1
|
Yang C, Wang Y, Wu X, Gong M, Li Y. Reduced levels of A20 protein prompted RIPK1-dependent apoptosis and blood-brain barrier breakdown during cerebral ischemia reperfusion injury. PLoS One 2023; 18:e0290015. [PMID: 37578944 PMCID: PMC10424866 DOI: 10.1371/journal.pone.0290015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/01/2023] [Indexed: 08/16/2023] Open
Abstract
Blood-brain barrier (BBB) leakage is an important cause of the exacerbation of pathological features of cerebral ischemia reperfusion injury (CIRI). However, the specific mechanism of BBB leakage is not clear. It was found that the CIRI resulted in RIPK1 activation and subsequent RIPK1-dependent apoptosis (RDA). Inhibition of RIPK1 significantly reduced BBB breakdown and brain damage. The aim of this study is to investigate the mechanism of RIPK1 in the BBB leakage during CIRI. It was discovered by proteomics that autophagy activation resulting from ischemia and reperfusion significantly downregulated the level of A20 protein. A20 is an important protein that regulates RIPK1 and RDA. It was hypothesized that activation of autophagy caused by ischemic reperfusion led to a decrease in A20 protein, which, in turn, caused the activation of RIPK1 and the occurrence of RDA, leading to leakage of the BBB. The findings in this study revealed the role of RIPK1 in the cell death and BBB leakage upon cerebral ischemia reperfusion injury, and these findings provide a novel perspective for the treatment of ischemic reperfusion.
Collapse
Affiliation(s)
- Chaonan Yang
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yongjiao Wang
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xiaohui Wu
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Min Gong
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
- WIMM, University of Oxford, Oxford, United Kingdom
| | - Ying Li
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Ding R, Su D, Zhao Q, Wang Y, Wang JY, Lv S, Ji X. The role of microRNAs in depression. Front Pharmacol 2023; 14:1129186. [PMID: 37063278 PMCID: PMC10090555 DOI: 10.3389/fphar.2023.1129186] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Major depressive disorder (MDD) is a psychiatric disorder with increasing prevalence worldwide. It is a leading cause of disability and suicide, severely affecting physical and mental health. However, the study of depression remains at an exploratory stage in terms of diagnostics and treatment due to the complexity of its pathogenesis. MicroRNAs are endogenous short-stranded non-coding RNAs capable of binding to the 3’untranslated region of mRNAs. Because of their ability to repress translation process of genes and are found at high levels in brain tissues, investigation of their role in depression has gradually increased recently. This article summarizes recent research progress on the relationship between microRNAs and depression. The microRNAs play a regulatory role in the pathophysiology of depression, involving dysregulation of monoamines, abnormalities in neuroplasticity and neurogenesis, hyperactivity of the HPA axis, and dysregulation of inflammatory responses. These microRNAs might provide new clue for the diagnosis and treatment of MDD, and the development of antidepressant drugs.
Collapse
Affiliation(s)
- Ruidong Ding
- Institute of Molecular Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| | - Dingyuan Su
- Institute of Molecular Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| | - Qian Zhao
- Institute of Molecular Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| | - Yu Wang
- Institute of Molecular Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| | - Jia-Yi Wang
- San-Quan College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Shuangyu Lv
- Institute of Molecular Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
- *Correspondence: Shuangyu Lv, ; Xinying Ji,
| | - Xinying Ji
- Institute of Molecular Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
- Kaifeng Key Laboratory for Infectious Diseases and Biosafety, Kaifeng, Henan, China
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China
- *Correspondence: Shuangyu Lv, ; Xinying Ji,
| |
Collapse
|
3
|
Helman TJ, Headrick JP, Stapelberg NJC, Braidy N. The sex-dependent response to psychosocial stress and ischaemic heart disease. Front Cardiovasc Med 2023; 10:1072042. [PMID: 37153459 PMCID: PMC10160413 DOI: 10.3389/fcvm.2023.1072042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Stress is an important risk factor for modern chronic diseases, with distinct influences in males and females. The sex specificity of the mammalian stress response contributes to the sex-dependent development and impacts of coronary artery disease (CAD). Compared to men, women appear to have greater susceptibility to chronic forms of psychosocial stress, extending beyond an increased incidence of mood disorders to include a 2- to 4-fold higher risk of stress-dependent myocardial infarction in women, and up to 10-fold higher risk of Takotsubo syndrome-a stress-dependent coronary-myocardial disorder most prevalent in post-menopausal women. Sex differences arise at all levels of the stress response: from initial perception of stress to behavioural, cognitive, and affective responses and longer-term disease outcomes. These fundamental differences involve interactions between chromosomal and gonadal determinants, (mal)adaptive epigenetic modulation across the lifespan (particularly in early life), and the extrinsic influences of socio-cultural, economic, and environmental factors. Pre-clinical investigations of biological mechanisms support distinct early life programming and a heightened corticolimbic-noradrenaline-neuroinflammatory reactivity in females vs. males, among implicated determinants of the chronic stress response. Unravelling the intrinsic molecular, cellular and systems biological basis of these differences, and their interactions with external lifestyle/socio-cultural determinants, can guide preventative and therapeutic strategies to better target coronary heart disease in a tailored sex-specific manner.
Collapse
Affiliation(s)
- Tessa J. Helman
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
- Correspondence: Tessa J. Helman
| | - John P. Headrick
- Schoolof Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | | | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
| |
Collapse
|
4
|
Weinekötter J, Gurtner C, Protschka M, von Bomhard W, Böttcher D, Schlinke A, Alber G, Rösch S, Steiner JM, Seeger J, Oechtering GU, Heilmann RM. Tissue S100/calgranulin expression and blood neutrophil-to-lymphocyte ratio (NLR) in dogs with lower urinary tract urothelial carcinoma. BMC Vet Res 2022; 18:412. [PMID: 36411489 PMCID: PMC9680134 DOI: 10.1186/s12917-022-03513-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Urothelial carcinoma (UC) is the most common neoplasm of the canine lower urinary tract, affecting approximately 2% of dogs. Elderly female patients of certain breeds are predisposed, and clinical signs of UC can easily be confused with urinary tract infection or urolithiasis. Diagnosis and treatment are challenging given the lack of disease-specific markers and treatments. The S100A8/A9 complex and S100A12 protein are Ca2+-binding proteins expressed by cells of the innate immune system and have shown promise as urinary screening markers for UC. The neutrophil-to-lymphocyte ratio (NLR) can also aid in distinguishing certain neoplastic from inflammatory conditions. Our study aimed to evaluate the tissue expression of S100/calgranulins and the blood NLR in dogs with UC. Urinary bladder and/or urethral tissue samples from dogs with UC (n = 10), non-neoplastic inflammatory lesions (NNUTD; n = 6), and no histologic changes (n = 11) were evaluated using immunohistochemistry. Blood NLRs were analyzed in dogs with UC (n = 22) or NNUTD (n = 26). RESULTS Tissue S100A12-positive cell counts were significantly higher in dogs with lower urinary tract disease than healthy controls (P = 0.0267 for UC, P = 0.0049 for NNUTD), with no significant difference between UC and NNUTD patients. Tissue S100A8/A9-positivity appeared to be higher with NNUTD than UC, but this difference did not reach statistical significance. The S100A8/A9+-to-S100A12+ ratio was significantly decreased in neoplastic and inflamed lower urinary tract tissue compared to histologically normal specimens (P = 0.0062 for UC, P = 0.0030 for NNUTD). NLRs were significantly higher in dogs with UC than in dogs with NNUTD, and a cut-off NLR of ≤ 2.83 distinguished UC from NNUTD with 41% sensitivity and 100% specificity. Higher NLRs were also associated with a poor overall survival time (P = 0.0417). CONCLUSIONS These results confirm that the S100/calgranulins play a role in the immune response to inflammatory and neoplastic lower urinary tract diseases in dogs, but the tissue expression of these proteins appears to differ from their concentrations reported in urine samples. Further investigations of the S100/calgranulin pathways in UC and their potential as diagnostic or prognostic tools and potential therapeutic targets are warranted. The NLR as a routinely available marker might be a useful surrogate to distinguish UC from inflammatory conditions.
Collapse
Affiliation(s)
- Jana Weinekötter
- grid.9647.c0000 0004 7669 9786Department for Small Animals, College of Veterinary Medicine, Leipzig University, An den Tierkliniken 23, 04103 Leipzig, SN Germany
| | - Corinne Gurtner
- grid.5734.50000 0001 0726 5157Institute of Animal Pathology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, CH-3001 Bern, BE Switzerland
| | - Martina Protschka
- grid.9647.c0000 0004 7669 9786Institute of Immunology, College of Veterinary Medicine, Biotechnological-Biomedical Center, Leipzig University, Deutscher Platz 5, 04103 Leipzig, SN Germany
| | - Wolf von Bomhard
- Specialty Center for Veterinary Pathology, Hartelstrasse 30, E80689 Munich, BY Germany
| | - Denny Böttcher
- grid.9647.c0000 0004 7669 9786Institute for Veterinary Pathology, College of Veterinary Medicine, Leipzig University, An Den Tierkliniken 33, E04103 Leipzig, SN Germany
| | - Annika Schlinke
- grid.9647.c0000 0004 7669 9786Department for Small Animals, College of Veterinary Medicine, Leipzig University, An den Tierkliniken 23, 04103 Leipzig, SN Germany
| | - Gottfried Alber
- grid.9647.c0000 0004 7669 9786Institute of Immunology, College of Veterinary Medicine, Biotechnological-Biomedical Center, Leipzig University, Deutscher Platz 5, 04103 Leipzig, SN Germany
| | - Sarah Rösch
- grid.9647.c0000 0004 7669 9786Department for Small Animals, College of Veterinary Medicine, Leipzig University, An den Tierkliniken 23, 04103 Leipzig, SN Germany ,grid.412970.90000 0001 0126 6191Small Animal Clinic, University of Veterinary Medicine Hannover Foundation, Bünteweg 9, 30559 Hannover, NI Germany
| | - Joerg M. Steiner
- grid.264756.40000 0004 4687 2082Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TAMU 4474, College Station, TX 77843-4474 USA
| | - Johannes Seeger
- grid.9647.c0000 0004 7669 9786Institute of Anatomy, Histology and Embryology, College of Veterinary Medicine, Leipzig University, An den Tierkliniken 43, 04103 Leipzig, SN Germany
| | - Gerhard U. Oechtering
- grid.9647.c0000 0004 7669 9786Department for Small Animals, College of Veterinary Medicine, Leipzig University, An den Tierkliniken 23, 04103 Leipzig, SN Germany
| | - Romy M. Heilmann
- grid.9647.c0000 0004 7669 9786Department for Small Animals, College of Veterinary Medicine, Leipzig University, An den Tierkliniken 23, 04103 Leipzig, SN Germany
| |
Collapse
|
5
|
Fornaro R, Actis GC, Caviglia GP, Pitoni D, Ribaldone DG. Inflammatory Bowel Disease: Role of Vagus Nerve Stimulation. J Clin Med 2022; 11:5690. [PMID: 36233558 PMCID: PMC9572047 DOI: 10.3390/jcm11195690] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/19/2022] [Accepted: 09/24/2022] [Indexed: 11/19/2022] Open
Abstract
Vagus nerve stimulation (VNS) is an accepted therapy for the treatment of refractory forms of epilepsy and depression. The brain-gut axis is increasingly being studied as a possible etiological factor of chronic inflammatory diseases, including inflammatory bowel diseases (IBD). A significant percentage of IBD patients lose response to treatments or experience side effects. In this perspective, VNS has shown the first efficacy data. The aim of this narrative review is to underline the biological plausibility of the use of VNS in patients affected by IBD, collect all clinical data in the literature, and hypothesize a target IBD population on which to focus the next clinical study.
