1
|
de Gois AM, Bispo JMM, Santos ER, Souza MF, Melo JEC, Mendonça MS, Almeida-Souza TH, Camargo EA, Medeiros KAAL, Leal PC, Santos HF, Lins LCRF, Ribeiro AM, Silva RH, Santos JR. β-2 agonist and antagonist adrenoceptors induce neuroprotection in a progressive model of parkinsonism. Neuropharmacology 2025; 271:110386. [PMID: 40023440 DOI: 10.1016/j.neuropharm.2025.110386] [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: 09/25/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by progressive dopaminergic dysfunction in the nigrostriatal pathway, as well as alterations in other monoamines systems. Research indicates that the use of β-adrenergic agonist and antagonists influences the risk of PD. This study evaluated the effects of salbutamol and propranolol on motor and neurochemical parameters in a progressive model of parkinsonism induced by reserpine (RES). Male Wistar rats were chronically treated with 12 subcutaneous injections of RES (0,1 mg/kg) given every other day for 24 days. From the 16th day onwards, the animals were daily treated with salbutamol (5 mg/kg) or propranolol (20 mg/kg) intraperitoneally for 8 days. Salbutamol reduced the motor deficit caused by RES in the catalepsy test, while propranolol reduced the damages caused by RES in the vacuous chewing movements evaluation. In immunohistochemical analysis both salbutamol and propranolol prevented dopaminergic damage in the substantia nigra pars compacta (SNpc), ventral tegmental area (VTA), striatum and noradrenergic damage in locus coeruleus (LC). In addition, salbutamol and propranolol prevented the increase in α-synuclein immunoreactivity caused by RES in the substantia nigra pars reticulata (SNr), striatum, prefrontal cortex (mPFC) and hippocampus. These data show that salbutamol and propranolol promote neuroprotective effects against reserpine-induced parkinsonism. However, further studies are needed to understand the mechanisms involved in β-adrenoceptors role in PD development.
Collapse
Affiliation(s)
- Auderlan M de Gois
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil.
| | - José M M Bispo
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | - Edson R Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | - Marina F Souza
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | - João E C Melo
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | - Mylaine S Mendonça
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | | | - Enilton A Camargo
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Katty A A L Medeiros
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil; Nursing Department, Federal University of Sergipe, Lagarto, SE, Brazil
| | - Pollyana C Leal
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil; Department of Medicine, Federal University of Sergipe, Lagarto, SE, Brazil
| | - Heitor F Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil; Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Lívia C R F Lins
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil; Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | - Regina H Silva
- Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - José R Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| |
Collapse
|
2
|
Jin S, Bellier J, Wells A, LIopis PM, Anekal PV, Tresback JS, Caldarone BJ, Liu L, Li S, Dettmer U, Ramalingam N, Selkoe DJ. Inhibition of hippocampal mossy fiber plasticity and episodic memory by human Aβ oligomers is prevented by enhancing cAMP signaling in Alzheimer's mice. Alzheimers Dement 2025; 21:e70194. [PMID: 40302031 PMCID: PMC12040739 DOI: 10.1002/alz.70194] [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: 02/05/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 05/01/2025]
Abstract
INTRODUCTION Early episodic memory impairment in Alzheimer's disease (AD) is linked to synaptic dysfunction from amyloid β-protein oligomers (oAβ), particularly affecting the dentate gyrus mossy fiber-CA3 pathway. The APPNL-G-F mouse model exhibits early deficits in mossy fiber long-term potentiation (mf-LTP). METHODS We administered the β-adrenergic receptor agonist isoproterenol (ISO) in vivo and phosphodiesterase type 4 inhibitor GSK356278 in vitro to assess their impact on mf-LTP and contextual fear memory. Fluorescence lifetime imaging (FLIM)-Förster resonance energy transfer (FRET) microscopy was used to visualize impaired and rescued cyclic adenosine monophosphate (cAMP) signaling in dentate gyrus neurons. RESULTS ISO prevented mf-LTP impairment at 3-4 mo and improved memory by 7 mo. GSK356278 inhibited mf-LTP deficits in a dose-dependent manner. ISO also reduced hyperphosphorylation of synapsin I and microgliosis. DISCUSSION These findings suggest that β-AR activation and phosphodiesterase 4 (PDE4) inhibition mitigate oAβ-induced memory deficits, supporting enhanced cAMP signaling as a therapeutic target for early AD. HIGHLIGHTS Early episodic memory deficits in AD linked to oAβ-induced synaptic dysfunction. Isoproterenol and GSK356278 improve mossy fiber-LTP and fear memory deficits. FLIM-FRET shows treatments restore cAMP signaling in dentate gyrus neurons. Isoproterenol reduces synapsin I hyperphosphorylation and microgliosis. Enhancing cAMP signaling may help mitigate early memory deficits in AD.
Collapse
Affiliation(s)
- Shan‐Xue Jin
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Jean‐Pierre Bellier
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | | | | | | | - Jason S. Tresback
- Center for Nanoscale SystemsHarvard UniversityCambridgeMassachusettsUSA
| | | | - Lei Liu
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Shaomin Li
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Dennis J. Selkoe
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
3
|
Li Q, Xie Y, Lin J, Li M, Gu Z, Xin T, Zhang Y, Lu Q, Guo Y, Xing Y, Wang W. Microglia Sing the Prelude of Neuroinflammation-Associated Depression. Mol Neurobiol 2025; 62:5311-5332. [PMID: 39535682 DOI: 10.1007/s12035-024-04575-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: 06/14/2023] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Major depressive disorder (MDD) is a psychiatric condition characterized by sadness and anhedonia and is closely linked to chronic low-grade neuroinflammation, which is primarily induced by microglia. Nonetheless, the mechanisms by which microglia elicit depressive symptoms remain uncertain. This review focuses on the mechanism linking microglia and depression encompassing the breakdown of the blood-brain barrier, the hypothalamic-pituitary-adrenal axis, the gut-brain axis, the vagus and sympathetic nervous systems, and the susceptibility influenced by epigenetic modifications on microglia. These pathways may lead to the alterations of microglia in cytokine levels, as well as increased oxidative stress. Simultaneously, many antidepressant treatments can alter the immune phenotype of microglia, while anti-inflammatory treatments can also have antidepressant effects. This framework linking microglia, neuroinflammation, and depression could serve as a reference for targeting microglia to treat depression.
Collapse
Affiliation(s)
- Qingqing Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Ying Xie
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Jinyi Lin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Miaomiao Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Ziyan Gu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Tianli Xin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yang Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Qixia Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yihui Guo
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yanhong Xing
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China.
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
4
|
Zou H, Pu W, Zhou J, Li J, Ma L, Wang S, Liu C, Mou J, Liu X, Yu T, Wei Y, Xie H, Cao S. Noradrenergic Locus Coeruleus-CA3 Activation Alleviates Neuropathic Pain and Anxiety- and Depression-Like Behaviors by Suppressing Microglial Neuroinflammation in SNI Mice. CNS Neurosci Ther 2025; 31:e70360. [PMID: 40130433 PMCID: PMC11933858 DOI: 10.1111/cns.70360] [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: 01/27/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/26/2025] Open
Abstract
OBJECTIVE Neuropathic pain (NP) arises from neuroimmune interactions following nerve injury and is often accompanied by anxiety and depression. The aim of the study is to evaluate the effects of the noradrenergic locus coeruleus (LC), a key regulator of pain and emotional states, projects extensively to the hippocampus. METHOD We investigated the effects of chronic NP on LC integrity and its projections to the hippocampal CA3 region in spared nerve injury (SNI) mice with behavioral tests, immunohistochemistry, neurochemical analyses, and Gq-DREADD. RESULTS Chronic NP induced LC neuronal loss, reduced hippocampal norepinephrine (NE) release, and triggered microglial activation and neuroinflammation in CA3. Selective activation of LC-CA3 noradrenergic neurons using Gq-DREADD chemogenetics alleviated NP and comorbid anxiety- and depression-like behaviors. This intervention suppressed microglial activation, decreased proinflammatory cytokines (TNF-α and IL-1β), and restored NE levels in CA3. CONCLUSION Our findings highlighted the therapeutic potential of targeting LC-CA3 projections to mitigate chronic NP and its neuropsychiatric comorbidities via modulation of hippocampal neuroinflammation.
Collapse
Affiliation(s)
- Helin Zou
- Department of Anesthesiology, the Tenth Affiliated HospitalSouthern Medical University (Dongguan People's Hospital)DongguanGuangdongChina
- Dongguan Key Laboratory of Anesthesia and Organ ProtectionDongguanGuangdongChina
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
- Department of AnesthesiologyMianyang Hospital of Traditional Chinese MedicineMianyangSichuanChina
| | - Weiyu Pu
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
| | - Junli Zhou
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
| | - Juan Li
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
| | - Lulin Ma
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
| | - Shuxian Wang
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
| | - Chengxi Liu
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
| | - Jing Mou
- Department of Anesthesiology, the Tenth Affiliated HospitalSouthern Medical University (Dongguan People's Hospital)DongguanGuangdongChina
- Dongguan Key Laboratory of Anesthesia and Organ ProtectionDongguanGuangdongChina
- Department of Pain Medicine, the Tenth Affiliated HospitalSouthern Medical University (Dongguan People's Hospital)DongguanGuangdongChina
| | - Xingfeng Liu
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
| | - Tian Yu
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
| | - Yiyong Wei
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City)ShenzhenGuangdongChina
| | - Haihui Xie
- Department of Anesthesiology, the Tenth Affiliated HospitalSouthern Medical University (Dongguan People's Hospital)DongguanGuangdongChina
- Dongguan Key Laboratory of Anesthesia and Organ ProtectionDongguanGuangdongChina
| | - Song Cao
- Department of Anesthesiology, the Tenth Affiliated HospitalSouthern Medical University (Dongguan People's Hospital)DongguanGuangdongChina
- Dongguan Key Laboratory of Anesthesia and Organ ProtectionDongguanGuangdongChina
- Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation)Zunyi Medical UniversityZunyiGuizhouChina
- Department of Pain Medicine, the Tenth Affiliated HospitalSouthern Medical University (Dongguan People's Hospital)DongguanGuangdongChina
| |
Collapse
|
5
|
Harvey ME, Shi M, Oh Y, Mitchell DA, Slayden OD, MacLean JA, Hayashi K. Multiple lesion inductions intensify central sensitization driven by neuroinflammation in a mouse model of endometriosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634555. [PMID: 39896574 PMCID: PMC11785222 DOI: 10.1101/2025.01.23.634555] [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: 02/04/2025]
Abstract
Introduction Endometriosis is an inflammatory disease associated with chronic pelvic pain (CPP). Growing evidence indicates that endometriotic lesions are not the sole source of pain. Instead, central nervous system (CNS) dysfunction created by prolonged peripheral and central sensitization plays a role in developing endometriosis-associated CPP. This study investigated how CPP is established using a multiple lesion induction mouse model of endometriosis, as repeated retrograde menstruation is considered underlying endometriosis pathogenesis. Methods We generated endometriosis-like lesions by injecting endometrial tissue fragments into the peritoneal cavity in mice. The mice received a single (1x) or multiple inductions (6x) to simulate recurrent retrograde menstruation. Lesion development, hyperalgesia by behavioral testing, signs of peripheral sensitization, chronic inflammation, and neuroinflammation were examined with lesions, peritoneal fluids, dorsal root ganglia (DRG), spinal codes, and brain. Results Multiple lesion inductions increased lesion numbers and elevated abdominal and hind paw hypersensitivity compared to single induction mice. Elevated persistent glial cell activation across several brain regions and/or spinal cords was found in the multiple induction mice. Specifically, IBA1+ microglial soma size was increased in the hippocampus and thalamus. IBA1+ cells were abundant in the cortex, hippocampus, thalamus, and hypothalamus of the multiple induction mice. GFAP+ astrocytes were mainly elevated in the hippocampus. Elevated TRPV1, SP, and CGRP expressions in the DRG were persistent in the multiple induction mice. Furthermore, multiple inductions induced the severe disappearance of TIM4hi MHCIIlo residential macrophages and the influx of increased proinflammatory TIM4lo MHCIIhi macrophages in the peritoneal cavity. The single and multiple inductions elevated secreted TNFα, IL-1β, and IL-6 levels in the peritoneal cavity at 2 weeks. Elevated cytokine levels returned to the pre-induction levels in the single induction mice at 6 weeks; however, they remained elevated in the multiple induction mice. Conclusions Our results indicate that the repeatedly occurring lesion inductions (=mimic retrograde menstruation) can be a peripheral stimulus that induces nociceptive pain and creates composite chronic inflammatory stimuli to cause neuroinflammation and sensitize the CNS. The circuits of neuroplasticity and stimulation of peripheral organs via a feedback loop of neuroinflammation may mediate widespread endometriosis-associated CPP.
