotransmission in TMS-EEG responses by evaluating the effects of mAChR antagonists on TEPs and TIOs in twenty-four healthy participants using a randomized, placebo-controlled crossover design. TEPs and TIOs were measured before and after administering a single oral dose of scopolamine (a non-selective mAChR antagonist), biperiden (an M1 mAChR antagonist), or placebo, with TMS targeting the left medial prefrontal cortex (mPFC), angular gyrus (AG), and supplementary motor area (SMA). The results indicated that mAChR-mediated cholinergic neurotransmission played a role in TEPs, but not TIOs, in a target-specific manner. Specifically, scopolamine significantly increased the amplitude of a local TEP component between approximately 40 and 63 ms post-stimulus when TMS was applied to the SMA, but not the mPFC or AG. Biperiden produced a similar but less pronounced effect. Importantly, the effects of these mAChR antagonists on TEPs were independent of those on sensory-evoked EEG potentials caused by TMS-associated sensory stimulation. These findings expand our understanding of TMS-EEG physiology, providing insights for its application in physiological and clinical research.
Collapse
Affiliation(s)
- Yufei Song
- Department of Neurology & Stroke, University of Tübingen, Germany; Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Pedro C Gordon
- Department of Neurology & Stroke, University of Tübingen, Germany; Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Olivier Roy
- Department of Neurology & Stroke, University of Tübingen, Germany; Hertie Institute for Clinical Brain Research, University of Tübingen, Germany; CERVO Brain Research Centre, Quebec, Canada; Department of Psychiatry and Neurosciences, Université Laval, Quebec, Canada
| | - Johanna Metsomaa
- Department of Neuroscience and Biomedical Engineering, Aalto University School of Science, Finland
| | - Paolo Belardinelli
- Department of Neurology & Stroke, University of Tübingen, Germany; Hertie Institute for Clinical Brain Research, University of Tübingen, Germany; CIMeC, Center for Mind/Brain Sciences, University of Trento, Italy
| | - Maryam Rostami
- Faculty of Electrical and Computer Engineering, University of Tehran, Iran
| | - Ulf Ziemann
- Department of Neurology & Stroke, University of Tübingen, Germany; Hertie Institute for Clinical Brain Research, University of Tübingen, Germany.
| |
Collapse
|
6
|
van der Horn HJ, Vakhtin AA, Julio K, Nitschke S, Shaff N, Dodd AB, Erhardt E, Phillips JP, Pirio Richardson S, Deligtisch A, Stewart M, Suarez Cedeno G, Meles SK, Mayer AR, Ryman SG. Parkinson's disease cerebrovascular reactivity pattern: A feasibility study. J Cereb Blood Flow Metab 2024; 44:1774-1786. [PMID: 38578669 PMCID: PMC11494834 DOI: 10.1177/0271678x241241895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
A mounting body of research points to cerebrovascular dysfunction as a fundamental element in the pathophysiology of Parkinson's disease (PD). In the current feasibility study, blood-oxygen-level-dependent (BOLD) MRI was used to measure cerebrovascular reactivity (CVR) in response to hypercapnia in 26 PD patients and 16 healthy controls (HC), and aimed to find a multivariate pattern specific to PD. Whole-brain maps of CVR amplitude (i.e., magnitude of response to CO2) and latency (i.e., time to reach maximum amplitude) were computed, which were further analyzed using scaled sub-profile model principal component analysis (SSM-PCA) with leave-one-out cross-validation. A meaningful pattern based on CVR latency was identified, which was named the PD CVR pattern (PD-CVRP). This pattern was characterized by relatively increased latency in basal ganglia, sensorimotor cortex, supplementary motor area, thalamus and visual cortex, as well as decreased latency in the cerebral white matter, relative to HC. There were no significant associations with clinical measures, though sample size may have limited our ability to detect significant associations. In summary, the PD-CVRP highlights the importance of cerebrovascular dysfunction in PD, and may be a potential biomarker for future clinical research and practice.
Collapse
Affiliation(s)
- Harm Jan van der Horn
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Andrei A Vakhtin
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Kayla Julio
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Stephanie Nitschke
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Nicholas Shaff
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Andrew B Dodd
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Erik Erhardt
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, USA
| | - John P Phillips
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Sarah Pirio Richardson
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
- New Mexico VA Health Care System, Albuquerque, NM, USA
| | - Amanda Deligtisch
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Melanie Stewart
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Gerson Suarez Cedeno
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Sanne K Meles
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrew R Mayer
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Sephira G Ryman
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
7
|
Corbo I, Favieri F, Forte G, Casagrande M. Decision-making under uncertainty in healthy and cognitively impaired aging: A systematic review and meta-analysis. Arch Gerontol Geriatr 2024; 129:105643. [PMID: 39369563 DOI: 10.1016/j.archger.2024.105643] [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: 07/19/2024] [Revised: 09/07/2024] [Accepted: 09/22/2024] [Indexed: 10/08/2024]
Abstract
Decision-making (DM) is a complex cognitive behavior that involves gathering information and assessing options to identify choices under risky and uncertain conditions. Mild Cognitive Impairment (MCI) is a construct that includes a constellation of symptoms ranging from behavioral to cognitive impairments. This cluster of symptoms is frequently associated with poor decision-making. This study aimed to examine decision-making in pathological aging, specifically MCI. Therefore, we conducted a systematic review and meta-analysis to evaluate these relationships. According to the PRISMA 2020 Statement, nine studies were selected for the systematic review and eight for the meta-analysis. The results highlighted that MCI is associated with impaired decision-making in risky and ambiguous situations. The systematic review reported that MCI was associated with impaired decision-making in ambiguous and in risky conditions. In contrast, the meta-analysis showed significant differences in overall decision-making and particularly in ambiguous conditions. This difficulty may be due to different impairments that affect MCI. The difficulty in advantageous decision-making could be due to different brain alterations in MCI, which could lead to problems in tasks requiring feedback-based responses. These findings advance our understanding of decision-making in aging and suggest how decision-making alterations in MCI would affect the totality of executive functions and daily activities.
Collapse
Affiliation(s)
- Ilaria Corbo
- Department of Dynamic and Clinical Psychology, and Health Studies - "Sapienza" University of Rome, Italy.
| | - Francesca Favieri
- Department of Dynamic and Clinical Psychology, and Health Studies - "Sapienza" University of Rome, Italy.
| | - Giuseppe Forte
- Department of Dynamic and Clinical Psychology, and Health Studies - "Sapienza" University of Rome, Italy.
| | - Maria Casagrande
- Department of Dynamic and Clinical Psychology, and Health Studies - "Sapienza" University of Rome, Italy.
| |
Collapse
|
8
|
Dasgupta S, Pandya MA, Zanin JP, Liu T, Sun Q, Li H, Friedman WJ. ProNGF elicits retrograde axonal degeneration of basal forebrain neurons through p75 NTR and induction of amyloid precursor protein. Sci Signal 2024; 17:eadn2616. [PMID: 39316663 PMCID: PMC11487763 DOI: 10.1126/scisignal.adn2616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 08/15/2024] [Indexed: 09/26/2024]
Abstract
Basal forebrain cholinergic neurons (BFCNs) extend long projections to multiple regions in the brain to regulate cognitive functions. Degeneration of BFCNs is seen with aging, after brain injury, and in neurodegenerative disorders. An increase in the amount of the immature proform of nerve growth factor (proNGF) in the cerebral cortex results in retrograde degeneration of BFCNs through activation of proNGF receptor p75NTR. Here, we investigated the signaling cascades initiated at the axon terminal that mediate proNGF-induced retrograde degeneration. We found that local axonal protein synthesis and retrograde transport mediated proNGF-induced degeneration initiated from the axon terminal. Analysis of the nascent axonal proteome revealed that proNGF stimulation of axonal terminals triggered the synthesis of numerous proteins within the axon, and pathway analysis showed that amyloid precursor protein (APP) was a key upstream regulator in cultured BFCNs and in mice. Our findings reveal a functional role for APP in mediating BFCN axonal degeneration and cell death induced by proNGF.
Collapse
Affiliation(s)
- Srestha Dasgupta
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Mansi A. Pandya
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Juan P. Zanin
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Tong Liu
- New Jersey Medical School, Medical Science Building, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Qian Sun
- New Jersey Medical School, Medical Science Building, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Hong Li
- New Jersey Medical School, Medical Science Building, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Wilma J. Friedman
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| |
Collapse
|
9
|
Hosny EN, Sawie HG, El-Gizawy MM, Mohammed HS, Faraag AR, Khadrawy YA. Therapeutic Effects of Alpha Lipoic Acid and/or Caffeine-Loaded Chitosan Nanoparticles on Memory Impairment and Neurochemical Changes in High-Fat Diet-Induced Obese Rats. Physiol Behav 2024:114697. [PMID: 39288867 DOI: 10.1016/j.physbeh.2024.114697] [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: 10/07/2023] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
The therapeutic effects of alpha lipoic acid (LA) and/or caffeine-loaded chitosan nanoparticles (CCNPs) on obesity-induced memory impairment were evaluated in the present study. Rats were divided into control rats, obese rats induced by high fat diet (HFD) and obese rats treated with LA and/or CCNPs. Obesity was confirmed by measuring the body mass index (BMI). Memory and cognitive functions were evaluated by novel object recognition test (NORT). The levels of serotonin (5-HT), dopamine (DA), norepinephrine (NE), lipid peroxidation (MDA), nitric oxide (NO), reduced glutathione (GSH), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), leptin (LEP) and ghrelin (GHR) and the activities of monoamine oxidase (MAO), acetylcholinesterase (AchE) and Na+,K+,ATPase were determined in the cortex and hippocampus. The cerebral histopathological alterations were examined in obese rats. Obese rats showed impaired memory and exhibited significant neurochemical changes, including decreased levels of 5-HT, DA, GSH, GHR, and Na+,K+-ATPase activity, as well as an increase in AchE, MAO, MDA, NO, IL-1β, TNF-α, and LEP. LA and/or CCNPs treatment reduced BMI and improved memory. LA or CCNPs alleviated the cortical and hippocampal neurochemical changes and histopathological changes induced by obesity. Furthermore, LA and CCNPs exhibited antioxidant and anti-inflammatory properties, which likely contributed to their effects. However, no synergistic effect was observed between LA and CCNPs. These findings suggest that LA or CCNPs may be a potential therapy against obesity and its adverse effects on memory, mediated by their ability to restore monoamine levels and exhibit antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Eman N Hosny
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Hussein G Sawie
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Mayada M El-Gizawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Haitham S Mohammed
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Abdel Razik Faraag
- Pathology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt.
| |
Collapse
|
10
|
Moyano P, Flores A, San Juan J, García J, Anadón MJ, Plaza JC, Naval MV, Fernández MDLC, Guerra-Menéndez L, Del Pino J. Imidacloprid unique and repeated treatment produces cholinergic transmission disruption and apoptotic cell death in SN56 cells. Food Chem Toxicol 2024; 193:114988. [PMID: 39251036 DOI: 10.1016/j.fct.2024.114988] [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: 06/30/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Imidacloprid (IMI), the most widely used worldwide neonicotinoid biocide, produces cognitive disorders after repeated and single treatment. However, little was studied about the possible mechanisms that produce this effect. Cholinergic neurotransmission regulates cognitive function. Most cholinergic neuronal bodies are present in the basal forebrain (BF), regulating memory and learning process, and their dysfunction or loss produces cognition decline. BF SN56 cholinergic wild-type or acetylcholinesterase (AChE), β-amyloid-precursor-protein (βAPP), Tau, glycogen-synthase-kinase-3-beta (GSK3β), beta-site-amyloid-precursor-protein-cleaving enzyme 1 (BACE1), and/or nuclear-factor-erythroid-2-related-factor-2 (NRF2) silenced cells were treated for 1 and 14 days with IMI (1 μM-800 μM) with or without recombinant heat-shock-protein-70 (rHSP70), recombinant proteasome 20S (rP20S) and with or without N-acetyl-cysteine (NAC) to determine the possible mechanisms that mediate this effect. IMI treatment for 1 and 14 days altered cholinergic transmission through AChE inhibition, and triggered cell death partially through oxidative stress generation, AChE-S overexpression, HSP70 downregulation, P20S inhibition, and Aβ and Tau peptides accumulation. IMI produced oxidative stress through reactive oxygen species production and antioxidant NRF2 pathway downregulation, and induced Aβ and Tau accumulation through BACE1, GSK3β, HSP70, and P20S dysfunction. These results may assist in determining the mechanisms that produce cognitive dysfunction observed following IMI exposure and provide new therapeutic tools.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Andrea Flores
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Javier San Juan
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jimena García
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - María José Anadón
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Jose Carlos Plaza
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Maria Victoria Naval
- Department of Pharmacology, Pharmacognosy and Bothanic, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - María de la Cabeza Fernández
- Department of Chemistry and Pharmaceutical Sciences, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Lucía Guerra-Menéndez
- Department of Physiology, Medicine School, San Pablo CEU University, 28003 Madrid, Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain.
| |
Collapse
|
11
|
Görgülü I, Jagannath V, Pons S, Koniuszewski F, Groszer M, Maskos U, Huck S, Scholze P. The human-specific nicotinic receptor subunit CHRFAM7A reduces α7 receptor function in human induced pluripotent stem cells-derived and transgenic mouse neurons. Eur J Neurosci 2024; 60:4893-4906. [PMID: 39073048 DOI: 10.1111/ejn.16474] [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: 06/12/2024] [Accepted: 07/07/2024] [Indexed: 07/30/2024]
Abstract
We investigated the impact of the human-specific gene CHRFAM7A on the function of α7 nicotinic acetylcholine receptors (α7 nAChRs) in two different types of neurons: human-induced pluripotent stem cell (hiPSC)-derived cortical neurons, and superior cervical ganglion (SCG) neurons, taken from transgenic mice expressing CHRFAM7A. dupα7, the gene product of CHRFAM7A, which lacks a major part of the extracellular N-terminal ligand-binding domain, co-assembles with α7, the gene product of CHRNA7. We assessed the receptor function in hiPSC-derived cortical and SCG neurons with Fura-2 calcium imaging and three different α7-specific ligands: PNU282987, choline, and 4BP-TQS. Given the short-lived open state of α7 receptors, we combined the two orthosteric agonists PNU282987 and choline with the type-2 positive allosteric modulator (PAM II) PNU120596. In line with different cellular models used previously, we demonstrate that CHRFAM7A has a major impact on nicotinic α7 nAChRs by reducing calcium transients in response to all three agonists.
