1
|
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
|
2
|
Laurencin C, Lancelot S, Brosse S, Mérida I, Redouté J, Greusard E, Lamberet L, Liotier V, Le Bars D, Costes N, Thobois S, Boulinguez P, Ballanger B. Noradrenergic alterations in Parkinson's disease: a combined 11C-yohimbine PET/neuromelanin MRI study. Brain 2024; 147:1377-1388. [PMID: 37787503 PMCID: PMC10994534 DOI: 10.1093/brain/awad338] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023] Open
Abstract
Degeneration of the noradrenergic system is now considered a pathological hallmark of Parkinson's disease, but little is known about its consequences in terms of parkinsonian manifestations. Here, we evaluated two aspects of the noradrenergic system using multimodal in vivo imaging in patients with Parkinson's disease and healthy controls: the pigmented cell bodies of the locus coeruleus with neuromelanin sensitive MRI; and the density of α2-adrenergic receptors (ARs) with PET using 11C-yohimbine. Thirty patients with Parkinson's disease and 30 age- and sex-matched healthy control subjects were included. The characteristics of the patients' symptoms were assessed using the Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS). Patients showed reduced neuromelanin signal intensity in the locus coeruleus compared with controls and diminished 11C-yohimbine binding in widespread cortical regions, including the motor cortex, as well as in the insula, thalamus and putamen. Clinically, locus coeruleus neuronal loss was correlated with motor (bradykinesia, motor fluctuations, tremor) and non-motor (fatigue, apathy, constipation) symptoms. A reduction of α2-AR availability in the thalamus was associated with tremor, while a reduction in the putamen, the insula and the superior temporal gyrus was associated with anxiety. These results highlight a multifaceted alteration of the noradrenergic system in Parkinson's disease since locus coeruleus and α2-AR degeneration were found to be partly uncoupled. These findings raise important issues about noradrenergic dysfunction that may encourage the search for new drugs targeting this system, including α2-ARs, for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Chloé Laurencin
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
- Department of Neurology C, Expert Parkinson Centre, Hospices Civils de Lyon, Pierre Wertheimer Neurological Hospital, NS-Park/F-CRIN, 69500 Bron, France
| | - Sophie Lancelot
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Sarah Brosse
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
| | - Inés Mérida
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Jérôme Redouté
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Elise Greusard
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Ludovic Lamberet
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | | | - Didier Le Bars
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Nicolas Costes
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Stéphane Thobois
- Department of Neurology C, Expert Parkinson Centre, Hospices Civils de Lyon, Pierre Wertheimer Neurological Hospital, NS-Park/F-CRIN, 69500 Bron, France
- Institut des Sciences Cognitives Marc Jeannerod, UMR 5229, CNRS, 69500 Bron, France
| | - Philippe Boulinguez
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
| | - Bénédicte Ballanger
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
| |
Collapse
|
3
|
Yang D, Xu J, Xu K, Xu P. Skeletal interoception in osteoarthritis. Bone Res 2024; 12:22. [PMID: 38561376 PMCID: PMC10985098 DOI: 10.1038/s41413-024-00328-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/02/2024] [Accepted: 03/16/2024] [Indexed: 04/04/2024] Open
Abstract
The interoception maintains proper physiological conditions and metabolic homeostasis by releasing regulatory signals after perceving changes in the internal state of the organism. Among its various forms, skeletal interoception specifically regulates the metabolic homeostasis of bones. Osteoarthritis (OA) is a complex joint disorder involving cartilage, subchondral bone, and synovium. The subchondral bone undergoes continuous remodeling to adapt to dynamic joint loads. Recent findings highlight that skeletal interoception mediated by aberrant mechanical loads contributes to pathological remodeling of the subchondral bone, resulting in subchondral bone sclerosis in OA. The skeletal interoception is also a potential mechanism for chronic synovial inflammation in OA. In this review, we offer a general overview of interoception, specifically skeletal interoception, subchondral bone microenviroment and the aberrant subchondral remedeling. We also discuss the role of skeletal interoception in abnormal subchondral bone remodeling and synovial inflammation in OA, as well as the potential prospects and challenges in exploring novel OA therapies that target skeletal interoception.
Collapse
Affiliation(s)
- Dinglong Yang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Jiawen Xu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ke Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Peng Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| |
Collapse
|
4
|
Dmytriv TR, Tsiumpala SA, Semchyshyn HM, Storey KB, Lushchak VI. Mitochondrial dysfunction as a possible trigger of neuroinflammation at post-traumatic stress disorder (PTSD). Front Physiol 2023; 14:1222826. [PMID: 37942228 PMCID: PMC10628526 DOI: 10.3389/fphys.2023.1222826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a neuropsychiatric disorder that occurs in approximately 15% of people as a result of some traumatic events. The main symptoms are re-experiencing and avoidance of everything related to this event and hyperarousal. The main component of the pathophysiology of PTSD is an imbalance in the functioning of the hypothalamic-pituitary-adrenal axis (HPA) and development of neuroinflammation. In parallel with this, mitochondrial dysfunction is observed, as in many other diseases. In this review, we focus on the question how mitochondria may be involved in the development of neuroinflammation and its maintaining at PTSD. First, we describe the differences in the operation of the neuro-endocrine system during stress versus PTSD. We then show changes in the activity/expression of mitochondrial proteins in PTSD and how they can affect the levels of hormones involved in PTSD development, as well as how mitochondrial damage/pathogen-associated molecule patterns (DAMPs/PAMPs) trigger development of inflammation. In addition, we examine the possibility of treating PTSD-related inflammation using mitochondria as a target.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Sviatoslav A. Tsiumpala
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Halyna M. Semchyshyn
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Kenneth B. Storey
- Department of Biology, Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
5
|
Krohn F, Lancini E, Ludwig M, Leiman M, Guruprasath G, Haag L, Panczyszyn J, Düzel E, Hämmerer D, Betts M. Noradrenergic neuromodulation in ageing and disease. Neurosci Biobehav Rev 2023; 152:105311. [PMID: 37437752 DOI: 10.1016/j.neubiorev.2023.105311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
The locus coeruleus (LC) is a small brainstem structure located in the lower pons and is the main source of noradrenaline (NA) in the brain. Via its phasic and tonic firing, it modulates cognition and autonomic functions and is involved in the brain's immune response. The extent of degeneration to the LC in healthy ageing remains unclear, however, noradrenergic dysfunction may contribute to the pathogenesis of Alzheimer's (AD) and Parkinson's disease (PD). Despite their differences in progression at later disease stages, the early involvement of the LC may lead to comparable behavioural symptoms such as preclinical sleep problems and neuropsychiatric symptoms as a result of AD and PD pathology. In this review, we draw attention to the mechanisms that underlie LC degeneration in ageing, AD and PD. We aim to motivate future research to investigate how early degeneration of the noradrenergic system may play a pivotal role in the pathogenesis of AD and PD which may also be relevant to other neurodegenerative diseases.
Collapse
Affiliation(s)
- F Krohn
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Lancini
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| | - M Ludwig
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - M Leiman
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - G Guruprasath
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - L Haag
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - J Panczyszyn
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Düzel
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - D Hämmerer
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany; Department of Psychology, University of Innsbruck, A-6020 Innsbruck, Austria
| | - M Betts
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
6
|
Tong RL, Kahn UN, Grafe LA, Hitti FL, Fried NT, Corbett BF. Stress circuitry: mechanisms behind nervous and immune system communication that influence behavior. Front Psychiatry 2023; 14:1240783. [PMID: 37706039 PMCID: PMC10495591 DOI: 10.3389/fpsyt.2023.1240783] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/16/2023] [Indexed: 09/15/2023] Open
Abstract
Inflammatory processes are increased by stress and contribute to the pathology of mood disorders. Stress is thought to primarily induce inflammation through peripheral and central noradrenergic neurotransmission. In healthy individuals, these pro-inflammatory effects are countered by glucocorticoid signaling, which is also activated by stress. In chronically stressed individuals, the anti-inflammatory effects of glucocorticoids are impaired, allowing pro-inflammatory effects to go unchecked. Mechanisms underlying this glucocorticoid resistance are well understood, but the precise circuits and molecular mechanisms by which stress increases inflammation are not as well known. In this narrative review, we summarize the mechanisms by which chronic stress increases inflammation and contributes to the onset and development of stress-related mood disorders. We focus on the neural substrates and molecular mechanisms, especially those regulated by noradrenergic signaling, that increase inflammatory processes in stressed individuals. We also discuss key knowledge gaps in our understanding of the communication between nervous and immune systems during stress and considerations for future therapeutic strategies. Here we highlight the mechanisms by which noradrenergic signaling contributes to inflammatory processes during stress and how this inflammation can contribute to the pathology of stress-related mood disorders. Understanding the mechanisms underlying crosstalk between the nervous and immune systems may lead to novel therapeutic strategies for mood disorders and/or provide important considerations for treating immune-related diseases in individuals suffering from stress-related disorders.