Collapse
Affiliation(s)
- Riccardo Fornaro
- Department of Neurosurgery, University Hospital “Maggiore Della Carità”, 28100 Novara, Italy
| | | | - Gian Paolo Caviglia
- Department of Medical Sciences, Division of Gastroenterology, University of Torino, 10126 Torino, Italy
| | - Demis Pitoni
- Department of Medical Sciences, Division of Gastroenterology, University of Torino, 10126 Torino, Italy
| | - Davide Giuseppe Ribaldone
- Department of Medical Sciences, Division of Gastroenterology, University of Torino, 10126 Torino, Italy
| |
Collapse
|
6
|
Koshko L, Scofield S, Mor G, Sadagurski M. Prenatal Pollutant Exposures and Hypothalamic Development: Early Life Disruption of Metabolic Programming. Front Endocrinol (Lausanne) 2022; 13:938094. [PMID: 35909533 PMCID: PMC9327615 DOI: 10.3389/fendo.2022.938094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Environmental contaminants in ambient air pollution pose a serious risk to long-term metabolic health. Strong evidence shows that prenatal exposure to pollutants can significantly increase the risk of Type II Diabetes (T2DM) in children and all ethnicities, even without the prevalence of obesity. The central nervous system (CNS) is critical in regulating whole-body metabolism. Within the CNS, the hypothalamus lies at the intersection of the neuroendocrine and autonomic systems and is primarily responsible for the regulation of energy homeostasis and satiety signals. The hypothalamus is particularly sensitive to insults during early neurodevelopmental periods and may be susceptible to alterations in the formation of neural metabolic circuitry. Although the precise molecular mechanism is not yet defined, alterations in hypothalamic developmental circuits may represent a leading cause of impaired metabolic programming. In this review, we present the current knowledge on the links between prenatal pollutant exposure and the hypothalamic programming of metabolism.
Collapse
Affiliation(s)
- Lisa Koshko
- Integrative Biosciences Center, Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Sydney Scofield
- Integrative Biosciences Center, Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology School of Medicine, Wayne State University, Detroit, MI, United States
| | - Marianna Sadagurski
- Integrative Biosciences Center, Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| |
Collapse
|
7
|
Bourhy L, Mazeraud A, Bozza FA, Turc G, Lledo PM, Sharshar T. Neuro-Inflammatory Response and Brain-Peripheral Crosstalk in Sepsis and Stroke. Front Immunol 2022; 13:834649. [PMID: 35464410 PMCID: PMC9022190 DOI: 10.3389/fimmu.2022.834649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/07/2022] [Indexed: 12/18/2022] Open
Abstract
Despite recent therapeutic advances, ischemic stroke is still a leading cause of death and disability. There is renewed attention on peripheral inflammatory signaling as a way of modulating the post-ischemic neuro-inflammatory process. The immune-brain crosstalk has long been the focus for understanding the mechanisms of sickness behavior, which is an adaptive autonomic, neuroendocrine, and behavioral response to a peripheral inflammation. It is mediated by humoral and neural pathways that mainly involve the circumventricular organs and vagal nerve, respectively. In this review we address the question of how sepsis and stroke can dysregulate this adaptive response, notably by impairing the central integration of peripheral signaling, but also by efferent control of the immune response. We highlight the potential role of gut-brain and brain-spleen signaling in stroke.
Collapse
Affiliation(s)
- Lena Bourhy
- Institut Pasteur, Université de Paris, Centre National de Recherche Scientifique, Unité Mixte de Recherche (CNRS UMR) 3571, Perception and Memory Unit, Paris, France
| | - Aurélien Mazeraud
- Institut Pasteur, Université de Paris, Centre National de Recherche Scientifique, Unité Mixte de Recherche (CNRS UMR) 3571, Perception and Memory Unit, Paris, France
- Neuro-Anesthesiology and Intensive Care Medicine, Groupe Hospitalier Universitaire (GHU) Paris Psychiatrie et Neurosciences, Université de Paris, Paris, France
| | - Fernando A. Bozza
- National Institute of Infectious Disease Evandro Chagas (INI), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Guillaume Turc
- Department of Neurology, GHU Paris Psychiatrie et Neurosciences, Université de Paris, Paris, France
| | - Pierre-Marie Lledo
- Institut Pasteur, Université de Paris, Centre National de Recherche Scientifique, Unité Mixte de Recherche (CNRS UMR) 3571, Perception and Memory Unit, Paris, France
| | - Tarek Sharshar
- Neuro-Anesthesiology and Intensive Care Medicine, Groupe Hospitalier Universitaire (GHU) Paris Psychiatrie et Neurosciences, Université de Paris, Paris, France
| |
Collapse
|
8
|
Kodali MC, Chen H, Liao FF. Temporal unsnarling of brain's acute neuroinflammatory transcriptional profiles reveals panendothelitis as the earliest event preceding microgliosis. Mol Psychiatry 2021; 26:3905-3919. [PMID: 33293688 PMCID: PMC7722246 DOI: 10.1038/s41380-020-00955-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is an acutely progressing brain dysfunction induced by systemic inflammation. The mechanism of initiation of neuroinflammation during SAE, which ultimately leads to delirium and cognitive dysfunction, remains elusive. We aimed to study the molecular events of SAE to capture its onset and progression into the central nervous system (CNS), and further identify the cellular players involved in mediating acute inflammatory signaling. Gene expression profiling on the cerebral vessels isolated from the brains of the mice treated with peripheral lipopolysaccharide (LPS) revealed that the cerebral vasculature responds within minutes to acute systemic inflammation by upregulating the expression of immediate early response genes, followed by activation of the nuclear factor-κB pathway. To identify the earliest responding cell type, we used fluorescence-activated cell sorting (FACS) to sort the glial and vascular cells from the brains of the mice treated with LPS at different time points, and RNA-seq was performed on microglia and cerebral endothelial cells (CECs). Bioinformatic analysis followed by further validation in all the cell types revealed that panendothelitis. i.e., the activation of CECs is the earliest event in the CNS during the inception of acute neuroinflammation. Microglial activation occurs later than that of CECs, suggesting that CECs are the most likely initial source of proinflammatory mediators, which could further initiate glial cell activation. This is then followed by the activation of apoptotic signaling in the CECs, which is known to lead to the blood-brain barrier disruption and allow peripheral cytokines to leak into the CNS, exacerbate the gliosis, and result in the vicious neuroinflammatory cascade. Together, our results model the earliest sequential events during the advancement of systemic inflammation into the CNS and facilitate to understand the interplay between the vascular and glial cells in initiating and driving acute neuroinflammation during SAE.
Collapse
Affiliation(s)
- Mahesh Chandra Kodali
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
- Integrated Biomedical Sciences Program, Molecular and Systems Pharmacology Track, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | - Hao Chen
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Francesca-Fang Liao
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
| |
Collapse
|
9
|
Wang FF, Ba J, Yu XJ, Shi XL, Liu JJ, Liu KL, Fu LY, Su Q, Li HB, Kang KB, Yi QY, Wang SQ, Gao HL, Qi J, Li Y, Zhu GQ, Kang YM. Central Blockade of E-Prostanoid 3 Receptor Ameliorated Hypertension Partially by Attenuating Oxidative Stress and Inflammation in the Hypothalamic Paraventricular Nucleus of Spontaneously Hypertensive Rats. Cardiovasc Toxicol 2021; 21:286-300. [PMID: 33165770 DOI: 10.1007/s12012-020-09619-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 10/24/2020] [Indexed: 12/27/2022]
Abstract
Hypertension, as one of the major risk factors for cardiovascular disease, significantly affects human health. Prostaglandin E2 (PGE2) and the E3-class prostanoid (EP3) receptor have previously been demonstrated to modulate blood pressure and hemodynamics in various animal models of hypertension. The PGE2-evoked pressor and biochemical responses can be blocked with the EP3 receptor antagonist, L-798106 (N-[(5-bromo-2methoxyphenyl)sulfonyl]-3-[2-(2-naphthalenylmethyl) phenyl]-2-propenamide). In the hypothalamic paraventricular nucleus (PVN), sympathetic excitation can be introduced by PGE2, which can activate EP3 receptors located in the PVN. In such a case, the central knockdown of EP3 receptor can be considered as a potential therapeutic modality for hypertension management. The present study examined the efficacy of the PVN infusion of L-798106, by performing experiments on spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto rats (WKYs). The rats were administered with chronic bilateral PVN infusion of L-798106 (10 μg/day) or the vehicle for 28 days. The results indicated that the SHRs had a higher mean arterial pressure (MAP), an increased Fra-like (Fra-LI) activity in the PVN, as well as a higher expression of gp91phox, mitogen-activated protein kinase (MAPK), and proinflammatory cytokines in the PVN compared with the WKYs. Additionally, the expression of Cu/Zn-SOD in the PVN of the SHRs was reduced compared with the WKYs. The bilateral PVN infusion of L-798106 significantly reduced MAP, as well as plasma norepinephrine (NE) levels in the SHRs. It also inhibited Fra-LI activity and reduced the expression of gp91phox, proinflammatory cytokines, and MAPK, whereas it increased the expression of Cu/Zn-SOD in the PVN of SHRs. In addition, L-798106 restored the balance of the neurotransmitters in the PVN. On the whole, the findings of the present study demonstrate that the PVN blockade of EP3 receptor can ameliorate hypertension and cardiac hypertrophy partially by attenuating ROS and proinflammatory cytokines, and modulating neurotransmitters in the PVN.
Collapse
Affiliation(s)
- Fang-Fang Wang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Functional Medicine, School of Basic Medical Sciences, Jiamusi University, Jiamusi, 154007, China
| | - Juan Ba
- Department of Anesthesiology, Center for Brian Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiao-Jing Yu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiao-Lian Shi
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jin-Jun Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Kai-Li Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Li-Yan Fu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qing Su
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Kai B Kang
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Qiu-Yue Yi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shu-Qiu Wang
- Department of Functional Medicine, School of Basic Medical Sciences, Jiamusi University, Jiamusi, 154007, China
| | - Hong-Li Gao
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jie Qi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ying Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Guo-Qing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
10
|
Bonaz B, Sinniger V, Pellissier S. Therapeutic Potential of Vagus Nerve Stimulation for Inflammatory Bowel Diseases. Front Neurosci 2021; 15:650971. [PMID: 33828455 PMCID: PMC8019822 DOI: 10.3389/fnins.2021.650971] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
The vagus nerve is a mixed nerve, comprising 80% afferent fibers and 20% efferent fibers. It allows a bidirectional communication between the central nervous system and the digestive tract. It has a dual anti-inflammatory properties via activation of the hypothalamic pituitary adrenal axis, by its afferents, but also through a vago-vagal inflammatory reflex involving an afferent (vagal) and an efferent (vagal) arm, called the cholinergic anti-inflammatory pathway. Indeed, the release of acetylcholine at the end of its efferent fibers is able to inhibit the release of tumor necrosis factor (TNF) alpha by macrophages via an interneuron of the enteric nervous system synapsing between the efferent vagal endings and the macrophages and releasing acetylcholine. The vagus nerve also synapses with the splenic sympathetic nerve to inhibit the release of TNF-alpha by splenic macrophages. It can also activate the spinal sympathetic system after central integration of its afferents. This anti-TNF-alpha effect of the vagus nerve can be used in the treatment of chronic inflammatory bowel diseases, represented by Crohn’s disease and ulcerative colitis where this cytokine plays a key role. Bioelectronic medicine, via vagus nerve stimulation, may have an interest in this non-drug therapeutic approach as an alternative to conventional anti-TNF-alpha drugs, which are not devoid of side effects feared by patients.