Collapse
Affiliation(s)
- Ilayda Görgülü
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Vinita Jagannath
- Institut du Fer à Moulin, Sorbonne University, UMR-S 1270, Paris, France
- MSD R&D Innovation Centre, London, UK
| | - Stephanie Pons
- Integrative Neurobiology of Cholinergic Systems, Institut Pasteur, Université Paris Cité, UMR 3571, Paris, France
| | - Filip Koniuszewski
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Matthias Groszer
- Institut du Fer à Moulin, Sorbonne University, UMR-S 1270, Paris, France
| | - Uwe Maskos
- Integrative Neurobiology of Cholinergic Systems, Institut Pasteur, Université Paris Cité, UMR 3571, Paris, France
| | - Sigismund Huck
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Petra Scholze
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Zhang Q, Singh P, Peng DW, Peng EY, Burns JM, Swerdlow RH, Suo WZ. Proactive M2 blockade prevents cognitive decline in GRK5-deficient APP transgenic mice via enhancing cholinergic neuronal resilience. J Biol Chem 2024; 300:107619. [PMID: 39098530 PMCID: PMC11400976 DOI: 10.1016/j.jbc.2024.107619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/09/2024] [Accepted: 07/14/2024] [Indexed: 08/06/2024] Open
Abstract
Alzheimer's disease (AD) poses an immense challenge in healthcare, lacking effective therapies. This study investigates the potential of anthranilamide derivative (AAD23), a selective M2 receptor antagonist, in proactively preventing cognitive impairments and cholinergic neuronal degeneration in G protein-coupled receptor kinase-5-deficient Swedish APP (GAP) mice. GAP mice manifest cognitive deficits by 7 months and develop senile plaques by 9 months. A 6-month AAD23 treatment was initiated at 5 months and stopped at 11 months before behavioral assessments without the treatment. AAD23-treated mice exhibited preserved cognitive abilities and improved cholinergic axonal health in the nucleus basalis of Meynert akin to wildtype mice. Conversely, vehicle-treated GAP mice displayed memory deficits and pronounced cholinergic axonal swellings in the nucleus basalis of Meynert. Notably, AAD23 treatment did not alter senile plaques and microgliosis. These findings highlight AAD23's efficacy in forestalling AD-related cognitive decline in G protein-coupled receptor kinase-5-deficient subjects, attributing its success to restoring cholinergic neuronal integrity and resilience, enhancing resistance against diverse degenerative insults.
Collapse
Affiliation(s)
- Qiang Zhang
- Laboratory for Alzheimer's Disease and Aging Research, Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, USA
| | - Prabhakar Singh
- Laboratory for Alzheimer's Disease and Aging Research, Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, USA
| | - David W Peng
- Laboratory for Alzheimer's Disease and Aging Research, Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, USA
| | - Evelyn Y Peng
- Laboratory for Alzheimer's Disease and Aging Research, Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, USA
| | - Jeffery M Burns
- Department of Neurology, University of Kansas Medical College, Kansas City, Kansas, USA; Department of Physiology, University of Kansas Medical College, Kansas City, Kansas, USA; The University of Kansas Alzheimer's Disease Center, Kansas City, Kansas, USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas Medical College, Kansas City, Kansas, USA; Department of Physiology, University of Kansas Medical College, Kansas City, Kansas, USA; The University of Kansas Alzheimer's Disease Center, Kansas City, Kansas, USA
| | - William Z Suo
- Laboratory for Alzheimer's Disease and Aging Research, Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, USA; Department of Neurology, University of Kansas Medical College, Kansas City, Kansas, USA; Department of Physiology, University of Kansas Medical College, Kansas City, Kansas, USA; The University of Kansas Alzheimer's Disease Center, Kansas City, Kansas, USA.
| |
Collapse
|
13
|
Damborsky JC, Yakel JL. Regulation of Hippocamposeptal Synaptic Transmission by GABA BRs Is Altered in 5XFAD Mice in a Sex- and Age-Dependent Manner. J Mol Neurosci 2024; 74:82. [PMID: 39212758 PMCID: PMC11364565 DOI: 10.1007/s12031-024-02260-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Hippocamposeptal (HS) neurons send GABAergic projections from the hippocampus to the medial septum/diagonal band of Broca (MS/DBB) as part of a reciprocal loop that is critical for memory. HS neurons are proposed to be particularly sensitive to the deleterious effects of pathological exposure to amyloid-β (Aβ), as would occur during Alzheimer's disease (AD). However, it is not known how HS GABA release in the MS/DBB is altered during the progression of AD. To target HS neurons in a mouse model of AD, we crossed SST-Cre mice to 5XFAD mice and performed stereotaxic injections of Cre-dependent AAV containing mCherry/channelrhodopsin-2 (ChR2) into the hippocampus of offspring at 4, 6, 9, and 12 months. We used optogenetics to selectively stimulate HS terminals while performing whole-cell patch-clamp recordings from MS/DBB neurons in slices. There was a transient reduction in HS-inhibitory postsynaptic current (IPSC) amplitude in female 5XFAD mice at 6 months, but no difference in males at any age, and no difference in paired-pulse ratio in either sex at any age. When bath applying the GABABR agonist, baclofen, we found a larger decrease in HS-IPSC amplitude in 5XFAD females at 9 months and 5XFAD males at 12 months. In 12-month-old 5XFAD females, response to baclofen was significantly reduced. These data suggest that there is a transient increase in responsiveness to GABABR activation in 5XFAD mice that occurs earlier in females than in males. These sex-specific changes to HS function are likely to impact the relay of information between the hippocampus and MS/DBB.
Collapse
Affiliation(s)
- Joanne C Damborsky
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 TW Alexander Dr., P.O. Box 12233, Mail Drop F2-08, Research Triangle Park, NC, 27709, USA
| | - Jerrel L Yakel
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 TW Alexander Dr., P.O. Box 12233, Mail Drop F2-08, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
14
|
Mei F, Zhao C, Li S, Xue Z, Zhao Y, Xu Y, Ye R, You H, Yu P, Han X, Carr GV, Weinberger DR, Yang F, Lu B. Ngfr + cholinergic projection from SI/nBM to mPFC selectively regulates temporal order recognition memory. Nat Commun 2024; 15:7342. [PMID: 39187496 PMCID: PMC11347598 DOI: 10.1038/s41467-024-51707-w] [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: 09/10/2023] [Accepted: 08/13/2024] [Indexed: 08/28/2024] Open
Abstract
Acetylcholine regulates various cognitive functions through broad cholinergic innervation. However, specific cholinergic subpopulations, circuits and molecular mechanisms underlying recognition memory remain largely unknown. Here we show that Ngfr+ cholinergic neurons in the substantia innominate (SI)/nucleus basalis of Meynert (nBM)-medial prefrontal cortex (mPFC) circuit selectively underlies recency judgements. Loss of nerve growth factor receptor (Ngfr-/- mice) reduced the excitability of cholinergic neurons in the SI/nBM-mPFC circuit but not in the medial septum (MS)-hippocampus pathway, and impaired temporal order memory but not novel object and object location recognition. Expression of Ngfr in Ngfr-/- SI/nBM restored defected temporal order memory. Fiber photometry revealed that acetylcholine release in mPFC not only predicted object encounters but also mediated recency judgments of objects, and such acetylcholine release was absent in Ngfr-/- mPFC. Chemogenetic and optogenetic inhibition of SI/nBM projection to mPFC in ChAT-Cre mice diminished mPFC acetylcholine release and deteriorated temporal order recognition. Impaired cholinergic activity led to a depolarizing shift of GABAergic inputs to mPFC pyramidal neurons, due to disturbed KCC2-mediated chloride gradients. Finally, potentiation of acetylcholine signaling upregulated KCC2 levels, restored GABAergic driving force and rescued temporal order recognition deficits in Ngfr-/- mice. Thus, NGFR-dependent SI/nBM-mPFC cholinergic circuit underlies temporal order recognition memory.
Collapse
Affiliation(s)
- Fan Mei
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Chen Zhao
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Shangjin Li
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Zeping Xue
- Basic and Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- School of Basic Medicine, Capital Medical University, Beijing, China
- Laboratory of Cognitive and Behavioral Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yueyang Zhao
- Basic and Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yihua Xu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Rongrong Ye
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - He You
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Peng Yu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Xinyu Han
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Gregory V Carr
- Department of Pharmacology and Molecular Sciences, Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel R Weinberger
- Department of Pharmacology and Molecular Sciences, Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Feng Yang
- Basic and Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
- Laboratory of Cognitive and Behavioral Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Conley AC, Vega JN, Johnson JV, Dumas JA, Newhouse PA. Effect of estradiol with or without micronized progesterone on cholinergic-related cognitive performance in postmenopausal women. Front Neurosci 2024; 18:1428675. [PMID: 39184322 PMCID: PMC11342399 DOI: 10.3389/fnins.2024.1428675] [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: 05/06/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction Women are at a higher risk of developing Alzheimer's disease (AD), and the decline in estrogens post-menopause is thought of as a factor increasing this risk. Estradiol (E2) is important in supporting cholinergic neuronal integrity, and cholinergic functioning may be negatively impacted following the loss of E2 post-menopause. The use of exogenous E2 has been observed to enhance cholinergically mediated cognitive performance in healthy post-menopausal women, which indicates a potentially protective mechanism. However, E2 is often co-administered with progestin or progesterone to prevent endometrial proliferation. Progesterone/progestins have previously been shown to have a detrimental effect on E2-mediated biological and cognitive effects mediated by cholinergic systems in preclinical models, therefore the present study aimed to assess whether progesterone would modify the effect of E2 to influence cognition during cholinergic blockade. Methods Twenty participants completed 3-months of oral E2 treatment with micronized progesterone (mPRO) or with placebo (PLC) in a repeated-measures within-subjects crossover design, in which they also completed five anticholinergic challenge days per hormone treatment condition. During the challenge participants were administered low or high doses of the nicotinic cholinergic antagonist mecamylamine, the muscarinic cholinergic antagonist scopolamine, or placebo. Following drug administration participants performed cognitive tests sensitive to cholinergic tone, assessing attention, episodic memory, and working memory. Results Significant decrements were found on some tasks when participants were taking E2+mPRO compared to E2 alone. Specifically, under more challenging task conditions and larger anticholinergic doses, participants showed poorer performance on the Critical Flicker Fusion task and the Stroop test and responded more conservatively on the N-back working memory task. Other tasks showed no differences between treatments under cholinergic blockade. Discussion The findings show that mPRO when taken in concert with E2, was detrimental to effortful cognitive performance, in the presence of cholinergic blockade. These results are important for assessing the impact of combined postmenopausal hormone treatment on cognitive performance that is dependent on cholinergic functioning after menopause.