Collapse
Affiliation(s)
- Rose L. Tong
- Corbett Laboratory, Department of Biology, Rutgers University, Camden, NJ, United States
| | - Ubaidah N. Kahn
- Fried Laboratory, Department of Biology, Rutgers University, Camden, NJ, United States
| | - Laura A. Grafe
- Grafe Laboratory, Department of Psychology, Bryn Mawr College, Bryn Mawr, PA, United States
| | - Frederick L. Hitti
- Hitti Laboratory, Department of Neurological Surgery and Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Nathan T. Fried
- Fried Laboratory, Department of Biology, Rutgers University, Camden, NJ, United States
| | - Brian F. Corbett
- Corbett Laboratory, Department of Biology, Rutgers University, Camden, NJ, United States
| |
Collapse
|
7
|
Laurencin C, Lancelot S, Merida I, Costes N, Redouté J, Le Bars D, Boulinguez P, Ballanger B. Distribution of α 2-Adrenergic Receptors in the Living Human Brain Using [ 11C]yohimbine PET. Biomolecules 2023; 13:biom13050843. [PMID: 37238713 DOI: 10.3390/biom13050843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The neurofunctional basis of the noradrenergic (NA) system and its associated disorders is still very incomplete because in vivo imaging tools in humans have been missing up to now. Here, for the first time, we use [11C]yohimbine in a large sample of subjects (46 healthy volunteers, 23 females, 23 males; aged 20-50) to perform direct quantification of regional alpha 2 adrenergic receptors' (α2-ARs) availability in the living human brain. The global map shows the highest [11C]yohimbine binding in the hippocampus, the occipital lobe, the cingulate gyrus, and the frontal lobe. Moderate binding was found in the parietal lobe, thalamus, parahippocampus, insula, and temporal lobe. Low levels of binding were found in the basal ganglia, the amygdala, the cerebellum, and the raphe nucleus. Parcellation of the brain into anatomical subregions revealed important variations in [11C]yohimbine binding within most structures. Strong heterogeneity was found in the occipital lobe, the frontal lobe, and the basal ganglia, with substantial gender effects. Mapping the distribution of α2-ARs in the living human brain may prove useful not only for understanding the role of the NA system in many brain functions, but also for understanding neurodegenerative diseases in which altered NA transmission with specific loss of α2-ARs is suspected.
Collapse
Affiliation(s)
- Chloé Laurencin
- Université de Lyon, 69622 Lyon, France
- Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
- INSERM U1028, Lyon Neuroscience Research Center (CRNL), 69000 Lyon, France
- CNRS UMR5292, Lyon Neuroscience Research Center (CRNL), 69000 Lyon, France
- Hôpital Neurologique Pierre Wertheimer, Service de Neurologie C, Centre Expert Parkinson, Hospices Civils de Lyon, 69677 Bron, France
| | - Sophie Lancelot
- Université de Lyon, 69622 Lyon, France
- Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
- INSERM U1028, Lyon Neuroscience Research Center (CRNL), 69000 Lyon, France
- CNRS UMR5292, Lyon Neuroscience Research Center (CRNL), 69000 Lyon, France
- CERMEP-Imagerie du Vivant, 69500 Bron, France
- Hospices Civils de Lyon, 69677 Bron, France
| | - Inès Merida
- CERMEP-Imagerie du Vivant, 69500 Bron, France
| | | | | | - Didier Le Bars
- CERMEP-Imagerie du Vivant, 69500 Bron, France
- Hospices Civils de Lyon, 69677 Bron, France
| | - Philippe Boulinguez
- Université de Lyon, 69622 Lyon, France
- Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
- INSERM U1028, Lyon Neuroscience Research Center (CRNL), 69000 Lyon, France
- CNRS UMR5292, Lyon Neuroscience Research Center (CRNL), 69000 Lyon, France
| | - Bénédicte Ballanger
- Université de Lyon, 69622 Lyon, France
- Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
- INSERM U1028, Lyon Neuroscience Research Center (CRNL), 69000 Lyon, France
- CNRS UMR5292, Lyon Neuroscience Research Center (CRNL), 69000 Lyon, France
| |
Collapse
|
8
|
Hosomoto K, Sasaki T, Yasuhara T, Kameda M, Sasada S, Kin I, Kuwahara K, Kawauchi S, Okazaki Y, Yabuno S, Sugahara C, Kawai K, Nagase T, Tanimoto S, Borlongan CV, Date I. Continuous vagus nerve stimulation exerts beneficial effects on rats with experimentally induced Parkinson's disease: Evidence suggesting involvement of a vagal afferent pathway. Brain Stimul 2023; 16:594-603. [PMID: 36914065 DOI: 10.1016/j.brs.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Vagus nerve stimulation (VNS) exerts neuroprotective and anti-inflammatory effects in preclinical models of central nervous system disorders, including Parkinson's disease (PD). VNS setting applied for experimental models is limited into single-time or intermittent short-duration stimulation. We developed a VNS device which could deliver continuous stimulation for rats. To date, the effects of vagal afferent- or efferent-selective stimulation on PD using continuous electrical stimulation remains to be determined. OBJECTIVE To investigate the effects of continuous and selective stimulation of vagal afferent or efferent fiber on Parkinsonian rats. METHODS Rats were divided into 5 group: intact VNS, afferent VNS (left VNS in the presence of left caudal vagotomy), efferent VNS (left VNS in the presence of left rostral vagotomy), sham, vagotomy. Rats underwent the implantation of cuff-electrode on left vagus nerve and 6-hydroxydopamine administration into the left striatum simultaneously. Electrical stimulation was delivered just after 6-OHDA administration and continued for 14 days. In afferent VNS and efferent VNS group, the vagus nerve was dissected at distal or proximal portion of cuff-electrode to imitate the selective stimulation of afferent or efferent vagal fiber respectively. RESULTS Intact VNS and afferent VNS reduced the behavioral impairments in cylinder test and methamphetamine-induced rotation test, which were accompanied by reduced inflammatory glial cells in substantia nigra with the increased density of the rate limiting enzyme in locus coeruleus. In contrast, efferent VNS did not exert any therapeutic effects. CONCLUSION Continuous VNS promoted neuroprotective and anti-inflammatory effect in experimental PD, highlighting the crucial role of the afferent vagal pathway in mediating these therapeutic outcomes.
Collapse
Affiliation(s)
- Kakeru Hosomoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan.
