1
|
Blomqvist A. Inflammation-induced fever depends on prostaglandin E2 production by brain endothelial cells and EP3 receptors in the median preoptic nucleus of the hypothalamus. Acta Physiol (Oxf) 2024; 240:e14238. [PMID: 39352065 DOI: 10.1111/apha.14238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 09/14/2024] [Indexed: 11/10/2024]
Affiliation(s)
- Anders Blomqvist
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
2
|
Llansola M, Izquierdo-Altarejos P, Montoliu C, Mincheva G, Palomares-Rodriguez A, Pedrosa MA, Arenas YM, Felipo V. Role of peripheral inflammation in minimal hepatic encephalopathy. Metab Brain Dis 2024; 39:1667-1677. [PMID: 39177864 DOI: 10.1007/s11011-024-01417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
Many patients with liver cirrhosis show minimal hepatic encephalopathy (MHE) with mild cognitive impairment (MCI) and motor alterations that reduce their quality of life. Some patients with steatotic liver disease also suffer MCI. To design treatments to improve MHE/MCI it is necessary to understand the mechanisms by which liver disease induce them. This review summarizes studies showing that appearance of MHE/MCI is associated with a shift in the immunophenotype leading to an "autoimmune-like" form with increased pro-inflammatory monocytes, enhanced CD4 T and B lymphocytes activation and increased plasma levels of pro-inflammatory cytokines, including IL-17, IL-21, TNFα, IL-15 and CCL20. The contribution of peripheral inflammation to trigger MHE is supported by studies in animal models and by the fact that rifaximin treatment reverses MHE in around 60% of patients in parallel with reversal of the changes in peripheral inflammation. MHE does not improve in patients in which peripheral inflammation is not improved by rifaximin. The process by which peripheral inflammation induces MHE involves induction of neuroinflammation in brain, with activation of microglia and astrocytes and increased pro-inflammatory TNFα and IL-1β, which is observed in patients who died with steatotic liver disease (SLD) or liver cirrhosis and in animal models of MHE. Neuroinflammation alters glutamatergic and GABAergic neurotransmission, leading to cognitive and motor impairment. Transmission of peripheral alterations into the brain is mediated by infiltration in brain of extracellular vesicles from plasma and of cells from the peripheral immune system. Acting on any step of the process peripheral inflammation - neuroinflammation - altered neurotransmission may improve MHE.
Collapse
Affiliation(s)
- Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | - Carmina Montoliu
- Departamento de Patología, Facultad de Medicina, Universidad Valencia, Valencia, Spain
- Fundación de Investigación Hospital Clínico Universitario de Valencia-INCLIVA, Valencia, Spain
| | - Gergana Mincheva
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | - María A Pedrosa
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Yaiza M Arenas
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
- Departamento de Patología, Facultad de Medicina, Universidad Valencia, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| |
Collapse
|
3
|
Blomqvist A. Prostaglandin E 2 production in the brainstem parabrachial nucleus facilitates the febrile response. Temperature (Austin) 2024; 11:309-317. [PMID: 39583895 PMCID: PMC11583619 DOI: 10.1080/23328940.2024.2401674] [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: 06/04/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 11/26/2024] Open
Abstract
Our body temperature is normally kept within a narrow range of 1°C. For example, if our body temperature rises, such as in a hot environment or due to strenuous exercise, our thermoregulatory system will trigger a powerful heat defense response with vasodilation, sweating, and lowered metabolism. During fever, which often involves body temperatures of up to 41°C, this heat defense mechanism is apparently inhibited; otherwise, the rising body temperature would be immediately combated, and fever would not be allowed to develop. New evidence suggests how and where this inhibition takes place. In two consecutive studies from Cheng et al. and Xu et al., it has been shown that prostaglandin E2, which generates fever by acting on thermosensory neurons in the preoptic hypothalamus, also acts on neurons in the brainstem parabrachial nucleus, which receive temperature information from temperature-activated spinal cord neurons and relay this information to the thermoregulatory center in the hypothalamus to either induce cold or heat defenses. By acting on the same type of prostaglandin E2 receptor that is critical for fever generation in the preoptic hypothalamus, the EP3 receptor, prostaglandin E2 inhibits the signaling of the heat-responsive parabrachial neurons, while stimulating the cold-responsive neurons. These novel findings thus show that prostaglandin E2, by binding to the same receptor subtype in the parabrachial nucleus as in the preoptic hypothalamus, adjusts the sensitivity of the thermosensory system in a coordinated manner to allow the development of febrile body temperatures.