Collapse
Affiliation(s)
- Alexander C. Conley
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jennifer N. Vega
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Julia V. Johnson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Julie A. Dumas
- Clinical Neuroscience Research Unit, Department of Psychiatry, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Paul A. Newhouse
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, United States
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Tennessee Valley Health System, Nashville, TN, United States
| |
Collapse
|
16
|
Luo F, Jiang L, Desai NS, Bai L, Watkins GV, Eldridge MAG, Plotnikova A, Mohanty A, Cummins AC, Averbeck BB, Talmage DA, Role LW. Comparative physiology and morphology of BLA-projecting NBM/SI cholinergic neurons in mouse and macaque. RESEARCH SQUARE 2024:rs.3.rs-4824445. [PMID: 39149491 PMCID: PMC11326416 DOI: 10.21203/rs.3.rs-4824445/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Cholinergic projection neurons of the nucleus basalis and substantia innominata (NBM/SI) densely innervate the basolateral amygdala (BLA) and have been shown to contribute to the encoding of fundamental and life-threatening experiences. Given the vital importance of these circuits in the acquisition and retention of memories that are essential for survival in a changing environment, it is not surprising that the basic anatomical organization of the NBM/SI is well conserved across animal classes as diverse as teleost and mammal. What is not known is the extent to which the physiology and morphology of NBM/SI neurons have also been conserved. To address this issue, we made patch-clamp recordings from NBM/SI neurons in ex vivo slices of two widely divergent mammalian species, mouse and rhesus macaque, focusing our efforts on cholinergic neurons that project to the BLA. We then reconstructed most of these recorded neurons post hoc to characterize neuronal morphology. We found that rhesus macaque BLA-projecting cholinergic neurons were both more intrinsically excitable and less morphologically compact than their mouse homologs. Combining measurements of 18 physiological features and 13 morphological features, we illustrate the extent of the separation. Although macaque and mouse neurons both exhibited considerable within-group diversity and overlapped with each other on multiple individual metrics, a combined morpho-electric analysis demonstrates that they form two distinct neuronal classes. Given the shared purpose of the circuits in which these neurons participate, this finding raises questions about (and offers constraints on) how these distinct classes result in similar behavior.
Collapse
Affiliation(s)
- Feng Luo
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Li Jiang
- Section on Genetics of Neuronal Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Niraj S. Desai
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Li Bai
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Gabrielle V. Watkins
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Mark A. G. Eldridge
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - Anya Plotnikova
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - Arya Mohanty
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - Alex C. Cummins
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - Bruno B. Averbeck
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - David A. Talmage
- Section on Genetics of Neuronal Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Lorna W. Role
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| |
Collapse
|
17
|
Shanazz K, Xie K, Oliver T, Bogan J, Vale F, Sword J, Kirov SA, Terry A, O'Herron P, Blake DT. Cortical Acetylcholine Response to Deep Brain Stimulation of the Basal Forebrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605828. [PMID: 39131297 PMCID: PMC11312592 DOI: 10.1101/2024.07.30.605828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Background Deep brain stimulation (DBS), the direct electrical stimulation of neuronal tissue in the basal forebrain to enhance release of the neurotransmitter acetylcholine, is under consideration as a method to improve executive function in patients with dementia. While some small studies indicate a positive response in the clinical setting, the relationship between DBS and acetylcholine pharmacokinetics is incompletely understood. Objective We examined the cortical acetylcholine response to different stimulation parameters of the basal forebrain. Methods 2-photon imaging was combined with deep brain stimulation. Stimulating electrodes were implanted in the subpallidal basal forebrain, and the ipsilateral somatosensory cortex was imaged. Acetylcholine activity was determined using the GRABACh-3.0 muscarinic acetylcholine receptor sensor, and blood vessels were imaged with Texas red. Results Experiments manipulating pulse train frequency demonstrated that integrated acetylcholine induced fluorescence was insensitive to frequency, and that peak levels were achieved with frequencies from 60 to 130 Hz. Altering pulse train length indicated that longer stimulation resulted in higher peaks and more activation with sublinear summation. The acetylcholinesterase inhibitor donepezil increased the peak response to 10s of stimulation at 60Hz, and the integrated response increased 57% with the 2 mg/kg dose, and 126% with the 4 mg/kg dose. Acetylcholine levels returned to baseline with a time constant of 14 to 18 seconds in all experiments. Conclusions These data demonstrate that acetylcholine receptor activation is insensitive to frequency between 60 and 130 Hz. High peak responses are achieved with up to 900 pulses. Donepezil increases total acetylcholine receptor activation associated with DBS but did not change temporal kinetics. The long time constants observed in the cerebral cortex add to the evidence supporting volume in addition to synaptic transmission.
Collapse
Affiliation(s)
- Khadijah Shanazz
- Dept of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Kun Xie
- Dept of Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Tucker Oliver
- Dept of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jamal Bogan
- Dept of Science and Mathematics, Augusta University, Augusta, GA
| | - Fernando Vale
- Dept of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jeremy Sword
- Dept of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Sergei A Kirov
- Dept of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
- Dept of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Alvin Terry
- Dept of Pharmacology and Toxicology , Medical College of Georgia, Augusta University, Augusta, GA
| | - Philip O'Herron
- Dept of Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - David T Blake
- Dept of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| |
Collapse
|
18
|
Bennett HC, Zhang Q, Wu YT, Manjila SB, Chon U, Shin D, Vanselow DJ, Pi HJ, Drew PJ, Kim Y. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. Nat Commun 2024; 15:6398. [PMID: 39080289 PMCID: PMC11289283 DOI: 10.1038/s41467-024-50559-8] [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: 06/09/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Aging is frequently associated with compromised cerebrovasculature and pericytes. However, we do not know how normal aging differentially impacts vascular structure and function in different brain areas. Here we utilize mesoscale microscopy methods and in vivo imaging to determine detailed changes in aged murine cerebrovascular networks. Whole-brain vascular tracing shows an overall ~10% decrease in vascular length and branching density with ~7% increase in vascular radii in aged brains. Light sheet imaging with 3D immunolabeling reveals increased arteriole tortuosity of aged brains. Notably, vasculature and pericyte densities show selective and significant reductions in the deep cortical layers, hippocampal network, and basal forebrain areas. We find increased blood extravasation, implying compromised blood-brain barrier function in aged brains. Moreover, in vivo imaging in awake mice demonstrates reduced baseline and on-demand blood oxygenation despite relatively intact neurovascular coupling. Collectively, we uncover regional vulnerabilities of cerebrovascular network and physiological changes that can mediate cognitive decline in normal aging.
Collapse
Affiliation(s)
- Hannah C Bennett
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Qingguang Zhang
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Neurosurgery, Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Steffy B Manjila
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Uree Chon
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Neurosciences Graduate Program, Stanford University, Stanford, CA, 94305, USA
| | - Donghui Shin
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Daniel J Vanselow
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Hyun-Jae Pi
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Patrick J Drew
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, Biology, and Neurosurgery, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA.
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
19
|
Xia Y, Dore V, Fripp J, Bourgeat P, Laws SM, Fowler CJ, Rainey-Smith SR, Martins RN, Rowe C, Masters CL, Coulson EJ, Maruff P. Association of Basal Forebrain Atrophy With Cognitive Decline in Early Alzheimer Disease. Neurology 2024; 103:e209626. [PMID: 38885444 PMCID: PMC11254448 DOI: 10.1212/wnl.0000000000209626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/09/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVES In early Alzheimer disease (AD), β-amyloid (Aβ) deposition is associated with volume loss in the basal forebrain (BF) and cognitive decline. However, the extent to which Aβ-related BF atrophy manifests as cognitive decline is not understood. This study sought to characterize the relationship between BF atrophy and the decline in memory and attention in patients with early AD. METHODS Participants from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study who completed Aβ-PET imaging and repeated MRI and cognitive assessments were included. At baseline, participants were classified based on their clinical dementia stage and Aβ status, yielding groups that were cognitively unimpaired (CU) Aβ-, CU Aβ+, and mild cognitive impairment (MCI) Aβ+. Linear mixed-effects models were used to assess changes in volumetric measures of BF subregions and the hippocampus and changes in AIBL memory and attention composite scores for each group compared with CU Aβ- participants. Associations between Aβ burden, brain atrophy, and cognitive decline were evaluated and explored further using mediation analyses. RESULTS The cohort included 476 participants (72.6 ± 5.9 years, 55.0% female) with longitudinal data from a median follow-up period of 6.1 years. Compared with the CU Aβ- group (n = 308), both CU Aβ+ (n = 107) and MCI Aβ+ (n = 61) adults showed faster decline in BF and hippocampal volumes and in memory and attention (Cohen d = 0.73-1.74). Rates of atrophy in BF subregions and the hippocampus correlated with cognitive decline, and each individually mediated the impact of Aβ burden on memory and attention decline. When all mediators were considered simultaneously, hippocampal atrophy primarily influenced the effect of Aβ burden on memory decline (β [SE] = -0.139 [0.032], proportion mediated [PM] = 28.0%) while the atrophy of the posterior nucleus basalis of Meynert in the BF (β [SE] = -0.068 [0.029], PM = 13.1%) and hippocampus (β [SE] = -0.121 [0.033], PM = 23.4%) distinctively influenced Aβ-related attention decline. DISCUSSION These findings highlight the significant role of BF atrophy in the complex pathway linking Aβ to cognitive impairment in early stages of AD. Volumetric assessment of BF subregions could be essential in elucidating the relationships between the brain structure and behavior in AD.
Collapse
Affiliation(s)
- Ying Xia
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Vincent Dore
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Jurgen Fripp
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Pierrick Bourgeat
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Simon M Laws
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Christopher J Fowler
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Stephanie R Rainey-Smith
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Ralph N Martins
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Christopher Rowe
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Colin L Masters
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Elizabeth J Coulson
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Paul Maruff
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| |
Collapse
|
20
|
Sundman MH, Green JM, Fuglevand AJ, Chou YH. TMS-derived short afferent inhibition discriminates cognitive status in older adults without dementia. AGING BRAIN 2024; 6:100123. [PMID: 39132326 PMCID: PMC11315225 DOI: 10.1016/j.nbas.2024.100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 06/29/2024] [Accepted: 07/14/2024] [Indexed: 08/13/2024] Open
Abstract
Aging is a complex and diverse biological process characterized by progressive molecular, cellular, and tissue damage, resulting in a loss of physiological integrity and heightened vulnerability to pathology. This biological diversity corresponds with highly variable cognitive trajectories, which are further confounded by genetic and environmental factors that influence the resilience of the aging brain. Given this complexity, there is a need for neurophysiological indicators that not only discern physiologic and pathologic aging but also closely align with cognitive trajectories. Transcranial Magnetic Stimulation (TMS) may have utility in this regard as a non-invasive brain stimulation tool that can characterize features of cortical excitability. Particularly, as a proxy for central cholinergic function, short-afferent inhibition (SAI) dysfunction is robustly associated with cognitive deficits in the latter stages of Alzheimer's Disease and Related Dementia (ADRD). In this study, we evaluated SAI in healthy young adults and older adults who, though absent clinical diagnoses, were algorithmically classified as cognitively normal (CN) or cognitively impaired (CI) according to the Jak/Bondi actuarial criteria. We report that SAI is preserved in the Old-CN cohort relative to the young adults, and SAI is significantly diminished in the Old-CI cohort relative to both young and CN older adults. Additionally, diminished SAI was significantly associated with impaired sustained attention and working memory. As a proxy measure for central cholinergic deficits, we discuss the potential value of SAI for discerning physiological and pathological aging.