Collapse
Affiliation(s)
- Madeleine E. Harvey
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, 1770 NE Stadium Way, Pullman, WA, 99164, USA
| | - Mingxin Shi
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, 1770 NE Stadium Way, Pullman, WA, 99164, USA
| | - Yeongseok Oh
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, 1770 NE Stadium Way, Pullman, WA, 99164, USA
| | - Debra A. Mitchell
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, 1770 NE Stadium Way, Pullman, WA, 99164, USA
| | - Ov D. Slayden
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - James A. MacLean
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, 1770 NE Stadium Way, Pullman, WA, 99164, USA
| | - Kanako Hayashi
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, 1770 NE Stadium Way, Pullman, WA, 99164, USA
| |
Collapse
|
6
|
Miliotou AN, Kotsoni A, Zacharia LC. Deciphering the Role of Adrenergic Receptors in Alzheimer's Disease: Paving the Way for Innovative Therapies. Biomolecules 2025; 15:128. [PMID: 39858522 PMCID: PMC11764010 DOI: 10.3390/biom15010128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Neurodegenerative diseases are currently among the most devastating diseases with no effective disease-modifying drugs in the market, with Alzheimer's disease (AD) being the most prevalent. AD is a complex multifactorial neurodegenerative disorder characterized by progressive and severe cognitive impairment and memory loss. It is the most common cause of progressive memory loss (dementia) in the elderly, and to date, there is no effective treatment to cure or slow disease progression substantially. The role of adrenergic receptors in the pathogenesis of Alzheimer's disease and other tauopathies is poorly understood or investigated. Recently, some studies indicated a potential benefit of drugs acting on the adrenergic receptors for AD and dementias, although due to the heterogeneity of the drug classes used, the results on the whole remain inconclusive. The scope of this review article is to comprehensively review the literature on the possible role of adrenergic receptors in neurodegenerative diseases, stemming from the use of agonists and antagonists including antihypertensive and asthma drugs acting on the adrenergic receptors, but also from animal models and in vitro models where these receptors have been studied. Ultimately, we hope to obtain a better understanding of the role of these receptors, identify the gaps in knowledge, and explore the possibility of repurposing such drugs for AD, given their long history of use and safety.
Collapse
Affiliation(s)
- Androulla N. Miliotou
- Department of Health Sciences, School of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, 2417 Nicosia, Cyprus; (A.N.M.)
| | - Andria Kotsoni
- Department of Health Sciences, School of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, 2417 Nicosia, Cyprus; (A.N.M.)
| | - Lefteris C. Zacharia
- Department of Health Sciences, School of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, 2417 Nicosia, Cyprus; (A.N.M.)
- Bioactive Molecules Research Center, School of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, 2417 Nicosia, Cyprus
| |
Collapse
|
7
|
LIN PQ, PEI QW, LI B, YANG JM, ZOU LN, SU DZ, ZHANG JP, YIN HP, Nadine M, YANG JJ, A NV, Musawir Abbas K, JIANG ZL, LI JJ, YIN DC. The effects of hypothalamic microglial activation on ventricular arrhythmias in stress cardiomyopathy. J Geriatr Cardiol 2024; 21:1119-1132. [PMID: 39935443 PMCID: PMC11808488 DOI: 10.26599/1671-5411.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Background Stress cardiomyopathy (SCM) currently has a high incidence in older adults, and the theories regarding its causes include "catecholamine myocardial toxicity" and "sympathetic hyperactivation". However, the role of the central nervous system in the pathogenesis of SCM remains unknown. We investigated the role of microglia activation in the paraventricular hypothalamic nucleus (PVN) in the development of SCM. Methods An SCM model was created using male Sprague-Dawley (SD) rats, immobilized for 6 h every day for a week. Electrocardiogram, cardiac electrophysiology, and echocardiography examinations were performed to verify the changes in cardiac structure and function in rats with SCM. RNA sequencing was used to explore the changes in the hypothalamus during SCM. In addition, brain and heart tissues were collected to detect microglial activation and sympathetic activity. Results The main findings were as follows: (1) immobilization stress successfully induced SCM in SD rats; (2) microglia were significantly activated in the hypothalamus, as evidenced by cytosol thickening, increases in the number of microglial branches, and microglia enriched in the PVN; (3) in SCM, the microglia in the PVN exhibited increased central and peripheral cardiac sympathetic activity and increased the expression of neuroinflammatory factors; and (4) it is possible that inhibiting microglial activation could suppress the sympathetic activity of the central nervous system and heart and increase cardiac electrical stability in SCM rats. Conclusions SCM was induced in SD rats by immobilization stress, acting through the activation of the hypothalamic microglia. The activated microglia were specifically enriched in the PVN, increasing the activity of the central and peripheral sympathetic nervous systems by regulating the expression of neuro-inflammatory factors, mediating dysfunction of the left ventricle, and increasing the susceptibility to ventricular arrhythmias.
Collapse
Affiliation(s)
- Peng-Qi LIN
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Quan-Wei PEI
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bin LI
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jie-Mei YANG
- Department of Echocardiography, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li-Na ZOU
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - De-Zhan SU
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun-Pei ZHANG
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong-Peng YIN
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mbabazi Nadine
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun-Jie YANG
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Nevzorova Vera A
- Institute Therapy and Instrumental Diagnostic, Pacific State Medical University, Vladivostok, Russia
| | - Khan Musawir Abbas
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhao-Lei JIANG
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jing-Jie LI
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - De-Chun YIN
- Department of Geriatrics, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
8
|
Ishino F, Itoh J, Matsuzawa A, Irie M, Suzuki T, Hiraoka Y, Yoshikawa M, Kaneko-Ishino T. RTL4, a Retrovirus-Derived Gene Implicated in Autism Spectrum Disorder, Is a Microglial Gene That Responds to Noradrenaline in the Postnatal Brain. Int J Mol Sci 2024; 25:13738. [PMID: 39769499 PMCID: PMC11678650 DOI: 10.3390/ijms252413738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Retrotransposon Gag-like 4 (RTL4), a gene acquired from a retrovirus, is a causative gene in autism spectrum disorder. Its knockout mice exhibit increased impulsivity, impaired short-term spatial memory, failure to adapt to novel environments, and delayed noradrenaline (NA) recovery in the frontal cortex. However, due to its very low expression in the brain, it remains unknown which brain cells express RTL4 and its dynamics in relation to NA. We addressed these issues using knock-in mice carrying endogenous Rtl4 fused to Venus, which encodes a fluorescent protein. The RTL4-Venus fusion protein was detected as a secreted protein in the midbrain, hypothalamus, hippocampus and amygdala in the postnatal brain. Its signal intensity was high during critical periods of neonatal adaptation to novel environments. It was upregulated by various stimuli, including isoproterenol administration, whereas it was decreased by anesthesia but was maintained by milnacipran administration, suggesting its highly sensitive response to stressors, possible dependence on the arousal state and involvement in the NA reuptake process. In vitro mixed glial culture experiments demonstrated that Rtl4 is a microglial gene and suggested that RTL4 secretion responds rapidly to isoproterenol. Microglial RTL4 plays an important role in the NA response and possibly in the development of the NAergic neuronal network in the brain.
Collapse
Affiliation(s)
- Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (A.M.); (M.I.)
| | - Johbu Itoh
- Department of Neurology, School of Medicine, Tokai University School of Medicine, Isehara 259-1193, Japan;
| | - Ayumi Matsuzawa
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (A.M.); (M.I.)
| | - Masahito Irie
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (A.M.); (M.I.)
- Faculty of Nursing, Tokai University School of Medicine, Isehara 259-1193, Japan
| | - Toru Suzuki
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (T.S.); (Y.H.)
| | - Yuichi Hiraoka
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (T.S.); (Y.H.)
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Masanobu Yoshikawa
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara 259-1193, Japan;
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, Tokai University School of Medicine, Isehara 259-1193, Japan
| |
Collapse
|
9
|
Slavova D, Ortiz V, Blaise M, Bairachnaya M, Giros B, Isingrini E. Role of the locus coeruleus-noradrenergic system in stress-related psychopathology and resilience: Clinical and pre-clinical evidences. Neurosci Biobehav Rev 2024; 167:105925. [PMID: 39427811 DOI: 10.1016/j.neubiorev.2024.105925] [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: 07/05/2024] [Revised: 08/28/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Stressful events, from daily stressors to traumatic experiences, are common and occur at any age. Despite the high prevalence of trauma, not everyone develops stress-related disorders like major depressive disorder (MDD) and post-traumatic stress disorder (PTSD), a variation attributed to resilience, the ability to adapt and avoid negative consequences of significant stress. This review examines the locus coeruleus-norepinephrine (LC-NE) system, a critical component in the brain's stress response. It discusses the LC-NE system's anatomical and functional complexity and its role in individual variability in stress responses. How different etiological factors and stress modalities affect the LC-NE system, influencing both adaptive stress responses and psychopathologies, are discussed and supported by evidence from human and animal studies. It also explores molecular and cellular adaptations in the LC that contribute to resilience, including roles of neuropeptide, inflammatory cytokines, and genetic modulation, and addresses developmental and sex differences in stress vulnerability. The need for a multifaceted approach to understand stress-induced psychopathologies is emphasized and pave the way for more personalized interventions for stress-related disorders.
Collapse
Affiliation(s)
- Déa Slavova
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France
| | - Vanesa Ortiz
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France
| | - Maud Blaise
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France
| | - Marya Bairachnaya
- Douglas Research Center Institute, McGill University, Montreal, Canada
| | - Bruno Giros
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France; Douglas Research Center Institute, McGill University, Montreal, Canada
| | - Elsa Isingrini
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France.
| |
Collapse
|
10
|
Medina J, De Guzman RM, Workman JL. Prolactin mitigates chronic stress-induced maladaptive behaviors and physiology in ovariectomized female rats. Neuropharmacology 2024; 258:110095. [PMID: 39084597 DOI: 10.1016/j.neuropharm.2024.110095] [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: 03/22/2024] [Revised: 06/05/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Stress is a major risk factor for several neuropsychiatric disorders in women, including postpartum depression. During the postpartum period, diminished ovarian hormone secretion increases susceptibility to developing depressive symptoms. Pleiotropic peptide hormones, like prolactin, are markedly released during lactation and suppress hypothalamic-pituitary-adrenal axis responses in women and acute stress-induced behavioral responses in female rodents. However, the effects of prolactin on chronic stress-induced maladaptive behaviors remain unclear. Here, we used chronic variable stress to induce maladaptive physiology in ovariectomized female rats and concurrently administered prolactin to assess its effects on several depression-relevant behavioral, endocrine, and neural characteristics. We found that chronic stress increased sucrose anhedonia and passive coping in saline-treated, but not prolactin-treated rats. Prolactin treatment did not alter stress-induced thigmotaxis, corticosterone (CORT) concentrations, hippocampal cell activation or survival. However, prolactin treatment reduced basal CORT concentrations and increased dopaminergic cells in the ventral tegmental area. Further, prolactin-treated rats had reduced microglial activation in the ventral hippocampus following chronic stress exposure. Together, these data suggest prolactin mitigates chronic stress-induced maladaptive behaviors and physiology in hypogonadal females. Moreover, these findings imply neuroendocrine-immune mechanisms by which peptide hormones confer stress resilience during periods of low ovarian hormone secretion.
Collapse
Affiliation(s)
- Joanna Medina
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY, 12222, USA.
| | - Rose M De Guzman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY, 12222, USA
| | - Joanna L Workman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY, 12222, USA; Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY, 12222, USA
| |
Collapse
|
11
|
Ghasemi M, Mehranfard N. Neuroprotective actions of norepinephrine in neurological diseases. Pflugers Arch 2024; 476:1703-1725. [PMID: 39136758 DOI: 10.1007/s00424-024-02999-w] [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: 03/25/2024] [Revised: 06/24/2024] [Accepted: 07/24/2024] [Indexed: 10/09/2024]
Abstract
Precise control of norepinephrine (NE) levels and NE-receptor interaction is crucial for proper function of the brain. Much evidence for this view comes from experimental studies that indicate an important role for NE in the pathophysiology and treatment of various conditions, including cognitive dysfunction, Alzheimer's disease, Parkinson's disease, multiple sclerosis, and sleep disorders. NE provides neuroprotection against several types of insults in multiple ways. It abrogates oxidative stress, attenuates neuroinflammatory responses in neurons and glial cells, reduces neuronal and glial cell activity, promotes autophagy, and ameliorates apoptotic responses to a variety of insults. It is beneficial for the treatment of neurodegenerative diseases because it improves the generation of neurotrophic factors, promotes neuronal survival, and plays an important role in the regulation of adult neurogenesis. This review aims to present the evidence supporting a principal role for NE in neuroprotection, and molecular mechanisms of neuroprotection.
Collapse
Affiliation(s)
- Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasrin Mehranfard
- Nanokadeh Darooee Samen Private Joint Stock Company, Shafa Street, Urmia, 5715793731, Iran.
| |
Collapse
|
12
|
Lanshakov DA, Sukhareva EV, Bulygina VV, Khozyainova AA, Gerashchenko TS, Denisov EV, Kalinina TS. Brainstem transcriptomic changes in male Wistar rats after acute stress, comparing the use of duplex specific nuclease (DSN). Sci Rep 2024; 14:21856. [PMID: 39300279 DOI: 10.1038/s41598-024-73042-2] [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: 12/25/2023] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
In this work, we have analyzed the transcriptomic changes in the brainstem of male Wistar rats 2 h after an acute stress exposure. We performed duplex-specific nuclease normalization of cDNA libraries and compared the results back-to-back for the first time. Based on our RNAseq data, we selected reference genes for RT-qPCR that are best suited for acute stress experiments. Most genes were upregulated. We detected a massive shift in neuropeptide Crh, Trh,Cga, Tshb, Uts2b, Tac4, Lep and neuropeptide receptor Hcrtr1, Sstr5, Bdkrb2, Crhr2 signaling, as well as glutamate Grin3b, Grm2 and GABA Gpr156, acetylcholine Chrm4,Chrne, adrenergic Adra2b receptors expression. A strong increase in the expression of intermediate filaments Krt83/Krt86/Krt80/Krt84/Krt87/Krt4/Krt76 and motor proteins Myo7a, Klc3 was detected. Remarkably, in the absence of astrocyte activation, we also observed signs of microglial activation at this time point. Both expression of anti-inflammatory cytokines Il13, Ccl24 and pro-inflammatory cytokine receptors Il9r, Il12rb1, Tnfrsf14, Tnfrsf13c, Tnfrsf25, Tnfrsf1b were increased. In the Wnt signaling pathway, we observed increased expression of ligands-receptors Wnt1, Wnt11, Ror2 and also negative regulators Notum, Sfrp5, Sost. RNAseq results after DSN treatment correlated at a high level with RNAseq results without DSN, but there was a proportion of genes that shifted their logFC values. They are mostly rare transcripts TPM 1-10 with higher 0.5-0.9 GC content.