Collapse
Affiliation(s)
- Bruno Bonaz
- Division of Hepato-Gastroenterology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France.,Grenoble Institute of Neurosciences, Inserm U1216, University Grenoble Alpes, Grenoble, France
| | - Valérie Sinniger
- Division of Hepato-Gastroenterology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France.,Grenoble Institute of Neurosciences, Inserm U1216, University Grenoble Alpes, Grenoble, France
| | - Sonia Pellissier
- Laboratoire Inter-Universitaire de Psychologie Personnalité, Cognition, Changement Social, University Grenoble Alpes, University Savoie Mont Blanc, Grenoble, France
| |
Collapse
|
11
|
Lorenz D, Maurer F, Philipp D, Albrecht F, Hüppe T, Sessler DI, Wolf B, Volk T, Kreuer S, Fink T. Changes in volatile organic compounds provoked by lipopolysaccharide- or alpha toxin-induced inflammation in ventilated rats. J Breath Res 2020; 15:016003. [PMID: 33103661 DOI: 10.1088/1752-7163/abb449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Inflammation may alter volatile organic compounds (VOCs) in exhaled breath. We therefore used ion mobility spectrometry (IMS) to evaluate exhaled breath components in two non-infectious inflammatory models. Fifty male Sprague Dawley rats were anesthetized and ventilated for 24 h. Five treatments were randomly assigned: (1) lipopolysaccharide low dose [5 mg/kg]; (2) lipopolysaccharide high dose [10 mg/kg]; (3) alpha toxin low dose [40 µg/kg]; (4) alpha toxin high dose [80 µg/kg]; and, (5) NaCl 0.9% as control group. Gas was sampled from the expiratory line of the ventilator every 20 min and analyzed with IMS combined with a multi-capillary column. VOCs were identified by comparison with an established database. Survival analysis was performed by log-rank test, other analyses by one-way or paired ANOVA-tests and post-hoc analysis according to Holm-Sidak. Rats given NaCl and low-dose alpha toxin survived 24 h. The median survival time in alpha toxin high-dose group was 23 (95%-confidence interval (CI): 21, 24) h. In contrast, the median survival time in rats given high-dose lipopolysaccharide was 12 (95% CI: 9, 14) and only 13 (95% CI: 10, 16) h in those given high-dose lipopolysaccharide. 73 different VOCs were detected, of which 35 were observed only in the rats, 38 could be found both in the blank measurements of ventilator air and in the exhaled air of the rats. Forty-nine of the VOCs were identifiable from a registry of compounds. Exhaled volatile compounds were comparable in each group before injection of lipopolysaccharide and alpha toxin. In the LPS groups, 1-pentanol increased and 2-propanol decreased. After alpha toxin treatment, 1-butanol and 1-pentanol increased whereas butanal and isopropylamine decreased. Induction of a non-infectious systemic inflammation (niSI) by lipopolysaccharide and alpha toxin changes VOCs in exhaled breath. Exhalome analysis may help identify niSI.
Collapse
Affiliation(s)
- Dominik Lorenz
- CBR - Center of Breath Research, Department of Anaesthesiology, Intensive Care and Pain Therapy, Saarland University Medical Center and Saarland University Faculty of Medicine, Building 57, 66421, Homburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Sayd A, Vargas-Caraveo A, Perea-Romero I, Robledo-Montaña J, Caso JR, Madrigal JLM, Leza JC, Orio L, Garcia-Bueno B. Depletion of brain perivascular macrophages regulates acute restraint stress-induced neuroinflammation and oxidative/nitrosative stress in rat frontal cortex. Eur Neuropsychopharmacol 2020; 34:50-64. [PMID: 32245674 DOI: 10.1016/j.euroneuro.2020.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 01/30/2020] [Accepted: 03/06/2020] [Indexed: 12/30/2022]
Abstract
The central nervous system can respond to peripheral immune stimuli through the activation of the neurovascular unit. One of the cellular types implicated are perivascular macrophages (PVMs), hematopoietic-derived brain-resident cells located in the perivascular space. PVMs have been implicated in the immune surveillance and in the regulation of the accumulation/trafficking of macromolecules in brain-blood interfaces. Recent studies suggested that the role of PVMs could vary depending on the nature and duration of the immune challenge applied. Here, we investigate the role of PVMs in stress-induced neuroinflammation and oxidative/nitrosative consequences. The basal phagocytic activity of PVMs was exploited to selectively deplete them by ICV injection of liposomes encapsulating the pro-apoptotic drug clodronate. Acute restraint stress-induced neuroinflammation and oxidative/nitrosative stress in rat brain frontal cortex samples were assessed by western blot and RT-PCR analyses. The depletion of PVMs: (1) decreased tumor necrosis-α levels (2) prevented the Janus kinase/signal transducers and activators of transcription pathway and increased interleukin-6 receptor protein-expression in stress conditions; (3) prevented the stress-induced Toll-like receptor 4/Myeloid differentiation primary response 88 protein signaling pathway; (4) down-regulated the pro-inflammatory nuclear factor κB/cyclooxygenase-2 pathway; (5) prevented stress-induced lipid peroxidation and the concomitant increase of the endogenous antioxidant mediators nuclear factor (erythroid-derived 2)-like 2, glutathione reductase 1 and Parkinsonism-associated deglycase mRNA expression. Our results point to PVMs as regulators of stress-induced neuroinflammation and oxidative/nitrosative stress. Much more scientific effort is still needed to evaluate whether their selective manipulation is promising as a therapeutic strategy for the treatment of stress-related neuropsychopathologies.
Collapse
Affiliation(s)
- Aline Sayd
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, Madrid 28040, Spain
| | - Alejandra Vargas-Caraveo
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, Madrid 28040, Spain; Campus Lerma, Biological and Health Sciences Division, Metropolitan Autonomous University (UAM), Lerma 52005, Mexico
| | - Irene Perea-Romero
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, Madrid 28040, Spain
| | - Javier Robledo-Montaña
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, Madrid 28040, Spain
| | - Javier R Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, Madrid 28040, Spain
| | - Jose L M Madrigal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, Madrid 28040, Spain
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, Madrid 28040, Spain
| | - Laura Orio
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, Red de Trastornos Adictivos (RTA) del Instituto de Salud Carlos III (ISCIII), Spain
| | - Borja Garcia-Bueno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, Madrid 28040, Spain.
| |
Collapse
|
13
|
Hao WZ, Li XJ, Zhang PW, Chen JX. A review of antibiotics, depression, and the gut microbiome. Psychiatry Res 2020; 284:112691. [PMID: 31791704 DOI: 10.1016/j.psychres.2019.112691] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 12/16/2022]
Abstract
Emerging evidence indicates that disruption of the intestinal flora play an important role in the pathogenesis of depression. As one of the causes of such disturbances, the role of antibiotics in depression risk is gradually being revealed. Herein, we review recent findings showing that the use of both single and multiple antibiotic regimens may be related to depression by changing the gut microbiota and the brain-gut axis. Based on recent discoveries, we also suggest that several brain-gut interactive mechanisms (particularly those involving nerve and glial cells, neurotransmitters, brain neurotrophic factors, inflammatory factors, short-chain fatty acids, circulating metabolites, blood-brain barrier, and oxidative stress) may help understand the effects of antibiotics on intestinal flora and its relationship with depression.
Collapse
Affiliation(s)
- Wen-Zhi Hao
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Xiao-Juan Li
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Pei-Wen Zhang
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Jia-Xu Chen
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
14
|
Zenz G, Farzi A, Fröhlich EE, Reichmann F, Holzer P. Intranasal Neuropeptide Y Blunts Lipopolysaccharide-Evoked Sickness Behavior but Not the Immune Response in Mice. Neurotherapeutics 2019; 16:1335-1349. [PMID: 31338703 PMCID: PMC6985076 DOI: 10.1007/s13311-019-00758-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Neuropeptide Y (NPY) has been demonstrated to exert stress buffering effects and promote resilience. Non-invasive intranasal (IN) application of NPY to rodents is able to mitigate traumatic stress-induced behavioral changes as well as dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis. However, it is unknown whether IN NPY could prevent the behavioral, pro-inflammatory and neurochemical responses to peripheral immune activation by the Toll-like receptor 4 (TLR4) stimulant lipopolysaccharide (LPS). Therefore, we analyzed the effects of IN NPY (100 μg) on the behavioral sickness response (reduced locomotion and exploration) and the underlying molecular mechanisms, 3 h and 21 h after intraperitoneal injections of LPS (0.03 mg/kg) in male C57BL/6N mice. The acute behavioral sickness response was significantly dampened by pretreatment with IN NPY 3 h after LPS injection. This effect was accompanied by diminished weight loss and lowered plasma corticosterone (CORT) levels 21 h after LPS injection. In contrast, acute circulating cytokine levels and hypothalamic cytokine mRNA expression remained unaltered by IN NPY, which indicates that the peripheral and cerebral immune response to LPS was left undisturbed. Our findings are in agreement with the reported activity of NPY to dampen the response of the HPA axis to stress. We propose that IN NPY ablates sickness behavior at a site beyond the peripheral and cerebral cytokine response, an action that is associated with reduced activity of the HPA axis as determined by decreased plasma CORT.These results indicate that IN NPY administration may be relevant to the management of neuropsychiatric disorders arising from immune-induced neuroendocrine dysfunction.
Collapse
Affiliation(s)
- Geraldine Zenz
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria.
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria
| | - Esther E Fröhlich
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria
- BioTechMed-Graz, Mozartgasse 12, A-8010, Graz, Austria
| |
Collapse
|
15
|
Bonaz B, Sinniger V, Pellissier S. Vagus Nerve Stimulation at the Interface of Brain-Gut Interactions. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a034199. [PMID: 30201788 DOI: 10.1101/cshperspect.a034199] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The vagus nerve, a key component of the cross-communication between the gut and the brain, is a major element of homeostasis sensing the "milieu intérieur" and boosting the nervous and endocrine responses to maintain the gastrointestinal health status. This nerve has anti-inflammatory properties regulating the gut through the activation of the hypothalamic-pituitary-adrenal axis and the release of cortisol and through a vagovagal reflex, which has an anti-tumor necrosis factor (TNF) effect called the cholinergic anti-inflammatory pathway. Stimulating this nerve is an interesting tool as a nondrug therapy for the treatment of gastrointestinal diseases in which brain-gut communication is dysfunctional, such as inflammatory bowel disorders and others. This review presents the rationale of vagal gastrointestinal physiology and diseases and the most recent advances in vagus nerve stimulation. It also highlights the main issues to be addressed in the future to improve this bioelectronic therapy for gastrointestinal disorders.
Collapse
Affiliation(s)
- Bruno Bonaz
- Division of Hepato-Gastroenterology, Grenoble University Hospital, 38043 Grenoble Cedex 09, France.,U1216, INSERM, GIN, Grenoble Institute of Neurosciences, University Grenoble Alpes, Grenoble, France
| | - Valérie Sinniger
- Division of Hepato-Gastroenterology, Grenoble University Hospital, 38043 Grenoble Cedex 09, France.,U1216, INSERM, GIN, Grenoble Institute of Neurosciences, University Grenoble Alpes, Grenoble, France
| | - Sonia Pellissier
- University Grenoble Alpes, University Savoie Mont Blanc, 38000 Grenoble, France
| |
Collapse
|
16
|
Liu YW, Zhao L, Zhou M, Wang H, Yang N, Dai SS. Transplantation with mGluR5 deficiency bone marrow displays antidepressant-like effect in C57BL/6J mice. Brain Behav Immun 2019; 79:114-124. [PMID: 30682501 DOI: 10.1016/j.bbi.2019.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/17/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
Antidepressant-like effects of metabotropic glutamate receptor 5 (mGluR5) have been verified by specific antagonists or whole body knock-out (KO) mice. Previous experiments indicate that blocking mGluR5 exerts antidepressant-like effects through neuronal mechanisms, like modulating NMDA receptor activity or 5-HT system. Here we found that transplanting bone marrow from mGluR5 KO mice to WT mice could also show antidepressant-like effects, which were confirmed by sucrose preference test and tail suspension test. Furthermore, mGluR5 deficiency dramatically inhibits cytokines release from bone marrow cells, such as IL-1β, TNF-α and IL-6, alleviating proinflammatory responses in LPS-induced depression model. In addition, inhibited cytokines could decrease the activation of brain endothelial cells in ERK-dependent manner. These data provide the evidence that blocking mGluR5 could improve depression through inhibiting peripheral immune responses, confirming the causal relationship between peripheral immune phenotype and brain behavior.