Collapse
Affiliation(s)
- Mark H. Sundman
- Brain Imaging and TMS Laboratory, Department of Psychology, University of Arizona, Tucson, AZ 85721, USA
| | - Jacob M. Green
- Brain Imaging and TMS Laboratory, Department of Psychology, University of Arizona, Tucson, AZ 85721, USA
| | - Andrew J. Fuglevand
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ 85721, USA
- Department of Neuroscience, College of Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - Ying-hui Chou
- Brain Imaging and TMS Laboratory, Department of Psychology, University of Arizona, Tucson, AZ 85721, USA
- Evelyn F McKnight Brain Institute, Arizona Center on Aging, and BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
21
|
Moschonas EH, Capeci HE, Annas EM, Domyslawski VB, Steber JA, Donald HM, Genkinger NR, Rennerfeldt PL, Bittner RA, Vozzella VJ, Cheng JP, Kline AE, Bondi CO. Evaluating the Efficacy of Chronic Galantamine on Sustained Attention and Cholinergic Neurotransmission in A Pre-Clinical Model of Traumatic Brain Injury. J Neurotrauma 2024. [PMID: 38994598 DOI: 10.1089/neu.2024.0173] [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: 07/13/2024] Open
Abstract
Cholinergic disruptions underlie attentional deficits following traumatic brain injury (TBI). Yet, drugs specifically targeting acetylcholinesterase (AChE) inhibition have yielded mixed outcomes. Therefore, we hypothesized that galantamine (GAL), a dual-action competitive AChE inhibitor and α7 nicotinic acetylcholine receptor (nAChR) positive allosteric modulator, provided chronically after injury, will attenuate TBI-induced deficits of sustained attention and enhance ACh efflux in the medial prefrontal cortex (mPFC), as assessed by in vivo microdialysis. In Experiment 1, adult male rats (n = 10-15/group) trained in the 3-choice serial reaction time (3-CSRT) test were randomly assigned to controlled cortical impact (CCI) or sham surgery and administered GAL (0.5, 2.0, or 5.0 mg/kg; i.p.) or saline vehicle (VEH; 1 mL/kg; i.p) beginning 24-h post-surgery and once daily thereafter for 27 days. Measures of sustained attention and distractibility were assessed on post-operative days 21-25 in the 3-CSRT, following which cortical lesion volume and basal forebrain cholinergic cells were quantified on day 27. In Experiment 2, adult male rats (n = 3-4/group) received a CCI and 24 h later administered (i.p.) one of the three doses of GAL or VEH for 21 days to quantify the dose-dependent effect of GAL on in vivo ACh efflux in the mPFC. Two weeks after the CCI, a guide cannula was implanted in the right mPFC. On post-surgery day 21, baseline and post-injection dialysate samples were collected in a temporally matched manner with the cohort undergoing behavior. ACh levels were analyzed using reverse phase high-performance liquid chromatography (HPLC) coupled to an electrochemical detector. Cortical lesion volume was quantified on day 22. The data were subjected to ANOVA, with repeated measures where appropriate, followed by Newman-Keuls post hoc analyses. All TBI groups displayed impaired sustained attention versus the pooled SHAM controls (p's < 0.05). Moreover, the highest dose of GAL (5.0 mg/kg) exacerbated attentional deficits relative to VEH and the two lower doses of GAL (p's < 0.05). TBI significantly reduced cholinergic cells in the right basal forebrain, regardless of treatment condition, versus SHAM (p < 0.05). In vivo microdialysis revealed no differences in basal ACh in the mPFC; however, GAL (5.0 mg/kg) significantly increased ACh efflux 30 min following injection compared to the VEH and the other GAL (0.5 and 2.0 mg/kg) treated groups (p's < 0.05). In both experiments, there were no differences in cortical lesion volume across treatment groups (p's > 0.05). In summary, albeit the higher dose of GAL increased ACh release, it did not improve measures of sustained attention or histopathological markers, thereby partially supporting the hypothesis and providing the impetus for further investigations into alternative cholinergic pharmacotherapies such as nAChR positive allosteric modulators.
Collapse
Affiliation(s)
- Eleni H Moschonas
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Haley E Capeci
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ellen M Annas
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Veronica B Domyslawski
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jade A Steber
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hailey M Donald
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nicholas R Genkinger
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Piper L Rennerfeldt
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rachel A Bittner
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Vincent J Vozzella
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jeffrey P Cheng
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anthony E Kline
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Corina O Bondi
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
22
|
Xu D, Yu X, Hu J, Yu Y, Wang L, Jiang B, Zhang M. White matter hyperintensities in cholinergic pathways correlates of cognitive impairment in moyamoya disease. Eur Radiol 2024; 34:4596-4606. [PMID: 38092950 DOI: 10.1007/s00330-023-10489-3] [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: 05/18/2023] [Revised: 09/24/2023] [Accepted: 10/28/2023] [Indexed: 06/29/2024]
Abstract
OBJECTIVE To investigate the effect of cholinergic pathways damage caused by white matter hyperintensities (WMHs) on cognitive function in moyamoya disease (MMD). METHODS We included 62 patients with MMD from a prospectively enrolled cohort. We evaluated the burden of cholinergic pathways damage caused by WMHs using the Cholinergic Pathways Hyperintensities Scale (CHIPS). Cognitive function was evaluated with the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA). Cognitive impairment was determined according to the cut-off of MMSE and education. Multivariate linear and logistic regression models were used to analyze whether CHIPS was independently associated with cognition. Receiver operating characteristic curve analysis was performed to identify the ability of CHIPS in discriminating cognitive impairment and normal cognition. RESULTS CHIPS was associated with both MMSE and MoCA (β = - 0.601 and β = - 0.672, both p < 0.001). After correcting age, sex, education, volumes of limbic areas, and other factors, CHIPS remained to be independently associated with both MMSE and MoCA (β = - 0.388 and β = - 0.334, both p < 0.001). In the logistic regression, only CHIPS was associated with cognitive impairment (odds ratio = 1.431, 95% confidence interval = 1.103 to 1.856, p = 0.007). The optimal cut-off of CHIPS score was 10, yielding a sensitivity of 87.5% and a specificity of 78.3% in identifying MMD patients with cognitive impairment. CONCLUSIONS The damage of cholinergic pathways caused by WMHs plays an independent effect on cognition and CHIPS could be a useful method in identifying MMD patients likely to be cognitive impairment. CLINICAL RELEVANCE STATEMENT This study shows that Cholinergic Pathways Hyperintensities Scale (CHIPS) could be a simple and reliable method in identifying cognitive impairment for patients with moyamoya disease. CHIPS could be helpful in clinical practice, such as guiding treatment decisions and predicting outcome. KEY POINTS • Cholinergic Pathways Hyperintensities Scale was significantly associated with cognitive screening tests in patients with moyamoya disease. • Cholinergic Pathways Hyperintensities Scale plays an independent effect on cognitive impairment in patients with moyamoya disease. • Cholinergic Pathways Hyperintensities Scale shows higher accuracy than education, volumes of limbic areas, and sex in identifying cognitive impairment in moyamoya disease.
Collapse
Affiliation(s)
- Duo Xu
- Department of Radiology, The 2nd Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinfeng Yu
- Department of Radiology, The 2nd Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junwen Hu
- Department of Neurosurgery, The 2nd Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yannan Yu
- Department of Radiology, UCSF, San Francisco, CA, USA
| | - Lin Wang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Biao Jiang
- Department of Radiology, The 2nd Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The 2nd Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
23
|
Voegtle A, Mohrbutter C, Hils J, Schulz S, Weuthen A, Brämer U, Ullsperger M, Sweeney-Reed CM. Cholinergic modulation of motor sequence learning. Eur J Neurosci 2024; 60:3706-3718. [PMID: 38716689 DOI: 10.1111/ejn.16374] [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: 10/12/2023] [Revised: 04/04/2024] [Accepted: 04/16/2024] [Indexed: 07/06/2024]
Abstract
The cholinergic system plays a key role in motor function, but whether pharmacological modulation of cholinergic activity affects motor sequence learning is unknown. The acetylcholine receptor antagonist biperiden, an established treatment in movement disorders, reduces attentional modulation, but whether it influences motor sequence learning is not clear. Using a randomized, double-blind placebo-controlled crossover design, we tested 30 healthy young participants and showed that biperiden impairs the ability to learn sequential finger movements, accompanied by widespread oscillatory broadband power changes (4-25 Hz) in the motor sequence learning network after receiving biperiden, with greater power in the theta, alpha and beta bands over ipsilateral motor and bilateral parietal-occipital areas. The reduced early theta power during a repeated compared with random sequence, likely reflecting disengagement of top-down attention to sensory processes, was disrupted by biperiden. Alpha synchronization during repeated sequences reflects sensory gating and lower visuospatial attention requirements compared with visuomotor responses to random sequences. After biperiden, alpha synchronization was greater, potentially reflecting excessive visuospatial attention reduction, affecting visuomotor responding required to enable sequence learning. Beta oscillations facilitate sequence learning by integrating visual and somatosensory inputs, stabilizing repeated sequences and promoting prediction of the next stimulus. The beta synchronization after biperiden fits with a disruption of the selective visuospatial attention enhancement associated with initial sequence learning. These findings highlight the role of cholinergic processes in motor sequence learning.
Collapse
Affiliation(s)
- Angela Voegtle
- Neurocybernetics and Rehabilitation, Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Catharina Mohrbutter
- Neurocybernetics and Rehabilitation, Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Jonathan Hils
- Institute of Psychology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Steve Schulz
- Institute of Psychology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Alexander Weuthen
- Institute of Psychology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Uwe Brämer
- Institute of Psychology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Markus Ullsperger
- Institute of Psychology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Catherine M Sweeney-Reed
- Neurocybernetics and Rehabilitation, Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
24
|
Oliver Goral R, Lamb PW, Yakel JL. Acetylcholine Neurons Become Cholinergic during Three Time Windows in the Developing Mouse Brain. eNeuro 2024; 11:ENEURO.0542-23.2024. [PMID: 38942474 PMCID: PMC11253243 DOI: 10.1523/eneuro.0542-23.2024] [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: 12/19/2023] [Revised: 06/12/2024] [Accepted: 06/22/2024] [Indexed: 06/30/2024] Open
Abstract
Acetylcholine (ACh) neurons in the central nervous system are required for the coordination of neural network activity during higher brain functions, such as attention, learning, and memory, as well as locomotion. Disturbed cholinergic signaling has been described in many neurodevelopmental and neurodegenerative disorders. Furthermore, cotransmission of other signaling molecules, such as glutamate and GABA, with ACh has been associated with essential roles in brain function or disease. However, it is unknown when ACh neurons become cholinergic during development. Thus, understanding the timeline of how the cholinergic system develops and becomes active in the healthy brain is a crucial part of understanding brain development. To study this, we used transgenic mice to selectively label ACh neurons with tdTomato. We imaged serial sectioned brains and generated whole-brain reconstructions at different time points during pre- and postnatal development. We found three crucial time windows-two in the prenatal and one in the postnatal brain-during which most ACh neuron populations become cholinergic in the brain. We also found that cholinergic gene expression is initiated in cortical ACh interneurons, while the cerebral cortex is innervated by cholinergic projection neurons from the basal forebrain. Taken together, we show that ACh neuron populations are present and become cholinergic before postnatal day 12, which is the onset of major sensory processes, such as hearing and vision. We conclude that the birth of ACh neurons and initiation of cholinergic gene expression are temporally separated during development but highly coordinated by brain anatomical structure.
Collapse
Affiliation(s)
- Rene Oliver Goral
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
- Center on Compulsive Behaviors, National Institutes of Health, Bethesda, Maryland 20892
| | - Patricia W Lamb
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Jerrel L Yakel
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| |
Collapse
|
25
|
Nguyen HTM, van der Westhuizen ET, Langmead CJ, Tobin AB, Sexton PM, Christopoulos A, Valant C. Opportunities and challenges for the development of M 1 muscarinic receptor positive allosteric modulators in the treatment for neurocognitive deficits. Br J Pharmacol 2024; 181:2114-2142. [PMID: 36355830 DOI: 10.1111/bph.15982] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/22/2022] [Accepted: 10/18/2022] [Indexed: 11/12/2022] Open
Abstract
Targeting allosteric sites of M1 muscarinic acetylcholine receptors (M1 receptors) is a promising strategy to treat neurocognitive disorders, such as Alzheimer's disease and schizophrenia. Indeed, the last two decades have seen an impressive body of work focussing on the design and development of positive allosteric modulators (PAMs) for the M1 receptor. This has led to the identification of a structurally diverse range of highly selective M1 PAMs. In preclinical models, M1 PAMs have shown rescue of cognitive deficits and improvement of endpoints predictive of symptom domains of schizophrenia. Yet, to date only a few M1 PAMs have reached early-stage clinical trials, with many of them failing to progress further due to on-target mediated cholinergic adverse effects that have plagued the development of this class of ligand. This review covers the recent preclinical and clinical studies in the field of M1 receptor drug discovery for the treatment of Alzheimer's disease and schizophrenia, with a specific focus on M1 PAM, highlighting both the undoubted potential but also key challenges for the successful translation of M1 PAMs from bench-side to bedside. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Huong T M Nguyen
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- Department of Biochemistry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | | | - Christopher J Langmead
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Melbourne, VIC, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, Melbourne, VIC, Australia
| | - Andrew B Tobin
- Centre for Translational Pharmacology, University of Glasgow, Glasgow, UK
| | - Patrick M Sexton
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, Melbourne, VIC, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Melbourne, VIC, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, Melbourne, VIC, Australia
| | - Celine Valant
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Melbourne, VIC, Australia
| |
Collapse
|
26
|
Kang Y, Toyoda H, Saito M. Search for unknown neural link between the masticatory and cognitive brain systems to clarify the involvement of its impairment in the pathogenesis of Alzheimer's disease. Front Cell Neurosci 2024; 18:1425645. [PMID: 38994328 PMCID: PMC11236757 DOI: 10.3389/fncel.2024.1425645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
Brain degenerations in sporadic Alzheimer's disease (AD) are observed earliest in the locus coeruleus (LC), a population of noradrenergic neurons, in which hyperphosphorylated tau protein expression and β-amyloid (Aβ) accumulation begin. Along with this, similar changes occur in the basal forebrain cholinergic neurons, such as the nucleus basalis of Meynert. Neuronal degeneration of the two neuronal nuclei leads to a decrease in neurotrophic factors such as brain-derived neurotrophic factor (BDNF) in the hippocampus and cerebral cortex, which results in the accumulation of Aβ and hyperphosphorylated tau protein and ultimately causes neuronal cell death in those cortices. On the other hand, a large number of epidemiological studies have shown that tooth loss or masticatory dysfunction is a risk factor for dementia including AD, and numerous studies using experimental animals have also shown that masticatory dysfunction causes brain degeneration in the basal forebrain, hippocampus, and cerebral cortex similar to those observed in human AD, and that learning and memory functions are impaired accordingly. However, it remains unclear how masticatory dysfunction can induce such brain degeneration similar to AD, and the neural mechanism linking the trigeminal nervous system responsible for mastication and the cognitive and memory brain system remains unknown. In this review paper, we provide clues to the search for such "missing link" by discussing the embryological, anatomical, and physiological relationship between LC and its laterally adjoining mesencephalic trigeminal nucleus which plays a central role in the masticatory functions.