Collapse
Affiliation(s)
- Anders Blomqvist
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
4
|
Ji W, Liu X, Liu P, He Y, Zhao Y, Zheng K, Cui J, Li W. The efficacy of fat-free mass index and appendicular skeletal muscle mass index in cancer malnutrition: a propensity score match analysis. Front Nutr 2023; 10:1172610. [PMID: 37492594 PMCID: PMC10364448 DOI: 10.3389/fnut.2023.1172610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/19/2023] [Indexed: 07/27/2023] Open
Abstract
Background Reduced muscle mass (RMM) is a phenotypic criterion for malnutrition; the appendicular skeletal muscle mass index (ASMI) and fat-free mass index (FFMI) are both applicable indicators in the global leadership initiative on malnutrition (GLIM) guideline. However, their sensitivity and prognostic effect remain unclear. Methods Clinical data of 2,477 patients with malignant tumors were collected. Multi-frequency bioelectrical impedance analysis was used to obtain ASMI and FFMI. RMM was confirmed by ASMI (< 7.0 kg/m2 for men and < 5.7 kg/m2 for women) or FFMI (< 17 kg/m2 for men and < 15 kg/m2 for women). Propensity score match analysis and logistic regression analysis were used to evaluate the efficacy of FFMI and ASMI in diagnosing severe malnutrition and multivariate Cox regression analysis to determine the efficacy of RMM in predicting survival. Results In total, 546 (22.0%) and 659 (26.6%) participants were diagnosed with RMM by ASMI (RMM.ASMI group) and FFMI (RMM.FFMI group); 375 cases overlapped. Body mass index (BMI), midarm circumference, triceps skinfold thickness, and maximum calf circumference were all significantly larger in the RMM.FFMI group for both sexes (P < 0.05). A 1:1 matched dataset constructed by propensity score match contained 810 cases. RMM.FFMI was an influential factor of severe malnutrition with HR = 3.033 (95% CI 2.068-4.449, P < 0.001), and RMM.ASMI was a predictive factor of overall survival (HR = 1.318, 95% CI 1.060-1.639, P = 0.013 in the RMM.ASMI subgroup, HR = 1.315, 95% CI 1.077-1.607, P = 0.007 in the RMM.FFMI subgroup). Conclusion In general, RMM indicates negative clinical outcomes; when defined by FFMI, it predicts nutritional status, and when defined by ASMI, it is related to poor survival in cancer patients.