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Masahiro Kameda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan; Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Susumu Sasada
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Ittetsu Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Ken Kuwahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Yosuke Okazaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Satoru Yabuno
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Chiaki Sugahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Koji Kawai
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Takayuki Nagase
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Shun Tanimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Cesario V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL, 33611, USA
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| |
Collapse
|
9
|
Torrente D, Su EJ, Schielke GP, Warnock M, Mann K, Lawrence DA. Opposing effects of β-2 and β-1 adrenergic receptor signaling on neuroinflammation and dopaminergic neuron survival in α-synuclein-mediated neurotoxicity. J Neuroinflammation 2023; 20:56. [PMID: 36864439 PMCID: PMC9983231 DOI: 10.1186/s12974-023-02748-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Noradrenergic neurons in the locus coeruleus (LC) are the primary source of norepinephrine (NE) in the brain and degeneration of these neurons is reported in the early stages of Parkinson's disease (PD), even prior to dopaminergic neuron degeneration in the substantia nigra (SN), which is a hallmark of PD pathology. NE depletion is generally associated with increased PD pathology in neurotoxin-based PD models. The effect of NE depletion in other models of PD-like α-synuclein-based models is largely unexplored. In PD models and in human patients, β-adrenergic receptors' (AR) signaling is associated with a reduction of neuroinflammation and PD pathology. However, the effect of NE depletion in the brain and the extent of NE and β-ARs signaling involvement in neuroinflammation, and dopaminergic neuron survival is poorly understood. METHODS Two mouse models of PD, a 6OHDA neurotoxin-based model and a human α-synuclein (hα-SYN) virus-based model of PD, were used. DSP-4 was used to deplete NE levels in the brain and its effect was confirmed by HPLC with electrochemical detection. A pharmacological approach was used to mechanistically understand the impact of DSP-4 in the hα-SYN model of PD using a norepinephrine transporter (NET) and a β-AR blocker. Epifluorescence and confocal imaging were used to study changes in microglia activation and T-cell infiltration after β1-AR and β2-AR agonist treatment in the hα-SYN virus-based model of PD. RESULTS Consistent with previous studies, we found that DSP-4 pretreatment increased dopaminergic neuron loss after 6OHDA injection. In contrast, DSP-4 pretreatment protected dopaminergic neurons after hα-SYN overexpression. DSP-4-mediated protection of dopaminergic neurons after hα-SYN overexpression was dependent on β-AR signaling since using a β-AR blocker prevented DSP-4-mediated dopaminergic neuron protection in this model of PD. Finally, we found that the β-2AR agonist, clenbuterol, reduced microglia activation, T-cell infiltration, and dopaminergic neuron degeneration, whereas xamoterol a β-1AR agonist showed increased neuroinflammation, blood brain barrier permeability (BBB), and dopaminergic neuron degeneration in the context of hα-SYN-mediated neurotoxicity. CONCLUSIONS Our data demonstrate that the effects of DSP-4 on dopaminergic neuron degeneration are model specific, and suggest that in the context of α-SYN-driven neuropathology, β2-AR specific agonists may have therapeutic benefit in PD.
Collapse
Affiliation(s)
- Daniel Torrente
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Enming J Su
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gerald P Schielke
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mark Warnock
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kris Mann
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel A Lawrence
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA.
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Wu N, Li X, Ma H, Zhang X, Liu B, Wang Y, Zheng Q, Fan X. The role of the gut microbiota and fecal microbiota transplantation in neuroimmune diseases. Front Neurol 2023; 14:1108738. [PMID: 36816570 PMCID: PMC9929158 DOI: 10.3389/fneur.2023.1108738] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
The gut microbiota plays a key role in the function of the host immune system and neuroimmune diseases. Alterations in the composition of the gut microbiota can lead to pathology and altered formation of microbiota-derived components and metabolites. A series of neuroimmune diseases, such as myasthenia gravis (MG), multiple sclerosis (MS), neuromyelitis optica spectrum disorders (NMOSDs), Guillain-Barré syndrome (GBS), and autoimmune encephalitis (AIE), are associated with changes in the gut microbiota. Microecological therapy by improving the gut microbiota is expected to be an effective measure for treating and preventing some neuroimmune diseases. This article reviews the research progress related to the roles of gut microbiota and fecal microbiota transplantation (FMT) in neuroimmune diseases.
Collapse
Affiliation(s)
- Nan Wu
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Xizhi Li
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - He Ma
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Xue Zhang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Bin Liu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Yuan Wang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China,*Correspondence: Yuan Wang ✉
| | - Qi Zheng
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China,Qi Zheng ✉
| | - Xueli Fan
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China,Xueli Fan ✉
| |
Collapse
|
11
|
Sancandi M, De Caro C, Cypaite N, Marascio N, Avagliano C, De Marco C, Russo E, Constanti A, Mercer A. Effects of a probiotic suspension Symprove™ on a rat early-stage Parkinson's disease model. Front Aging Neurosci 2023; 14:986127. [PMID: 36742204 PMCID: PMC9890174 DOI: 10.3389/fnagi.2022.986127] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
An increasing number of studies in recent years have focused on the role that the gut may play in Parkinson's Disease (PD) pathogenesis, suggesting that the maintenance of a healthy gut may lead to potential treatments of the disease. The health of microbiota has been shown to be directly associated with parameters that play a potential role in PD including gut barrier integrity, immunity, function, metabolism and the correct functioning of the gut-brain axis. The gut microbiota (GM) may therefore be employed as valuable indicators for early diagnosis of PD and potential targets for preventing or treating PD symptoms. Preserving the gut homeostasis using probiotics may therefore lead to a promising treatment strategy due to their known benefits in improving constipation, motor impairments, inflammation, and neurodegeneration. However, the mechanisms underlying the effects of probiotics in PD are yet to be clarified. In this project, we have tested the efficacy of an oral probiotic suspension, Symprove™, on an established animal model of PD. Symprove™, unlike many commercially available probiotics, has been shown to be resistant to gastric acidity, improve symptoms in gastrointestinal diseases and improve gut integrity in an in vitro PD model. In this study, we used an early-stage PD rat model to determine the effect of Symprove™ on neurodegeneration and neuroinflammation in the brain and on plasma cytokine levels, GM composition and short chain fatty acid (SCFA) release. Symprove™ was shown to significantly influence both the gut and brain of the PD model. It preserved the gut integrity in the PD model, reduced plasma inflammatory markers and changed microbiota composition. The treatment also prevented the reduction in SCFAs and striatal inflammation and prevented tyrosine hydroxylase (TH)-positive cell loss by 17% compared to that observed in animals treated with placebo. We conclude that Symprove™ treatment may have a positive influence on the symptomology of early-stage PD with obvious implications for the improvement of gut integrity and possibly delaying/preventing the onset of neuroinflammation and neurodegeneration in human PD patients.
Collapse
Affiliation(s)
- Marco Sancandi
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom
| | - Carmen De Caro
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
| | - Neringa Cypaite
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom
| | - Nadia Marascio
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Carmela De Marco
- Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Emilio Russo
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
| | - Andrew Constanti
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom
| | - Audrey Mercer
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom,*Correspondence: Audrey Mercer,
| |
Collapse
|
12
|
Oshaghi M, Kourosh-Arami M, Roozbehkia M. Role of neurotransmitters in immune-mediated inflammatory disorders: a crosstalk between the nervous and immune systems. Neurol Sci 2023; 44:99-113. [PMID: 36169755 DOI: 10.1007/s10072-022-06413-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/14/2022] [Indexed: 02/07/2023]
Abstract
Immune-mediated inflammatory diseases (IMIDs) are a group of common heterogeneous disorders, characterized by an alteration of cellular homeostasis. Primarily, it has been shown that the release and diffusion of neurotransmitters from nervous tissue could result in signaling through lymphocyte cell-surface receptors and the modulation of immune function. This finding led to the idea that the neurotransmitters could serve as immunomodulators. It is now manifested that neurotransmitters can also be released from leukocytes and act as autocrine or paracrine modulators. Increasing data indicate that there is a crosstalk between inflammation and alterations in neurotransmission. The primary goal of this review is to demonstrate how these two pathways may converge at the level of the neuron and glia to involve in IMID. We review the role of neurotransmitters in IMID. The different effects that these compounds exert on a variety of immune cells are also reviewed. Current and future developments in understanding the cross-talk between the immune and nervous systems will undoubtedly identify new ways for treating immune-mediated diseases utilizing agonists or antagonists of neurotransmitter receptors.