Collapse
Affiliation(s)
- Dmitriy A Lanshakov
- Postgenomics Neurobiology Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation.
- Natural Science Department, Novosibirsk State University, Novosibirsk, Russian Federation.
| | - Ekaterina V Sukhareva
- Postgenomics Neurobiology Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation
| | - Veta V Bulygina
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation
| | - Anna A Khozyainova
- Laboratory of Cancer Progression Biology, Tomsk National Research Medical Center, Cancer Research Institute, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Tatiana S Gerashchenko
- Laboratory of Cancer Progression Biology, Tomsk National Research Medical Center, Cancer Research Institute, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Evgeny V Denisov
- Laboratory of Cancer Progression Biology, Tomsk National Research Medical Center, Cancer Research Institute, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Tatyana S Kalinina
- Natural Science Department, Novosibirsk State University, Novosibirsk, Russian Federation
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation
| |
Collapse
|
13
|
Zhao Y, Li X, Yu W, Lin W, Wei W, Zhang L, Liu D, Ma H, Chen J. Differential expression of ADRB1 causes different responses to norepinephrine in adipocytes of Duroc-Landrace-Yorkshire pigs and min pigs. J Therm Biol 2024; 123:103906. [PMID: 38970835 DOI: 10.1016/j.jtherbio.2024.103906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/11/2024] [Accepted: 06/23/2024] [Indexed: 07/08/2024]
Abstract
Research has shown that pigs from different regions exhibit varying responses to cold stimuli. Typically, cold stimuli induce browning of white adipose tissue mediated by adrenaline, promoting non-shivering thermogenesis. However, the molecular mechanisms underlying differential response of pig breeds to norepinephrine are unclear. The aim of this study was to investigate the differences and molecular mechanisms of the effects of norepinephrine (NE) treatment on adipocytes of Min pigs (a cold-resistant pig breed) and Duroc-Landrace-Yorkshire (DLY) pigs. Real time-qPCR, western blot, and immunofluorescence were performed following NE treatment on cell cultures of adipocytes originating from Min pigs (n = 3) and DLY pigs (n = 3) to assess the expressions of adipogenesis markers, beige fat markers, and mitochondrial biogenesis markers. The results showed that NE did not affect browning of adipocytes in DLY pigs, whereas promoted browning of adipocytes in Min pigs. Further, the expression of ADRB1 (Adrenoceptor Beta 1, ADRB1) was higher in subcutaneous adipose tissue and adipocytes of Min pigs than those of DLY pigs. Overexpression of ADRB1 in DLY pig adipocytes enhanced sensitivity to NE, exhibiting decreased adipogenesis markers, upregulated beige fat markers, and increased mitochondrial biogenesis. Conversely, adipocytes treated with ADRB1 antagonist in Min pigs resulted in decreased cellular sensitivity to NE. Further studies revealed differential CpG island methylation in ADRB1 promoter region, with lower methylation levels in Min pigs compared to DLY pigs. In conclusion, differential methylation of the ADRB1 promoter region leads to different ADRB1 expression, resulting in varying responsiveness to NE in adipocytes of two pig breeds. Our results provide new insights for further analysis of the differential cold responsiveness in pig breeds from different regions.
Collapse
Affiliation(s)
- Yuelei Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xuexin Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wensai Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Weimin Lin
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Di Liu
- Institute of Animal Husbandry Research, HeiLongJiang Academy of Agricultural Sciences, Harbi, 150086, China
| | - Hong Ma
- Institute of Animal Husbandry Research, HeiLongJiang Academy of Agricultural Sciences, Harbi, 150086, China
| | - Jie Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
14
|
Birkle TJ, Willems HM, Skidmore J, Brown GC. Disease phenotypic screening in neuron-glia cocultures identifies blockers of inflammatory neurodegeneration. iScience 2024; 27:109454. [PMID: 38550989 PMCID: PMC10973195 DOI: 10.1016/j.isci.2024.109454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/09/2023] [Accepted: 03/06/2024] [Indexed: 01/30/2025] Open
Abstract
Neuropathology is often mediated by interactions between neurons and glia that cannot be modeled by monocultures. However, cocultures are difficult to use and analyze for high-content screening. Here, we perform compound screening using primary neuron-glia cultures to model inflammatory neurodegeneration, live-cell stains, and automated classification of neurons, astrocytes or microglia using open-source software. Out of 227 compounds with known bioactivities, 29 protected against lipopolysaccharide-induced neuronal loss, including drugs affecting adrenergic, steroid, inflammatory and MAP kinase signaling. The screen also identified physiological compounds, such as noradrenaline and progesterone, that protected and identified neurotoxic compounds, such as a TLR7 agonist, that induced microglial proliferation. Most compounds used here have not been tested in a neuron-glia coculture neurodegeneration assay previously. Thus, combining a complex cellular disease model with high-content screening of known compounds and automated image analysis allows identification of important biology, as well as potential targets and drugs for treatment.
Collapse
Affiliation(s)
| | | | - John Skidmore
- ALBORADA Drug Discovery Institute, Cambridge CB2 0AH, UK
| | - Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| |
Collapse
|
15
|
Li S, Yang D, Zhou X, Chen L, Liu L, Lin R, Li X, Liu Y, Qiu H, Cao H, Liu J, Cheng Q. Neurological and metabolic related pathophysiologies and treatment of comorbid diabetes with depression. CNS Neurosci Ther 2024; 30:e14497. [PMID: 37927197 PMCID: PMC11017426 DOI: 10.1111/cns.14497] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND The comorbidity between diabetes mellitus and depression was revealed, and diabetes mellitus increased the prevalence of depressive disorder, which ranked 13th in the leading causes of disability-adjusted life-years. Insulin resistance, which is common in diabetes mellitus, has increased the risk of depressive symptoms in both humans and animals. However, the mechanisms behind the comorbidity are multi-factorial and complicated. There is still no causal chain to explain the comorbidity exactly. Moreover, Selective serotonin reuptake inhibitors, insulin and metformin, which are recommended for treating diabetes mellitus-induced depression, were found to be a risk factor in some complications of diabetes. AIMS Given these problems, many researchers made remarkable efforts to analyze diabetes complicating depression from different aspects, including insulin resistance, stress and Hypothalamic-Pituitary-Adrenal axis, neurological system, oxidative stress, and inflammation. Drug therapy, such as Hydrogen Sulfide, Cannabidiol, Ascorbic Acid and Hesperidin, are conducive to alleviating diabetes mellitus and depression. Here, we reviewed the exact pathophysiology underlying the comorbidity between depressive disorder and diabetes mellitus and drug therapy. METHODS The review refers to the available literature in PubMed and Web of Science, searching critical terms related to diabetes mellitus, depression and drug therapy. RESULTS In this review, we found that brain structure and function, neurogenesis, brain-derived neurotrophic factor and glucose and lipid metabolism were involved in the pathophysiology of the comorbidity. Obesity might lead to diabetes mellitus and depression through reduced adiponectin and increased leptin and resistin. In addition, drug therapy displayed in this review could expand the region of potential therapy. CONCLUSIONS The review summarizes the mechanisms underlying the comorbidity. It also overviews drug therapy with anti-diabetic and anti-depressant effects.
Collapse
Affiliation(s)
- Sixin Li
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Dong Yang
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Xuhui Zhou
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Lu Chen
- Department of Gastroenterology, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of GastroenterologyBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Lini Liu
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Ruoheng Lin
- Department of Psychiatry, National Clinical Research Center for Mental DisordersThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Xinyu Li
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Ying Liu
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Huiwen Qiu
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Hui Cao
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Jian Liu
- Center for Medical Research and Innovation, The First Hospital, Hunan University of Chinese MedicineChangshaHunanChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
16
|
Wee IC, Arulsamy A, Corrigan F, Collins-Praino L. Long-Term Impact of Diffuse Traumatic Brain Injury on Neuroinflammation and Catecholaminergic Signaling: Potential Relevance for Parkinson's Disease Risk. Molecules 2024; 29:1470. [PMID: 38611750 PMCID: PMC11013319 DOI: 10.3390/molecules29071470] [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: 01/05/2024] [Revised: 02/11/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Traumatic brain injury (TBI) is associated with an increased risk of developing Parkinson's disease (PD), though the exact mechanisms remain unclear. TBI triggers acute neuroinflammation and catecholamine dysfunction post-injury, both implicated in PD pathophysiology. The long-term impact on these pathways following TBI, however, remains uncertain. In this study, male Sprague-Dawley rats underwent sham surgery or Marmarou's impact acceleration model to induce varying TBI severities: single mild TBI (mTBI), repetitive mild TBI (rmTBI), or moderate-severe TBI (msTBI). At 12 months post-injury, astrocyte reactivity (GFAP) and microglial levels (IBA1) were assessed in the striatum (STR), substantia nigra (SN), and prefrontal cortex (PFC) using immunohistochemistry. Key enzymes and receptors involved in catecholaminergic transmission were measured via Western blot within the same regions. Minimal changes in these markers were observed, regardless of initial injury severity. Following mTBI, elevated protein levels of dopamine D1 receptors (DRD1) were noted in the PFC, while msTBI resulted in increased alpha-2A adrenoceptors (ADRA2A) in the STR and decreased dopamine beta-hydroxylase (DβH) in the SN. Neuroinflammatory changes were subtle, with a reduced number of GFAP+ cells in the SN following msTBI. However, considering the potential for neurodegenerative outcomes to manifest decades after injury, longer post-injury intervals may be necessary to observe PD-relevant alterations within these systems.
Collapse
Affiliation(s)
- Ing Chee Wee
- Cognition, Ageing and Neurodegenerative Disease Laboratory, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia;
| | - Frances Corrigan
- Head Injury Lab, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Lyndsey Collins-Praino
- Cognition, Ageing and Neurodegenerative Disease Laboratory, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia;
| |
Collapse
|
17
|
Chen D, Lou Q, Song XJ, Kang F, Liu A, Zheng C, Li Y, Wang D, Qun S, Zhang Z, Cao P, Jin Y. Microglia govern the extinction of acute stress-induced anxiety-like behaviors in male mice. Nat Commun 2024; 15:449. [PMID: 38200023 PMCID: PMC10781988 DOI: 10.1038/s41467-024-44704-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Anxiety-associated symptoms following acute stress usually become extinct gradually within a period of time. However, the mechanisms underlying how individuals cope with stress to achieve the extinction of anxiety are not clear. Here we show that acute restraint stress causes an increase in the activity of GABAergic neurons in the CeA (GABACeA) in male mice, resulting in anxiety-like behaviors within 12 hours; meanwhile, elevated GABACeA neuronal CX3CL1 secretion via MST4 (mammalian sterile-20-like kinase 4)-NF-κB-CX3CL1 signaling consequently activates microglia in the CeA. Activated microglia in turn inhibit GABACeA neuronal activity via the engulfment of their dendritic spines, ultimately leading to the extinction of anxiety-like behaviors induced by restraint stress. These findings reveal a dynamic molecular and cellular mechanism in which microglia drive a negative feedback to inhibit GABACeA neuronal activity, thus facilitating maintenance of brain homeostasis in response to acute stress.
Collapse
Grants
- 32025017 National Natural Science Foundation of China (National Science Foundation of China)
- 32121002 National Natural Science Foundation of China (National Science Foundation of China)
- 82101300 National Natural Science Foundation of China (National Science Foundation of China)
- U22A20305 National Natural Science Foundation of China (National Science Foundation of China)
- the National Key Research and Development Program of China (STI2030-Major Projects 2021ZD0203100), Plans for Major Provincial Science & Technology Projects (202303a07020002), the CAS Project for Young Scientists in Basic Research (YSBR-013), the Innovative Research Team of High-level Local Universities in Shanghai (SHSMU-ZDCX20211902), the Institute of Health and Medicine (OYZD20220007)
- the China National Postdoctoral Program for Innovative Talents (BX20220283), the China Postdoctoral Science Foundation (2023M733395)
- Youth Innovation Promotion Association CAS, CAS Collaborative Innovation Program of Hefei Science Center (2021HSC-CIP013), the Fundamental Research Funds for the Central Universities (WK9100000030), USTC Research Funds of the Double First-Class Initiative (YD9100002018), the Natural Science Foundation of Anhui Province (2208085J30), and USTC Tang Scholar.
Collapse
Affiliation(s)
- Danyang Chen
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Qianqian Lou
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xiang-Jie Song
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Fang Kang
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - An Liu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230022, China
| | - Changjian Zheng
- Department of Anesthesiology, the First Affiliated Hospital of Wannan Medical College, Wuhu, 241002, China
| | - Yanhua Li
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Di Wang
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Sen Qun
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Zhi Zhang
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- The Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| | - Peng Cao
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| | - Yan Jin
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
18
|
Stanford SC, Heal DJ. Adrenoceptors: A Focus on Psychiatric Disorders and Their Treatments. Handb Exp Pharmacol 2024; 285:507-554. [PMID: 37495853 DOI: 10.1007/164_2023_675] [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] [Indexed: 07/28/2023]
Abstract
Research into the involvement of adrenoceptor subtypes in the cause(s) of psychiatric disorders is particularly challenging. This is partly because of difficulties in developing animal models that recapitulate the human condition but also because no evidence for any causal links has emerged from studies of patients. These, and other obstacles, are outlined in this chapter. Nevertheless, many drugs that are used to treat psychiatric disorders bind to adrenoceptors to some extent. Direct or indirect modulation of the function of specific adrenoceptor subtypes mediates all or part of the therapeutic actions of drugs in various psychiatric disorders. On the other hand, interactions with central or peripheral adrenoceptors can also explain their side effects. This chapter discusses both aspects of the field, focusing on disorders that are prevalent: depression, schizophrenia, anxiety, attention-deficit hyperactivity disorder, binge-eating disorder, and substance use disorder. In so doing, we highlight some unanswered questions that need to be resolved before it will be feasible to explain how changes in the function of any adrenoceptor subtype affect mood and behavior in humans and other animals.