Collapse
Affiliation(s)
- Wei Ji
- Center of Cancer, The First Affiliated Hospital of Jilin University, Changchun, China
| | - XiangLiang Liu
- Center of Cancer, The First Affiliated Hospital of Jilin University, Changchun, China
| | - Pengfei Liu
- Cancer Department, Longyan First Hospital, Fujian, Longyan, China
| | - YuWei He
- Center of Cancer, The First Affiliated Hospital of Jilin University, Changchun, China
| | - YiXin Zhao
- Center of Cancer, The First Affiliated Hospital of Jilin University, Changchun, China
| | - Kaiwen Zheng
- Center of Cancer, The First Affiliated Hospital of Jilin University, Changchun, China
| | - JiuWei Cui
- Center of Cancer, The First Affiliated Hospital of Jilin University, Changchun, China
| | - Wei Li
- Center of Cancer, The First Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Kang Y, Gao Y, Li X, Guo X, Liu Z, Li W, Wei J, Qi Y. Bupleurum chinense exerts a mild antipyretic effect on LPS-induced pyrexia rats involving inhibition of peripheral TNF-α production. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116375. [PMID: 36934787 DOI: 10.1016/j.jep.2023.116375] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bupleuri Radix, the dried roots of Bupleurum chinense DC. (BC) or Bupleurum scorzonerifolium Willd., is one of the most frequently used traditional Chinese medicines. As the species in Xiao-Chai-Hu decoction, BC has been used as an antipyretic medicine with a long history. However, its antipyretic characteristics and underlying mechanism(s) remain unclear. AIM OF THE STUDY To elucidate the antipyretic characteristics and mechanism(s) of BC used in its traditional way. METHODS The water extract of BC (BCE) was prepared according to the traditional decocting mode. Murine fever and endotoxemia models were induced by intravenous injection of lipopolysaccharide (LPS). In vitro complement activation assay and the levels of TNF-α, IL-6, IL-1β, and C5a were determined by ELISA. RESULTS BCE exerted a confirmed but mild antipyretic effect on LPS-induced fever of rat. In vitro, it significantly lowered LPS-elevated TNF-α in the supernatant of rat complete blood cells and THP-1 cells, but failed to decrease IL-6 and IL-1β. In murine endotoxemia models, BCE markedly decreased serum TNF-α, but had no impact on IL-6 and IL-1β. BCE also restricted complement activation in vitro and in vivo. Nevertheless, the mixture of saikosaponin A and D could not suppress supernatant TNF-α of monocytes and serum TNF-α of endotoxemia mice. CONCLUSIONS The present study dissects the peripheral mechanism for the antipyretic effect of BC used in the traditional way. Our findings indicate that BCE directly suppresses monocyte-produced TNF-α, thus decreasing circulating TNF-α, which may be responsible for its mild but confirmed antipyretic action.
Collapse
Affiliation(s)
- Yuan Kang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Yuan Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Ximeng Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Xinwei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Zhuangzhuang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Wenjing Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Jianhe Wei
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Yun Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
6
|
Blomqvist A. Prostaglandin E 2 Production by Brain Endothelial Cells and the Generation of Fever. DNA Cell Biol 2023; 42:107-112. [PMID: 36720071 PMCID: PMC10024267 DOI: 10.1089/dna.2022.0662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We recently demonstrated that prostaglandin production in brain endothelial cells is both necessary and sufficient for the generation of fever during systemic immune challenge. I here discuss this finding in light of the previous literature and point to some unresolved issues.
Collapse
Affiliation(s)
- Anders Blomqvist
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
7
|
Bezerra LFG, Silva APSD, Cunha RXD, Oliveira JRSD, Barros MDD, Silva VMDMAD, Lima VLDM. Antioxidant, anti-inflammatory and analgesic activity of Mimosa acutistipula (Mart.) Benth. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:115964. [PMID: 36436717 DOI: 10.1016/j.jep.2022.115964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Medicinal plants belonging to the genus Mimosa, such as Mimosa tenuiflora, M. caesalpinifolia, and M. verrucosa are known for their popular use for asthma, bronchitis and fever. Ethnopharmacological studies report that Mimosa acutistipula is used to treat alopecia and pharyngitis, conditions that can be related to oxidative stress, inflammatory processes and painful limitations. However, there is no studies on its efficacy and mechanism of action. AIM OF THE STUDY To elucidate the antioxidant, anti-inflammatory, analgesic and antipyretic activity of M. acutistipula leaves. MATERIALS AND METHODS Phytochemical profile of M. acutistipula extracts was evaluated by several reaction-specific methods. Secondary metabolites such as tannins, phenols and flavonoids were quantified with colorimetric assays. In