Collapse
Affiliation(s)
- Youngnam Kang
- Department of Behavioral Physiology, Osaka University Graduate School of Human Sciences, Osaka, Japan
| | - Hiroki Toyoda
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Mitsuru Saito
- Department of Oral Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
27
|
Zaborszky L, Varsanyi P, Alloway K, Chavez C, Gielow M, Gombkoto P, Kondo H, Nadasdy Z. Functional architecture of the forebrain cholinergic system in rodents. RESEARCH SQUARE 2024:rs.3.rs-4504727. [PMID: 38947053 PMCID: PMC11213185 DOI: 10.21203/rs.3.rs-4504727/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The basal forebrain cholinergic system (BFCS) participates in functions that are global across the brain, such as sleep-wake cycles, but also participates in capacities that are more behaviorally and anatomically specific, including sensory perception. To better understand the underlying organization principles of the BFCS, more and higher quality anatomical data and analysis is needed. Here, we created a "virtual Basal Forebrain", combining data from numerous rats with cortical retrograde tracer injections into a common 3D reference coordinate space and developed a "spatial density correlation" methodology to analyze patterns in BFCS cortical projection targets, revealing that the BFCS is organized into three principal networks: somatosensory-motor, auditory, and visual. Within each network, clusters of cholinergic cells with increasing complexity innervate cortical targets. These networks represent hierarchically organized building blocks that may enable the BFCS to coordinate spatially selective signaling, including parallel modulation of multiple functionally interconnected yet diverse groups of cortical areas.
Collapse
Affiliation(s)
| | | | | | | | | | - Peter Gombkoto
- Swiss Federal Institute of Technology in Zurich (ETH Zurich)
| | | | | |
Collapse
|
28
|
van Velthoven CTJ, Gao Y, Kunst M, Lee C, McMillen D, Chakka AB, Casper T, Clark M, Chakrabarty R, Daniel S, Dolbeare T, Ferrer R, Gloe J, Goldy J, Guzman J, Halterman C, Ho W, Huang M, James K, Nguy B, Pham T, Ronellenfitch K, Thomas ED, Torkelson A, Pagan CM, Kruse L, Dee N, Ng L, Waters J, Smith KA, Tasic B, Yao Z, Zeng H. The transcriptomic and spatial organization of telencephalic GABAergic neuronal types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599583. [PMID: 38948843 PMCID: PMC11212977 DOI: 10.1101/2024.06.18.599583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The telencephalon of the mammalian brain comprises multiple regions and circuit pathways that play adaptive and integrative roles in a variety of brain functions. There is a wide array of GABAergic neurons in the telencephalon; they play a multitude of circuit functions, and dysfunction of these neurons has been implicated in diverse brain disorders. In this study, we conducted a systematic and in-depth analysis of the transcriptomic and spatial organization of GABAergic neuronal types in all regions of the mouse telencephalon and their developmental origins. This was accomplished by utilizing 611,423 single-cell transcriptomes from the comprehensive and high-resolution transcriptomic and spatial cell type atlas for the adult whole mouse brain we have generated, supplemented with an additional single-cell RNA-sequencing dataset containing 99,438 high-quality single-cell transcriptomes collected from the pre- and postnatal developing mouse brain. We present a hierarchically organized adult telencephalic GABAergic neuronal cell type taxonomy of 7 classes, 52 subclasses, 284 supertypes, and 1,051 clusters, as well as a corresponding developmental taxonomy of 450 clusters across different ages. Detailed charting efforts reveal extraordinary complexity where relationships among cell types reflect both spatial locations and developmental origins. Transcriptomically and developmentally related cell types can often be found in distant and diverse brain regions indicating that long-distance migration and dispersion is a common characteristic of nearly all classes of telencephalic GABAergic neurons. Additionally, we find various spatial dimensions of both discrete and continuous variations among related cell types that are correlated with gene expression gradients. Lastly, we find that cortical, striatal and some pallidal GABAergic neurons undergo extensive postnatal diversification, whereas septal and most pallidal GABAergic neuronal types emerge simultaneously during the embryonic stage with limited postnatal diversification. Overall, the telencephalic GABAergic cell type taxonomy can serve as a foundational reference for molecular, structural and functional studies of cell types and circuits by the entire community.
Collapse
Affiliation(s)
| | - Yuan Gao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mike Huang
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Beagan Nguy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
29
|
Li Z, Ma Q, Deng Y, Rolls ET, Shen C, Li Y, Zhang W, Xiang S, Langley C, Sahakian BJ, Robbins TW, Yu JT, Feng J, Cheng W. Irritable Bowel Syndrome Is Associated With Brain Health by Neuroimaging, Behavioral, Biochemical, and Genetic Analyses. Biol Psychiatry 2024; 95:1122-1132. [PMID: 38199582 DOI: 10.1016/j.biopsych.2023.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/14/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) interacts with psychopathology in a complex way; however, little is known about the underlying brain, biochemical, and genetic mechanisms. METHODS To clarify the phenotypic and genetic associations between IBS and brain health, we performed a comprehensive retrospective cohort study on a large population. Our study included 171,104 participants from the UK Biobank who underwent a thorough assessment of IBS, with the majority also providing neuroimaging, behavioral, biochemical, and genetic information. Multistage linked analyses were conducted, including phenome-wide association analysis, polygenic risk score calculation, and 2-sample Mendelian randomization analysis. RESULTS The phenome-wide association analysis showed that IBS was linked to brain health problems, including anxiety and depression, and poor cognitive performance. Significantly lower brain volumes associated with more severe IBS were found in key areas related to emotional regulation and higher-order cognition, including the medial orbitofrontal cortex/ventromedial prefrontal cortex, anterior insula, anterior and mid-cingulate cortices, dorsolateral prefrontal cortex, and hippocampus. Higher triglycerides, lower high-intensity lipoprotein, and lower platelets were also related (p < 1 × 10-10) to more severe IBS. Finally, Mendelian randomization analyses demonstrated potential causal relationships between IBS and brain health and indicated possible mediating effects of dyslipidemia and inflammation. CONCLUSIONS For the first time, this study provides a comprehensive understanding of the relationship between IBS and brain health phenotypes, integrating perspectives from neuroimaging, behavioral performance, biochemical factors, and genetics, which is of great significance for clinical applications to potentially address brain health impairments in patients with IBS.
Collapse
Affiliation(s)
- Zeyu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qing Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yueting Deng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Edmund T Rolls
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Department of Computer Science, University of Warwick, Coventry, United Kingdom; Oxford Centre for Computational Neuroscience, Oxford, United Kingdom.
| | - Chun Shen
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yuzhu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shitong Xiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Christelle Langley
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Barbara J Sahakian
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Trevor W Robbins
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| | - Jin-Tai Yu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Department of Computer Science, University of Warwick, Coventry, United Kingdom; Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, China.
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, China.
| |
Collapse
|
30
|
Liu D, Hsueh SC, Tweedie D, Price N, Glotfelty E, Lecca D, Telljohann R, deCabo R, Hoffer BJ, Greig NH. Chronic inflammation with microglia senescence at basal forebrain: impact on cholinergic deficit in Alzheimer's brain haemodynamics. Brain Commun 2024; 6:fcae204. [PMID: 38978722 PMCID: PMC11228546 DOI: 10.1093/braincomms/fcae204] [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: 06/06/2023] [Revised: 04/23/2024] [Accepted: 06/11/2024] [Indexed: 07/10/2024] Open
Abstract
Cholinergic innervation in the brain is involved in modulating neurovascular function including cerebral blood flow haemodynamics in response to neuronal activity. Cholinergic deficit is associated with pathophysiology in Alzheimer's disease, albeit the aetiology remains to be clarified. In the current study, neocortex cerebral blood flow response to acetylcholine was evaluated by Laser-Doppler Flowmetry (LDF) in 3xTgAD Alzheimer's disease model) and wild-type mice of two age groups. The peak of cerebral blood flow to acetylcholine (i.v.) from baseline levels (% ΔrCBF) was higher in young 3xTgAD versus in wild-type mice (48.35; 95% CI:27.03-69.67 versus 22.70; CI:15.5-29.91, P < 0.05); this was reversed in old 3xTgAD mice (21.44; CI:2.52-40.35 versus 23.25; CI:23.25-39). Choline acetyltransferase protein was reduced in neocortex, while cerebrovascular reactivity to acetylcholine was preserved in young 3×TgAD mice. This suggests endogenous acetylcholine deficit and possible cholinergic denervation from selected cholinergic nuclei within the basal forebrain. The early deposition of tauopathy moieties (mutant hTau and pTau181) and its coincidence in cholinergic cell clusters (occasionaly), were observed at the basal forebrain of 3xTgAD mice including substantia innominate, nucleus Basalis of Meynert and nucleus of horizontal limb diagonal band of Broca. A prominent feature was microglia interacting tauopathy and demonstrated a variety of morphology changes particularly when located in proximity to tauopathy. The microglia ramified phenotype was reduced as evaluated by the ramification index and Fractal analysis. Increased microglia senescence, identified as SASP (senescence-associated secretory phenotype), was colocalization with p16Ink4ɑ, a marker of irreversible cell-cycle arrest in old 3xTgAD versus wild-type mice (P = 0.001). The p16Ink4ɑ was also observed in neuronal cells bearing tauopathy within the basal forebrain of 3xTgAD mice. TNF-ɑ, the pro-inflammatory cytokine elevated persistently in microglia (Pearson's correlation coefficient = 0.62) and the loss of cholinergic cells in vulnerable basal forebrain environment, was indicated by image analysis in 3xTgAD mice, which linked to the cholinergic deficits in neocortex rCBF haemodynamics. Our study revealed the early change of CBF haemodynamics to acetylcholine in 3xTgAD model. As a major effector of brain innate immune activation, microglia SASP with age-related disease progression is indicative of immune cell senescence, which contributes to chronic inflammation and cholinergic deficits at the basal forebrain. Targeting neuroinflammation and senescence may mitigate cholinergic pathophysiology in Alzheimer's disease.
Collapse
Affiliation(s)
- Dong Liu
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Shih Chang Hsueh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
- Department of Pediatrics, Columbia University Irving Medical Center, Columbia University Vagelos Physicians & Surgeons College of Medicine, New York City, NY 10032, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Nate Price
- Experimental Gerontology Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Elliot Glotfelty
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
- Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, USA
| | - Daniela Lecca
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
- Shock, Trauma & Anesthesiology Research Center, University of Maryland, Baltimore, MD 21201, USA
| | - Richard Telljohann
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Rafael deCabo
- Department of Pediatrics, Columbia University Irving Medical Center, Columbia University Vagelos Physicians & Surgeons College of Medicine, New York City, NY 10032, USA
| | - Barry J Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, University Hospitals, Cleveland, OH 44106, USA
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
31
|
Ye Q, Nunez J, Zhang X. Multiple cholinergic receptor subtypes coordinate dual modulation of acetylcholine on anterior and posterior paraventricular thalamic neurons. J Neurochem 2024; 168:995-1018. [PMID: 38664195 PMCID: PMC11136594 DOI: 10.1111/jnc.16115] [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: 09/28/2023] [Revised: 03/31/2024] [Accepted: 04/05/2024] [Indexed: 05/31/2024]
Abstract
Paraventricular thalamus (PVT) plays important roles in the regulation of emotion and motivation through connecting many brain structures including the midbrain and the limbic system. Although acetylcholine (ACh) neurons of the midbrain were reported to send projections to PVT, little is known about how cholinergic signaling regulates PVT neurons. Here, we used both RNAscope and slice patch-clamp recordings to characterize cholinergic receptor expression and ACh modulation of PVT neurons in mice. We found ACh excited a majority of anterior PVT (aPVT) neurons but predominantly inhibited posterior PVT (pPVT) neurons. Compared to pPVT with more inhibitory M2 receptors, aPVT expressed higher levels of all excitatory receptor subtypes including nicotinic α4, α7, and muscarinic M1 and M3. The ACh-induced excitation was mimicked by nicotine and antagonized by selective blockers for α4β2 and α7 nicotinic ACh receptor (nAChR) subtypes as well as selective antagonists for M1 and M3 muscarinic ACh receptors (mAChR). The ACh-induced inhibition was attenuated by selective M2 and M4 mAChR receptor antagonists. Furthermore, we found ACh increased the frequency of excitatory postsynaptic currents (EPSCs) on a majority of aPVT neurons but decreased EPSC frequency on a larger number of pPVT neurons. In addition, ACh caused an acute increase followed by a lasting reduction in inhibitory postsynaptic currents (IPSCs) on PVT neurons of both subregions. Together, these data suggest that multiple AChR subtypes coordinate a differential modulation of ACh on aPVT and pPVT neurons.