Collapse
Affiliation(s)
- Yang-Wuyue Liu
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, PR China
| | - Li Zhao
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, PR China
| | - Mi Zhou
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, PR China
| | - Hao Wang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Nan Yang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Shuang-Shuang Dai
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, PR China; Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China.
| |
Collapse
|
17
|
Barrientos RM, Brunton PJ, Lenz KM, Pyter L, Spencer SJ. Neuroimmunology of the female brain across the lifespan: Plasticity to psychopathology. Brain Behav Immun 2019; 79:39-55. [PMID: 30872093 PMCID: PMC6591071 DOI: 10.1016/j.bbi.2019.03.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/25/2019] [Accepted: 03/09/2019] [Indexed: 02/06/2023] Open
Abstract
The female brain is highly dynamic and can fundamentally remodel throughout the normal ovarian cycle as well as in critical life stages including perinatal development, pregnancy and old-age. As such, females are particularly vulnerable to infections, psychological disorders, certain cancers, and cognitive impairments. We will present the latest evidence on the female brain; how it develops through the neonatal period; how it changes through the ovarian cycle in normal individuals; how it adapts to pregnancy and postpartum; how it responds to illness and disease, particularly cancer; and, finally, how it is shaped by old age. Throughout, we will highlight female vulnerability to and resilience against disease and dysfunction in the face of environmental challenges.
Collapse
Affiliation(s)
- R M Barrientos
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH 43210, United States
| | - P J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, Scotland, UK; Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University School of Medicine, International Campus, Haining, Zhejiang 314400, PR China
| | - K M Lenz
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychology, Department of Neuroscience, The Ohio State University, Columbus, OH 43210, United States
| | - L Pyter
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States
| | - S J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic. 3083, Australia.
| |
Collapse
|
18
|
Macedo F, Dos Santos LS, Glezer I, da Cunha FM. Brain Innate Immune Response in Diet-Induced Obesity as a Paradigm for Metabolic Influence on Inflammatory Signaling. Front Neurosci 2019; 13:342. [PMID: 31068773 PMCID: PMC6491681 DOI: 10.3389/fnins.2019.00342] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/25/2019] [Indexed: 12/19/2022] Open
Abstract
Obesity is a predisposing factor for numerous morbidities, including those affecting the central nervous system. Hypothalamic inflammation is a hallmark of obesity and is believed to participate in the onset and progression of the obese phenotype, by promoting changes in neuronal functions involved in the control of metabolism. The activation of brain immune cells in the hypothalamus, which are represented by microglia and brain macrophages, is associated with obesity and has been the focus of intense research. Despite the significant body of knowledge gathered on this topic, obesity-induced metabolic changes in brain cells involved in innate immune responses are still poorly characterized due, at least in part, to limitations in the existing experimental methods. Since the metabolic state influences immune responses of microglia and other myeloid cells, the understanding and characterization of the effects of cellular metabolism on the functions of these cells, and their impact on brain integrity, are crucial for the development of efficient therapeutic interventions for individuals exposed to a long-term high fat diet (HFD). Here we review and speculate on the cellular basis that may underlie the observed changes in the reactivity and metabolism of the innate immune cells of the brain in diet-induced obesity (DIO), and discuss important points that deserve further investigation.
Collapse
Affiliation(s)
- Felipe Macedo
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lucas Souza Dos Santos
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Isaias Glezer
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda Marques da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Russell JA, Brunton PJ. Giving a good start to a new life via maternal brain allostatic adaptations in pregnancy. Front Neuroendocrinol 2019; 53:100739. [PMID: 30802468 DOI: 10.1016/j.yfrne.2019.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/29/2019] [Accepted: 02/21/2019] [Indexed: 12/23/2022]
Abstract
Successful pregnancy requires adjustments to multiple maternal homeostatic mechanisms, governed by the maternal brain to support and enable survival of the growing fetus and placenta. Such adjustments fit the concept of allostasis (stability through change) and have a cost: allostatic load. Allostasis is driven by ovarian, anterior pituitary, placental and feto-placental hormones acting on the maternal brain to promote adaptations that support the pregnancy and protect the fetus. Many women carry an existing allostatic load into pregnancy, from socio-economic circumstances, poor mental health and in 'developed' countries, also from obesity. These pregnancies have poorer outcomes indicating negative interactions (failing allostasis) between pre-pregnancy and pregnancy allostatic loads. Use of animal models, such as adult prenatally stressed female offspring with abnormal neuroendocrine, metabolic and behavioural phenotypes, to probe gene expression changes, and epigenetic mechanisms in the maternal brain in adverse pregnancies are discussed, with the prospect of ameliorating poor pregnancy outcomes.
Collapse
Affiliation(s)
- John A Russell
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Paula J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK; Zhejiang University-University of Edinburgh Institute, Haining, Zhejiang, PR China.
| |
Collapse
|
20
|
Feng SYS, Hollis JH, Samarasinghe T, Phillips DJ, Rao S, Yu VYH, Walker AM. Endotoxin-induced cerebral pathophysiology: differences between fetus and newborn. Physiol Rep 2019; 7:e13973. [PMID: 30785235 PMCID: PMC6381816 DOI: 10.14814/phy2.13973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023] Open
Abstract
As the comparative pathophysiology of perinatal infection in the fetus and newborn is uncertain, this study contrasted the cerebral effects of endotoxemia in conscious fetal sheep and newborn lambs. Responses to intravenous bacterial endotoxin (lipopolysaccharide, LPS) or normal saline were studied on three consecutive days in fetal sheep (LPS 1 μg/kg, n = 5; normal saline n = 5) and newborn lambs (LPS 2 μg/kg, n = 10; normal saline n = 5). Cerebro-vascular function was assessed by monitoring cerebral blood flow (CBF) and cerebral vascular resistance (CVR) over 12 h each day, and inflammatory responses were assessed by plasma TNF alpha (TNF-α), nitrate and nitrite concentrations. Brain injury was quantified by counting both resting and active macrophages in the caudate nucleus and periventricular white matter (PVWM). An acute cerebral vasoconstriction (within 1 h of LPS injection) occurred in both the fetus (ΔCVR +53%) and newborn (ΔCVR +63%); subsequently prolonged cerebral vasodilatation occurred in the fetus (ΔCVR -33%) in association with double plasma nitrate/nitrite concentrations, but not in the newborn. Abundant infiltration of activated macrophages was observed in both CN and PVWM at each age, with the extent being 2-3 times greater in the fetus (P < 0.001). In conclusion, while the fetus and newborn experience a similar acute disruption of the cerebral circulation after LPS, the fetus suffers a more prolonged circulatory disruption, a greater infiltration of activated macrophages, and an exaggerated susceptibility to brain injury.
Collapse
Affiliation(s)
- Susan Y. S. Feng
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Neonatal DirectorateKing Edward Memorial HospitalPerth Children's HospitalSubiacoWestern AustraliaAustralia
| | - Jacob H. Hollis
- Department of PhysiologyMonash UniversityClaytonVictoriaAustralia
| | | | - David J. Phillips
- Academic & Medical PortfolioEpworth HealthCareRichmondVictoriaAustralia
| | - Shripada Rao
- Neonatal DirectorateKing Edward Memorial HospitalPerth Children's HospitalSubiacoWestern AustraliaAustralia
| | - Victor Y. H. Yu
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Monash NewbornMonash Medical CentreClaytonVictoriaAustralia
| | - Adrian M. Walker
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
| |
Collapse
|
21
|
Huang S, Hu H, Cai YH, Hua F. Effect of parecoxib in the treatment of postoperative cognitive dysfunction: A systematic review and meta-analysis. Medicine (Baltimore) 2019; 98:e13812. [PMID: 30608392 PMCID: PMC6344118 DOI: 10.1097/md.0000000000013812] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Parecoxib is a selective cyclooxygenase (COX)-2 inhibitor widely used as an analgesia technique in perioperative period for its potent anti-inflammatory and analgesic effects. However, litter is known about its effect on postoperative cognitive dysfunction (POCD). The purpose of this meta-analysis of randomized controlled trials (RCTs) was to evaluate the effect of parecoxib in the treatment of postoperative cognitive dysfunction. METHODS We searched PubMed, Cochrane Library and Embase databases for relevant studies up to October 2017. We selected fixed-effect model for analysis of data heterogeneity. Statistical analyses were performed by using Review Manager Version 5.3 for Windows. RESULTS Four RCTs with 904 patients that underwent surgical operations were included. The meta-analysis demonstrated parecoxib could significantly decrease the incidence of POCD on postoperative day 1, day 3, day 5, and day 7 when compared with control treatment; IL-6 and S100β concentrations were lower up to postoperative day 2. The consumption of morphine, fentanyl and tramadol in parecoxib groups were lower than control groups. CONCLUSION Our meta-analysis suggested that the administration of Parecoxib was effective in treating early POCD within 7 days and reducing IL-6 and S100β concentrations within 2 days after operations. Nevertheless, our current study with some limitations such as the small sample size only provided limited quality of evidence, confirmation from further meta-analysis with large-scale, well-designed RCTs is required.
Collapse
Affiliation(s)
- Song Huang
- Anesthesia Department, The Second Affiliated Hospital of Nanchang University, Min De Road
| | - Haijun Hu
- Anesthesia Department, The Second Affiliated Hospital of Nanchang University, Min De Road
| | - Yue-Hong Cai
- Ophthalmology Department, Jiangxi Provincial People's Hospital, Nanchang, PR China
| | - Fuzhou Hua
- Anesthesia Department, The Second Affiliated Hospital of Nanchang University, Min De Road
| |
Collapse
|
22
|
Two hit induced acute lung injury impairs cognitive function in mice: A potential model to study cross talk between lung and brain. Brain Behav Immun 2018; 73:633-642. [PMID: 30026058 DOI: 10.1016/j.bbi.2018.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/06/2018] [Accepted: 07/14/2018] [Indexed: 01/12/2023] Open
Abstract
Acute lung injury (ALI), a pulmonary inflammatory disorder, is associated with high morbidity and mortality rates. Interestingly, ALI survivors have been reported for some neurocognitive deterioration at/after discharge. However, the molecular factors behind such extra pulmonary manifestation are not clearly known. The present work was designed to investigate lung-brain cross talk in experimental mice for deciphering primary molecular factors that may be involved in ALI-mediated cognitive impairment. ALI was induced in Balb/c mice by intra-tracheal administration of either 0.1 N HCl (2 ml/kg) or LPS (1 mg/kg) as single hits or both agents were administered successively to mimic the 'two hit' model. Interestingly two hit-mediated ALI resulted in exaggerated inflammatory response as reflected by increased pulmonary neutrophils and inflammatory factors (TNF-α/IL-1β/IL-6). Additionally, two hits resulted in delayed resolution of lung inflammation and was coupled with persistent decline in memory, as assessed by Morris water maze test. Further, two hits elevate serum levels of TNF-α/IL-1β which was associated with compromised blood brain barrier (BBB), as evident by decreased expression of occludin/claudin-5 and consequent Evans-blue extravasation in hippocampus 1 week post injury. Finally, dexamethasone protects against the two hit mediated cognitive impairment by lowering the pro-inflammatory factors (TNF-α/IL-1β) both in lungs and blood. Overall, we report for the first time that 'two hit' mediated ALI cause persistent cognitive impairment in mice partly via up-regulating systemic expression of TNF-α/IL-1β that may disrupt BBB and hence the model may be a useful tool to examine the lung-brain cross-talk at the molecular level for exploring newer therapeutics.
Collapse
|
23
|
Waclawiková B, El Aidy S. Role of Microbiota and Tryptophan Metabolites in the Remote Effect of Intestinal Inflammation on Brain and Depression. Pharmaceuticals (Basel) 2018; 11:ph11030063. [PMID: 29941795 PMCID: PMC6160932 DOI: 10.3390/ph11030063] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/15/2022] Open
Abstract
The human gastrointestinal tract is inhabited by trillions of commensal bacteria collectively known as the gut microbiota. Our recognition of the significance of the complex interaction between the microbiota, and its host has grown dramatically over the past years. A balanced microbial community is a key regulator of the immune response, and metabolism of dietary components, which in turn, modulates several brain processes impacting mood and behavior. Consequently, it is likely that disruptions within the composition of the microbiota would remotely affect the mental state of the host. Here, we discuss how intestinal bacteria and their metabolites can orchestrate gut-associated neuroimmune mechanisms that influence mood and behavior leading to depression. In particular, we focus on microbiota-triggered gut inflammation and its implications in shifting the tryptophan metabolism towards kynurenine biosynthesis while disrupting the serotonergic signaling. We further investigate the gaps to be bridged in this exciting field of research in order to clarify our understanding of the multifaceted crosstalk in the microbiota–gut–brain interphase, bringing about novel, microbiota-targeted therapeutics for mental illnesses.