Collapse
Affiliation(s)
- Mojgan Oshaghi
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maryam Roozbehkia
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Chen W, Zhang XN, Su YS, Wang XY, Li HC, Liu YH, Wan HY, Qu ZY, Jing XH, He W. Electroacupuncture activated local sympathetic noradrenergic signaling to relieve synovitis and referred pain behaviors in knee osteoarthritis rats. Front Mol Neurosci 2023; 16:1069965. [PMID: 36959872 PMCID: PMC10028095 DOI: 10.3389/fnmol.2023.1069965] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
Introduction Recent research has focused on the local control of articular inflammation through neuronal stimulation to avoid the systemic side effects of conventional pharmacological therapies. Electroacupuncture (EA) has been proven to be useful for inflammation suppressing and pain reduction in knee osteoarthritis (KOA) patients, yet its mechanism remains unclear. Methods In the present study, the KOA model was established using the intra-articular injection of sodium monoiodoacetate (MIA) (1 mg/50 μL) into the knee cavity. EA was delivered at the ipsilateral ST36-GB34 acupoints. Hind paw weight-bearing and withdrawl thresholds were measured. On day 9, the histology, dep enrichment proteins, cytokines contents, immune cell population of the synovial membrane of the affected limbs were measured using HE staining, Masson staining, DIA quantitative proteomic analysis, flow cytometry, immunofluorescence staining, ELISA, and Western Blot. The ultrastructure of the saphenous nerve of the affected limb was observed using transmission electron microscopy on the 14th day after modeling. Results The result demonstrated that EA intervention during the midterm phase of the articular inflammation alleviated inflammatory pain behaviors and cartilage damage, but not during the early phase. Mid-term EA suppressed the levels of proinflammatory cytokines TNF-α, IL-1β, and IL-6 in the synovium on day 9 after MIA by elevating the level of sympathetic neurotransmitters Norepinephrine (NE) in the synovium but not systemic NE or systemic adrenaline. Selective blocking of the sympathetic function (6-OHDA) and β2-adrenergic receptor (ICI 118,551) prevented the anti-inflammatory effects of EA. EA-induced increment of the NE in the synovium inhibited the CXCL1-CXCR2 dependent overexpression of IL-6 in the synovial macrophages in a β2-adrenergic receptor (AR)-mediated manner. Discussion These results revealed that EA activated sympathetic noradrenergic signaling to control local inflammation in KOA rats and contributed to the development of novel therapeutic neurostimulation strategies for inflammatory diseases.
Collapse
|
14
|
Matsumoto H, Tagai K, Endo H, Matsuoka K, Takado Y, Kokubo N, Shimada H, Goto T, Goto TK, Higuchi M. Association of Tooth Loss with Alzheimer's Disease Tau Pathologies Assessed by Positron Emission Tomography. J Alzheimers Dis 2023; 96:1253-1265. [PMID: 37980663 PMCID: PMC10741329 DOI: 10.3233/jad-230581] [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] [Accepted: 09/24/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Deterioration of the oral environment is one of the risk factors for dementia. A previous study of an Alzheimer's disease (AD) model mouse suggests that tooth loss induces denervation of the mesencephalic trigeminal nucleus and neuroinflammation, possibly leading to accelerated tau dissemination from the nearby locus coeruleus (LC). OBJECTIVE To elucidate the relevance of oral conditions and amyloid-β (Aβ) and tau pathologies in human participants. METHODS We examined the number of remaining teeth and the biofilm-gingival interface index in 24 AD-spectrum patients and 19 age-matched healthy controls (HCs). They also underwent positron emission tomography (PET) imaging of Aβ and tau with specific radiotracers, 11C-PiB and 18F-PM-PBB3, respectively. All AD-spectrum patients were Aβ-positive, and all HCs were Aβ-negative. We analyzed the correlation between the oral parameters and radiotracer retention. RESULTS No differences were found in oral conditions between the AD and HC groups. 11C-PiB retentions did not correlate with the oral indices in either group. In AD-spectrum patients, brain-wide, voxel-based image analysis highlighted several regions, including the LC and associated brainstem substructures, as areas where 18F-PM-PBB3 retentions negatively correlated with the remaining teeth and revealed the correlation of tau deposits in the LC (r = -0.479, p = 0.018) primarily with the hippocampal and neighboring areas. The tau deposition in none of the brain regions was associated with the periodontal status. CONCLUSIONS Our findings with previous preclinical evidence imply that tooth loss may enhance AD tau pathogenesis, promoting tau spreading from LC to the hippocampal formation.
Collapse
Affiliation(s)
- Hideki Matsumoto
- Department of Oral and Maxillofacial Radiology, Tokyo Dental College, Tokyo, Japan
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Kenji Tagai
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
- Department of Psychiatry, The Jikei University of Medicine, Tokyo, Japan
| | - Hironobu Endo
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Kiwamu Matsuoka
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Naomi Kokubo
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
- Department of Functional Neurology & Neurosurgery, Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tetsuya Goto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Tazuko K. Goto
- Department of Oral and Maxillofacial Radiology, Tokyo Dental College, Tokyo, Japan
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
- Tokyo Dental College Research Branding Project, Tokyo Dental College, Tokyo, Japan
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| |
Collapse
|
15
|
Tonelli Enrico V, Vo N, Methe B, Morris A, Sowa G. An unexpected connection: A narrative review of the associations between Gut Microbiome and Musculoskeletal Pain. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2022; 31:3603-3615. [PMID: 36308543 PMCID: PMC9617047 DOI: 10.1007/s00586-022-07429-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Multiple diverse factors contribute to musculoskeletal pain, a major cause of physical dysfunction and health-related costs worldwide. Rapidly growing evidence demonstrates that the gut microbiome has overarching influences on human health and the body's homeostasis and resilience to internal and external perturbations. This broad role of the gut microbiome is potentially relevant and connected to musculoskeletal pain, though the literature on the topic is limited. Thus, the literature on the topic of musculoskeletal pain and gut microbiome was explored. METHODS This narrative review explores the vast array of reported metabolites associated with inflammation and immune-metabolic response, which are known contributors to musculoskeletal pain. Moreover, it covers known modifiable (e.g., diet, lifestyle choices, exposure to prescription drugs, pollutants, and chemicals) and non-modifiable factors (e.g., gut architecture, genetics, age, birth history, and early feeding patterns) that are known to contribute to changes to the gut microbiome. Particular attention is devoted to modifiable factors, as the ultimate goal of researching this topic is to implement gut microbiome health interventions into clinical practice. RESULTS Overall, numerous associations exist in the literature that could converge on the gut microbiome's pivotal role in musculoskeletal health. Particularly, a variety of metabolites that are either directly produced or indirectly modulated by the gut microbiome have been highlighted. CONCLUSION The review highlights noticeable connections between the gut and musculoskeletal health, thus warranting future research to focus on the gut microbiome's role in musculoskeletal conditions.
Collapse
Affiliation(s)
- Valerio Tonelli Enrico
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, University of Pittsburgh, 200 Lothrop Street, Room E1612, BST, Pittsburgh, PA, 15261, USA.
- Department of Physical Therapy, University of Pittsburgh, 100 Technology Dr, Pittsburgh, PA, 15219, USA.
| | - Nam Vo
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, University of Pittsburgh, 200 Lothrop Street, Room E1612, BST, Pittsburgh, PA, 15261, USA
| | - Barbara Methe
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, University of Pittsburgh, 1218 Scaife Hall 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Alison Morris
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, University of Pittsburgh, 1218 Scaife Hall 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Gwendolyn Sowa
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, University of Pittsburgh, 200 Lothrop Street, Room E1612, BST, Pittsburgh, PA, 15261, USA
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, Kaufmann Medical Building, Suite 910, 3471 Fifth Avenue, Pittsburgh, PA, 15213, USA
| |
Collapse
|
16
|
Zakaria FH, Samhani I, Mustafa MZ, Shafin N. Pathophysiology of Depression: Stingless Bee Honey Promising as an Antidepressant. Molecules 2022; 27:molecules27165091. [PMID: 36014336 PMCID: PMC9416360 DOI: 10.3390/molecules27165091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/30/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Depression is a debilitating psychiatric disorder impacting an individual’s quality of life. It is the most prevalent mental illness across all age categories, incurring huge socio-economic impacts. Most depression treatments currently focus on the elevation of neurotransmitters according to the monoamine hypothesis. Conventional treatments include tricyclic antidepressants (TCAs), norepinephrine–dopamine reuptake inhibitors (NDRIs), monoamine oxidase inhibitors (MAOIs), and serotonin reuptake inhibitors (SSRIs). Despite numerous pharmacological strategies utilising conventional drugs, the discovery of alternative medicines from natural products is a must for safer and beneficial brain supplement. About 30% of patients have been reported to show resistance to drug treatments coupled with functional impairment, poor quality of life, and suicidal ideation with a high relapse rate. Hence, there is an urgency for novel discoveries of safer and highly effective depression treatments. Stingless bee honey (SBH) has been proven to contain a high level of antioxidants compared to other types of honey. This is a comprehensive review of the potential use of SBH as a new candidate for antidepressants from the perspective of the monoamine, inflammatory and neurotrophin hypotheses.