Collapse
Affiliation(s)
- Qiying Ye
- Department of Psychology, Florida State University, Tallahassee, Florida, USA
| | - Jeremiah Nunez
- Department of Psychology, Florida State University, Tallahassee, Florida, USA
| | - Xiaobing Zhang
- Department of Psychology, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
32
|
Chen H, Gu X, Mao Z, Zeng Q, Jin M, Wang W, Martyniuk CJ. Molecular, behavioral, and growth responses of juvenile yellow catfish (Tachysurus fulvidraco) exposed to carbamazepine. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 271:106929. [PMID: 38663201 DOI: 10.1016/j.aquatox.2024.106929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/21/2024]
Abstract
Carbamazepine (CBZ) is an anticonvulsant medication used to treat epilepsy and bipolar disorder. Due to its persistence and low removal rate in wastewater treatment plants, it is frequently detected in the environment, raising concerns regarding its potential adverse effects on aquatic organisms and ecosystems. In this study, we aimed to assess the impact of CBZ on the behavior and growth of juvenile yellow catfish Tachysurus fulvidraco, a native and economically important species in China. Fish were exposed to CBZ at three concentrations of 1, 10, or 100 µg/L for 14 days. The fish exposed to 10 and 100 μg/L of CBZ exhibited decreased feeding, and a significant increase in cannibalistic tendencies was observed in fish exposed to 100 μg/L CBZ. Acetylcholinesterase activity was increased in the brain of fish exposed to 100 μg/L CBZ. CBZ also inhibited the growth of yellow catfish. To better elucidate mechanisms of toxicity, transcriptomics was conducted in both the brain and liver. In the brain, gene networks associated with neurotransmitter dysfunction were altered by CBZ, as well as networks associated with mitochondrial dysfunction and metabolism. In the liver, gene networks associated with the immune system were altered by CBZ. The current study improves comprehension of the sub-lethal effects of CBZ and reveals novel insight into molecular and biochemical pathways disrupted by CBZ, identifying putative key events associated with reduced growth and altered behavior. This study emphasizes the necessity for improved comprehension of the effects of pharmaceutical contaminants on fish at environmentally relevant levels.
Collapse
Affiliation(s)
- Huihui Chen
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; Department of Organismal Biology, Uppsala University, Uppsala 75236, Sweden
| | - Xiaohong Gu
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China.
| | - Zhigang Mao
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China.
| | - Qingfei Zeng
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Miao Jin
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Wenxia Wang
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; College of Life Sciences, Linyi University, Linyi 276000, China
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611 United States
| |
Collapse
|
33
|
Tsotsokou G, Trompoukis G, Papatheodoropoulos C. Muscarinic Modulation of Synaptic Transmission and Short-Term Plasticity in the Dorsal and Ventral Hippocampus. Mol Cell Neurosci 2024; 129:103935. [PMID: 38703973 DOI: 10.1016/j.mcn.2024.103935] [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: 09/29/2023] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
Muscarinic neurotransmission is fundamentally involved in supporting several brain functions by modulating flow of information in brain neural circuits including the hippocampus which displays a remarkable functional segregation along its longitudinal axis. However, how muscarinic neuromodulation contributes to the functional segregation along the hippocampus remains unclear. In this study we show that the nonselective muscarinic receptor agonist carbachol similarly suppresses basal synaptic transmission in the dorsal and ventral CA1 hippocampal field, in a concentration-depended manner. Furthermore, using a ten-pulse stimulation train of varying frequency we found that carbachol changes the frequency filtering properties more in ventral than dorsal hippocampus by facilitating synaptic inputs at a wide range of input frequencies in the ventral compared with dorsal hippocampus. Using the M2 receptor antagonist gallamine and the M4 receptor antagonist tropicamide, we found that M2 receptors are involved in controlling basal synaptic transmission and short-term synaptic plasticity (STSP) in the ventral but not the dorsal hippocampus, while M4 receptors participate in modulating basal synaptic transmission and STSP in both segments of the hippocampus. These results were corroborated by the higher protein expression levels of M2 receptors in the ventral compared with dorsal hippocampus. We conclude that muscarinic transmission modulates excitatory synaptic transmission and short-term synaptic plasticity along the entire rat hippocampus by acting through M4 receptors and recruiting M2 receptors only in the ventral hippocampus. Furthermore, M4 receptors appear to exert a permissive role on the actions of M2 receptors on STSP in the ventral hippocampus. This dorsoventral differentiation of muscarinic modulation is expected to have important implications in information processing along the endogenous hippocampal circuitry.
Collapse
Affiliation(s)
- Giota Tsotsokou
- Laboratory of Physiology, University of Patras, Department of Medicine, Rion, Greece
| | - George Trompoukis
- Laboratory of Physiology, University of Patras, Department of Medicine, Rion, Greece
| | | |
Collapse
|
34
|
Özçete ÖD, Banerjee A, Kaeser PS. Mechanisms of neuromodulatory volume transmission. Mol Psychiatry 2024:10.1038/s41380-024-02608-3. [PMID: 38789677 DOI: 10.1038/s41380-024-02608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
A wealth of neuromodulatory transmitters regulate synaptic circuits in the brain. Their mode of signaling, often called volume transmission, differs from classical synaptic transmission in important ways. In synaptic transmission, vesicles rapidly fuse in response to action potentials and release their transmitter content. The transmitters are then sensed by nearby receptors on select target cells with minimal delay. Signal transmission is restricted to synaptic contacts and typically occurs within ~1 ms. Volume transmission doesn't rely on synaptic contact sites and is the main mode of monoamines and neuropeptides, important neuromodulators in the brain. It is less precise than synaptic transmission, and the underlying molecular mechanisms and spatiotemporal scales are often not well understood. Here, we review literature on mechanisms of volume transmission and raise scientific questions that should be addressed in the years ahead. We define five domains by which volume transmission systems can differ from synaptic transmission and from one another. These domains are (1) innervation patterns and firing properties, (2) transmitter synthesis and loading into different types of vesicles, (3) architecture and distribution of release sites, (4) transmitter diffusion, degradation, and reuptake, and (5) receptor types and their positioning on target cells. We discuss these five domains for dopamine, a well-studied monoamine, and then compare the literature on dopamine with that on norepinephrine and serotonin. We include assessments of neuropeptide signaling and of central acetylcholine transmission. Through this review, we provide a molecular and cellular framework for volume transmission. This mechanistic knowledge is essential to define how neuromodulatory systems control behavior in health and disease and to understand how they are modulated by medical treatments and by drugs of abuse.
Collapse
Affiliation(s)
- Özge D Özçete
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Cui A, Zhang J, Liu Z, Mu X, Zhong X, Xu H, Shan G. Patterned Au@Ag nanoarrays with electrically stimulated laccase-mimicking activity for dual-mode detection of epinephrine. Talanta 2024; 272:125821. [PMID: 38412753 DOI: 10.1016/j.talanta.2024.125821] [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: 11/10/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
Epinephrine (EP) is a crucial neurotransmitter in the central nervous system. However, an abnormal level of EP in biological fluids can lead to various diseases. Therefore, it is essential to rapidly and accurately detect EP content. Herein, electrically stimulated patterned Au@Ag nanoarrays with laccase-mimicking activity were designed for the dual-mode detection of EP concentration. The patterned Au@Ag nanoarrays exhibit excellent electrochemical properties and electrically stimulated laccase-mimicking activity. They provide sensitive electrochemical responses for detecting EP content. Simultaneously, the Au@Ag nanoarrays can catalyze the oxidation of EP, enabling its detection through a colorimetric process. This dual-mode approach achieves the detection of EP content over a wide linear range of 0.5-200 μM, with a low detection limit of 0.152 μM. Furthermore, the utility of these nanoarrays for sensing EP in human serum was evaluated. This work provides a convenient method using patterned nanozyme array for the visible, rapid and accurate detection of EP content. It provides the important implication for the development of portable and reliable on-site analytical instruments.
Collapse
Affiliation(s)
- Anni Cui
- Centre for Advanced Optoelectronic Functional Materials Research, Key Laboratory for UV Light-Emitting Materials and Technology of the Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Jialu Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zhifei Liu
- High School Attached to Northeast Normal University International Division, Changchun, 130021, China
| | - Xin Mu
- Centre for Advanced Optoelectronic Functional Materials Research, Key Laboratory for UV Light-Emitting Materials and Technology of the Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Xiahua Zhong
- Centre for Advanced Optoelectronic Functional Materials Research, Key Laboratory for UV Light-Emitting Materials and Technology of the Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Haitao Xu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Guiye Shan
- Centre for Advanced Optoelectronic Functional Materials Research, Key Laboratory for UV Light-Emitting Materials and Technology of the Ministry of Education, Northeast Normal University, Changchun, 130024, China.
| |
Collapse
|
36
|
Wang J, Yang Q, Liu X, Li J, Wen YL, Hu Y, Xu TL, Duan S, Xu H. The basal forebrain to lateral habenula circuitry mediates social behavioral maladaptation. Nat Commun 2024; 15:4013. [PMID: 38740778 DOI: 10.1038/s41467-024-48378-y] [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: 10/06/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Elucidating the neural basis of fear allows for more effective treatments for maladaptive fear often observed in psychiatric disorders. Although the basal forebrain (BF) has an essential role in fear learning, its function in fear expression and the underlying neuronal and circuit substrates are much less understood. Here we report that BF glutamatergic neurons are robustly activated by social stimulus following social fear conditioning in male mice. And cell-type-specific inhibition of those excitatory neurons largely reduces social fear expression. At the circuit level, BF glutamatergic neurons make functional contacts with the lateral habenula (LHb) neurons and these connections are potentiated in conditioned mice. Moreover, optogenetic inhibition of BF-LHb glutamatergic pathway significantly reduces social fear responses. These data unravel an important function of the BF in fear expression via its glutamatergic projection onto the LHb, and suggest that selective targeting BF-LHb excitatory circuitry could alleviate maladaptive fear in relevant disorders.
Collapse
Affiliation(s)
- Jun Wang
- Department of Neurobiology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China.
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China.
| | - Qian Yang
- Department of Neurobiology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xue Liu
- Department of Neurobiology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Lingang Laboratory, Shanghai, 200031, China
| | - Jie Li
- Department of Neurobiology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ya-Lan Wen
- Department of Neurobiology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuzheng Hu
- Department of Psychology and Behavioral Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Tian-Le Xu
- Center for Brain Science and Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shumin Duan
- Department of Neurobiology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| | - Han Xu
- Department of Neurobiology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China.
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China.
- Lingang Laboratory, Shanghai, 200031, China.
| |
Collapse
|
37
|
Ni S, Harris B, Gong P. Distributed and dynamical communication: a mechanism for flexible cortico-cortical interactions and its functional roles in visual attention. Commun Biol 2024; 7:550. [PMID: 38719883 PMCID: PMC11078951 DOI: 10.1038/s42003-024-06228-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
Perceptual and cognitive processing relies on flexible communication among cortical areas; however, the underlying neural mechanism remains unclear. Here we report a mechanism based on the realistic spatiotemporal dynamics of propagating wave patterns in neural population activity. Using a biophysically plausible, multiarea spiking neural circuit model, we demonstrate that these wave patterns, characterized by their rich and complex dynamics, can account for a wide variety of empirically observed neural processes. The coordinated interactions of these wave patterns give rise to distributed and dynamic communication (DDC) that enables flexible and rapid routing of neural activity across cortical areas. We elucidate how DDC unifies the previously proposed oscillation synchronization-based and subspace-based views of interareal communication, offering experimentally testable predictions that we validate through the analysis of Allen Institute Neuropixels data. Furthermore, we demonstrate that DDC can be effectively modulated during attention tasks through the interplay of neuromodulators and cortical feedback loops. This modulation process explains many neural effects of attention, underscoring the fundamental functional role of DDC in cognition.