Collapse
Affiliation(s)
- Barbora Waclawiková
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands.
| | - Sahar El Aidy
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands.
| |
Collapse
|
24
|
Liberman AC, Budziñski ML, Sokn C, Gobbini RP, Steininger A, Arzt E. Regulatory and Mechanistic Actions of Glucocorticoids on T and Inflammatory Cells. Front Endocrinol (Lausanne) 2018; 9:235. [PMID: 29867767 PMCID: PMC5964134 DOI: 10.3389/fendo.2018.00235] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/25/2018] [Indexed: 12/24/2022] Open
Abstract
Glucocorticoids (GCs) play an important role in regulating the inflammatory and immune response and have been used since decades to treat various inflammatory and autoimmune disorders. Fine-tuning the glucocorticoid receptor (GR) activity is instrumental in the search for novel therapeutic strategies aimed to reduce pathological signaling and restoring homeostasis. Despite the primary anti-inflammatory actions of GCs, there are studies suggesting that under certain conditions GCs may also exert pro-inflammatory responses. For these reasons the understanding of the GR basic mechanisms of action on different immune cells in the periphery (e.g., macrophages, dendritic cells, neutrophils, and T cells) and in the brain (microglia) contexts, that we review in this chapter, is a continuous matter of interest and may reveal novel therapeutic targets for the treatment of immune and inflammatory response.
Collapse
Affiliation(s)
- Ana C. Liberman
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Maia L. Budziñski
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Clara Sokn
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Romina Paula Gobbini
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Anja Steininger
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Eduardo Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- *Correspondence: Eduardo Arzt,
| |
Collapse
|
25
|
Senegenin exerts anti-depression effect in mice induced by chronic un-predictable mild stress via inhibition of NF-κB regulating NLRP3 signal pathway. Int Immunopharmacol 2017; 53:24-32. [DOI: 10.1016/j.intimp.2017.10.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/22/2017] [Accepted: 10/01/2017] [Indexed: 01/19/2023]
|
26
|
Holzer P, Farzi A, Hassan AM, Zenz G, Jačan A, Reichmann F. Visceral Inflammation and Immune Activation Stress the Brain. Front Immunol 2017; 8:1613. [PMID: 29213271 PMCID: PMC5702648 DOI: 10.3389/fimmu.2017.01613] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022] Open
Abstract
Stress refers to a dynamic process in which the homeostasis of an organism is challenged, the outcome depending on the type, severity, and duration of stressors involved, the stress responses triggered, and the stress resilience of the organism. Importantly, the relationship between stress and the immune system is bidirectional, as not only stressors have an impact on immune function, but alterations in immune function themselves can elicit stress responses. Such bidirectional interactions have been prominently identified to occur in the gastrointestinal tract in which there is a close cross-talk between the gut microbiota and the local immune system, governed by the permeability of the intestinal mucosa. External stressors disturb the homeostasis between microbiota and gut, these disturbances being signaled to the brain via multiple communication pathways constituting the gut-brain axis, ultimately eliciting stress responses and perturbations of brain function. In view of these relationships, the present article sets out to highlight some of the interactions between peripheral immune activation, especially in the visceral system, and brain function, behavior, and stress coping. These issues are exemplified by the way through which the intestinal microbiota as well as microbe-associated molecular patterns including lipopolysaccharide communicate with the immune system and brain, and the mechanisms whereby overt inflammation in the GI tract impacts on emotional-affective behavior, pain sensitivity, and stress coping. The interactions between the peripheral immune system and the brain take place along the gut-brain axis, the major communication pathways of which comprise microbial metabolites, gut hormones, immune mediators, and sensory neurons. Through these signaling systems, several transmitter and neuropeptide systems within the brain are altered under conditions of peripheral immune stress, enabling adaptive processes related to stress coping and resilience to take place. These aspects of the impact of immune stress on molecular and behavioral processes in the brain have a bearing on several disturbances of mental health and highlight novel opportunities of therapeutic intervention.
Collapse
Affiliation(s)
- Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Ahmed M Hassan
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Geraldine Zenz
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Angela Jačan
- CBmed GmbH-Center for Biomarker Research in Medicine, Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| |
Collapse
|
27
|
Murta V, Ferrari C. Peripheral Inflammation and Demyelinating Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 949:263-285. [PMID: 27714694 DOI: 10.1007/978-3-319-40764-7_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In recent decades, several neurodegenerative diseases have been shown to be exacerbated by systemic inflammatory processes. There is a wide range of literature that demonstrates a clear but complex relationship between the central nervous system (CNS) and the immunological system, both under naïve or pathological conditions. In diseased brains, peripheral inflammation can transform "primed" microglia into an "active" state, which can trigger stronger pathological responses. Demyelinating diseases are a group of neurodegenerative diseases characterized by inflammatory lesions associated with demyelination, which in turn induces axonal damage, neurodegeneration, and progressive loss of function. Among them, the most important are multiple sclerosis (MS) and neuromyelitis optica (NMO). In this review, we will analyze the effect of specific peripheral inflammatory stimuli in the progression of demyelinating diseases and discuss their animal models. In most cases, peripheral immune stimuli are exacerbating.
Collapse
Affiliation(s)
- Verónica Murta
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carina Ferrari
- Instituto de Ciencias Básicas y Medicina Experimental, Instituto Universitario del Hospital Italiano, Buenos Aires, Argentina.
| |
Collapse
|
28
|
Yu Y, Wei SG, Weiss RM, Felder RB. TNF-α receptor 1 knockdown in the subfornical organ ameliorates sympathetic excitation and cardiac hemodynamics in heart failure rats. Am J Physiol Heart Circ Physiol 2017; 313:H744-H756. [PMID: 28710070 DOI: 10.1152/ajpheart.00280.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/21/2017] [Accepted: 07/08/2017] [Indexed: 02/07/2023]
Abstract
In systolic heart failure (HF), circulating proinflammatory cytokines upregulate inflammation and renin-angiotensin system (RAS) activity in cardiovascular regions of the brain, contributing to sympathetic excitation and cardiac dysfunction. Important among these is the subfornical organ (SFO), a forebrain circumventricular organ that lacks an effective blood-brain barrier and senses circulating humors. We hypothesized that the tumor necrosis factor-α (TNF-α) receptor 1 (TNFR1) in the SFO contributes to sympathetic excitation and cardiac dysfunction in HF rats. Rats received SFO microinjections of a TNFR1 shRNA or a scrambled shRNA lentiviral vector carrying green fluorescent protein, or vehicle. One week later, some rats were euthanized to confirm the accuracy of the SFO microinjections and the transfection potential of the lentiviral vector. Other rats underwent coronary artery ligation (CL) to induce HF or a sham operation. Four weeks after CL, vehicle- and scrambled shRNA-treated HF rats had significant increases in TNFR1 mRNA and protein, NF-κB activity, and mRNA for inflammatory mediators, RAS components and c-Fos protein in the SFO and downstream in the hypothalamic paraventricular nucleus, along with increased plasma norepinephrine levels and impaired cardiac function, compared with vehicle-treated sham-operated rats. In HF rats treated with TNFR1 shRNA, TNFR1 was reduced in the SFO but not paraventricular nucleus, and the central and peripheral manifestations of HF were ameliorated. In sham-operated rats treated with TNFR1 shRNA, TNFR1 expression was also reduced in the SFO but there were no other effects. These results suggest a key role for TNFR1 in the SFO in the pathophysiology of systolic HF.NEW & NOTEWORTHY Activation of TNF-α receptor 1 in the subfornical organ (SFO) contributes to sympathetic excitation in heart failure rats by increasing inflammation and renin-angiotensin system activity in the SFO and downstream in the hypothalamic paraventricular nucleus. Cytokine receptors in the SFO may be a target for central intervention in cardiovascular conditions characterized by peripheral inflammation.
Collapse
Affiliation(s)
- Yang Yu
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Shun-Guang Wei
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Robert M Weiss
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Robert B Felder
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and .,Research Service, Veterans Affairs Medical Center, Iowa City, Iowa
| |
Collapse
|
29
|
Vore AS, Doremus-Fitzwater T, Gano A, Deak T. Adolescent Ethanol Exposure Leads to Stimulus-Specific Changes in Cytokine Reactivity and Hypothalamic-Pituitary-Adrenal Axis Sensitivity in Adulthood. Front Behav Neurosci 2017; 11:78. [PMID: 28522965 PMCID: PMC5415566 DOI: 10.3389/fnbeh.2017.00078] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/12/2017] [Indexed: 12/23/2022] Open
Abstract
Adolescent alcohol use comprises a significant public health concern and is often characterized by binge-like consumption patterns. While ethanol exposure in adulthood has been shown to alter the stress response, including the Hypothalamic–Pituitary–Adrenal (HPA) axis, few studies have examined whether binge-like ethanol exposure during adolescence results in enduring changes in HPA axis sensitivity in adulthood. In the present studies, adolescent Sprague-Dawley rats were given intragastric (i.g.) intubations of ethanol (4 g/kg) or vehicle once per day for three consecutive days, beginning on postnatal day (P) 30 (±1). This exposure was followed by a 2-day period of rest/withdrawal. Rats received a total of either two (Experiments 1, 2 and 3) or four (Experiment 4) cycles of ethanol exposure and were subsequently allowed to age normally until adulthood. In Experiment 1, adult, (P71–75), ethanol- or vehicle-exposed rats received a 60 min restraint stress challenge. In Experiment 2, rats received a 50 μg/kg injection of lipopolysaccharide (LPS). In Experiment 3, rats received a challenge of 2.5 g/kg ethanol (intraperitoneally; i.p.). In Experiment 4, male and female ethanol- or vehicle- exposed rats received a 50 μg/kg injection of LPS. In all experiments, blood samples were collected for later assessment of corticosterone (CORT), blood ethanol concentrations (BECs), and the cellular fraction of blood was analyzed for cytokine gene expression. As expected, all three challenges led to a time-dependent surge in CORT. Gene expression analyses of cytokines (Interleukin [IL]-6, IL-1β, and Tumor necrosis factor alpha [TNFα]) from the cellular fraction of blood revealed unique, time-dependent patterns of cytokine expression depending upon the nature of the adult challenge incurred (restraint, LPS, or EtOH). Importantly, adolescent ethanol exposure led to attenuated restraint and LPS-induced cytokine expression in males, whereas female rats displayed an absence of cytokine alterations, and a tendency toward heightened HPA axis reactivity. These findings suggest that adolescent ethanol exposure may cause lasting alterations in cytokine regulation and HPA axis sensitivity that (a) persist into adulthood; (b) may vary depending on the nature of the challenge incurred during adulthood; and that (c) are sex-specific.
Collapse
Affiliation(s)
- Andrew S Vore
- Behavioral Neuroscience Program, Department of Psychology, Binghamton UniversityBinghamton, NY, USA
| | | | - Anny Gano
- Behavioral Neuroscience Program, Department of Psychology, Binghamton UniversityBinghamton, NY, USA
| | - Terrence Deak
- Behavioral Neuroscience Program, Department of Psychology, Binghamton UniversityBinghamton, NY, USA
| |
Collapse
|
30
|
D'Mello C, Swain MG. Immune-to-Brain Communication Pathways in Inflammation-Associated Sickness and Depression. Curr Top Behav Neurosci 2017; 31:73-94. [PMID: 27677781 DOI: 10.1007/7854_2016_37] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
A growing body of evidence now highlights a key role for inflammation in mediating sickness behaviors and depression. Systemic inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, and chronic liver disease have high comorbidity with depression. How the periphery communicates with the brain to mediate changes in neurotransmission and thereby behavior is not completely understood. Traditional routes of communication between the periphery and the brain involve neural and humoral pathways with TNFα, IL-1β, and IL-6 being the three main cytokines that have primarily been implicated in mediating signaling via these pathways. However, in recent years communication via peripheral immune-cell-to-brain and the gut-microbiota-to-brain routes have received increasing attention for their ability to modulate brain function. In this chapter we discuss periphery-to-brain communication pathways and their potential role in mediating inflammation-associated sickness behaviors and depression.