Collapse
Affiliation(s)
- Fatin Haniza Zakaria
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
| | - Ismail Samhani
- Faculty of Medicine, Universiti Sultan Zainal Abidin (UniSZA), Medical Campus, Jalan Sultan Mahmud, Kuala Terengganu 20400, Malaysia
| | - Mohd Zulkifli Mustafa
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
- Correspondence: (M.Z.M.); (N.S.); Tel.: +609-7673000 (M.Z.M. & N.S.)
| | - Nazlahshaniza Shafin
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
- Correspondence: (M.Z.M.); (N.S.); Tel.: +609-7673000 (M.Z.M. & N.S.)
| |
Collapse
|
17
|
Gu C, Chen Y, Chen Y, Liu CF, Zhu Z, Wang M. Role of G Protein-Coupled Receptors in Microglial Activation: Implication in Parkinson's Disease. Front Aging Neurosci 2021; 13:768156. [PMID: 34867296 PMCID: PMC8635063 DOI: 10.3389/fnagi.2021.768156] [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: 08/31/2021] [Accepted: 10/23/2021] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease (PD) is one of the prevalent neurodegenerative diseases associated with preferential loss of dopaminergic (DA) neurons in the substantia nigra compacta (SNc) and accumulation of α-synuclein in DA neurons. Even though the precise pathogenesis of PD is not clear, a large number of studies have shown that microglia-mediated neuroinflammation plays a vital role in the process of PD development. G protein-coupled receptors (GPCRs) are widely expressed in microglia and several of them act as regulators of microglial activation upon corresponding ligands stimulations. Upon α-synuclein insults, microglia would become excessively activated through some innate immune receptors. Presently, as lack of ideal drugs for treating PD, certain GPCR which is highly expressed in microglia of PD brain and mediates neuroinflammation effectively could be a prospective source for PD therapeutic intervention. Here, six kinds of GPCRs and two types of innate immune receptors were introduced, containing adenosine receptors, purinergic receptors, metabotropic glutamate receptors, adrenergic receptors, cannabinoid receptors, and melatonin receptors and their roles in neuroinflammation; we highlighted the relationship between these six GPCRs and microglial activation in PD. Based on the existing findings, we tried to expound the implication of microglial GPCRs-regulated neuroinflammation to the pathophysiology of PD and their potential to become a new expectation for clinical therapeutics.
Collapse
Affiliation(s)
- Chao Gu
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| | - Yajing Chen
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| | - Yan Chen
- Department of Child and Adolescent Healthcare, Children’s Hospital of Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology, Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zengyan Zhu
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| | - Mei Wang
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| |
Collapse
|
18
|
Tseng CT, Gaulding SJ, Dancel CLE, Thorn CA. Local activation of α2 adrenergic receptors is required for vagus nerve stimulation induced motor cortical plasticity. Sci Rep 2021; 11:21645. [PMID: 34737352 PMCID: PMC8568982 DOI: 10.1038/s41598-021-00976-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022] Open
Abstract
Vagus nerve stimulation (VNS) paired with rehabilitation training is emerging as a potential treatment for improving recovery of motor function following stroke. In rats, VNS paired with skilled forelimb training results in significant reorganization of the somatotopic cortical motor map; however, the mechanisms underlying this form of VNS-dependent plasticity remain unclear. Recent studies have shown that VNS-driven cortical plasticity is dependent on noradrenergic innervation of the neocortex. In the central nervous system, noradrenergic α2 receptors (α2-ARs) are widely expressed in the motor cortex and have been critically implicated in synaptic communication and plasticity. In current study, we examined whether activation of cortical α2-ARs is necessary for VNS-driven motor cortical reorganization to occur. Consistent with previous studies, we found that VNS paired with motor training enlarges the map representation of task-relevant musculature in the motor cortex. Infusion of α2-AR antagonists into M1 blocked VNS-driven motor map reorganization from occurring. Our results suggest that local α2-AR activation is required for VNS-induced cortical reorganization to occur, providing insight into the mechanisms that may underlie the neuroplastic effects of VNS therapy.
Collapse
Affiliation(s)
- Ching-Tzu Tseng
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA
| | - Solomon J Gaulding
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA
| | - Canice Lei E Dancel
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA
| | - Catherine A Thorn
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA.
| |
Collapse
|
19
|
Semyachkina-Glushkovskaya O, Mamedova A, Vinnik V, Klimova M, Saranceva E, Ageev V, Yu T, Zhu D, Penzel T, Kurths J. Brain Mechanisms of COVID-19-Sleep Disorders. Int J Mol Sci 2021; 22:6917. [PMID: 34203143 PMCID: PMC8268116 DOI: 10.3390/ijms22136917] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
2020 and 2021 have been unprecedented years due to the rapid spread of the modified severe acute respiratory syndrome coronavirus around the world. The coronavirus disease 2019 (COVID-19) causes atypical infiltrated pneumonia with many neurological symptoms, and major sleep changes. The exposure of people to stress, such as social confinement and changes in daily routines, is accompanied by various sleep disturbances, known as 'coronasomnia' phenomenon. Sleep disorders induce neuroinflammation, which promotes the blood-brain barrier (BBB) disruption and entry of antigens and inflammatory factors into the brain. Here, we review findings and trends in sleep research in 2020-2021, demonstrating how COVID-19 and sleep disorders can induce BBB leakage via neuroinflammation, which might contribute to the 'coronasomnia' phenomenon. The new studies suggest that the control of sleep hygiene and quality should be incorporated into the rehabilitation of COVID-19 patients. We also discuss perspective strategies for the prevention of COVID-19-related BBB disorders. We demonstrate that sleep might be a novel biomarker of BBB leakage, and the analysis of sleep EEG patterns can be a breakthrough non-invasive technology for diagnosis of the COVID-19-caused BBB disruption.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Aysel Mamedova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Valeria Vinnik
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Maria Klimova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Elena Saranceva
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Vasily Ageev
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Tingting Yu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dan Zhu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Thomas Penzel
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Sleep Medicine Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Kurths
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
20
|
Inhibition of α-Synuclein Accumulation Improves Neuronal Apoptosis and Delayed Postoperative Cognitive Recovery in Aged Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5572899. [PMID: 34194605 PMCID: PMC8181110 DOI: 10.1155/2021/5572899] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/29/2021] [Accepted: 04/22/2021] [Indexed: 11/18/2022]
Abstract
Delayed neurocognitive recovery (dNCR) is a major complication after anesthesia and surgery in older adults. Alpha-synuclein (α-syn; encoded by the gene, SNCA) has recently been shown to play an important role in hippocampus-dependent working memory. Aggregated forms of α-syn are associated with multiple neurotoxic mechanisms, such as mitochondrial dysfunction and cell death. In this study, we found that blocking α-syn improved both mitochondrial function and mitochondria-dependent neuronal apoptosis in a mouse model of dNCR. Various forms of α-syn (including total α-syn, phosphorylated-Ser129-α-syn, and oligomers) were upregulated in hippocampal tissue and extracted mitochondria after surgical challenge. Clenbuterol is a novel transcription modulator of Scna. Clenbuterol significantly attenuated surgery-induced progressive accumulation of various toxic α-syn forms in the hippocampus, as well as mitochondrial damage and memory deficits in aged mice following surgery. We also observed excessive mitochondrial α-syn accumulation and increased mitochondria-mediated apoptosis in vitro using nerve growth factor-differentiated PC12 cells and primary hippocampal neurons exposed to lipopolysaccharide. To further validate the neuroprotective effect of α-syn inhibition, we used a lentiviral Snca-shRNA (Lv-shSnca) to knockdown Snca. Of note, Lv-shSnca transfection significantly inhibited neuronal apoptosis mediated by the mitochondrial apoptosis pathway in neurons exposed to lipopolysaccharide. This α-syn inhibition improved the disruption to mitochondrial morphology and function, as well as decreased levels of apoptosis. Our results suggest that targeting pathological α-syn may achieve neuroprotection through regulation of mitochondrial homeostasis and suppression of apoptosis in the aged hippocampus, further strengthening the therapeutic potential of targeting α-syn for dNCR.