Collapse
Affiliation(s)
- Shencong Ni
- School of Physics, University of Sydney, Sydney, NSW, Australia
| | - Brendan Harris
- School of Physics, University of Sydney, Sydney, NSW, Australia
| | - Pulin Gong
- School of Physics, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
38
|
Wang Y, Wang X, Wang L, Zheng L, Meng S, Zhu N, An X, Wang L, Yang J, Zheng C, Ming D. Dynamic prediction of goal location by coordinated representation of prefrontal-hippocampal theta sequences. Curr Biol 2024; 34:1866-1879.e6. [PMID: 38608677 DOI: 10.1016/j.cub.2024.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/20/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024]
Abstract
Prefrontal (PFC) and hippocampal (HPC) sequences of neuronal firing modulated by theta rhythms could represent upcoming choices during spatial memory-guided decision-making. How the PFC-HPC network dynamically coordinates theta sequences to predict specific goal locations and how it is interrupted in memory impairments induced by amyloid beta (Aβ) remain unclear. Here, we detected theta sequences of firing activities of PFC neurons and HPC place cells during goal-directed spatial memory tasks. We found that PFC ensembles exhibited predictive representation of the specific goal location since the starting phase of memory retrieval, earlier than the hippocampus. High predictive accuracy of PFC theta sequences existed during successful memory retrieval and positively correlated with memory performance. Coordinated PFC-HPC sequences showed PFC-dominant prediction of goal locations during successful memory retrieval. Furthermore, we found that theta sequences of both regions still existed under Aβ accumulation, whereas their predictive representation of goal locations was weakened with disrupted spatial representation of HPC place cells and PFC neurons. These findings highlight the essential role of coordinated PFC-HPC sequences in successful memory retrieval of a precise goal location.
Collapse
Affiliation(s)
- Yimeng Wang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Xueling Wang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Ling Wang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin 300072, China
| | - Li Zheng
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Shuang Meng
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Nan Zhu
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Xingwei An
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin 300072, China
| | - Lei Wang
- School of Statistics and Data Science, Nankai University, Tianjin 300071, China.
| | - Jiajia Yang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300072, China.
| | - Chenguang Zheng
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300072, China.
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300072, China.
| |
Collapse
|
39
|
Chang YB, Jung EJ, Jo K, Suh HJ, Choi HS. Neuroprotective effect of whey protein hydrolysate containing leucine-aspartate-isoleucine-glutamine-lysine on HT22 cells in hydrogen peroxide-induced oxidative stress. J Dairy Sci 2024; 107:2620-2632. [PMID: 38101744 DOI: 10.3168/jds.2023-24284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023]
Abstract
This study aimed to investigate the neuroprotective effects of whey protein hydrolysate (WPH) containing the pentapeptide leucine-aspartate-isoleucine-glutamine-lysine (LDIQK). Whey protein hydrolysate (50, 100, and 200 µg/mL) demonstrated the ability to restore the viability of HT22 cells subjected to 300 µM hydrogen peroxide (H2O2)-induced oxidative stress. Furthermore, at a concentration of 200 µg/mL, it significantly reduced the increase in reactive oxygen species production and calcium ion (Ca2+) influx induced by H2O2 by 46.1% and 46.2%, respectively. Similarly, the hydrolysate significantly decreased the levels of p-tau, a hallmark of tauopathy, and BCL2 associated X (BAX), a proapoptosis factor, while increasing the protein levels of choline acetyltransferase (ChAT), an enzyme involved in acetylcholine synthesis, brain-derived neurotrophic factor (BDNF), a nerve growth factor, and B-cell lymphoma 2 (BCL2, an antiapoptotic factor. Furthermore, it increased nuclear factor erythroid 2-related factor 2 (Nrf2)-hemoxygenase-1(HO-1) signaling, which is associated with the antioxidant response, while reducing the activation of mitogen-activated protein kinase (MAPK) signaling pathway components, namely phosphor-extracellular signal-regulated kinases (p-ERK), phosphor-c-Jun N-terminal kinases (p-JNK), and p-p38. Column chromatography and tandem mass spectrometry analysis identified LDIQK as a compound with neuroprotective effects in WPH; it inhibited Ca2+ influx and regulated the BAX/BCL2 ratio. Collectively, WPH containing LDIQK demonstrated neuroprotective effects against H2O2-induced neuronal cell damage, suggesting that WPH or its active peptide, LDIQK, may serve as a potential edible agent for improving cognitive dysfunction.
Collapse
Affiliation(s)
- Yeok B Chang
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Eun-Jin Jung
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea
| | - Kyungae Jo
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hyung J Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University, Seoul 02841, Republic of Korea.
| | - Hyeon-Son Choi
- Department of Food Nutrition, Sangmyung University, Seoul 03016, Republic of Korea.
| |
Collapse
|
40
|
Kniffin A, Bangasser DA, Parikh V. Septohippocampal cholinergic system at the intersection of stress and cognition: Current trends and translational implications. Eur J Neurosci 2024; 59:2155-2180. [PMID: 37118907 PMCID: PMC10875782 DOI: 10.1111/ejn.15999] [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: 09/27/2022] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 04/30/2023]
Abstract
Deficits in hippocampus-dependent memory processes are common across psychiatric and neurodegenerative disorders such as depression, anxiety and Alzheimer's disease. Moreover, stress is a major environmental risk factor for these pathologies and it exerts detrimental effects on hippocampal functioning via the activation of hypothalamic-pituitary-adrenal (HPA) axis. The medial septum cholinergic neurons extensively innervate the hippocampus. Although, the cholinergic septohippocampal pathway (SHP) has long been implicated in learning and memory, its involvement in mediating the adaptive and maladaptive impact of stress on mnemonic processes remains less clear. Here, we discuss current research highlighting the contributions of cholinergic SHP in modulating memory encoding, consolidation and retrieval. Then, we present evidence supporting the view that neurobiological interactions between HPA axis stress response and cholinergic signalling impact hippocampal computations. Finally, we critically discuss potential challenges and opportunities to target cholinergic SHP as a therapeutic strategy to improve cognitive impairments in stress-related disorders. We argue that such efforts should consider recent conceptualisations on the dynamic nature of cholinergic signalling in modulating distinct subcomponents of memory and its interactions with cellular substrates that regulate the adaptive stress response.
Collapse
Affiliation(s)
- Alyssa Kniffin
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA 19122
| | - Debra A. Bangasser
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA
| | - Vinay Parikh
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA 19122
| |
Collapse
|
41
|
Chen WJ, Chen H, Li ZM, Huang WY, Wu JL. Acetylcholine muscarinic M1 receptors in the rodent prefrontal cortex modulate cognitive abilities to establish social hierarchy. Neuropsychopharmacology 2024; 49:974-982. [PMID: 38135842 PMCID: PMC11039707 DOI: 10.1038/s41386-023-01785-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/29/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
In most social species, the attainment of social dominance is strongly affected by personality traits. Dominant individuals show better cognitive abilities, however, whether an individual's cognition can determine its social status has remained inconclusive. We found that mice show better cognitive abilities tend to possess a higher social rank after cohousing. The dynamic release of acetylcholine (ACh) in the prelimbic cortex (PL) is correlated with mouse dominance behavior. ACh enhanced the excitability of the PL neurons via acetylcholine muscarinic M1 receptors (M1). Inhibition of M1 impaired mice cognitive performance and induced losing in social competition. Mice with M1 deficiency in the PL performed worse on cognitive behavioral tests, and exhibited lower status when re-grouped with others. Elevating ACh level in the PL of subordinate mice induced winning. These results provide direct evidence for the involvement of M1 in social hierarchy and suggest that social rank can be tuned by altering cognition through cholinergic system.
Collapse
Affiliation(s)
- Wen-Jun Chen
- Medical Research and Experimental Center, Meizhou People's Hospital, Meizhou, 514031, China
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, 514031, China
| | - Hao Chen
- Department of Neurobiology, Southern Medical University, Guangzhou, 510515, China
| | - Zi-Ming Li
- Department of Neurobiology, Southern Medical University, Guangzhou, 510515, China
| | - Wei-Yuan Huang
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Jian-Lin Wu
- Medical Research and Experimental Center, Meizhou People's Hospital, Meizhou, 514031, China.
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, 514031, China.
| |
Collapse
|
42
|
Lozovaya N, Moumen A, Hammond C. Basal Forebrain Cholinergic Neurons Have Specific Characteristics during the Perinatal Period. eNeuro 2024; 11:ENEURO.0538-23.2024. [PMID: 38755010 PMCID: PMC11137802 DOI: 10.1523/eneuro.0538-23.2024] [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: 11/13/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024] Open
Abstract
Cholinergic neurons of the basal forebrain represent the main source of cholinergic innervation of large parts of the neocortex and are involved in adults in the modulation of attention, memory, and arousal. During the first postnatal days, they play a crucial role in the development of cortical neurons and cortical cytoarchitecture. However, their characteristics, during this period have not been studied. To understand how they can fulfill this role, we investigated the morphological and electrophysiological maturation of cholinergic neurons of the substantia innominata-nucleus basalis of Meynert (SI/NBM) complex in the perinatal period in mice. We show that cholinergic neurons, whether or not they express gamma-aminobutyric acid (GABA) as a cotransmitter, are already functional at Embryonic Day 18. Until the end of the first postnatal week, they constitute a single population of neurons with a well developed dendritic tree, a spontaneous activity including bursting periods, and a short-latency response to depolarizations (early-firing). They are excited by both their GABAergic and glutamatergic afferents. During the second postnatal week, a second, less excitable, neuronal population emerges, with a longer delay response to depolarizations (late-firing), together with the hyperpolarizing action of GABAA receptor-mediated currents. This classification into early-firing (40%) and late-firing (60%) neurons is again independent of the coexpression of GABAergic markers. These results strongly suggest that during the first postnatal week, the specific properties of developing SI/NBM cholinergic neurons allow them to spontaneously release acetylcholine (ACh), or ACh and GABA, into the developing cortex.
Collapse
|
43
|
Vidal M, Onderdijk KE, Aguilera AM, Six J, Maes PJ, Fritz TH, Leman M. Cholinergic-related pupil activity reflects level of emotionality during motor performance. Eur J Neurosci 2024; 59:2193-2207. [PMID: 37118877 DOI: 10.1111/ejn.15998] [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: 10/13/2022] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 04/30/2023]
Abstract
Pupil size covaries with the diffusion rate of the cholinergic and noradrenergic neurons throughout the brain, which are essential to arousal. Recent findings suggest that slow pupil fluctuations during locomotion are an index of sustained activity in cholinergic axons, whereas phasic dilations are related to the activity of noradrenergic axons. Here, we investigated movement induced arousal (i.e., by singing and swaying to music), hypothesising that actively engaging in musical behaviour will provoke stronger emotional engagement in participants and lead to different qualitative patterns of tonic and phasic pupil activity. A challenge in the analysis of pupil data is the turbulent behaviour of pupil diameter due to exogenous ocular activity commonly encountered during motor tasks and the high variability typically found between individuals. To address this, we developed an algorithm that adaptively estimates and removes pupil responses to ocular events, as well as a functional data methodology, derived from Pfaffs' generalised arousal, that provides a new statistical dimension on how pupil data can be interpreted according to putative neuromodulatory signalling. We found that actively engaging in singing enhanced slow cholinergic-related pupil dilations and having the opportunity to move your body while performing amplified the effect of singing on pupil activity. Phasic pupil oscillations during motor execution attenuated in time, which is often interpreted as a measure of sense of agency over movement.
Collapse
Affiliation(s)
- Marc Vidal
- IPEM, Ghent University, Ghent, Belgium
- Department of Statistics and Operations Research, Institute of Mathematics, University of Granada, Granada, Spain
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | | | - Ana M Aguilera
- Department of Statistics and Operations Research, Institute of Mathematics, University of Granada, Granada, Spain
| | - Joren Six
- IPEM, Ghent University, Ghent, Belgium
| | | | - Thomas Hans Fritz
- IPEM, Ghent University, Ghent, Belgium
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | | |
Collapse
|
44
|
Pastor V, Medina JH. α7 nicotinic acetylcholine receptor in memory processing. Eur J Neurosci 2024; 59:2138-2154. [PMID: 36634032 DOI: 10.1111/ejn.15913] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
Information storage in the brain involves different memory types and stages that are processed by several brain regions. Cholinergic pathways through acetylcholine receptors actively participate on memory modulation, and their disfunction is associated with cognitive decline in several neurological disorders. During the last decade, the role of α7 subtype of nicotinic acetylcholine receptors in different memory stages has been studied. However, the information about their role in memory processing is still scarce. In this review, we attempt to identify brain areas where α7 nicotinic receptors have an essential role in different memory types and stages. In addition, we discuss recent work implicating-or not-α7 nicotinic receptors as promising pharmacological targets for memory impairment associated with neurological disorders.