Collapse
Affiliation(s)
- Charlotte D'Mello
- Immunology Research Group, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, 3280 Hospital Dr. NW, Calgary, AB, Canada, T2N 4N1
| | - Mark G Swain
- Immunology Research Group, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, 3280 Hospital Dr. NW, Calgary, AB, Canada, T2N 4N1.
| |
Collapse
|
31
|
Calderón-Garcidueñas L, Maronpot RR, Torres-Jardon R, Henríquez-Roldán C, Schoonhoven R, Acuña-Ayala H, Villarreal-Calderón A, Nakamura J, Fernando R, Reed W, Azzarelli B, Swenberg JA. DNA Damage in Nasal and Brain Tissues of Canines Exposed to Air Pollutants Is Associated with Evidence of Chronic Brain Inflammation and Neurodegeneration. Toxicol Pathol 2016; 31:524-38. [PMID: 14692621 DOI: 10.1080/01926230390226645] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acute, subchronic, or chronic exposures to particulate matter (PM) and pollutant gases affect people in urban areas and those exposed to fires, disasters, and wars. Respiratory tract inflammation, production of mediators of inflammation capable of reaching the brain, systemic circulation of PM, and disruption of the nasal respiratory and olfactory barriers are likely in these populations. DNA damage is crucial in aging and in age-associated diseases such as Alzheimer's disease. We evaluated apurinic/apyrimidinic (AP) sites in nasal and brain genomic DNA, and explored by immunohistochemistry the expression of nuclear factor NF κB p65, inducible nitric oxide synthase (iNOS), cyclo-oxygenase 2 (COX2), metallothionein I and II, apolipoprotein E, amyloid precursor protein (APP), and beta-amyloid1-42 in healthy dogs naturally exposed to urban pollution in Mexico City. Nickel (Ni) and vanadium (V) were measured by inductively coupled plasma mass spectrometry (ICP-MS). Forty mongrel dogs, ages 7 days—10 years were studied (14 controls from Tlaxcala and 26 exposed to urban pollution in South West Metropolitan Mexico City (SWMMC)). Nasal respiratory and olfactory epithelium were found to be early pollutant targets. Olfactory bulb and hippocampal AP sites were significantly higher in exposed than in control age matched animals. Ni and V were present in a gradient from olfactory mucosa > olfactory bulb > frontal cortex. Exposed dogs had (a) nuclear neuronal NF κB p65, (b) endothelial, glial and neuronal iNOS, (c) endothelial and glial COX2, (d) ApoE in neuronal, glial and vascular cells, and (e) APP and β amyloid1-42 in neurons, diffuse plaques (the earliest at age 11 months), and in subarachnoid blood vessels. Increased AP sites and the inflammatory and stress protein brain responses were early and significant in dogs exposed to urban pollution. Oil combustion PM-associated metals Ni and V were detected in the brain. There was an acceleration of Alzheimer's-type pathology in dogs chronically exposed to air pollutants. Respiratory tract inflammation and deteriorating olfactory and respiratory barriers may play a role in the observed neuropathology. These data suggest that Alzheimer's disease may be the sequela of air pollutant exposures and the resulting systemic inflammation.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- Environmental Pathology Program, University of North Carolina at Chapel Hill, North Carolina 27599-7310, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
OBJECTIVES The objectives of this review are to discuss the pathophysiology of the pro-inflammatory response to the cardiopulmonary bypass circuit, the impact of ischemia reperfusion injury on post-operative organ function, the compensatory anti-inflammatory response and the evidence for immune-modulatory strategies and their impact on outcomes. DATA SOURCE MEDLINE, PubMed. CONCLUSION Innovations such as the development of more biocompatible surfaces and miniaturized circuits, as well as the increasing expertise of surgeons, anesthesiologists and perfusionists has transformed cardiac surgery and the use of cardiopulmonary bypass into a relatively routine procedure with favorable outcomes. Despite these refinements, the attendant inflammatory response to bypass, ischemia reperfusion injury and the compensatory anti-inflammatory response contribute to post-operative morbidity and mortality. Additional studies are needed to further delineate the impact of immunomodulatory strategies on outcomes.
Collapse
|
33
|
Lu J, Shao RH, Hu L, Tu Y, Guo JY. Potential antiinflammatory effects of acupuncture in a chronic stress model of depression in rats. Neurosci Lett 2016; 618:31-38. [PMID: 26921452 DOI: 10.1016/j.neulet.2016.02.040] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 10/18/2015] [Accepted: 02/22/2016] [Indexed: 02/05/2023]
Abstract
Accumulating evidence indicates that inflammation may contribute to the pathophysiology of depression. Acupuncture in traditional Chinese medicine has been considered an effective treatment for depression. However, whether the mechanisms that underlie the antidepressant effect of acupuncture are related to its antiinflammatory properties remains unclear. In the present study, rats were exposed to chronic unpredictable mild stress (CUMS) for 28 days to induce depressive-like behavior. Body weight, sucrose preference, and locomotor activity in the open field were measured. After the behavioral tests, reverse transcription polymerase chain reaction was used to determine the mRNA expression of proinflammatory cytokines (interleukin-1β [IL-1β], interleukin-6 [IL-6], and tumor necrosis factor-α [TNF-α]), and enzyme-linked immunosorbent assay was used to detect cytokine concentrations. CUMS rats exhibited decrease in body weight, sucrose preference, and locomotor activity in the open field test. Chronic acupuncture and fluoxetine treatment reversed CUMS-induced depressive-like behavior. Compared with control rats, the mRNA and protein expression of IL-1β, IL-6, and TNF-α in the hippocampus and prefrontal cortex and cytokine concentrations in serum significantly increased in CUMS rats. Acupuncture and fluoxetine treatment significantly decreased the levels of proinflammatory cytokines in the hippocampus, prefrontal cortex, and serum. These results suggest that acupuncture has antidepressant-like effects, and its mechanism of action appears to involve the inhibition of proinflammatory cytokines.
Collapse
Affiliation(s)
- Jun Lu
- College of Acupuncture-Moxibustion and Tui Na, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Run-Hui Shao
- College of Acupuncture-Moxibustion and Tui Na, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Li Hu
- Tangshan Feng-Run Hospital of TCM, Tangshan 064000, China
| | - Ya Tu
- College of Acupuncture-Moxibustion and Tui Na, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jian-You Guo
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
34
|
Fischer CW, Elfving B, Lund S, Wegener G. Behavioral and systemic consequences of long-term inflammatory challenge. J Neuroimmunol 2015; 288:40-6. [PMID: 26531693 DOI: 10.1016/j.jneuroim.2015.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/14/2015] [Accepted: 08/19/2015] [Indexed: 12/24/2022]
Abstract
Inflammatory reactions are involved in a diversity of diseases, including major depressive disorder. Cytokines act as intercellular signaling molecules and mediators of inflammation between the periphery and the brain. Within the brain, evidence from animal studies of acute inflammation has shown that elevated cytokine levels are linked to behavioral responses of sickness and depression-like behavior. Although chronic inflammation is more translational to human depression than acute studies, little is known on central cytokine expression and associated behavioral responses following chronic immune challenges. The present study assessed behavioral changes and a selection of cytokines in the brain and in the blood in rats randomized to receive a single or 8week administration with either lipopolysaccharide (LPS, 600μg/kg, i.p.) or saline. Acute and long-term LPS treatments caused similar sickness and depression-like behavior. Chronic LPS administration did not have an effect on blood cytokine levels, indicating endotoxin tolerance, whereas increased fasting blood glucose was observed, indicating insulin resistance, a metabolic consequence of chronic inflammation. While a single LPS injection produced a generalized cytokine response in the brain, long-term LPS administration produced a specific central cytokine response with increased interleukin (IL)-1β and interferon (IFN)-γ. These cytokines can explain the behavioral changes observed, and could indicate microglia activation, although future studies are needed to uncover this assumption. Taken together, although the behavioral outcome was similar between acute and chronic LPS administration, the central cytokine response was distinct. As the long-term LPS paradigm also posed a metabolic demand, this setting may reflect a more translational insight into inflammatory reactions in human depression, and could prove useful for assessing cytokine down-stream effects and experimental antidepressant drug products.
Collapse
Affiliation(s)
- Christina W Fischer
- Translational Neuropsychiatry Unit, Aarhus University, Skovagervej 2, 8240 Risskov, Denmark.
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Aarhus University, Skovagervej 2, 8240 Risskov, Denmark.
| | - Sten Lund
- Department of Internal Medicine and Endocrinology, Aarhus University Hospital, Aarhus, Denmark.
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Aarhus University, Skovagervej 2, 8240 Risskov, Denmark.
| |
Collapse
|
35
|
Wahab F, Santos-Junior NN, de Almeida Rodrigues RP, Costa LHA, Catalão CHR, Rocha MJA. Interleukin-1 Receptor Antagonist Decreases Hypothalamic Oxidative Stress During Experimental Sepsis. Mol Neurobiol 2015; 53:3992-3998. [DOI: 10.1007/s12035-015-9338-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/01/2015] [Indexed: 01/08/2023]
|
36
|
Wahab F, Tazinafo LF, Cárnio EC, Aguila FA, Batalhão ME, Rocha MJA. Interleukin-1 receptor antagonist decreases cerebrospinal fluid nitric oxide levels and increases vasopressin secretion in the late phase of sepsis in rats. Endocrine 2015; 49:215-21. [PMID: 25338201 DOI: 10.1007/s12020-014-0452-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 10/09/2014] [Indexed: 12/24/2022]
Abstract
The aim of this study was to analyze the effect of IL-1ra (an Interleukin-1 receptor antagonist) on sepsis-induced alterations in vasopressin (AVP) and nitric oxide (NO) levels. In addition, IL-1ra effect on the hypothalamic nitric oxide synthase (NOS) activities and survival rate was also analyzed. After Wistar rats were intracerebroventricular injected with IL-1ra (9 pmol) or vehicle (PBS 0.01 M), sepsis was induced by cecal-ligation and puncture (CLP). Blood, CSF, and hypothalamic samples were collected from different groups of rats (n = 8/group) after 4, 6, and 24 h. AVP and NO levels were greatly increased in CLP. Both total NOS and inducible NOS (iNOS) activities were also greatly increased in CLP rats. These changes in AVP, NO, and NOS were not observed in sham-operated control rats. IL-1ra administration did not alter plasma AVP levels after 4 and 6 h as compared to vehicle in CLP animals but after 24 h were significantly (P < 0.01) higher in IL-1ra-treated animals. IL-1ra administration significantly (P < 0.01) decreased NO concentration in CSF but not in plasma. Both total NOS and iNOS activities were also significantly decreased by IL-1ra at 24 h in CLP animals. Moreover, the 24 h survival rate of IL-1ra-treated rats increased by 38 % in comparison to vehicle administered animals. The central administration of IL-1ra increased AVP secretion in the late phase of sepsis which was beneficial for survival. We believe that one of the mechanisms for this effect of IL-1ra is through reduction of NO concentration in CSF and hence lower hypothalamic iNOS activities in the septic rats.