Collapse
Affiliation(s)
- S Clare Stanford
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - David J Heal
- DevelRx Ltd, BioCity, Nottingham, UK
- Department of Life Sciences, University of Bath, Bath, UK
| |
Collapse
|
19
|
Chen K, Qi X, Zhu LL, Li ML, Cong B, Li YM. Quantitative analysis of microglia morphological changes in the hypothalamus of chronically stressed rats. Brain Res Bull 2024; 206:110861. [PMID: 38141789 DOI: 10.1016/j.brainresbull.2023.110861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 12/25/2023]
Abstract
Based on the successful establishment of a rat model of chronic restraint stress, we used multiple algorithms to quantify the morphological changes of rat hypothalamic microglia from various perspectives, providing a pathomorphological basis for the subsequent study of molecular mechanisms of hypothalamic stress injury, such as neuroinflammation. To verify the successful establishment of the chronic stress model, an enzyme-linked immunosorbent assay was performed to detect serum glucocorticoid levels. Microglia labeled with Iba1 in frozen sections of rat hypothalamus were scanned and photographed at multiple levels using confocal microscopy. Subsequently, images were processed for external contouring and skeletonization, and morphological indices of microglia were calculated and analyzed using fractal, skeleton, and Sholl analysis. In addition, the co-expression of CD68 (a marker that can reflect phagocytic activity) and Iba1 was observed by immunofluorescence technique. Compared with the control group, microglia in the chronic stress group displayed reduced fractal dimension and lacunarity, increased density and circularity, enlarged soma areas, and shortened and reduced branches. Sholl analysis confirmed the reduced complexity of microglia following chronic stress. Meanwhile, microglia CD68 increased significantly, indicating that the microglia in the chronic stress group have greater phagocytosis activity. In summary, chronic restraint stress promoted the conversion of microglia in the rat hypothalamus to a less complex form, manifested as larger soma, shorter and fewer branches, more uniform and dense texture, and increased circularity; indeed, the shape of these microglia resembled that of amoeba and they displayed strong phagocytosis activity.
Collapse
Affiliation(s)
- Ke Chen
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China
| | - Xin Qi
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China
| | - Lin-Lin Zhu
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China
| | - Mei-Li Li
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China
| | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China.
| | - Ying-Min Li
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China.
| |
Collapse
|
20
|
Davis AB, Lloyd KR, Bollinger JL, Wohleb ES, Reyes TM. Adolescent high fat diet alters the transcriptional response of microglia in the prefrontal cortex in response to stressors in both male and female mice. Stress 2024; 27:2365864. [PMID: 38912878 PMCID: PMC11228993 DOI: 10.1080/10253890.2024.2365864] [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: 10/31/2023] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Both obesity and high fat diets (HFD) have been associated with an increase in inflammatory gene expression within the brain. Microglia play an important role in early cortical development and may be responsive to HFD, particularly during sensitive windows, such as adolescence. We hypothesized that HFD during adolescence would increase proinflammatory gene expression in microglia at baseline and potentiate the microglial stress response. Two stressors were examined, a physiological stressor [lipopolysaccharide (LPS), IP] and a psychological stressor [15 min restraint (RST)]. From 3 to 7 weeks of age, male and female mice were fed standard control diet (SC, 20% energy from fat) or HFD (60% energy from fat). On P49, 1 h before sacrifice, mice were randomly assigned to either stressor exposure or control conditions. Microglia from the frontal cortex were enriched using a Percoll density gradient and isolated via fluorescence-activated cell sorting (FACS), followed by RNA expression analysis of 30 genes (27 target genes, three housekeeping genes) using Fluidigm, a medium throughput qPCR platform. We found that adolescent HFD induced sex-specific transcriptional response in cortical microglia, both at baseline and in response to a stressor. Contrary to our hypothesis, adolescent HFD did not potentiate the transcriptional response to stressors in males, but rather in some cases, resulted in a blunted or absent response to the stressor. This was most apparent in males treated with LPS. However, in females, potentiation of the LPS response was observed for select proinflammatory genes, including Tnfa and Socs3. Further, HFD increased the expression of Itgam, Ikbkb, and Apoe in cortical microglia of both sexes, while adrenergic receptor expression (Adrb1 and Adra2a) was changed in response to stressor exposure with no effect of diet. These data identify classes of genes that are uniquely affected by adolescent exposure to HFD and different stressor modalities in males and females.
Collapse
Affiliation(s)
- Alyshia B Davis
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Kelsey R Lloyd
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Teresa M Reyes
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
21
|
Gu X, Zhao Z, Chen X, Zhang L, Fang H, Zhao T, Ju S, Gao W, Qian X, Wang X, Zhang J, Cheng H. Imaging microglia surveillance during sleep-wake cycles in freely behaving mice. eLife 2023; 12:RP86749. [PMID: 38132088 PMCID: PMC10746140 DOI: 10.7554/elife.86749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Microglia surveillance manifests itself as dynamic changes in cell morphology and functional remodeling. Whether and how microglia surveillance is coupled to brain state switches during natural sleep-wake cycles remains unclear. To address this question, we used miniature two-photon microscopy (mTPM) to acquire time-lapse high-resolution microglia images of the somatosensory cortex, along with EEG/EMG recordings and behavioral video, in freely-behaving mice. We uncovered fast and robust brain state-dependent changes in microglia surveillance, occurring in parallel with sleep dynamics and early-onset phagocytic microglial contraction during sleep deprivation stress. We also detected local norepinephrine fluctuation occurring in a sleep state-dependent manner. We showed that the locus coeruleus-norepinephrine system, which is crucial to sleep homeostasis, is required for both sleep state-dependent and stress-induced microglial responses and β2-adrenergic receptor signaling plays a significant role in this process. These results provide direct evidence that microglial surveillance is exquisitely tuned to signals and stressors that regulate sleep dynamics and homeostasis so as to adjust its varied roles to complement those of neurons in the brain. In vivo imaging with mTPM in freely behaving animals, as demonstrated here, opens a new avenue for future investigation of microglia dynamics and sleep biology in freely behaving animals.
Collapse
Affiliation(s)
- Xiaochun Gu
- PKU-Nanjing Institute of Translational Medicine, Nanjing Raygen HealthNanjingChina
- National Platform for Medical Engineering Education Integration, Southeast UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast UniversityNanjingChina
- Key Laboratory of Developmental Genes and Human Diseases, Department of Histology Embryology, Medical School, Southeast UniversityNanjingChina
| | - Zhong Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational MedicineNanjingChina
| | - Xueli Chen
- Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational MedicineNanjingChina
| | - Lifeng Zhang
- Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational MedicineNanjingChina
| | - Huaqiang Fang
- Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational MedicineNanjingChina
| | - Ting Zhao
- PKU-Nanjing Institute of Translational Medicine, Nanjing Raygen HealthNanjingChina
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast UniversityNanjingChina
| | - Weizheng Gao
- Academy of Advanced Interdisciplinary Study, College of Engineering, Peking UniversityBeijingChina
| | - Xiaoyu Qian
- Academy of Advanced Interdisciplinary Study, College of Engineering, Peking UniversityBeijingChina
| | - Xianhua Wang
- Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational MedicineNanjingChina
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking UniversityBeijingChina
| | - Jue Zhang
- Academy of Advanced Interdisciplinary Study, College of Engineering, Peking UniversityBeijingChina
| | - Heping Cheng
- Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational MedicineNanjingChina
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking UniversityBeijingChina
- National Biomedical Imaging Center, State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, College of Future Technology, Peking UniversityBeijingChina
| |
Collapse
|
22
|
Zheng Y, Liu D, Guo H, Chen W, Liu Z, Li Z, Hu T, Zhang Y, Li X, Zhao Z, Cai Q, Ge F, Fan Y, Guan X. Paternal methamphetamine exposure induces higher sensitivity to methamphetamine in male offspring through driving ADRB1 on CaMKII-positive neurons in mPFC. Transl Psychiatry 2023; 13:324. [PMID: 37857642 PMCID: PMC10587075 DOI: 10.1038/s41398-023-02624-x] [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: 02/09/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023] Open
Abstract
Paternal abuse of drugs, such as methamphetamine (METH), elevates the risk of developing addiction in subsequent generations, however, its underlying molecular mechanism remains poorly understood. Male adult mice (F0) were exposed to METH for 30 days, followed by mating with naïve female mice to create the first-generation mice (F1). When growing to adulthood, F1 were subjected to conditioned place preference (CPP) test. Subthreshold dose of METH (sd-METH), insufficient to induce CPP normally, were used in F1. Selective antagonist (betaxolol) for β1-adrenergic receptor (ADRB1) or its knocking-down virus were administrated into mPFC to regulate ADRB1 function and expression on CaMKII-positive neurons. METH-sired male F1 acquired sd-METH-induced CPP, indicating that paternal METH exposure induce higher sensitivity to METH in male F1. Compared with saline (SAL)-sired male F1, CaMKII-positive neuronal activity was normal without sd-METH, but strongly evoked after sd-METH treatment in METH-sired male F1 during adulthood. METH-sired male F1 had higher ADRB1 levels without sd-METH, which was kept at higher levels after sd-METH treatment in mPFC. Either inhibiting ADRB1 function with betaxolol, or knocking-down ADRB1 level on CaMKII-positive neurons (ADRB1CaMKII) with virus transfection efficiently suppressed sd-METH -evoked mPFC activation, and ultimately blocked sd-METH-induced CPP in METH-sired male F1. In the process, the p-ERK1/2 and ΔFosB may be potential subsequent signals of mPFC ADRB1CaMKII. The mPFC ADRB1CaMKII mediates paternal METH exposure-induced higher sensitivity to drug addiction in male offspring, raising a promising pharmacological target for predicting or treating transgenerational addiction.
Collapse
Affiliation(s)
- Yanyan Zheng
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Dekang Liu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hao Guo
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wenwen Chen
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhaoyu Liu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhaosu Li
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tao Hu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuanyuan Zhang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiang Li
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ziheng Zhao
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qinglong Cai
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Feifei Ge
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
23
|
Diebold M, Fehrenbacher L, Frosch M, Prinz M. How myeloid cells shape experimental autoimmune encephalomyelitis: At the crossroads of outside-in immunity. Eur J Immunol 2023; 53:e2250234. [PMID: 37505465 DOI: 10.1002/eji.202250234] [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: 02/02/2023] [Revised: 05/21/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of central nervous system (CNS) autoimmunity. It is most commonly used to mimic aspects of multiple sclerosis (MS), a demyelinating disorder of the human brain and spinal cord. The innate immune response displays one of the core pathophysiological features linked to both the acute and chronic stages of MS. Hence, understanding and targeting the innate immune response is essential. Microglia and other CNS resident MUs, as well as infiltrating myeloid cells, diverge substantially in terms of both their biology and their roles in EAE. Recent advances in the field show that antigen presentation, as well as disease-propagating and regulatory interactions with lymphocytes, can be attributed to specific myeloid cell types and cell states in EAE lesions, following a distinct temporal pattern during disease initiation, propagation and recovery. Furthermore, single-cell techniques enable the assessment of characteristic proinflammatory as well as beneficial cell states, and identification of potential treatment targets. Here, we discuss the principles of EAE induction and protocols for varying experimental paradigms, the composition of the myeloid compartment of the CNS during health and disease, and systematically review effects on myeloid cells for therapeutic approaches in EAE.
Collapse
Affiliation(s)
- Martin Diebold
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Luca Fehrenbacher
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Maximilian Frosch
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| |
Collapse
|
24
|
Gui H, Chen X, Li L, Zhu L, Jing Q, Nie Y, Zhang X. Psychological distress influences lung cancer: Advances and perspectives on the immune system and immunotherapy. Int Immunopharmacol 2023; 121:110251. [PMID: 37348230 DOI: 10.1016/j.intimp.2023.110251] [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: 02/19/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 06/24/2023]
Abstract
Lung cancer has the highest incidence rate and mortality worldwide. Moreover, multiple factors may cause heterogeneity in the efficacy of immunotherapy for lung cancer, and preclinical studies have gradually uncovered the promotive effects of psychological distress (PD) on tumor hallmarks. Therefore, treatment targeted at PD may be a vital factor in adjusting and improving immunotherapy for lung cancer. Here, by focusing on the central nervous system, as well as stress-related crucial neurotransmitters and hormones, we highlight the effects of PD on the lung immune system, the lung tumor microenvironment (TME) and immunotherapy, which brings a practicable means and psychosocial perspective to lung cancer treatment.