Collapse
|
21
|
Jing L, Hou L, Zhang D, Li S, Ruan Z, Zhang X, Hong JS, Wang Q. Microglial Activation Mediates Noradrenergic Locus Coeruleus Neurodegeneration via Complement Receptor 3 in a Rotenone-Induced Parkinson's Disease Mouse Model. J Inflamm Res 2021; 14:1341-1356. [PMID: 33859489 PMCID: PMC8044341 DOI: 10.2147/jir.s299927] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
Background Chronic exposure to the insecticide rotenone can damage dopaminergic neurons and lead to an increased risk of Parkinson’s disease (PD). Whereas it is not clear whether rotenone induces neurodegeneration of noradrenergic locus coeruleus (LC/NE) neurons. Chronic neuroinflammation mediated by microglia has been involved in the pathogenesis of PD. Evidence shows that complement receptor 3 (CR3) is a crucial regulator of microglial activation and related neurodegeneration. However, it is not clear whether CR3 mediates rotenone-elicited degeneration of LC/NE neurons through microglia-mediated neuroinflammation. Materials and Methods Wild type (WT) and CR3 knockout (KO) mice were treated with rotenone. PLX3397 and minocycline were used to deplete or inactivate the microglia. Leukadherin-1 (LA-1) was used to modulate CR3. LC/NE neurodegeneration, microglial phenotype, and expression of CR3 were determined by using immunohistochemistry, Western blot and real-time polymerase chain reaction (PCR) techniques. The glutathione (GSH) and malondialdehyde (MDA) contents were measured by using commercial kits. Results Rotenone exposure led to dose- and time-dependent LC/NE neuronal loss and microglial activation in mice. Depletion of microglia by PLX3397 or inhibition of microglial activation by minocycline significantly reduced rotenone-induced LC/NE neurodegeneration. Mechanistic studies revealed that CR3 played an essential role in the rotenone-induced activation of microglia and neurodegeneration of LC/NE neurons. Rotenone elevated the expression of CR3, and genetic ablation of CR3 markedly reduced rotenone-induced microglial activation and M1 polarization. LA-1 also suppressed rotenone-induced toxic microglial M1 activation. Furthermore, lack of CR3 or treatment with LA-1 reduced oxidative stress in the brainstem of rotenone-intoxicated mice. Finally, we found that mice deficient in CR3 or treated with LA-1 were more resistant to rotenone-induced LC/NE neurodegeneration than WT or vehicle-treated mice, respectively. Conclusion Our results indicate that CR3-mediated microglial activation participates in rotenone-induced LC/NE neurodegeneration, providing novel insight into environmental toxin-induced neurotoxicity and related Parkinsonism.
Collapse
Affiliation(s)
- Lu Jing
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China.,Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Liyan Hou
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Dongdong Zhang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Sheng Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Zhengzheng Ruan
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Xiaomeng Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Qingshan Wang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China.,National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| |
Collapse
|
22
|
Idazoxan and Efaroxan Potentiate the Endurance Performances and the Antioxidant Activity of Ephedrine in Rats. ACTA ACUST UNITED AC 2021; 57:medicina57030194. [PMID: 33668888 PMCID: PMC7996498 DOI: 10.3390/medicina57030194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/19/2022]
Abstract
Background and objectives: The connections between the imidazoline system and multiple other neurotransmitter systems in the brain (adrenergic, dopaminergic, serotoninergic, glutamatergic, opioid) indicate the complexity of the mechanisms underlying motor activity and behavior. The aim of the present research was to investigate the effects of the combination of ephedrine (EPD) and imidazoline antagonists idazoxan (IDZ) and efaroxan (EFR) on the endurance performance in the treadmill test in rats. Materials and Methods: We used Wistar rats distributed as follows: Group 1 (Control) receiving distilled water 0.3 mL/100 g body weight; Group 2 (EPD) receiving 20 mg/kg ephedrine; Group 3 (EPD + IDZ) receiving 20 mg/kg ephedrine + 3 mg/kg idazoxan; Group 4 (EPD + EFR) receiving 20 mg/kg ephedrine + 1 mg/kg efaroxan. An additional group (C) of animals receiving 0.3 mL/100 g body weight distilled water (but not subjected to effort) was used. Endurance capacity was evaluated using a treadmill running PanLAB assay. The evaluation of the substances’ influence on oxidative stress was performed by spectrophotometric determination of superoxide dismutase (SOD) and glutathione peroxidase (GPX) activity. Results: Treatment with EPD-IDZ and EPD-EFR were correlated with a longer distance traveled on the belt and with a decrease in the necessary electric shocks to motivate the animal to continue running in the forced locomotion test. Additionally, an increase in the activity of antioxidant enzymes was found. Conclusions: Idazoxan and efaroxan potentiated the physical effort-related effects of ephedrine with regard to endurance capacity and antioxidant activity in rats.
Collapse
|
23
|
Clark I, Vissel B. Broader Insights into Understanding Tumor Necrosis Factor and Neurodegenerative Disease Pathogenesis Infer New Therapeutic Approaches. J Alzheimers Dis 2021; 79:931-948. [PMID: 33459706 PMCID: PMC7990436 DOI: 10.3233/jad-201186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
Proinflammatory cytokines such as tumor necrosis factor (TNF), with its now appreciated key roles in neurophysiology as well as neuropathophysiology, are sufficiently well-documented to be useful tools for enquiry into the natural history of neurodegenerative diseases. We review the broader literature on TNF to rationalize why abruptly-acquired neurodegenerative states do not exhibit the remorseless clinical progression seen in those states with gradual onsets. We propose that the three typically non-worsening neurodegenerative syndromes, post-stroke, post-traumatic brain injury (TBI), and post cardiac arrest, usually become and remain static because of excess cerebral TNF induced by the initial dramatic peak keeping microglia chronically activated through an autocrine loop of microglial activation through excess cerebral TNF. The existence of this autocrine loop rationalizes post-damage repair with perispinal etanercept and proposes a treatment for cerebral aspects of COVID-19 chronicity. Another insufficiently considered aspect of cerebral proinflammatory cytokines is the fitness of the endogenous cerebral anti-TNF system provided by norepinephrine (NE), generated and distributed throughout the brain from the locus coeruleus (LC). We propose that an intact LC, and therefore an intact NE-mediated endogenous anti-cerebral TNF system, plus the DAMP (damage or danger-associated molecular pattern) input having diminished, is what allows post-stroke, post-TBI, and post cardiac arrest patients a strong long-term survival advantage over Alzheimer's disease and Parkinson's disease sufferers. In contrast, Alzheimer's disease and Parkinson's disease patients remorselessly worsen, being handicapped by sustained, accumulating, DAMP and PAMP (pathogen-associated molecular patterns) input, as well as loss of the LC-origin, NE-mediated, endogenous anti-cerebral TNF system. Adrenergic receptor agonists may counter this.