Collapse
Affiliation(s)
- Fazal Wahab
- Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto, Avenida do Café s/n CEP, Ribeirão Preto, SP, 14040-904, Brazil
| | | | | | | | | | | |
Collapse
|
37
|
ALAMILI M, ROSENBERG J, GÖGENUR I. Day-night variation in heart rate variability changes induced by endotoxaemia in healthy volunteers. Acta Anaesthesiol Scand 2015; 59:457-64. [PMID: 25790066 DOI: 10.1111/aas.12472] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 12/20/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Morbidity and mortality in response to sepsis may be dependent on clock time for the initiation of sepsis. Endotoxaemia, an experimental model for systemic inflammation, induces alterations in sympatico-vagal balance in the autonomic nervous system (ANS). The activity of sympathetic and parasympathetic activity can be estimated by measuring heart rate variability (HRV). Based on the intimate link between ANS and the inflammatory response, we hypothesized, that HRV changes seen during endotoxaemia would be different based on time of the day the endotoxaemia is initiated. We investigated day/night variation in endotoxaemia-induced changes in HRV. METHODS A randomized, crossover study with 12 healthy men (age 18-31) was conducted. Endotoxaemia were induced by lipopolysaccharide (LPS) endotoxin 0.3 ng/kg b.w. in two visits (day visit and night visit). At the day visit, endotoxaemia were induced at 12:00 h, and at the night visit it was induced at 24:00 h. Holter recordings were started 1 h before administration of LPS, and continued for 10 h. Time-domain and frequency-domain parameters of HRV were analysed. RESULTS A total of nine persons finished the study with valid recordings. Endotoxaemia at both night and day resulted in a significant depression in HRV parameters high-frequency power (HF), low-frequency power (LF), standard deviation of normal-to-normal (NN) intervals, root mean square of successive differences and proportion of NN50 divided by total number of NNs (P<0.001). The ratio LF/HF and mean heart rate significantly increased by endotoxaemia (P<0.001). At night-time endotoxaemia, a more pronounced depression of LF, HF and SDNN (P<0.01) and a more pronounced increase in the ratio of LF/HF and mean heart rate (P<0.01) occurred compared with day-time endotoxaemia. CONCLUSION Endotoxaemia induced changes in HRV exhibit a day-night difference. This difference may have clinical consequences in patients with sepsis.
Collapse
Affiliation(s)
- M. ALAMILI
- Department of Surgery; Køge Hospital; Copenhagen University; Køge Denmark
| | - J. ROSENBERG
- Department of Surgery; Herlev Hospital; Copenhagen University; Copenhagen Denmark
| | - I. GÖGENUR
- Department of Surgery; Køge Hospital; Copenhagen University; Køge Denmark
| |
Collapse
|
38
|
Camargo CHF, Justus FF, Retzlaff G, Blood MRY, Schafranski MD. Action of anti-TNF-α drugs on the progression of Alzheimer's disease: A case report. Dement Neuropsychol 2015; 9:196-200. [PMID: 29213962 PMCID: PMC5619359 DOI: 10.1590/1980-57642015dn92000015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 03/15/2015] [Indexed: 02/23/2023] Open
Abstract
The aim of this study was to describe a clinical case of a patient with Alzheimer's disease (AD) in use of an anti-TNF-α agent for rheumatoid arthritis (RA). The patient reported is an 81-year-old Caucasian man and retired teacher, diagnosed with RA in 2008 and AD in 2011. Treatment with donepezil was started in 2011 and the use of etanercept introduced in 2012. He was previously treated with adalimumab in 2010 for 18 months. In 2013, the subject was engaged in a clinical trial to assess a complementary non-pharmacological approach for AD, presenting significant cognitive improvement during the follow-up period. We propose the hypothesis of a synergistic effect of anti-TNF-α medication used for the treatment of RA as the cause of the improvement in cognitive response observed. These findings could suggest a possible use of this drug class in the therapeutic management of AD.
Collapse
Affiliation(s)
| | - Filipe Fernandes Justus
- Neurology Service, Medicine Department, Hospital
Universitário – State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Giuliano Retzlaff
- Neurology Service, Medicine Department, Hospital
Universitário – State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Marcelo Rezende Young Blood
- Neurology Service, Medicine Department, Hospital
Universitário – State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Marcelo Derbli Schafranski
- Rheumatology Service, Medicine Department, Hospital
Universitário – State University of Ponta Grossa, Ponta Grossa, Brazil
| |
Collapse
|
39
|
Shah SC, Kornak J, Khalili M. Depression is not associated with peripheral insulin resistance in patients with chronic hepatitis C infection. J Viral Hepat 2015; 22:272-80. [PMID: 25196736 PMCID: PMC4386832 DOI: 10.1111/jvh.12306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Depression is common in individuals infected with hepatitis C virus (HCV), and both depression and HCV infection are independently associated with insulin resistance (IR). To evaluate the relationship between depression and IR, among other factors, in an HCV-infected cohort. In this cross-sectional analysis, seventy-four non-type 2 diabetic, noncirrhotic, HCV-infected patients underwent comprehensive clinical, histologic and metabolic evaluation. IR was assessed directly with an insulin suppression test by measuring steady-state plasma glucose (SSPG) levels during continuous infusions of octreotide, glucose and insulin. Logistic regression modelling was used to evaluate predictors associated with depression. Thirty-nine (53%) patients were depressed, and 21 (54%) depressed patients were on at least one antidepressant. A higher estimated proportion of depressed patients were Caucasian (51% vs 20%, P = 0.005), unemployed (69% vs 49%, P = 0.07), heavier smokers (18 pack-years vs 13 pack-years, P = 0.07), on substance abuse therapy (16% vs 3%, P = 0.06) and had lower HDL levels (1.2 mmol/L vs 1.4 mmol/L, P = 0.01). The mean SSPG levels in depressed and nondepressed patients were 7.3 and 8.3 mmol/L (P = 0.45), respectively. In multipredictor adjusted analysis, only Caucasian race (OR 4.19, 95% CI 1.42-12.35, P = 0.009) and lower HDL (OR 0.95, 95% CI 0.89-0.99, P = 0.046) were associated with depression. In conclusion, although prevalent, depression was not associated with peripheral IR in this HCV-infected cohort. Attention to other modifiable factors associated with depression in the HCV-infected population is warranted.
Collapse
Affiliation(s)
- S. C. Shah
- Department of Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, CA, USA
| | - J. Kornak
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - M. Khalili
- Department of Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, CA, USA,Liver Center, San Francisco General Hospital, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
40
|
Abstract
The present review assesses the current state of literature defining integrative autonomic-immune physiological processing, focusing on studies that have employed electrophysiological, pharmacological, molecular biological, and central nervous system experimental approaches. Central autonomic neural networks are informed of peripheral immune status via numerous communicating pathways, including neural and non-neural. Cytokines and other immune factors affect the level of activity and responsivity of discharges in sympathetic and parasympathetic nerves innervating diverse targets. Multiple levels of the neuraxis contribute to cytokine-induced changes in efferent parasympathetic and sympathetic nerve outflows, leading to modulation of peripheral immune responses. The functionality of local sympathoimmune interactions depends on the microenvironment created by diverse signaling mechanisms involving integration between sympathetic nervous system neurotransmitters and neuromodulators; specific adrenergic receptors; and the presence or absence of immune cells, cytokines, and bacteria. Functional mechanisms contributing to the cholinergic anti-inflammatory pathway likely involve novel cholinergic-adrenergic interactions at peripheral sites, including autonomic ganglion and lymphoid targets. Immune cells express adrenergic and nicotinic receptors. Neurotransmitters released by sympathetic and parasympathetic nerve endings bind to their respective receptors located on the surface of immune cells and initiate immune-modulatory responses. Both sympathetic and parasympathetic arms of the autonomic nervous system are instrumental in orchestrating neuroimmune processes, although additional studies are required to understand dynamic and complex adrenergic-cholinergic interactions. Further understanding of regulatory mechanisms linking the sympathetic nervous, parasympathetic nervous, and immune systems is critical for understanding relationships between chronic disease development and immune-associated changes in autonomic nervous system function.
Collapse
Affiliation(s)
- M J Kenney
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas
| | | |
Collapse
|
41
|
Neznanov NG, Kozlova SN, Mazo GE, Shlyakhto NG, Smirnov BI. Comorbidity of depressive disorders and coronary heart disease: general aspects of pathogenesis. Zh Nevrol Psikhiatr Im S S Korsakova 2015; 115:20-26. [DOI: 10.17116/jnevro20151155120-26] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
42
|
Abstract
Amyloid-β plaques and neurofibrillary tangles are the main neuropathological hallmarks in Alzheimer's disease (AD), the most common cause of dementia in the elderly. However, it has become increasingly apparent that neuroinflammation plays a significant role in the pathophysiology of AD. This review summarizes the current status of neuroinflammation research related to AD, focusing on the connections between neuroinflammation and some inflammation factors in AD. Among these connections, we discuss the dysfunctional blood-brain barrier and alterations in the functional responses of microglia and astrocytes in this process. In addition, we summarize and discuss the role of intracellular signaling pathways involved in inflammatory responses in astrocytes and microglia, including the mitogen-activated protein kinase pathways, nuclear factor-kappa B cascade, and peroxisome proliferator-activated receptor-gamma transcription factors. Finally, the dysregulation of the control and release of pro- and anti-inflammatory cytokines and classic AD pathology (amyloid plaques and neurofibrillary tangles) in AD is also reviewed.
Collapse
Affiliation(s)
- Fengjin Zhang
- Department of Pharmacy, General Hospital of Guangzhou Military Command, Guangzhou City, People's Republic of China ; School of Bioscience and Bioengineering, South China University of Technology, Guangzhou City, People's Republic of China
| | - Linlan Jiang
- Department of Pharmacy, General Hospital of Guangzhou Military Command, Guangzhou City, People's Republic of China
| |
Collapse
|
43
|
Ito K, Hirooka Y, Sunagawa K. Cardiac sympathetic afferent stimulation induces salt-sensitive sympathoexcitation through hypothalamic epithelial Na+ channel activation. Am J Physiol Heart Circ Physiol 2014; 308:H530-9. [PMID: 25527778 DOI: 10.1152/ajpheart.00586.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cardiac sympathetic afferent (CSA), which plays an important role in heart-brain communication for sympathoexcitation, is stimulated in heart failure. Additionally, high salt intake leads to further sympathoexcitation due to activation of hypothalamic epithelial Na(+) channels (ENaCs) in heart failure. In the present study, we stimulated the CSA in adult male mice by epicardial application of capsaicin and using ethanol as a control to determine whether CSA stimulation led to activation of hypothalamic ENaCs, resulting in salt-induced sympathoexcitation. Three days after capsaicin treatment, an upregulation of hypothalamic α-ENaCs, without activation of mineralocorticoid receptors, was observed. We also examined expression levels of the known ENaC activator TNF-α. Hypothalamic TNF-α increased in capsaicin-treated mice, whereas intracerebroventricular infusion of the TNF-α blocker etanercept prevented capsaicin-induced upregulation of α-ENaCs. To examine brain arterial pressure (AP) sensitivity toward Na(+), we performed an intracerebroventricular infusion of high Na(+)-containing (0.2 M) artificial cerebrospinal fluid. AP and heart rate were significantly increased in capsaicin-treated mice compared with control mice. CSA stimulation also caused excitatory responses with high salt intake. Compared with a regular salt diet, the high-salt diet augmented AP, heart rate, and 24-h urinary norepinephrine excretion, which is an indirect marker of sympathetic activity with mineralocorticoid receptor activation, in capsaicin-treated mice but not in ethanol-treated mice. Treatment with etanercept or the ENaC blocker benzamil prevented these salt-induced excitatory responses. In summary, we show that CSA stimulation leads to an upregulation of hypothalamic α-ENaCs mediated via an increase in TNF-α and results in increased salt sensitivity.
Collapse
Affiliation(s)
- Koji Ito
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and
| | - Yoshitaka Hirooka
- Department of Advanced Cardiovascular Regulation and Therapeutics, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kenji Sunagawa
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and
| |
Collapse
|
44
|
Gaur S, Agnihotri R. Alzheimer's disease and chronic periodontitis: is there an association? Geriatr Gerontol Int 2014; 15:391-404. [PMID: 25511390 DOI: 10.1111/ggi.12425] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2014] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease, an affliction of old age, is one of the leading causes for dementia worldwide. Various risk factors including family history, genetics and infections have been implicated in its pathogenesis. The cognitive decline in this condition is mainly a result of the formation of amyloid deposits that provoke neuroinflammation, ultimately resulting in cell death. Recently, an association between peripheral inflammation and Alzheimer's disease was hypothesized. It was suggested that chronic systemic inflammation worsened the inflammatory processes in the brain. This was mainly attributed to increased levels of pro-inflammatory mediators, such as interleukin-1, interleukin -6 and tumor necrosis factor-α in the plasma. As chronic periodontitis is a widespread peripheral immunoinflammatory condition, it has been proposed to play a significant role in the aggravation of Alzheimer's disease. With this background, the current review focuses on the relationship between Alzheimer's disease and chronic periodontitis, and its therapeutic implications.