Collapse
Affiliation(s)
- Huan Gui
- Department of Hyperbaric Oxygen, People`s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China; School of Medicine, Guizhou University, Guiyang 550025, China
| | - Xulong Chen
- School of Medicine, Guizhou University, Guiyang 550025, China; Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Linzhao Li
- School of Medicine, Guizhou University, Guiyang 550025, China
| | - Lan Zhu
- School of Medicine, Guizhou University, Guiyang 550025, China
| | - Qianyu Jing
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Yingjie Nie
- School of Medicine, Guizhou University, Guiyang 550025, China; NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| | - Xiangyan Zhang
- School of Medicine, Guizhou University, Guiyang 550025, China; NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| |
Collapse
|
25
|
Zhou H, Wang K, Xu Z, Liu D, Wang Y, Guo M. Chronic unpredictable stress induces depression/anxiety-related behaviors and alterations of hippocampal monoamine receptor mRNA expression in female mice at different ages. Heliyon 2023; 9:e18369. [PMID: 37539192 PMCID: PMC10393760 DOI: 10.1016/j.heliyon.2023.e18369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 08/05/2023] Open
Abstract
Depression and anxiety are the most common mental health disorders. Though they affect people at any age and occur more often in females, the pathophysiological changes under these conditions are less investigated. In the present study, we examined the effects of age and stress on depression- and anxiety-related behaviors in female mice. Saccharin preference and the open field test were carried out before and after chronic unpredictable stress in 4-, 14- and 25-month-old female mice. After behavioral tests, mRNA levels of monoamine receptors in the hippocampus were measured by real-time RT-PCR. Chronic unpredictable stress decreased saccharin preference in 4-, 14- and 25-month-old mice and the time spent in the center in the open field test in 25-month-old mice. For monoamine receptors, analysis of variance revealed significant effects of age on mRNA levels of Htr1a, Htr2a, Htr6, Adra1a, Adrb2, and Adrb3, significant effects of stress on mRNA levels of Htr4, Adra2c, Adrb1, and Adrb2, and interactions of age × stress on mRNA levels of Htr1a, Htr5b, Adra1d, Adra2a, Adra2c, and Adrb1. Chronic unpredictable stress decreased mRNA levels of Htr4, Htr5b, Adra2c, and Adrb1 in 4-month-old female mice. Correlations were observed between saccharin preference and mRNA levels of Htr4, Htr5b, Htr6, Adra1d, Adra2a, and Adra2c in 4-month-old mice and between the time spent in the center in the open field test and mRNA levels of Htr1b in 4-month-old mice, Htr3a, Htr7, and Adrb2 in 14-month-old mice, and Drd2 in 4- and 14-month-old mice. Our findings support that stress induces depression- and anxiety-related behaviors and the expression of hippocampal monoamine receptors in an age-dependent manner in female mice.
Collapse
Affiliation(s)
- Han Zhou
- Department of Psychology, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Kaixin Wang
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Zhicheng Xu
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Dunjiang Liu
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Yameng Wang
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Ming Guo
- Department of Psychology, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| |
Collapse
|
26
|
Sequeira MK, Bolton JL. Stressed Microglia: Neuroendocrine-Neuroimmune Interactions in the Stress Response. Endocrinology 2023; 164:bqad088. [PMID: 37279575 PMCID: PMC11491833 DOI: 10.1210/endocr/bqad088] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 06/08/2023]
Abstract
Stressful life experiences are associated with the development of neuropsychiatric disorders like depression. Emerging evidence indicates that microglia, the specialized resident macrophages of the brain, may be a key mediator of the relationship between psychosocial stressor exposure and adaptive or maladaptive responses at the level of synaptic, circuit, and neuroimmune alterations. Here, we review current literature regarding how psychosocial stressor exposure changes microglial structure and function, thereby altering behavioral and brain outcomes, with a particular focus on age- and sex-dependent effects. We argue that additional emphasis should be placed in future research on investigating sex differences and the impacts of stressor exposure during sensitive periods of development, as well as going beyond traditional morphological measurements to interrogate microglial function. The bidirectional relationship between microglia and the stress response, particularly the role of microglia in the neuroendocrine control of stress-related circuits, is also an important area for future investigation. Finally, we discuss emerging themes and future directions that point to the possibility of the development of novel therapeutics for stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
| | - Jessica L Bolton
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
27
|
Huang Z, Jordan JD, Zhang Q. Early life adversity as a risk factor for cognitive impairment and Alzheimer's disease. Transl Neurodegener 2023; 12:25. [PMID: 37173751 PMCID: PMC10182702 DOI: 10.1186/s40035-023-00355-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Neurological conditions, including cognitive impairment and Alzheimer's disease (AD), impose a huge burden on society, affecting millions of people globally. In addition to genetic factors, recent studies indicate that environmental and experiential factors may contribute to the pathogenesis of these diseases. Early life adversity (ELA) has a profound impact on brain function and health later in life. In rodent models, exposure to ELA results in specific cognitive deficits and aggravated AD pathology. Extensive concerns have been raised regarding the higher risk of developing cognitive impairments in people with a history of ELA. In this review, we scrutinize findings from human and animal studies focusing on the connection of ELA with cognitive impairment and AD. These discoveries suggest that ELA, especially at early postnatal stages, increases susceptibility to cognitive impairment and AD later in life. In terms of mechanisms, ELA could lead to dysregulation of the hypothalamus-pituitary-adrenal axis, altered gut microbiome, persistent inflammation, oligodendrocyte dysfunction, hypomyelination, and aberrant adult hippocampal neurogenesis. Crosstalks among these events may synergistically contribute to cognitive impairment later in life. Additionally, we discuss several interventions that may alleviate adverse consequences of ELA. Further investigation into this crucial area will help improve ELA management and reduce the burden of related neurological conditions.
Collapse
Affiliation(s)
- Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - J Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
28
|
Orso R, Creutzberg KC, Lumertz FS, Kestering-Ferreira E, Stocchero BA, Perrone MK, Begni V, Grassi-Oliveira R, Riva MA, Viola TW. A systematic review and multilevel meta-analysis of the prenatal and early life stress effects on rodent microglia, astrocyte, and oligodendrocyte density and morphology. Neurosci Biobehav Rev 2023; 150:105202. [PMID: 37116770 DOI: 10.1016/j.neubiorev.2023.105202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023]
Abstract
Exposure to stress during early development may lead to altered neurobiological functions, thus increasing the risk for psychiatric illnesses later in life. One potential mechanism associated with those outcomes is the disruption of glial density and morphology, despite results from rodent studies have been conflicting. To address that we performed a systematic review and meta-analysis of rodent studies that investigated the effects of prenatal stress (PNS) and early life stress (ELS) on microglia, astrocyte, and oligodendrocyte density and morphology within the offspring. Our meta-analysis demonstrates that animals exposed to PNS or ELS showed significant increase in microglia density, as well as decreased oligodendrocyte density. Moreover, ELS exposure induced an increase in microglia soma size. However, we were unable to identify significant effects on astrocytes. Meta-regression indicated that experimental stress protocol, sex, age, and type of tissue analyzed are important covariates that impact those results. Importantly, PNS microglia showed higher estimates in young animals, while the ELS effects were stronger in adult animals. This set of data reinforces that alterations in glial cells could play a role in stress-induced dysfunctions throughout development.
Collapse
Affiliation(s)
- Rodrigo Orso
- Department of Pharmacological and Biomolecular Sciences, University of Milan - Via Balzaretti 9, 20133 - Milan (Italy).
| | - Kerstin Camile Creutzberg
- Department of Pharmacological and Biomolecular Sciences, University of Milan - Via Balzaretti 9, 20133 - Milan (Italy).
| | - Francisco Sindermann Lumertz
- School of Medicine, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul - Avenida Ipiranga 6681, Building 12A, 90619-900 - Porto Alegre (Brazil).
| | - Erika Kestering-Ferreira
- School of Medicine, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul - Avenida Ipiranga 6681, Building 12A, 90619-900 - Porto Alegre (Brazil).
| | - Bruna Alvim Stocchero
- School of Medicine, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul - Avenida Ipiranga 6681, Building 12A, 90619-900 - Porto Alegre (Brazil).
| | - Mariana Kude Perrone
- School of Medicine, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul - Avenida Ipiranga 6681, Building 12A, 90619-900 - Porto Alegre (Brazil).
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan - Via Balzaretti 9, 20133 - Milan (Italy).
| | - Rodrigo Grassi-Oliveira
- Translational Neuropsychiatry Unit, Aarhus University - Entrance A, Palle Juul-Jenses Blvd. 11, 6(th) floor, 8200 - Aarhus (Denmark).
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan - Via Balzaretti 9, 20133 - Milan (Italy); Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli - Via Pilastroni 4, 25125- Brescia (Italy).
| | - Thiago Wendt Viola
- School of Medicine, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul - Avenida Ipiranga 6681, Building 12A, 90619-900 - Porto Alegre (Brazil).
| |
Collapse
|
29
|
Fujikawa R, Tsuda M. The Functions and Phenotypes of Microglia in Alzheimer's Disease. Cells 2023; 12:cells12081207. [PMID: 37190116 DOI: 10.3390/cells12081207] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide, but therapeutic strategies to slow down AD pathology and symptoms have not yet been successful. While attention has been focused on neurodegeneration in AD pathogenesis, recent decades have provided evidence of the importance of microglia, and resident immune cells in the central nervous system. In addition, new technologies, including single-cell RNA sequencing, have revealed heterogeneous cell states of microglia in AD. In this review, we systematically summarize the microglial response to amyloid-β and tau tangles, and the risk factor genes expressed in microglia. Furthermore, we discuss the characteristics of protective microglia that appear during AD pathology and the relationship between AD and microglia-induced inflammation during chronic pain. Understanding the diverse roles of microglia will help identify new therapeutic strategies for AD.
Collapse
Affiliation(s)
- Risako Fujikawa
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Kyushu University Institute for Advanced Study, Fukuoka 819-0395, Japan
| |
Collapse
|
30
|
Pilipović I, Stojić-Vukanić Z, Prijić I, Jasnić N, Djordjević J, Leposavić G. β-Adrenoceptor Blockade Moderates Neuroinflammation in Male and Female EAE Rats and Abrogates Sexual Dimorphisms in the Major Neuroinflammatory Pathways by Being More Efficient in Males. Cell Mol Neurobiol 2023; 43:1237-1265. [PMID: 35798933 PMCID: PMC11414456 DOI: 10.1007/s10571-022-01246-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/18/2022] [Indexed: 11/03/2022]
Abstract
Our previous studies showed more severe experimental autoimmune encephalomyelitis (EAE) in male compared with female adult rats, and moderating effect of propranolol-induced β-adrenoceptor blockade on EAE in females, the effect associated with transcriptional stimulation of Nrf2/HO-1 axis in spinal cord microglia. This study examined putative sexual dimorphism in propranolol action on EAE severity. Propranolol treatment beginning from the onset of clinical EAE mitigated EAE severity in rats of both sexes, but to a greater extent in males exhibiting higher noradrenaline levels and myeloid cell β2-adrenoceptor expression in spinal cord. This correlated with more prominent stimulatory effects of propranolol not only on CX3CL1/CX3CR1/Nrf2/HO-1 cascade, but also on Stat3/Socs3 signaling axis in spinal cord microglia/myeloid cells (mirrored in the decreased Stat3 and the increased Socs3 expression) from male rats compared with their female counterparts. Propranolol diminished the frequency of activated cells among microglia, increased their phagocyting/endocyting capacity, and shifted cytokine secretory profile of microglia/blood-borne myeloid cells towards an anti-inflammatory/neuroprotective phenotype. Additionally, it downregulated the expression of chemokines (CCL2, CCL19/21) driving T-cell/monocyte trafficking into spinal cord. Consequently, in propranolol-treated rats fewer activated CD4+ T cells and IL-17+ T cells, including CD4+IL17+ cells coexpressing IFN-γ/GM-CSF, were recovered from spinal cord of propranolol-treated rats compared with sex-matched saline-injected controls. All the effects of propranolol were more prominent in males. The study as a whole disclosed that sexual dimorphism in multiple molecular mechanisms implicated in EAE development may be responsible for greater severity of EAE in male rats and sexually dimorphic action of substances affecting them. Propranolol moderated EAE severity more effectively in male rats, exhibiting greater spinal cord noradrenaline (NA) levels and myeloid cell β2-adrenoceptor (β2-AR) expression than females. Propranolol affected CX3CR1/Nrf2/HO-1 and Stat3/Socs3 signaling axes in myeloid cells, favored their anti-inflammatory/neuroprotective phenotype and, consequently, reduced Th cell reactivation and differentiation into highly pathogenic IL-17/IFN-γ/GM-CSF-producing cells.
Collapse
Affiliation(s)
- Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Ivana Prijić
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Nebojša Jasnić
- Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Jelena Djordjević
- Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221, Belgrade, Serbia.
| |
Collapse
|
31
|
Fülöp B, Hunyady Á, Bencze N, Kormos V, Szentes N, Dénes Á, Lénárt N, Borbély É, Helyes Z. IL-1 Mediates Chronic Stress-Induced Hyperalgesia Accompanied by Microglia and Astroglia Morphological Changes in Pain-Related Brain Regions in Mice. Int J Mol Sci 2023; 24:ijms24065479. [PMID: 36982563 PMCID: PMC10052634 DOI: 10.3390/ijms24065479] [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: 01/30/2023] [Revised: 03/01/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Chronic stress causes several pain conditions including fibromyalgia. Its pathophysiological mechanisms are unknown, and the therapy is unresolved. Since the involvement of interleukin-1 (IL-1) has been described in stress and inflammatory pain but no data are available regarding stress-induced pain, we studied its role in a chronic restraint stress (CRS) mouse model. Female and male C57Bl/6J wild-type (WT) and IL-1αβ-deficient (knock-out: IL-1 KO) mice were exposed to 6 h of immobilization/day for 4 weeks. Mechanonociception, cold tolerance, behavioral alterations, relative thymus/adrenal gland weights, microglia ionized calcium-binding adaptor molecule 1 (IBA1) and astrocyte glial fibrillary acidic protein (GFAP) integrated density, number and morphological transformation in pain-related brain regions were determined. CRS induced 15–20% mechanical hyperalgesia after 2 weeks in WT mice in both sexes, which was significantly reduced in female but not in male IL-1 KOs. Increased IBA1+ integrated density in the central nucleus of amygdala, primary somatosensory cortex hind limb representation part, hippocampus cornu ammonis area 3 (CA3) and periaqueductal gray matter (PAG) was present, accompanied by a cell number increase in IBA1+ microglia in stressed female WTs but not in IL-1 KOs. CRS induced morphological changes of GFAP+ astrocytes in WT but not in KO mice. Stress evoked cold hypersensitivity in the stressed animals. Anxiety and depression-like behaviors, thymus and adrenal gland weight changes were detectable in all groups after 2 but not 4 weeks of CRS due to adaptation. Thus, IL-1 mediates chronic stress-induced hyperalgesia in female mice, without other major behavioral alterations, suggesting the analgesic potentials of IL-1 in blocking drugs in stress-related pain syndromes.