Collapse
Affiliation(s)
- I.A. Clark
- Research School of Biology, Australian National University, Canberra, Australia
| | - B. Vissel
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology, Sydney, Australia
- St. Vincent’s Centre for Applied Medical Research, Sydney, Australia
| |
Collapse
|
24
|
Tafseer S, Gupta R, Ahmad R, Jain S, Bhatia MS, Gupta LK. Bupropion monotherapy alters neurotrophic and inflammatory markers in patients of major depressive disorder. Pharmacol Biochem Behav 2020; 200:173073. [PMID: 33186562 DOI: 10.1016/j.pbb.2020.173073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 09/25/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Emerging hypotheses in the pathophysiology of major depressive disorder (MDD) indicate the role of neurotrophic factors and inflammation. This study assessed the association between therapeutic response of bupropion and serum brain-derived neurotrophic factor (BDNF) and tumour necrosis factor-α (TNF-α) levels in patients with MDD. METHODS Thirty patients (aged 18 to 60 years) with MDD diagnosed by DSM-5 criteria, with Hamilton Depression Rating scale (HAM-D) score ≥ 20 were included in the study. Patients were given bupropion sustained release (SR) in the doses of 150 mg once daily. All patients were followed up for 12 weeks. RESULTS HAM-D score at the start of the treatment was 25.57 ± 1.85 which significantly reduced to 10.8 ± 4.24 at 12 weeks of treatment. The serum BDNF level increased significantly (p < 0.05) from 2.42 ± 0.19 ng/ml to 2.97 ± 0.10 ng/ml and the levels of serum TNF-α reduced significantly (p < 0.05) from 4.45 ± 0.95 pg/ml to 2.11 ± 0.84 pg/ml at 12 weeks of treatment, in responders to treatment. CONCLUSION The results of our study suggest that bupropion SR monotherapy is effective and well tolerated in MDD patients with moderate to severe depression, and its therapeutic efficacy is accompanied by an increase in serum BDNF levels and a decrease in serum TNF-α levels.
Collapse
Affiliation(s)
- Sana Tafseer
- Department of Pharmacology, University College of Medical Sciences & Guru Teg Bahadur Hospital, New Delhi 110095, India
| | - Rachna Gupta
- Department of Pharmacology, University College of Medical Sciences & Guru Teg Bahadur Hospital, New Delhi 110095, India.
| | - Rafat Ahmad
- Department of Biochemistry, University College of Medical Sciences & Guru Teg Bahadur Hospital, New Delhi 110095, India
| | - Seema Jain
- Department of Pharmacology, University College of Medical Sciences & Guru Teg Bahadur Hospital, New Delhi 110095, India
| | - M S Bhatia
- Department of Psychiatry, University College of Medical Sciences & Guru Teg Bahadur Hospital, New Delhi 110095, India
| | - Lalit K Gupta
- Department of Pharmacology, Lady Hardinge Medical College & Smt. S.K. Hospital, New Delhi, India
| |
Collapse
|
25
|
Ryan KM, Harkin A. Regulation of β 2-adrenoceptors in brain glia: implications for neuroinflammatory and degenerative disorders. Neural Regen Res 2020; 15:2035-2036. [PMID: 32394954 PMCID: PMC7716048 DOI: 10.4103/1673-5374.282255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Karen M Ryan
- Trinity College Institute of Neuroscience, Trinity College Dublin; Department of Psychiatry, St. Patrick's University Hospital, Trinity College Dublin, Dublin, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
26
|
Norepinephrine, neurodevelopment and behavior. Neurochem Int 2020; 135:104706. [PMID: 32092327 DOI: 10.1016/j.neuint.2020.104706] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 02/06/2023]
Abstract
Neurotransmitters play critical roles in the developing nervous system. Among the neurotransmitters, norepinephrine (NE) is in particular postulated to be an important regulator of brain development. NE is expressed during early stages of development and is known to regulate both the development of noradrenergic neurons and the development of target areas. NE participates in the shaping and the wiring of the nervous system during the critical periods of development, and perturbations in this process can alter the brain's developmental trajectory, which in turn can cause long-lasting and even permanent changes in the brain function and behavior later in life. Here we will briefly review evidence for the role of noradrenergic system in neurodevelopmental processes and will discuss about the potential disruptors of noradrenergic system during development and their behavioral consequences.
Collapse
|
27
|
O'Neill E, Yssel JD, McNamara C, Harkin A. Pharmacological targeting of β 2 -adrenoceptors is neuroprotective in the LPS inflammatory rat model of Parkinson's disease. Br J Pharmacol 2020; 177:282-297. [PMID: 31506926 PMCID: PMC6989960 DOI: 10.1111/bph.14862] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/20/2019] [Accepted: 08/27/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Chronic inflammation may play a role in the pathogenesis of Parkinson's disease (PD). Noradrenaline is an endogenous neurotransmitter with anti-inflammatory properties. In the present investigation, we assessed the immunomodulatory and neuroprotective efficacy of pharmacologically targeting the CNS noradrenergic system in a rat model of PD. EXPERIMENTAL APPROACH The impact of treatment with the β2 -adrenoceptor agonists clenbuterol and formoterol was assessed in the intranigral LPS rat model of PD. The immunomodulatory potential of formoterol to influence the CNS response to systemic inflammation was also assessed. KEY RESULTS LPS-induced deficits in motor function (akinesia and forelimb-use asymmetry) and nigrostriatal dopamine loss were rescued by both agents. Treatment with the noradrenaline reuptake inhibitor atomoxetine reduced striatal dopamine loss and motor deficits following intranigral LPS injection. Co-treatment with the β2 -adrenoceptor antagonist ICI 118,551 attenuated the protective effects of atomoxetine. Systemic LPS challenge exacerbated reactive microgliosis, IL-1β production, dopamine cell loss in the substantia nigra, nerve terminal degeneration in the striatum, and associated motor impairments in animals that previously received intranigral LPS. This exacerbation was attenuated by formoterol treatment. CONCLUSION AND IMPLICATIONS The results indicate that pharmacologically targeting β2 -adrenoceptors has the propensity to regulate the neuroinflammatory phenotype in vivo and may be a potential neuroprotective strategy where inflammation contributes to the progression of dopaminergic neurodegeneration. In accordance with this, clinical agents such as β2 -adrenoceptor agonists may prove useful as immunomodulatory agents in the treatment of neurodegenerative conditions associated with brain inflammation.
Collapse
Affiliation(s)
- Eoin O'Neill
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of NeuroscienceTrinity College DublinDublin 2Ireland
| | - Justin D. Yssel
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of NeuroscienceTrinity College DublinDublin 2Ireland
| | - Caoimhe McNamara
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of NeuroscienceTrinity College DublinDublin 2Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of NeuroscienceTrinity College DublinDublin 2Ireland
| |
Collapse
|
28
|
Zhu YF, Wang WP, Zheng XF, Chen Z, Chen T, Huang ZY, Jia LJ, Lei WL. Characteristic response of striatal astrocytes to dopamine depletion. Neural Regen Res 2020; 15:724-730. [PMID: 31638097 PMCID: PMC6975155 DOI: 10.4103/1673-5374.266917] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Astrocytes and astrocyte-related proteins play important roles in maintaining normal brain function, and also regulate pathological processes in brain diseases and injury. However, the role of astrocytes in the dopamine-depleted striatum remains unclear. A rat model of Parkinson’s disease was therefore established by injecting 10 μL 6-hydroxydopamine (2.5 μg/μL) into the right medial forebrain bundle. Immunohistochemical staining was used to detect the immunoreactivity of glial fibrillary acidic protein (GFAP), calcium-binding protein B (S100B), and signal transducer and activator of transcription 3 (STAT3) in the striatum, and to investigate the co-expression of GFAP with S100B and STAT3. Western blot assay was used to measure the protein expression of GFAP, S100B, and STAT3 in the striatum. Results demonstrated that striatal GFAP-immunoreactive cells had an astrocytic appearance under normal conditions, but that dopamine depletion induced a reactive phenotype with obvious morphological changes. The normal striatum also contained S100B and STAT3 expression. S100B-immunoreactive cells were uniform in the striatum, with round bodies and sparse, thin processes. STAT3-immunoreactive cells presented round cell bodies with sparse processes, or were darkly stained with a large cell body. Dopamine deprivation induced by 6-hydroxydopamine significantly enhanced the immunohistochemical positive reaction of S100B and STAT3. Normal striatal astrocytes expressed both S100B and STAT3. Striatal dopamine deprivation increased the number of GFAP/S100B and GFAP/STAT3 double-labeled cells, and increased the protein levels of GFAP, S100B, and STAT3. The present results suggest that morphological changes in astrocytes and changes in expression levels of astrocyte-related proteins are involved in the pathological process of striatal dopamine depletion. The study was approved by Animal Care and Use Committee of Sun Yat-sen University, China (Zhongshan Medical Ethics 2014 No. 23) on September 22, 2014.