Collapse
Affiliation(s)
- Verónica Pastor
- Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis" (IBCN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Facultad de Medicina, Departamento de Ciencias Fisiológicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jorge H Medina
- Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis" (IBCN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
| |
Collapse
|
45
|
Blanco I, Caccavano A, Wu JY, Vicini S, Glasgow E, Conant K. Coupling of Sharp Wave Events between Zebrafish Hippocampal and Amygdala Homologs. J Neurosci 2024; 44:e1467232024. [PMID: 38508712 PMCID: PMC11044098 DOI: 10.1523/jneurosci.1467-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 03/22/2024] Open
Abstract
The mammalian hippocampus exhibits spontaneous sharp wave events (1-30 Hz) with an often-present superimposed fast ripple oscillation (120-220 Hz) to form a sharp wave ripple (SWR) complex. During slow-wave sleep or quiet restfulness, SWRs result from the sequential spiking of hippocampal cell assemblies initially activated during learned or imagined experiences. Additional cortical/subcortical areas exhibit SWR events that are coupled to hippocampal SWRs, and studies in mammals suggest that coupling may be critical for the consolidation and recall of specific memories. In the present study, we have examined juvenile male and female zebrafish and show that SWR events are intrinsically generated and maintained within the telencephalon and that their hippocampal homolog, the anterodorsolateral lobe (ADL), exhibits SW events with ∼9% containing an embedded ripple (SWR). Single-cell calcium imaging coupled to local field potential recordings revealed that ∼10% of active cells in the dorsal telencephalon participate in any given SW event. Furthermore, fluctuations in cholinergic tone modulate SW events consistent with mammalian studies. Moreover, the basolateral amygdala (BLA) homolog exhibits SW events with ∼5% containing an embedded ripple. Computing the SW peak coincidence difference between the ADL and BLA showed bidirectional communication. Simultaneous coupling occurred more frequently within the same hemisphere, and in coupled events across hemispheres, the ADL more commonly preceded BLA. Together, these data suggest conserved mechanisms across species by which SW and SWR events are modulated, and memories may be transferred and consolidated through regional coupling.
Collapse
Affiliation(s)
- Ismary Blanco
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | - Adam Caccavano
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | - Jian-Young Wu
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Departments of Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | - Stefano Vicini
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057
| | - Eric Glasgow
- Oncology, Georgetown University Medical Center, Washington, DC 20057
| | - Katherine Conant
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Departments of Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| |
Collapse
|
46
|
Zou Y, Tong C, Peng W, Qiu Y, Li J, Xia Y, Pei M, Zhang K, Li W, Xu M, Liang Z. Cell-type-specific optogenetic fMRI on basal forebrain reveals functional network basis of behavioral preference. Neuron 2024; 112:1342-1357.e6. [PMID: 38359827 DOI: 10.1016/j.neuron.2024.01.017] [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: 07/31/2023] [Revised: 12/12/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024]
Abstract
The basal forebrain (BF) is a complex structure that plays key roles in regulating various brain functions. However, it remains unclear how cholinergic and non-cholinergic BF neurons modulate large-scale functional networks and their relevance in intrinsic and extrinsic behaviors. With an optimized awake mouse optogenetic fMRI approach, we revealed that optogenetic stimulation of four BF neuron types evoked distinct cell-type-specific whole-brain BOLD activations, which could be attributed to BF-originated low-dimensional structural networks. Additionally, optogenetic activation of VGLUT2, ChAT, and PV neurons in the BF modulated the preference for locomotion, exploration, and grooming, respectively. Furthermore, we uncovered the functional network basis of the above BF-modulated behavioral preference through a decoding model linking the BF-modulated BOLD activation, low-dimensional structural networks, and behavioral preference. To summarize, we decoded the functional network basis of differential behavioral preferences with cell-type-specific optogenetic fMRI on the BF and provided an avenue for investigating mouse behaviors from a whole-brain view.
Collapse
Affiliation(s)
- Yijuan Zou
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; International Center for Primate Brain Research, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 201602, China
| | - Chuanjun Tong
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; International Center for Primate Brain Research, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 201602, China
| | - Wanling Peng
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yue Qiu
- Cardiac Intensive Care Center, Zhongshan Hospital, Fudan University Shanghai, Shanghai 200032, China
| | - Jiangxue Li
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Ying Xia
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengchao Pei
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kaiwei Zhang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weishuai Li
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Min Xu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Zhifeng Liang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; International Center for Primate Brain Research, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 201602, China.
| |
Collapse
|
47
|
Lane RM, Darreh-Shori T, Junge C, Li D, Yang Q, Edwards AL, Graham DL, Moore K, Mummery CJ. Onset of Alzheimer disease in apolipoprotein ɛ4 carriers is earlier in butyrylcholinesterase K variant carriers. BMC Neurol 2024; 24:116. [PMID: 38594621 PMCID: PMC11003149 DOI: 10.1186/s12883-024-03611-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND The authors sought to examine the impact of the K-variant of butyrylcholinesterase (BCHE-K) carrier status on age-at-diagnosis of Alzheimer disease (AD) in APOE4 carriers. METHODS Patients aged 50-74 years with cerebrospinal fluid (CSF) biomarker-confirmed AD, were recruited to clinical trial (NCT03186989 since June 14, 2017). Baseline demographics, disease characteristics, and biomarkers were evaluated in 45 patients according to BCHE-K and APOE4 allelic status in this post-hoc study. RESULTS In APOE4 carriers (N = 33), the mean age-at-diagnosis of AD in BCHE-K carriers (n = 11) was 6.4 years earlier than in BCHE-K noncarriers (n = 22, P < .001, ANOVA). In APOE4 noncarriers (N = 12) there was no observed influence of BCHE-K. APOE4 carriers with BCHE-K also exhibited slightly higher amyloid and tau accumulations compared to BCHE-K noncarriers. A predominantly amyloid, limited tau, and limbic-amnestic phenotype was exemplified by APOE4 homozygotes with BCHE-K. In the overall population, multiple regression analyses demonstrated an association of amyloid accumulation with APOE4 carrier status (P < .029), larger total brain ventricle volume (P < .021), less synaptic injury (Ng, P < .001), and less tau pathophysiology (p-tau181, P < .005). In contrast, tau pathophysiology was associated with more neuroaxonal damage (NfL, P = .002), more synaptic injury (Ng, P < .001), and higher levels of glial activation (YKL-40, P = .01). CONCLUSION These findings have implications for the genetic architecture of prognosis in early AD, not the genetics of susceptibility to AD. In patients with early AD aged less than 75 years, the mean age-at-diagnosis of AD in APOE4 carriers was reduced by over 6 years in BCHE-K carriers versus noncarriers. The functional status of glia may explain many of the effects of APOE4 and BCHE-K on the early AD phenotype. TRIAL REGISTRATION NCT03186989 since June 14, 2017.
Collapse
Affiliation(s)
- Roger M Lane
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA.
| | - Taher Darreh-Shori
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatric, Karolinska Institutet, Stockholm, Sweden
| | - Candice Junge
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - Dan Li
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - Qingqing Yang
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | | | | | - Katrina Moore
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | | |
Collapse
|
48
|
Novakova L, Gajdos M, Barton M, Brabenec L, Zeleznikova Z, Moravkova I, Rektorova I. Striato-cortical functional connectivity changes in mild cognitive impairment with Lewy bodies. Parkinsonism Relat Disord 2024; 121:106031. [PMID: 38364623 DOI: 10.1016/j.parkreldis.2024.106031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Functional connectivity changes in clinically overt neurodegenerative diseases such as dementia with Lewy bodies have been described, but studies on connectivity changes in the pre-dementia phase are scarce. OBJECTIVES We concentrated on evaluating striato-cortical functional connectivity differences between patients with Mild Cognitive Impairment with Lewy bodies and healthy controls and on assessing the relation to cognition. METHODS Altogether, we enrolled 77 participants (47 patients, of which 35 met all the inclusion criteria for the final analysis, and 30 age- and gender-matched healthy controls, of which 28 met all the inclusion criteria for the final analysis) to study the seed-based connectivity of the dorsal, middle, and ventral striatum. We assessed correlations between functional connectivity in the regions of between-group differences and neuropsychological scores of interest (visuospatial and executive domains z-scores). RESULTS Subjects with Mild Cognitive Impairment with Lewy Bodies, as compared to healthy controls, showed increased connectivity from the dorsal part of the striatum particularly to the bilateral anterior part of the temporal cortex with an association with executive functions. CONCLUSIONS We were able to capture early abnormal connectivity within cholinergic and noradrenergic pathways that correlated with cognitive functions known to be linked to cholinergic/noradrenergic deficits. The knowledge of specific alterations may improve our understanding of early neural changes in pre-dementia stages and enhance research of disease modifying therapy.
Collapse
Affiliation(s)
- Lubomira Novakova
- Brain and Mind Research Program, CEITEC, Masaryk University, Brno, Czech Republic
| | - Martin Gajdos
- Brain and Mind Research Program, CEITEC, Masaryk University, Brno, Czech Republic
| | - Marek Barton
- Brain and Mind Research Program, CEITEC, Masaryk University, Brno, Czech Republic
| | - Lubos Brabenec
- Brain and Mind Research Program, CEITEC, Masaryk University, Brno, Czech Republic
| | - Zaneta Zeleznikova
- Brain and Mind Research Program, CEITEC, Masaryk University, Brno, Czech Republic; First Department of Neurology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Ivona Moravkova
- Brain and Mind Research Program, CEITEC, Masaryk University, Brno, Czech Republic; First Department of Neurology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Irena Rektorova
- Brain and Mind Research Program, CEITEC, Masaryk University, Brno, Czech Republic; First Department of Neurology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
49
|
Söylemez GK, Uzun S. The effect of nonpharmacological interventions applied by nurses to intensive care patients on the duration of delirium: a meta-analysis study. Ir J Med Sci 2024; 193:865-873. [PMID: 37624451 DOI: 10.1007/s11845-023-03504-5] [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: 07/31/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
OBJECTIVE It was aimed to determine the effect level of nonpharmacological methods applied by nurses to patients hospitalized in the intensive care unit on the duration of delirium. MATERIALS AND METHODS For this study, relevant studies were accessed by searching in July-October 2022. After the necessary exclusions were made, 14 studies were included in the study. The total sample size of the studies was 1123. RESULTS According to the results, nonpharmacological interventions applied by nurses to intensive care patients were found to reduce the duration of delirium (SMD: - 0.625, 95% CI: - 1.1040-0.210; Z = - 2.950, p = 0.003, I2 = 93.119%). The country of the study (SMD: - 0.047, p = 0.001) and the types of nonpharmacological interventions used (SMD: - 0.062, p = 0.000) influenced the effect size of modulators on the duration of delirium in ICU patients. CONCLUSION Nonpharmacological interventions applied by nurses, who have significant responsibilities in the protection and promotion of health, were found to reduce the duration of delirium in intensive care patients. This study shows that nurses, one of the main components of the multidisciplinary team in intensive care, are successful when they apply nonpharmacological interventions well.
Collapse
Affiliation(s)
- Gönül Kara Söylemez
- Nursing Department, Faculty of Health Sciences, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Sevda Uzun
- Department of Nursing, Gümüşhane University Faculty of Health Sciences, Gumushane, Turkey.
| |
Collapse
|
50
|
Mu R, Hou X, Liu Q, Wang W, Qin C, Li H. Up-regulation of GPR139 in the medial septum ameliorates cognitive impairment in two mouse models of Alzheimer's disease. Int Immunopharmacol 2024; 130:111786. [PMID: 38447415 DOI: 10.1016/j.intimp.2024.111786] [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: 01/11/2024] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
G-protein coupled receptors (GPCRs) constitute the largest class of cell surface receptors and present prominent drug targets. GPR139 is an orphan GPCR detected in the septum of the brain. However, its roles in cognition are still unclear. Here we first established a mouse model of cognitive impairment by a single intracerebroventricular injection of aggregated amyloid-beta peptide 1-42 (Aβ1-42). RNA-sequencing data analysis showed that Aβ1-42 induced a significant decrease of GPR139 mRNA in the basal forebrain. Using GPR139 agonist JNJ-63533054 and behavioral tests, we found that GPR139 activation in the brain ameliorated Aβ1-42-induced cognitive impairment. Using western blot, TUNEL apoptosis and Golgi staining assays, we showed that GPR139 activation alleviated Aβ1-42-induced apoptosis and synaptotoxicity in the basal forebrain rather than prefrontal cortex and hippocampus. The further study identified that GPR139 was widely expressed in cholinergic neurons of the medial septum (MS). Using the overexpression virus and transgenic animal model, we showed that up-regulation of GPR139 in MS cholinergic neurons ameliorated cognitive impairment, apoptosis and synaptotoxicity in APP/PS1 transgenic mice. These findings reveal that GPR139 of MS cholinergic neurons could be a critical node in cognition and potentially provides insight into the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Ronghao Mu
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China; Department of Child Developmental Behavior, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Xiaoying Hou
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Qi Liu
- Department of Child Developmental Behavior, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wan Wang
- Department of Child Developmental Behavior, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chi Qin
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Huixian Li
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|