Collapse
Affiliation(s)
- Barbara Fülöp
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Ágnes Hunyady
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- GSK Vaccines Institute for Global Health, I-53100 Siena, Italy
| | - Noémi Bencze
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Nikolett Szentes
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Ádám Dénes
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, H-1083 Budapest, Hungary
| | - Nikolett Lénárt
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, H-1083 Budapest, Hungary
| | - Éva Borbély
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- Correspondence:
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- Eotvos Lorand Research Network, Chronic Pain Research Group, University of Pécs, H-7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, H-1117 Budapest, Hungary
| |
Collapse
|
32
|
Bashir ST, Redden CR, Raj K, Arcanjo RB, Stasiak S, Li Q, Steelman AJ, Nowak RA. Endometriosis leads to central nervous system-wide glial activation in a mouse model of endometriosis. J Neuroinflammation 2023; 20:59. [PMID: 36879305 PMCID: PMC9987089 DOI: 10.1186/s12974-023-02713-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 01/31/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Chronic pelvic pain (CPP) is a common symptom of endometriosis. Women with endometriosis are also at a high risk of suffering from anxiety, depression, and other psychological disorders. Recent studies indicate that endometriosis can affect the central nervous system (CNS). Changes in the functional activity of neurons, functional magnetic resonance imaging signals, and gene expression have been reported in the brains of rat and mouse models of endometriosis. The majority of the studies thus far have focused on neuronal changes, whereas changes in the glial cells in different brain regions have not been studied. METHODS Endometriosis was induced in female mice (45-day-old; n = 6-11/timepoint) by syngeneic transfer of donor uterine tissue into the peritoneal cavity of recipient animals. Brains, spines, and endometriotic lesions were collected for analysis at 4, 8, 16, and 32 days post-induction. Sham surgery mice were used as controls (n = 6/timepoint). The pain was assessed using behavioral tests. Using immunohistochemistry for microglia marker ionized calcium-binding adapter molecule-1 (IBA1) and machine learning "Weka trainable segmentation" plugin in Fiji, we evaluated the morphological changes in microglia in different brain regions. Changes in glial fibrillary acidic protein (GFAP) for astrocytes, tumor necrosis factor (TNF), and interleukin-6 (IL6) were also evaluated. RESULTS We observed an increase in microglial soma size in the cortex, hippocampus, thalamus, and hypothalamus of mice with endometriosis compared to sham controls on days 8, 16, and 32. The percentage of IBA1 and GFAP-positive area was increased in the cortex, hippocampus, thalamus, and hypothalamus in mice with endometriosis compared to sham controls on day 16. The number of microglia and astrocytes did not differ between endometriosis and sham control groups. We observed increased TNF and IL6 expression when expression levels from all brain regions were combined. Mice with endometriosis displayed reduced burrowing behavior and hyperalgesia in the abdomen and hind-paw. CONCLUSION We believe this is the first report of central nervous system-wide glial activation in a mouse model of endometriosis. These results have significant implications for understanding chronic pain associated with endometriosis and other issues such as anxiety and depression in women with endometriosis.
Collapse
Affiliation(s)
- Shah Tauseef Bashir
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 W. Gregory Drive, Room 314 ASL, Urbana, IL, 61801, USA
| | - Catherine R Redden
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 W. Gregory Drive, Room 314 ASL, Urbana, IL, 61801, USA
| | - Kishori Raj
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 W. Gregory Drive, Room 314 ASL, Urbana, IL, 61801, USA
| | - Rachel B Arcanjo
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 W. Gregory Drive, Room 314 ASL, Urbana, IL, 61801, USA
| | - Sandra Stasiak
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 W. Gregory Drive, Room 314 ASL, Urbana, IL, 61801, USA
| | - Quanxi Li
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Andrew J Steelman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 W. Gregory Drive, Room 314 ASL, Urbana, IL, 61801, USA
| | - Romana A Nowak
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 W. Gregory Drive, Room 314 ASL, Urbana, IL, 61801, USA.
| |
Collapse
|
33
|
Pilipović I, Stojić-Vukanić Z, Leposavić G. Adrenoceptors as potential target for add-on immunomodulatory therapy in multiple sclerosis. Pharmacol Ther 2023; 243:108358. [PMID: 36804434 DOI: 10.1016/j.pharmthera.2023.108358] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
This review summarizes recent findings related to the role of the sympathetic nervous system (SNS) in pathogenesis of multiple sclerosis (MS) and its commonly used experimental model - experimental autoimmune encephalomyelitis (EAE). They indicate that noradrenaline, the key end-point mediator of the SNS, acting through β-adrenoceptor, has a contributory role in the early stages of MS/EAE development. This stage is characterized by the SNS hyperactivity (increased release of noradrenaline) reflecting the net effect of different factors, such as the disease-associated inflammation, stress, vitamin D hypovitaminosis, Epstein-Barr virus infection and dysbiosis. Thus, the administration of propranolol, a non-selective β-adrenoceptor blocker, readily crossing the blood-brain barrier, to experimental rats before the autoimmune challenge and in the early (preclinical/prodromal) phase of the disease mitigates EAE severity. This phenomenon has been ascribed to the alleviation of neuroinflammation (due to attenuation of primarily microglial activation/proinflammatory functions) and the diminution of the magnitude of the primary CD4+ T-cell autoimmune response (the effect associated with impaired autoantigen uptake by antigen presenting cells and their migration into draining lymph nodes). The former is partly related to breaking of the catecholamine-dependent self-amplifying microglial feed-forward loop and the positive feedback loop between microglia and the SNS, leading to down-regulation of the SNS hyperactivity and its enhancing influence on microglial activation/proinflammatory functions and the magnitude of autoimmune response. The effects of propranolol are shown to be more prominent in male EAE animals, the phenomenon important as males (like men) are likely to develop clinically more severe disease. Thus, these findings could serve as a firm scientific background for formulation of a new sex-specific immune-intervention strategy for the early phases of MS (characterized by the SNS hyperactivity) exploiting anti-(neuro)inflammatory and immunomodulatory properties of propranolol and other relatively cheap and safe adrenergic drugs with similar therapeutic profile.
Collapse
Affiliation(s)
- Ivan Pilipović
- Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- University of Belgrade-Faculty of Pharmacy, Department of Microbiology and Immunology, Belgrade, Serbia
| | - Gordana Leposavić
- University of Belgrade-Faculty of Pharmacy, Department of Pathobiology, Belgrade, Serbia.
| |
Collapse
|
34
|
Antoine D, Venigalla G, Truitt B, Roy S. Linking the gut microbiome to microglial activation in opioid use disorder. Front Neurosci 2022; 16:1050661. [PMID: 36590299 PMCID: PMC9800800 DOI: 10.3389/fnins.2022.1050661] [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: 09/22/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Substance use disorder (SUD) is a physical and psychological disorder globally prevalent today that has resulted in over 107,000 drug overdose deaths in 2021 in the United States alone. This manuscript reviews the potential relationship between opioid use disorder (OUD), a prevalent subset of SUD, and the microglia, the resident macrophages of the central nervous system (CNS), as they have been found to become significantly more activated during opioid exposure. The inflammatory response mediated by the microglia could contribute to the pathophysiology of SUDs, in particular OUD. Further understanding of the microglia and how they respond to not only signals in the CNS but also signals from other areas of the body, such as the gut microbiome, could explain how the microglia are involved in drug use. Several studies have shown extensive communication between the gut microbiome and the microglia, which may be an important factor in the initiation and development of OUD. Particularly, strategies seeking to manipulate and restore the gut microbiome have been shown to reduce microglial activation and attenuate inflammation. In this review, we discuss the evidence for a link between the microglia and OUD and how the gut microbiome might influence microglial activation to drive the disorder and its associated behaviors. Understanding this connection between microglia and the gut microbiome in the context of drug use may present additional therapeutic targets to treat the different stages of drug use.
Collapse
Affiliation(s)
- Danielle Antoine
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States,Department of Neuroscience, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Greeshma Venigalla
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Bridget Truitt
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States,Department of Neuroscience, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Sabita Roy
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States,*Correspondence: Sabita Roy,
| |
Collapse
|
35
|
Sleep and Neuroimmunomodulation for Maintenance of Optimum Brain Function: Role of Noradrenaline. Brain Sci 2022; 12:brainsci12121725. [PMID: 36552184 PMCID: PMC9776456 DOI: 10.3390/brainsci12121725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/03/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Immune function and sleep are two normal physiological processes to protect the living organism from falling sick. There is hardly any disease in which they remain unaffected, though the quantum of effect may differ. Therefore, we propose the existence of a strong correlation between sleep (quality or quantity) and immune response. This may be supported by the fact that sleep loss modulates many of the immunological molecules, which includes interferons; however, not much is known about their mechanism of action. Sleep is divided into rapid eye movement sleep (REMS) and non-REMS. For practical reasons, experimental studies have been conducted mostly by inducing loss of REMS. It has been shown that withdrawal of noradrenaline (NA) is a necessity for generation of REMS. Moreover, NA level increases in the brain upon REMS loss and the elevated NA is responsible for many of the sleep loss-associated symptoms. In this review, we describe how sleep (and its disturbance/loss) modulates the immune system by modulating the NA level in the brain or vice versa to maintain immune functions, physiological homeostasis, and normal healthy living. The increased levels of NA during REMS loss may cause neuroinflammation possibly by glial activation (as NA is a key modulator of microglia). Therefore, maintaining sleep hygiene plays a crucial role for a normal healthy living.
Collapse
|
36
|
Medina-Rodriguez EM, Rice KC, Jope RS, Beurel E. Comparison of inflammatory and behavioral responses to chronic stress in female and male mice. Brain Behav Immun 2022; 106:180-197. [PMID: 36058417 PMCID: PMC9561002 DOI: 10.1016/j.bbi.2022.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Major depressive disorder (MDD) is a debilitating disease with a high worldwide prevalence. Despite its greater prevalence in women, male animals are used in most preclinical studies of depression even though there are many sex differences in key components of depression, such as stress responses and immune system functions. In the present study, we found that chronic restraint stress-induced depressive-like behaviors are quite similar in male and female mice, with both sexes displaying increased immobility time in the tail suspension test and reduced social interactions, and both sexes exhibited deficits in working and spatial memories. However, in contrast to the similar depressive-like behaviors developed by male and female mice in response to stress, they displayed different patterns of pro-inflammatory cytokine increases in the periphery and the brain, different changes in microglia, and different changes in the expression of Toll-like receptor 4 in response to stress. Treatment with (+)-naloxone, a Toll-like receptor 4 antagonist that previously demonstrated anti-depressant-like effects in male mice, was more efficacious in male than female mice in reducing the deleterious effects of stress, and its effects were not microbiome-mediated. Altogether, these results suggest differential mechanisms to consider in potential sex-specific treatments of depression.
Collapse
Affiliation(s)
- Eva M Medina-Rodriguez
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33125, United States
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Richard S Jope
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33125, United States; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States.
| |
Collapse
|
37
|
Abe N, Tarumi M, Fujieda Y, Takahashi N, Karino K, Uchida M, Kono M, Tanaka Y, Hasebe R, Kato M, Amengual O, Arinuma Y, Oku K, Sato W, Tha KK, Yamasaki M, Watanabe M, Atsumi T, Murakami M. Pathogenic neuropsychiatric effect of stress-induced microglial interleukin 12/23 axis in systemic lupus erythematosus. Ann Rheum Dis 2022; 81:1564-1575. [PMID: 35817472 DOI: 10.1136/ard-2022-222566] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/14/2022] [Indexed: 12/18/2022]
Abstract
OBJECTIVES The central nervous system disorder in systemic lupus erythematosus (SLE), called neuropsychiatric lupus (NPSLE), is one of the most severe phenotypes with various clinical symptoms, including mood disorder, psychosis and delirium as diffuse neuropsychological manifestations (dNPSLE). Although stress is one of the aggravating factors for neuropsychiatric symptoms, its role in the pathogenesis of dNPSLE remains to be elucidated. We aimed to investigate stress effects on the neuropsychiatric pathophysiology in SLE using lupus-prone mice and patients' data. METHODS Sleep disturbance stress (SDS) for 2 weeks was placed on 6-8-week-old female MRL/lpr and control mice. Behavioural phenotyping, histopathological analyses and gene and protein expression analyses were performed to assess SDS-induced neuroimmunological alterations. We also evaluated cytokines of the cerebrospinal fluid and brain regional volumes in patients with dNPSLE and patients with non-dNPSLE. RESULTS SDS-subjected MRL/lpr mice exhibited less anxiety-like behaviour, whereas stressed control mice showed increased anxiety. Furthermore, stress strongly activated the medial prefrontal cortex (mPFC) in SDS-subjected MRL/lpr. A transcriptome analysis of the PFC revealed the upregulation of microglial activation-related genes, including Il12b. We confirmed that stress-induced microglial activation and the upregulation of interleukin (IL) 12/23p40 proteins and increased dendritic spines in the mPFC of stressed MRL/lpr mice. IL-12/23p40 neutralisation and tyrosine kinase 2 inhibition mitigated the stress-induced neuropsychiatric phenotypes of MRL/lpr mice. We also found a higher level of cerebrospinal fluid IL-12/23p40 and more atrophy in the mPFC of patients with dNPSLE than those with non-dNPSLE. CONCLUSIONS The microglial IL-12/23 axis in the mPFC might be associated with the pathogenesis and a promising therapeutic target for dNPSLE.