Collapse
Affiliation(s)
- Yao-Feng Zhu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province; Institute of Medicine, College of Medicine, Jishou University, Jishou, Hunan Province, China
| | - Wei-Ping Wang
- Periodical Center, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xue-Feng Zheng
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhi Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Tao Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zi-Yun Huang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lin-Ju Jia
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Wan-Long Lei
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
29
|
Ryan KM, Boyle NT, Harkin A, Connor TJ. Dexamethasone attenuates inflammatory-mediated suppression of β 2-adrenoceptor expression in rat primary mixed glia. J Neuroimmunol 2019; 338:577082. [PMID: 31707103 DOI: 10.1016/j.jneuroim.2019.577082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 10/04/2019] [Accepted: 10/04/2019] [Indexed: 11/29/2022]
Abstract
β2-adrenoceptors are G-protein coupled receptors expressed on both astrocytes and microglia that play a key role in mediating the anti-inflammatory actions of noradrenaline in the CNS. Here the effect of an inflammatory stimulus (LPS + IFN-γ) was examined on glial β2-adrenoceptor expression and function. Exposure of glia to LPS + IFN-γ decreased β2-adrenoceptor mRNA and agonist-stimulated production of the intracellular second messenger cAMP. Pre-treatment with the synthetic glucocorticoid and potent anti-inflammatory agent dexamethasone prevented the LPS + IFN-γ-induced suppression of β2-adrenoceptor mRNA expression. These results raise the possibility that inflammation-mediated β2-adrenoceptor downregulation in glia may dampen the innate anti-inflammatory properties of noradrenaline in the CNS.
Collapse
Affiliation(s)
- Karen M Ryan
- Neuroimmunology Research Group, Department of Physiology, Trinity College Institute of Neuroscience & School of Medicine, Trinity College, Dublin 2, Ireland
| | - Noreen T Boyle
- Neuroimmunology Research Group, Department of Physiology, Trinity College Institute of Neuroscience & School of Medicine, Trinity College, Dublin 2, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin 2, Ireland.
| | - Thomas J Connor
- Neuroimmunology Research Group, Department of Physiology, Trinity College Institute of Neuroscience & School of Medicine, Trinity College, Dublin 2, Ireland
| |
Collapse
|
30
|
Lesion of the Locus Coeruleus Damages Learning and Memory Performance in Paraquat and Maneb-induced Mouse Parkinson’s Disease Model. Neuroscience 2019; 419:129-140. [DOI: 10.1016/j.neuroscience.2019.09.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 11/18/2022]
|
31
|
Abstract
Noradrenergic system of brain supplies the neurotransmitter noradrenalin throughout the brain through widespread efferent projections and play pivotal role in cognitive activities and could be involve in motor and non-motor symptoms of Parkinson's disease (PD) pathology. Profound loss of noradrenergic pathways has been reported in both Parkinson's and Alzheimer's disease (AD) pathology however their employment in therapeutics is still scarce. Therefore the present review is providing the various aspects for involvement on noradrenergic pathways in PD and AD pathology as well as the imaging of locus coeruleus as indicative diagnostic marker for disease. The present review is describing about the role of tiny nucleus locus coeruleus located noradrenergic pathways in said pathologies and discussing the past research as well as lacunas in this regard.
Collapse
Affiliation(s)
- Sarika Singh
- Toxicology and Experimental Medicine Division, CDRI-CSIR, Lucknow, UP, India
| |
Collapse
|
32
|
Lamkin DM, Srivastava S, Bradshaw KP, Betz JE, Muy KB, Wiese AM, Yee SK, Waggoner RM, Arevalo JMG, Yoon AJ, Faull KF, Sloan EK, Cole SW. C/EBPβ regulates the M2 transcriptome in β-adrenergic-stimulated macrophages. Brain Behav Immun 2019; 80:839-848. [PMID: 31132458 PMCID: PMC6660400 DOI: 10.1016/j.bbi.2019.05.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 12/11/2022] Open
Abstract
At the M2 terminal of the macrophage activation spectrum, expression of genes is regulated by transcription factors that include STAT6, CREB, and C/EBPβ. Signaling through β-adrenergic receptors drives M2 activation of macrophages, but little is known about the transcription factors involved. In the present study, we found that C/EBPβ regulates the signaling pathway between β-adrenergic stimulation and expression of Arg1 and several other specific genes in the greater M2 transcriptome. β-adrenergic signaling induced Cebpb gene expression relatively early with a peak at 1 h post-stimulation, followed by peak Arg1 gene expression at 8 h. C/EBPβ transcription factor activity was elevated at the enhancer region for Arg 1 at both 4 and 8 h after stimulation but not near the more proximal promoter region. Knockdown of Cebpb suppressed the β-adrenergic-induced peak in Cebpb gene expression as well as subsequent accumulation of C/EBPβ protein in the nucleus, which resulted in suppression of β-adrenergic-induced Arg1 gene expression. Analysis of genome-wide transcriptional profiles identified 20 additional M2 genes that followed the same pattern of regulation by β-adrenergic- and C/EBPβ-signaling. Promoter-based bioinformatic analysis confirmed enrichment of binding motifs for C/EBPβ transcription factor across these M2 genes. These findings pinpoint a mechanism that may be targeted to redirect the deleterious influence of β-adrenergic signaling on macrophage involvement in M2-related diseases such as cancer.
Collapse
Affiliation(s)
- Donald M Lamkin
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States; Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles 90095, United States; Jonsson Comprehensive Cancer Center, University of California, Los Angeles 90095, United States.
| | - Shreyesi Srivastava
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Karen P Bradshaw
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Jenna E Betz
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Kevin B Muy
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Anna M Wiese
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Shelby K Yee
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Rebecca M Waggoner
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Jesusa M G Arevalo
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States; Divison of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles 90095, United States
| | - Alexander J Yoon
- Pasarow Mass Spectrometry Laboratory, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Kym F Faull
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles 90095, United States; Pasarow Mass Spectrometry Laboratory, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Erica K Sloan
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States; Jonsson Comprehensive Cancer Center, University of California, Los Angeles 90095, United States; Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, VIC 3002, Australia
| | - Steve W Cole
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States; Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles 90095, United States; Jonsson Comprehensive Cancer Center, University of California, Los Angeles 90095, United States; Divison of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles 90095, United States
| |
Collapse
|
33
|
Sancandi M, Schul EV, Economides G, Constanti A, Mercer A. Structural Changes Observed in the Piriform Cortex in a Rat Model of Pre-motor Parkinson's Disease. Front Cell Neurosci 2018; 12:479. [PMID: 30618629 PMCID: PMC6296349 DOI: 10.3389/fncel.2018.00479] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/22/2018] [Indexed: 12/11/2022] Open
Abstract
Early diagnosis of Parkinson’s disease (PD) offers perhaps, the most promising route to a successful clinical intervention, and the use of an animal model exhibiting symptoms comparable to those observed in PD patients in the early stage of the disease, may facilitate screening of novel therapies for delaying the onset of more debilitating motor and behavioral abnormalities. In this study, a rat model of pre-motor PD was used to study the etiology of hyposmia, a non-motor symptom linked to the early stage of the disease when the motor symptoms have yet to be experienced. The study focussed on determining the effect of a partial reduction of both dopamine and noradrenaline levels on the olfactory cortex. Neuroinflammation and striking structural changes were observed in the model. These changes were prevented by treatment with a neuroprotective drug, a glucagon-like peptide-1 (GLP1) receptor agonist, exendin-4 (EX-4).
Collapse
|