vitro antioxidant potential was evaluated using DPPH and ABTS + as free radical scavenging tests, FRAP and phosphomolybdenum as oxide-reduction assays, and anti-hemolytic for lipid peroxidation evaluation. In vivo anti-inflammatory evaluation was performed by paw edema, and peritonitis induced by carrageenan. Analgesic effect and its possible mechanisms were determined by acetic acid-induced abdominal writhing and the formalin test. Antipyretic activity was evaluated by yeast-induced fever. RESULTS Cyclohexane, chloroform, ethyl acetate and methanol extracts of leaves had presence of tannins, flavonoids, phenol, alkaloids, terpenes (except methanolic extract), and saponins (only for methanolic and chloroformic extracts). In phenols, flavonoids and tannins quantification, methanolic and ethyl acetate extract had higher amounts of this phytocompounds. Ethyl acetate extract, due to its more expressive quantity of phenols and flavonoids, was chosen for carrying out the in vivo tests. Due to the relationship between oxidative stress and inflammation, antioxidant tests were performed, showing that ethyl acetate extract had a high total antioxidant activity (70.18%), moderate activity in DPPH radical scavenging, and a moderate ABTS + radical inhibition (33.61%), and FRAP assay (112.32 μg Fe2+/g). M. acutistipula showed anti-inflammatory activity, with 54.43% of reduction in paw edema (50 mg/kg) when compared to the vehicle. In peritonitis test, a reduction in the concentration of NO could be seen, which is highly involved in the anti-inflammatory activity and is responsible for the increase in permeability. In the analgesic evaluation, most significant results in writhing test were seen at 100 mg/kg, with a 34.7% reduction of writhing. A dual mechanism of action was confirmed with the formalin test, both neurogenic and inflammatory pain were reduced, with a mechanism via opioid route. In the antipyretic test, results were significantly decreased at all concentrations tested. CONCLUSION M. acutistipula leaves ethyl acetate extract showed expressive concentrations of phenolic compounds and antioxidant activity. It also exhibited anti-inflammatory and analgesic activity, besides its antipyretic effect. Thus, these results provide information regarding its popular use and might help future therapeutics involving this specimen.
Collapse
Affiliation(s)
- Layza Fernanda Gomes Bezerra
- Laboratory of Lipids and Applications of Biomolecules in Prevalent and Neglected Diseases, Center of Biosciences, Federal University of Pernambuco, Professor Moraes Rego Avenue 1235, Recife, PE, 50670-90, Brazil
| | - Ana Paula Sant'Anna da Silva
- Laboratory of Lipids and Applications of Biomolecules in Prevalent and Neglected Diseases, Center of Biosciences, Federal University of Pernambuco, Professor Moraes Rego Avenue 1235, Recife, PE, 50670-90, Brazil
| | - Rebeca Xavier da Cunha
- Laboratory of Lipids and Applications of Biomolecules in Prevalent and Neglected Diseases, Center of Biosciences, Federal University of Pernambuco, Professor Moraes Rego Avenue 1235, Recife, PE, 50670-90, Brazil
| | - João Ricardhis Saturnino de Oliveira
- Laboratory of Lipids and Applications of Biomolecules in Prevalent and Neglected Diseases, Center of Biosciences, Federal University of Pernambuco, Professor Moraes Rego Avenue 1235, Recife, PE, 50670-90, Brazil
| | - Mateus Domingues de Barros
- Laboratory of Lipids and Applications of Biomolecules in Prevalent and Neglected Diseases, Center of Biosciences, Federal University of Pernambuco, Professor Moraes Rego Avenue 1235, Recife, PE, 50670-90, Brazil
| | - Vycttor Mateus de Melo Alves da Silva
- Laboratory of Lipids and Applications of Biomolecules in Prevalent and Neglected Diseases, Center of Biosciences, Federal University of Pernambuco, Professor Moraes Rego Avenue 1235, Recife, PE, 50670-90, Brazil
| | - Vera Lúcia de Menezes Lima
- Laboratory of Lipids and Applications of Biomolecules in Prevalent and Neglected Diseases, Center of Biosciences, Federal University of Pernambuco, Professor Moraes Rego Avenue 1235, Recife, PE, 50670-90, Brazil.
| |
Collapse
|
8
|
Eskilsson A, Shionoya K, Blomqvist A. Prostaglandin production in brain endothelial cells during the initiation of fever. Commun Integr Biol 2023; 16:2166237. [PMID: 36644132 PMCID: PMC9839369 DOI: 10.1080/19420889.2023.2166237] [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] [Indexed: 01/13/2023] Open
Abstract
The initiation of fever has been a matter of controversy. Based on observations of little or no induction of prostaglandin synthesizing enzymes in the brain during the first phase of fever it was suggested that fever is initiated by prostaglandin released into the circulation from cells in the liver and lungs. Here we show in the mouse that prostaglandin synthesis is rapidly induced in the brain after immune challenge. These data are consistent with our recent findings in functional experiments that prostaglandin production in brain endothelial cells is both necessary and sufficient for the generation of all phases of fever.