Collapse
Affiliation(s)
- Nobuya Abe
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masato Tarumi
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuichiro Fujieda
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Nobuhiko Takahashi
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kohei Karino
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mona Uchida
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Michihito Kono
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Tanaka
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Inage, Japan
| | - Rie Hasebe
- Center for Infectious Cancers, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
| | - Masaru Kato
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Olga Amengual
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshiyuki Arinuma
- Department of Rheumatology and Infectious Diseases, School of Medicine, Kitasato University, Sagamihara, Japan
| | - Kenji Oku
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Rheumatology and Infectious Diseases, School of Medicine, Kitasato University, Sagamihara, Japan
| | - Wakiro Sato
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Khin Khin Tha
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Miwako Yamasaki
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Inage, Japan
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
| |
Collapse
|
38
|
Miyata S, Ishino Y, Shimizu S, Tohyama M. Involvement of inflammatory responses in the brain to the onset of major depressive disorder due to stress exposure. Front Aging Neurosci 2022; 14:934346. [PMID: 35936767 PMCID: PMC9354609 DOI: 10.3389/fnagi.2022.934346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
Major depressive disorder (MDD) is a multifactorial disease affected by several environmental factors. Although several potential onset hypotheses have been identified, the molecular mechanisms underlying the pathogenesis of this disorder remain unclear. Several recent studies have suggested that among many environmental factors, inflammation and immune abnormalities in the brain or the peripheral tissues are associated with the onset of MDDs. Furthermore, several stress-related hypotheses have been proposed to explain the onset of MDDs. Thus, inflammation or immune abnormalities can be considered stress responses that occur within the brain or other tissues and are regarded as one of the mechanisms underlying the stress hypothesis of MDDs. Therefore, we introduce several current advances in inflammation studies in the brain that might be related to the pathophysiology of MDD due to stress exposure in this review.
Collapse
Affiliation(s)
- Shingo Miyata
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kindai University, Osaka, Japan
- *Correspondence: Shingo Miyata
| | - Yugo Ishino
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kindai University, Osaka, Japan
| | - Shoko Shimizu
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kindai University, Osaka, Japan
| | - Masaya Tohyama
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kindai University, Osaka, Japan
- Osaka Prefectural Hospital Organization, Osaka, Japan
| |
Collapse
|
39
|
Carnac T. Schizophrenia Hypothesis: Autonomic Nervous System Dysregulation of Fetal and Adult Immune Tolerance. Front Syst Neurosci 2022; 16:844383. [PMID: 35844244 PMCID: PMC9283579 DOI: 10.3389/fnsys.2022.844383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
The autonomic nervous system can control immune cell activation via both sympathetic adrenergic and parasympathetic cholinergic nerve release of norepinephrine and acetylcholine. The hypothesis put forward in this paper suggests that autonomic nervous system dysfunction leads to dysregulation of immune tolerance mechanisms in brain-resident and peripheral immune cells leading to excessive production of pro-inflammatory cytokines such as Tumor Necrosis Factor alpha (TNF-α). Inactivation of Glycogen Synthase Kinase-3β (GSK3β) is a process that takes place in macrophages and microglia when a toll-like receptor 4 (TLR4) ligand binds to the TLR4 receptor. When Damage-Associated Molecular Patterns (DAMPS) and Pathogen-Associated Molecular Patterns (PAMPS) bind to TLR4s, the phosphatidylinositol-3-kinase (PI3K)-protein kinase B (Akt) pathway should be activated, leading to inactivation of GSK3β. This switches the macrophage from producing pro-inflammatory cytokines to anti-inflammatory cytokines. Acetylcholine activation of the α7 subunit of the nicotinic acetylcholine receptor (α7 nAChR) on the cell surface of immune cells leads to PI3K/Akt pathway activation and can control immune cell polarization. Dysregulation of this pathway due to dysfunction of the prenatal autonomic nervous system could lead to impaired fetal immune tolerance mechanisms and a greater vulnerability to Maternal Immune Activation (MIA) resulting in neurodevelopmental abnormalities. It could also lead to the adult schizophrenia patient’s immune system being more vulnerable to chronic stress-induced DAMP release. If a schizophrenia patient experiences chronic stress, an increased production of pro-inflammatory cytokines such as TNF-α could cause significant damage. TNF-α could increase the permeability of the intestinal and blood brain barrier, resulting in lipopolysaccharide (LPS) and TNF-α translocation to the brain and consequent increases in glutamate release. MIA has been found to reduce Glutamic Acid Decarboxylase mRNA expression, resulting in reduced Gamma-aminobutyric acid (GABA) synthesis, which combined with an increase of glutamate release could result in an imbalance of glutamate and GABA neurotransmitters. Schizophrenia could be a “two-hit” illness comprised of a genetic “hit” of autonomic nervous system dysfunction and an environmental hit of MIA. This combination of factors could lead to neurotransmitter imbalance and the development of psychotic symptoms.
Collapse
|
40
|
Bekała A, Płotek W, Siwicka-Gieroba D, Sołek-Pastuszka J, Bohatyrewicz R, Biernawska J, Kotfis K, Bielacz M, Jaroszyński A, Dabrowski W. Melatonin and the Brain-Heart Crosstalk in Neurocritically Ill Patients-From Molecular Action to Clinical Practice. Int J Mol Sci 2022; 23:7094. [PMID: 35806098 PMCID: PMC9267006 DOI: 10.3390/ijms23137094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 01/27/2023] Open
Abstract
Brain injury, especially traumatic brain injury (TBI), may induce severe dysfunction of extracerebral organs. Cardiac dysfunction associated with TBI is common and well known as the brain-heart crosstalk, which broadly refers to different cardiac disorders such as cardiac arrhythmias, ischemia, hemodynamic insufficiency, and sudden cardiac death, which corresponds to acute disorders of brain function. TBI-related cardiac dysfunction can both worsen the brain damage and increase the risk of death. TBI-related cardiac disorders have been mainly treated symptomatically. However, the analysis of pathomechanisms of TBI-related cardiac dysfunction has highlighted an important role of melatonin in the prevention and treatment of such disorders. Melatonin is a neurohormone released by the pineal gland. It plays a crucial role in the coordination of the circadian rhythm. Additionally, melatonin possesses strong anti-inflammatory, antioxidative, and antiapoptotic properties and can modulate sympathetic and parasympathetic activities. Melatonin has a protective effect not only on the brain, by attenuating its injury, but on extracranial organs, including the heart. The aim of this study was to analyze the molecular activity of melatonin in terms of TBI-related cardiac disorders. Our article describes the benefits resulting from using melatonin as an adjuvant in protection and treatment of brain injury-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Artur Bekała
- Department of Anesthesiology in Obstetrics and Gynecology, Gynecology and Obstetrics Clinical Hospital, Poznań University of Medical Sciences, 60-535 Poznan, Poland;
| | - Włodzimierz Płotek
- Department of Anesthesiology in Obstetrics and Gynecology, Gynecology and Obstetrics Clinical Hospital, Poznań University of Medical Sciences, 60-535 Poznan, Poland;
| | - Dorota Siwicka-Gieroba
- Department of Anesthesiology and Intensive Care, Medical University of Lublin, Jaczewskiego Street 8, 20-954 Lublin, Poland; (D.S.-G.); (M.B.)
| | - Joanna Sołek-Pastuszka
- Department of Anaesthesiology and Intensive Therapy, Pomeranian Medical University, 71-242 Szczecin, Poland; (J.S.-P.); (R.B.); (J.B.)
| | - Romuald Bohatyrewicz
- Department of Anaesthesiology and Intensive Therapy, Pomeranian Medical University, 71-242 Szczecin, Poland; (J.S.-P.); (R.B.); (J.B.)
| | - Jowita Biernawska
- Department of Anaesthesiology and Intensive Therapy, Pomeranian Medical University, 71-242 Szczecin, Poland; (J.S.-P.); (R.B.); (J.B.)
| | - Katarzyna Kotfis
- Department of Anaesthesiology, Intensive therapy and Acute Intoxications, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
| | - Magdalena Bielacz
- Department of Anesthesiology and Intensive Care, Medical University of Lublin, Jaczewskiego Street 8, 20-954 Lublin, Poland; (D.S.-G.); (M.B.)
| | - Andrzej Jaroszyński
- Department of Nephrology, Institute of Medical Science, Jan Kochanowski University of Kielce, 25-735 Kielce, Poland;
| | - Wojciech Dabrowski
- Department of Anesthesiology and Intensive Care, Medical University of Lublin, Jaczewskiego Street 8, 20-954 Lublin, Poland; (D.S.-G.); (M.B.)
| |
Collapse
|
41
|
Liu G, Cheng J, Zhang T, Shao Y, Chen X, Han L, Zhou R, Wu B. Inhibition of Microbiota-dependent Trimethylamine N-Oxide Production Ameliorates High Salt Diet-Induced Sympathetic Excitation and Hypertension in Rats by Attenuating Central Neuroinflammation and Oxidative Stress. Front Pharmacol 2022; 13:856914. [PMID: 35359866 PMCID: PMC8961329 DOI: 10.3389/fphar.2022.856914] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/14/2022] [Indexed: 12/18/2022] Open
Abstract
Excessive dietary salt intake induces neuroinflammation and oxidative stress in the brain, which lead to sympathetic excitation, contributing to hypertension. However, the underlying mechanisms remain elusive. Accumulating evidence reveals that trimethylamine-N-oxide (TMAO), a gut microbiota-derived metabolite, is implicated in the pathogenesis of multiple cardiovascular diseases. The present study sought to determine whether central TMAO is elevated and associated with neuroinflammation and oxidative stress in the brain after long-term high salt (HS) diet intake and, if so, whether inhibition of TMAO generation ameliorates HS-induced sympathetic excitation and hypertension. Sprague-Dawley rats were fed either a HS diet or a normal salt (NS) diet and simultaneously treated with vehicle (VEH) or 1.0% 3,3-Dimethyl-1-butanol (DMB, an inhibitor of trimethylamine formation) for 8 weeks. HS + VEH rats, compared with NS + VEH rats, had elevated TMAO in plasma and cerebrospinal fluid (CSF), increased blood pressure (BP), and increased sympathetic drive as indicated by the BP response to ganglionic blockade and plasma norepinephrine levels. HS-induced these changes were attenuated by DMB, which significantly reduced TMAO in plasma and CSF. Neuroinflammation as assessed by proinflammatory cytokine expression and NF-κB activity and microglial activity, and oxidative stress as measured by NAD(P)H oxidase subunit expression and NAD(P)H activity and reactive oxygen species (ROS) production in the hypothalamic paraventricular nucleus (PVN) were increased in HS + VEH rats but were decreased by DMB. DMB had no effects on above measured parameters in NS rats. The results suggest that long-term HS diet intake causes elevation in TMAO in the circulation and brain, which is associated with increased neuroinflammation and oxidative stress in the PVN, an important cardiovascular regulatory center. Inhibition of TMAO generation ameliorates HS-induced sympathetic excitation and hypertension by reducing neuroinflammation and oxidative stress in the PVN.
Collapse
Affiliation(s)
- Gang Liu
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Jiayin Cheng
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Tianhao Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yingxin Shao
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| | - Xiangxu Chen
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| | - Lihong Han
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| | - Ru Zhou
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| | - Bin Wu
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
42
|
Stress induced microglial activation contributes to depression. Pharmacol Res 2022; 179:106145. [DOI: 10.1016/j.phrs.2022.106145] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023]
|
43
|
Wang J, Zhou Y, Li K, Li X, Guo M, Peng M. A Noradrenergic Lesion Attenuates Surgery-Induced Cognitive Impairment in Rats by Suppressing Neuroinflammation. Front Mol Neurosci 2021; 14:752838. [PMID: 34916906 PMCID: PMC8671038 DOI: 10.3389/fnmol.2021.752838] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/25/2021] [Indexed: 11/22/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a common postoperative neurocognitive complication in elderly patients. However, the specific pathogenesis is unknown, and it has been demonstrated that neuroinflammation plays a key role in POCD. Recently, increasing evidence has proven that the locus coeruleus noradrenergic (LCNE) system participates in regulating neuroinflammation in some neurodegenerative disorders. We hypothesize that LCNE plays an important role in the neuroinflammation of POCD. In this study, 400 μg of N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) was injected intracerebroventricularly into each rat 7 days before anesthesia/surgery to deplete the locus coeruleus (LC) noradrenaline (NE). We applied a simple laparotomy and brief upper mesenteric artery clamping surgery as the rat POCD model. The open field test, novel objection and novel location (NL) recognition, and Morris water maze (MWM) were performed to assess postoperative cognition. High-performance liquid chromatography (HPLC) was used to measure the level of NE in plasma and brain tissues, and immunofluorescence staining was applied to evaluate the activation of microglia and astrocytes. We also used enzyme-linked immune-sorbent assay (ELISA) to assess the levels of inflammatory cytokines and brain-derived neurotrophic factor (BDNF). Pretreatment with DSP-4 decreased the levels of systemic and central NE, increased the level of interleukin-6 (IL-6) in the plasma at 6 h after the surgery, decreased the concentration of IL-6 in the prefrontal cortex and hippocampus, and decreased the level of interleukin-1β (IL-1β) in the plasma, prefrontal cortex, and hippocampus at 1 week postoperatively. In addition, DSP-4 treatment attenuated hippocampal-dependent learning and memory impairment in rats with POCD, with a downregulation of the activation of microglia and astrocytes in the prefrontal cortex and hippocampus. In conclusion, these findings provide evidence of the effects of LCNE in modulating neuroinflammation in rats with POCD and provide a new perspective in the prevention and treatment of POCD.