Collapse
Affiliation(s)
- Sumit Gaur
- Department of Pedodontics and Preventive Dentistry, Manipal College of Dental Sciences, Manipal University, Manipal, Karnataka, India
| | | |
Collapse
|
45
|
Marques AH, Bjørke-Monsen AL, Teixeira AL, Silverman MN. Maternal stress, nutrition and physical activity: Impact on immune function, CNS development and psychopathology. Brain Res 2014; 1617:28-46. [PMID: 25451133 DOI: 10.1016/j.brainres.2014.10.051] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 10/20/2014] [Accepted: 10/25/2014] [Indexed: 12/14/2022]
Abstract
Evidence suggests that maternal and fetal immune dysfunction may impact fetal brain development and could play a role in neurodevelopmental disorders, although the definitive pathophysiological mechanisms are still not completely understood. Stress, malnutrition and physical inactivity are three maternal behavioral lifestyle factors that can influence immune and central nervous system (CNS) functions in both the mother and fetus, and may therefore, increase risk for neurodevelopmental/psychiatric disorders. First, we will briefly review some aspects of maternal-fetal immune system interactions and development of immune tolerance. Second, we will discuss the bidirectional communication between the immune system and CNS and the pathways by which immune dysfunction could contribute to neurodevelopmental disorders. Third, we will discuss the effects of prenatal stress and malnutrition (over and undernutrition) on perinatal programming of the CNS and immune system, and how this might influence neurodevelopment. Finally, we will discuss the beneficial impact of physical fitness during pregnancy on the maternal-fetal unit and infant and how regular physical activity and exercise can be an effective buffer against stress- and inflammatory-related disorders. Although regular physical activity has been shown to promote neuroplasticity and an anti-inflammatory state in the adult, there is a paucity of studies evaluating its impact on CNS and immune function during pregnancy. Implementing stress reduction, proper nutrition and ample physical activity during pregnancy and the childbearing period may be an efficient strategy to counteract the impact of maternal stress and malnutrition/obesity on the developing fetus. Such behavioral interventions could have an impact on early development of the CNS and immune system and contribute to the prevention of neurodevelopmental and psychiatric disorders. Further research is needed to elucidate this relationship and the underlying mechanisms of protection. This article is part of a Special Issue entitled SI: Neuroimmunology in Health And Disease.
Collapse
Affiliation(s)
- Andrea Horvath Marques
- Obsessive--Compulsive Spectrum Disorders Program, Department & Institute of Psychiatry, University of São Paulo, Medical School, São Paulo, Brazil.
| | | | - Antônio L Teixeira
- School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marni N Silverman
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
46
|
Raju GS, Moghal MMR, Hossain MS, Hassan MM, Billah MM, Ahamed SK, Rana SMM. Assessment of pharmacological activities of two medicinal plant of Bangladesh: Launaea sarmentosa and Aegialitis rotundifolia roxb in the management of pain, pyrexia and inflammation. Biol Res 2014; 47:55. [PMID: 25418519 PMCID: PMC4416252 DOI: 10.1186/0717-6287-47-55] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 10/11/2014] [Indexed: 11/29/2022] Open
Abstract
Background The current study aims at evaluating the analgesic, anti-pyretic and anti-inflammatory properties of methanolic extract of the stem, bark and leaves of Launaea sarmentosa and Aegialitis rotundifolia roxb. Results The AELS and AEAR extract presented a significant (***p < 0.001) dose dependent increase in reaction time in writhing method and showed inhibition of 63.1% and 57.1% respectively at the doses of 400 mg/kg body weight while standard drug showed (P < 0.001) inhibition of 69.23%. In tail immersion method, AELS and AEAR showed maximum time of tail retention at 30 min in hot water i.e. 6.93 sec and 6.54 sec respectively at highest doses of 400 mg/kg body weight than lower dose while standard pentazocine showed reaction time of 7.62 sec. The AELS and AEAR extract also exhibited promising anti-inflammatory effect as demonstrated by statistically significant inhibition of paw volume by 32.48% and 26.75% respectively at the dose of 400 mg/kg body weight while the value at the dose of 200 mg/kg body weight were linear to higher dose at the 3rd hour of study. On the other hand, Standard indomethacin inhibited 40.13% of inflammation (***P < 0.001). In Cotton-pellet granuloma method, AELS and AEAR extract at the dose of 400 mg/kg body weight exhibited inhibition of inflammation of 34.7% and 29.1% respectively while standard drug showed (P < 0.001) inhibition of 63.22%. Intraperitoneal administration of AELS and AEAR showed dose dependent decrease in body temperature in brewer’s yeast induced hyperthermia in rats at both doses. However, AELS significantly decreased body temperature (***p < 0.001) at 400 mg/kg compared to control. Conclusions Present work propose that the methanolic extract of Launaea sarmentosa and Aegialitis rotundifolia roxb possesses dose dependent pharmacological action which supports its therapeutic use in folk medicine possibly mediated through the inhibition or blocking of release of prostaglandin and/or actions of vasoactive substances such as histamine, serotonin and kinins.
Collapse
|
47
|
Rossetti C, Halfon O, Boutrel B. Controversies about a common etiology for eating and mood disorders. Front Psychol 2014; 5:1205. [PMID: 25386150 PMCID: PMC4209809 DOI: 10.3389/fpsyg.2014.01205] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 10/06/2014] [Indexed: 12/25/2022] Open
Abstract
Obesity and depression represent a growing health concern worldwide. For many years, basic science and medicine have considered obesity as a metabolic illness, while depression was classified a psychiatric disorder. Despite accumulating evidence suggesting that obesity and depression may share commonalities, the causal link between eating and mood disorders remains to be fully understood. This etiology is highly complex, consisting of multiple environmental and genetic risk factors that interact with each other. In this review, we sought to summarize the preclinical and clinical evidence supporting a common etiology for eating and mood disorders, with a particular emphasis on signaling pathways involved in the maintenance of energy balance and mood stability, among which orexigenic and anorexigenic neuropeptides, metabolic factors, stress responsive hormones, cytokines, and neurotrophic factors.
Collapse
Affiliation(s)
- Clara Rossetti
- Center for Psychiatric Neuroscience, Lausanne University Hospital Lausanne, Switzerland
| | - Olivier Halfon
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital Lausanne, Switzerland
| | - Benjamin Boutrel
- Center for Psychiatric Neuroscience, Lausanne University Hospital Lausanne, Switzerland ; Division of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital Lausanne, Switzerland
| |
Collapse
|
48
|
Early activation of STAT3 regulates reactive astrogliosis induced by diverse forms of neurotoxicity. PLoS One 2014; 9:e102003. [PMID: 25025494 PMCID: PMC4098997 DOI: 10.1371/journal.pone.0102003] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/13/2014] [Indexed: 12/31/2022] Open
Abstract
Astrogliosis, a cellular response characterized by astrocytic hypertrophy and accumulation of GFAP, is a hallmark of all types of central nervous system (CNS) injuries. Potential signaling mechanisms driving the conversion of astrocytes into “reactive” phenotypes differ with respect to the injury models employed and can be complicated by factors such as disruption of the blood-brain barrier (BBB). As denervation tools, neurotoxicants have the advantage of selective targeting of brain regions and cell types, often with sparing of the BBB. Previously, we found that neuroinflammation and activation of the JAK2-STAT3 pathway in astrocytes precedes up regulation of GFAP in the MPTP mouse model of dopaminergic neurotoxicity. Here we show that multiple mechanistically distinct mouse models of neurotoxicity (MPTP, AMP, METH, MDA, MDMA, KA, TMT) engender the same neuroinflammatory and STAT3 activation responses in specific regions of the brain targeted by each neurotoxicant. The STAT3 effects seen for TMT in the mouse could be generalized to the rat, demonstrating cross-species validity for STAT3 activation. Pharmacological antagonists of the neurotoxic effects blocked neuroinflammatory responses, pSTAT3tyr705 and GFAP induction, indicating that damage to neuronal targets instigated astrogliosis. Selective deletion of STAT3 from astrocytes in STAT3 conditional knockout mice markedly attenuated MPTP-induced astrogliosis. Monitoring STAT3 translocation in GFAP-positive cells indicated that effects of MPTP, METH and KA on pSTAT3tyr705 were localized to astrocytes. These findings strongly implicate the STAT3 pathway in astrocytes as a broadly triggered signaling pathway for astrogliosis. We also observed, however, that the acute neuroinflammatory response to the known inflammogen, LPS, can activate STAT3 in CNS tissue without inducing classical signs of astrogliosis. Thus, acute phase neuroinflammatory responses and neurotoxicity-induced astrogliosis both signal through STAT3 but appear to do so through different modules, perhaps localized to different cell types.
Collapse
|
49
|
Strunk T, Inder T, Wang X, Burgner D, Mallard C, Levy O. Infection-induced inflammation and cerebral injury in preterm infants. THE LANCET. INFECTIOUS DISEASES 2014; 14:751-762. [PMID: 24877996 DOI: 10.1016/s1473-3099(14)70710-8] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Preterm birth and infectious diseases are the most common causes of neonatal and early childhood deaths worldwide. The rates of preterm birth have increased over recent decades and account for 11% of all births worldwide. Preterm infants are at significant risk of severe infection in early life and throughout childhood. Bacteraemia, inflammation, or both during the neonatal period in preterm infants is associated with adverse outcomes, including death, chronic lung disease, and neurodevelopmental impairment. Recent studies suggest that bacteraemia could trigger cerebral injury even without penetration of viable bacteria into the CNS. Here we review available evidence that supports the concept of a strong association between bacteraemia, inflammation, and cerebral injury in preterm infants, with an emphasis on the underlying biological mechanisms, clinical correlates, and translational opportunities.
Collapse
Affiliation(s)
- Tobias Strunk
- Centre for Neonatal Research and Education, School of Paediatrics and Child Health, The University of Western Australia, Perth, WA, Australia; Neonatal Clinical Care Unit, King Edward Memorial Hospital, Perth, WA, Australia.
| | - Terrie Inder
- Department of Pediatrics, Neurology and Radiology, Washington University, St Louis, USA
| | - Xiaoyang Wang
- Perinatal Center, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Shangjie, Henan, China
| | - David Burgner
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Carina Mallard
- Perinatal Center, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
50
|
Sotelo JL, Musselman D, Nemeroff C. The biology of depression in cancer and the relationship between depression and cancer progression. Int Rev Psychiatry 2014; 26:16-30. [PMID: 24716498 DOI: 10.3109/09540261.2013.875891] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The prevalence of depressive symptoms in patients with cancer exceeds that observed in the general population and depression is associated with a poorer prognosis in cancer patients. The increased prevalence is not solely explained by the psychosocial stress associated with the diagnosis. Pro-inflammatory cytokines, which induce sickness behaviour with symptoms overlapping those of clinical depression, are validated biomarkers of increased inflammation in patients with cancer. A growing literature reveals that chronic inflammatory processes associated with stress may also underlie depression symptoms in general, and in patients with cancer in particular. Therapeutic modalities, which are frequently poorly tolerated, are used in the treatment of cancer. These interventions are associated with inflammatory reactions, which may help to explain their toxicity. There is evidence that antidepressants can effectively treat symptoms of depression in cancer patients though the database is meager. Novel agents with anti-inflammatory properties may be effective alternatives for patients with treatment-resistant depression who exhibit evidence of increased inflammation.
Collapse
Affiliation(s)
- Jorge Luis Sotelo
- Department of Psychiatry and Behavioral Sciences, Leonard M. Miller School of Medicine, University of Miami Hospital , Miami, Florida
| | | | | |
Collapse
|