Collapse
Affiliation(s)
- Jiayu Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Zhou
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ke Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaofeng Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Meimei Guo
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mian Peng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
44
|
Zou H, Li J, Zhou J, Yi X, Cao S. Effects of norepinephrine on microglial neuroinflammation and neuropathic pain. IBRAIN 2021; 7:309-317. [PMID: 37786561 PMCID: PMC10528971 DOI: 10.1002/ibra.12001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 10/04/2023]
Abstract
Norepinephrine (NE) is an important neurotransmitter in the central nervous system. NE is released from locus coeruleus neurons and is involved in a variety of physiological and pathological processes. Neuroinflammation is a common manifestation of many kinds of neurological diseases. The activation of microglia directly affects the status of neuroinflammation. Several kinds of adrenergic receptors, which anchor on microglia and can be regulated by NE, affect the activation of microglia and neuroinflammation. NE influences chronic pain, anxiety, and depression by regulating the activation of microglia.
Collapse
Affiliation(s)
- He‐Lin Zou
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiGuizhouChina
| | - Juan Li
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiGuizhouChina
| | - Jun‐Li Zhou
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiGuizhouChina
| | - Xi Yi
- Department of Pain MedicineAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Song Cao
- Department of Pain MedicineAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
45
|
Abstract
BACKGROUND Propranolol, a nonselective β-adrenergic receptor antagonist, is approved by the U.S. Food and Drug Administration to treat problematic infantile hemangiomas, but a subset of patients experience treatment complications. Parents wary of long-term use and side effects consult plastic surgeons on surgical options or as a second opinion. Understanding the mechanism(s) of action of propranolol will allow plastic surgeons to better inform parents. METHODS A systemic literature search was performed to query published translational and basic science studies on propranolol effects on infantile hemangiomas and cells derived from these lesions. RESULTS In experimental studies, propranolol was antiproliferative and cytotoxic against hemangioma endothelial and stem cells and affected infantile hemangioma perivascular cell contractility. Propranolol inhibited migration, network formation, vascular endothelial growth factor A production, and vascular endothelial growth factor receptor 2 activation and down-regulated PI3K/AKT and mitogen-activated protein kinase signaling in hemangioma endothelial cells, but it increased ERK1/2 activity in hemangioma stem cells. At effective clinical doses, measured propranolol plasma concentration is 100 times higher than necessary for complete β-adrenergic receptor blockade, yet was 10 to 100 times less than required to induce hemangioma stem cell death. CONCLUSIONS Propranolol targets multiple cell types in infantile hemangiomas by means of β-adrenergic receptor-dependent and -independent mechanisms. Plasma concentration played a significant role. At clinically relevant doses, incomplete infantile hemangioma suppression may explain the rebound phenomenon and worsening ulceration, and propranolol off target effects may lead to commonly reported adverse effects, such as sleep and gastrointestinal disturbances. Propranolol limitations and complications underscore the importance of surgical treatment options in cases of rebound and severe adverse effects. Surgical intervention remains an important treatment choice when parents are hesitant to use propranolol.
Collapse
|
46
|
Mather M. Noradrenaline in the aging brain: Promoting cognitive reserve or accelerating Alzheimer's disease? Semin Cell Dev Biol 2021; 116:108-124. [PMID: 34099360 PMCID: PMC8292227 DOI: 10.1016/j.semcdb.2021.05.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022]
Abstract
Many believe that engaging in novel and mentally challenging activities promotes brain health and prevents Alzheimer's disease in later life. However, mental stimulation may also have risks as well as benefits. As neurons release neurotransmitters, they often also release amyloid peptides and tau proteins into the extracellular space. These by-products of neural activity can aggregate into the tau tangle and amyloid plaque signatures of Alzheimer's disease. Over time, more active brain regions accumulate more pathology. Thus, increasing brain activity can have a cost. But the neuromodulator noradrenaline, released during novel and mentally stimulating events, may have some protective effects-as well as some negative effects. Via its inhibitory and excitatory effects on neurons and microglia, noradrenaline sometimes prevents and sometimes accelerates the production and accumulation of amyloid-β and tau in various brain regions. Both α2A- and β-adrenergic receptors influence amyloid-β production and tau hyperphosphorylation. Adrenergic activity also influences clearance of amyloid-β and tau. Furthermore, some findings suggest that Alzheimer's disease increases noradrenergic activity, at least in its early phases. Because older brains clear the by-products of synaptic activity less effectively, increased synaptic activity in the older brain risks accelerating the accumulation of Alzheimer's pathology more than it does in the younger brain.
Collapse
Affiliation(s)
- Mara Mather
- Leonard Davis School of Gerontology, Department of Psychology, & Department of Biomedical Engineering, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089, United States.
| |
Collapse
|
47
|
Microglia react to partner loss in a sex- and brain site-specific manner in prairie voles. Brain Behav Immun 2021; 96:168-186. [PMID: 34058309 PMCID: PMC8319132 DOI: 10.1016/j.bbi.2021.05.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/21/2022] Open
Abstract
Positive social relationships are paramount for the survival of mammals and beneficial for mental and physical health, buffer against stressors, and even promote appropriate immune system functioning. By contrast, impaired social relationships, social isolation, or the loss of a bonded partner lead to aggravated physical and mental health. For example, in humans partner loss is detrimental for the functioning of the immune system and heightens the susceptibility for the development of post-traumatic stress disorders, anxiety disorders, and major depressive disorders. To understand potential underlying mechanisms, the monogamous prairie vole can provide important insights. In the present study, we separated pair bonded male and female prairie voles after five days of co-housing, subjected them to the forced swim test on the fourth day following separation, and studied their microglia morphology and activation in specific brain regions. Partner loss increased passive stress-coping in male, but not female, prairie voles. Moreover, partner loss was associated with microglial priming within the parvocellular region of the paraventricular nucleus of the hypothalamus (PVN) in male prairie voles, whereas in female prairie voles the morphological activation within the whole PVN and the prelimbic cortex (PrL) was decreased, marked by a shift towards ramified microglial morphology. Expression of the immediate early protein c-Fos following partner loss was changed within the PrL of male, but not female, prairie voles. However, the loss of a partner did not affect the investigated aspects of the peripheral immune response. These data suggest a potential sex-dependent mechanism for the regulation of microglial activity following the loss of a partner, which might contribute to the observed differences in passive stress-coping. This study furthers our understanding of the effects of partner loss and its short-term impact on the CNS as well as the CNS immune system and the peripheral innate immune system in both male and female prairie voles.
Collapse
|
48
|
Warfield AE, Prather JF, Todd WD. Systems and Circuits Linking Chronic Pain and Circadian Rhythms. Front Neurosci 2021; 15:705173. [PMID: 34276301 PMCID: PMC8284721 DOI: 10.3389/fnins.2021.705173] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/10/2021] [Indexed: 12/15/2022] Open
Abstract
Research over the last 20 years regarding the link between circadian rhythms and chronic pain pathology has suggested interconnected mechanisms that are not fully understood. Strong evidence for a bidirectional relationship between circadian function and pain has been revealed through inflammatory and immune studies as well as neuropathic ones. However, one limitation of many of these studies is a focus on only a few molecules or cell types, often within only one region of the brain or spinal cord, rather than systems-level interactions. To address this, our review will examine the circadian system as a whole, from the intracellular genetic machinery that controls its timing mechanism to its input and output circuits, and how chronic pain, whether inflammatory or neuropathic, may mediate or be driven by changes in these processes. We will investigate how rhythms of circadian clock gene expression and behavior, immune cells, cytokines, chemokines, intracellular signaling, and glial cells affect and are affected by chronic pain in animal models and human pathologies. We will also discuss key areas in both circadian rhythms and chronic pain that are sexually dimorphic. Understanding the overlapping mechanisms and complex interplay between pain and circadian mediators, the various nuclei they affect, and how they differ between sexes, will be crucial to move forward in developing treatments for chronic pain and for determining how and when they will achieve their maximum efficacy.
Collapse
Affiliation(s)
| | | | - William D. Todd
- Program in Neuroscience, Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| |
Collapse
|
49
|
Doppler CEJ, Kinnerup MB, Brune C, Farrher E, Betts M, Fedorova TD, Schaldemose JL, Knudsen K, Ismail R, Seger AD, Hansen AK, Stær K, Fink GR, Brooks DJ, Nahimi A, Borghammer P, Sommerauer M. Regional locus coeruleus degeneration is uncoupled from noradrenergic terminal loss in Parkinson's disease. Brain 2021; 144:2732-2744. [PMID: 34196700 DOI: 10.1093/brain/awab236] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/18/2021] [Accepted: 06/06/2021] [Indexed: 11/13/2022] Open
Abstract
Previous studies have reported substantial involvement of the noradrenergic system in Parkinson's disease. Neuromelanin-sensitive MRI sequences and PET tracers have become available to visualize the cell bodies in the locus coeruleus and the density of noradrenergic terminal transporters. Combining these methods, we investigated the relationship of neurodegeneration in these distinct compartments in Parkinson's disease. We examined 93 subjects (40 healthy controls and 53 Parkinson's disease patients) with neuromelanin-sensitive turbo spin-echo MRI and calculated locus coeruleus-to-pons signal contrasts. Voxels with the highest intensities were extracted from published locus coeruleus coordinates transformed to individual MRI. To also investigate a potential spatial pattern of locus coeruleus degeneration, we extracted the highest signal intensities from the rostral, middle, and caudal third of the locus coeruleus. Additionally, a study-specific probabilistic map of the locus coeruleus was created and used to extract mean MRI contrast from the entire locus coeruleus and each rostro-caudal subdivision. Locus coeruleus volumes were measured using manual segmentations. A subset of 73 subjects had 11C-MeNER PET to determine noradrenaline transporter density, and distribution volume ratios of noradrenaline transporter-rich regions were computed. Parkinson's disease patients showed reduced locus coeruleus MRI contrast independently of the selected method (voxel approaches: p < 0.0001, p < 0.001; probabilistic map: p < 0.05), specifically on the clinically-defined most affected side (p < 0.05), and reduced locus coeruleus volume (p < 0.0001). Reduced MRI contrast was confined to the middle and caudal locus coeruleus (voxel approach-rostral: p = 0.48, middle: p < 0.0001, and caudal: p < 0.05; probabilistic map-rostral: p = 0.90, middle: p < 0.01, and caudal: p < 0.05). The noradrenaline transporter density was lower in Parkinson's disease patients in all examined regions (group effect p < 0.0001). No significant correlation was observed between locus coeruleus MRI contrast and noradrenaline transporter density. In contrast, the individual ratios of noradrenaline transporter density and locus coeruleus MRI contrast were lower in Parkinson's disease patients in all examined regions (group effect p < 0.001). Our multimodal imaging approach revealed pronounced noradrenergic terminal loss relative to cellular locus coeruleus degeneration in Parkinson's disease; the latter followed a distinct spatial pattern with the middle-caudal portion being more affected than the rostral part. The data shed first light on the interaction between the axonal and cell body compartments and their differential susceptibility to neurodegeneration in Parkinson's disease, which may eventually direct research toward potential novel treatment approaches.
Collapse
Affiliation(s)
- Christopher E J Doppler
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, D-52425 Jülich, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, D-50937 Köln, Germany
| | - Martin B Kinnerup
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Corinna Brune
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, D-50937 Köln, Germany
| | - Ezequiel Farrher
- Institute of Neuroscience and Medicine (INM-4), Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Matthew Betts
- German Center for Neurodegenerative Diseases (DZNE), D-39120 Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany.,Center for Behavioral Brain Sciences, University of Magdeburg, D-39120 Magdeburg, Germany
| | - Tatyana D Fedorova
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Jeppe L Schaldemose
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Karoline Knudsen
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Rola Ismail
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Aline D Seger
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, D-52425 Jülich, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, D-50937 Köln, Germany
| | - Allan K Hansen
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Kristian Stær
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Gereon R Fink
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, D-52425 Jülich, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, D-50937 Köln, Germany
| | - David J Brooks
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark.,Division of Brain Sciences, Imperial College London, London SW7 2AZ, UK.,Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle upon Tyne NE1 7RU, UK
| | - Adjmal Nahimi
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Per Borghammer
- Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Michael Sommerauer
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, D-52425 Jülich, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, D-50937 Köln, Germany.,Department of Nuclear Medicine and PET, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| |
Collapse
|
50
|
Ohgomori T, Iinuma K, Yamada J, Jinno S. A unique subtype of ramified microglia associated with synapses in the rat hippocampus. Eur J Neurosci 2021; 54:4740-4754. [PMID: 34110047 DOI: 10.1111/ejn.15330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/13/2022]
Abstract
To date, a number of studies have reported the heterogeneity of activated microglia. However, there is increasing evidence suggests that ramified, so-called resting, microglia may also be heterogeneous, and they may play diverse roles in normal brain homeostasis. Here, we found that both 5D4 keratan sulfate epitope-positive (5D4+ ) and 5D4-negative (5D4- ) microglia coexisted in the hippocampus of normal rats, while all microglia were negative for the 5D4 epitope in the hippocampus of normal mice. We thus aimed to determine the potential heterogeneity of microglia related to the 5D4 epitope in the normal rat hippocampus. The optical disector analysis showed that the densities of 5D4+ microglia were higher in the stratum oriens of the CA3 region than in other layers and regions. Although both 5D4+ and 5D4- microglia exhibited a ramified morphology, the three-dimensional reconstruction analysis showed that the node numbers, end numbers, and complexity of processes were higher in 5D4+ than in 5D4- microglia. The linear discriminant analysis showed that 5D4+ and 5D4- microglia can be classified into distinct morphometric subtypes. The ratios of contact between synaptic boutons and microglial processes were higher in 5D4+ than in 5D4- microglia. The gene expressions of pro-inflammatory cytokine interleukin-1β and purinergic receptor P2Y12 (P2Y12 R) were higher in 5D4+ than in 5D4- microglia. Together, these results indicate that at least two different subtypes of ramified microglia coexist in the normal rat hippocampus and also suggest that 5D4+ microglia may represent a unique subtype associated with synapses.
Collapse
Affiliation(s)
- Tomohiro Ohgomori
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, Kaizuka, Japan
| | - Kyoko Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|