Collapse
Affiliation(s)
- Anna Eskilsson
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Kiseko Shionoya
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Anders Blomqvist
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden,CONTACT Anders Blomqvist Division of Neurobiology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, SE-581 85Linköping, Sweden
| |
Collapse
|
9
|
Hu A, Yuan H, Qin Y, Zhu Y, Zhang L, Chen Q, Wu L. Lipopolysaccharide (LPS) increases susceptibility to epilepsy via interleukin-1 type 1 receptor signaling. Brain Res 2022; 1793:148052. [PMID: 35970265 DOI: 10.1016/j.brainres.2022.148052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/27/2022] [Accepted: 08/09/2022] [Indexed: 11/18/2022]
Abstract
Epilepsy is the most common disease of the nervous system, characterized by aberrant normal brain activity. Neuroinflammation is a prominent feature in the brain in epileptic humans and animal models of epilepsy. However, it remains elusive as to how peripheral inflammation affects epilepsy. Herein we demonstrated significantly greater seizure susceptibility and severity of epilepsy under kainic acid (KA) via intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) in mouse model of epilepsy. Nissl staining was employed for assessment of the neuronal damage, immunofluorescence for staining of the microglial cells and astrocytes in the mouse brain slices, and ELISA for detection of the changes of inflammatory factors. We observed a smaller population of viable neurons in CA1 and CA3 regions, a greater population of IBA-1-positive and GFAP-positive cells, with a significant upregulation of IL-1β and IL-6 in hippocampus of epileptic mice when treated with LPS, indicating that LPS aggravates hippocampal neuron injury in epilepsy, and induces neuroinflammation in the hippocampus. In addition, we provide an evident increase in BrdU+/DCX+ and Nestin+ cell populations in dentate gyrus (DG) in LPS-treated group, versus saline group on epileptic mouse model, which demonstrated LPS treatment enhanced hippocampal neurogenesis. In order to investigate whether interleukin-1 type 1 (IL-1R1) signaling is involved in this process, we adopted IL-1R1 globally restored mice (IL-1R1GR/GR) as an IL-1R1 reporter to visualize labeling of IL-1R1 mRNA and protein by means of RFP staining. Strikingly, the RFP immunofluorescence revealed increased IL-1R1 expression in LPS-treated group, versus saline group. Further, blockage of central IL-1R1 alleviated seizure susceptibility and severity of epilepsy. In summary, our findings suggested that LPS could enhance central inflammatory response and aggravate the susceptibility to epileptic seizure, which we postulated to be mediated by IL-1R1.
Collapse
Affiliation(s)
- Ankang Hu
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Honghua Yuan
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Ying Qin
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yuhua Zhu
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lingzhi Zhang
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Quangang Chen
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lianlian Wu
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
10
|
Nemeth DP, Liu X, McKim DB, DiSabato DJ, Oliver B, Herd A, Katta A, Negray CE, Floyd J, McGovern S, Pruden PS, Zhutang F, Smirnova M, Godbout JP, Sheridan J, Quan N. Dynamic Interleukin-1 Receptor Type 1 Signaling Mediates Microglia-Vasculature Interactions Following Repeated Systemic LPS. J Inflamm Res 2022; 15:1575-1590. [PMID: 35282272 PMCID: PMC8906862 DOI: 10.2147/jir.s350114] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/16/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction Lipopolysaccharide (LPS) preconditioning involves repeated, systemic, and sub-threshold doses of LPS, which induces a neuroprotective state within the CNS, thus preventing neuronal death and functional losses. Recently, proinflammatory cytokine, Interleukin-1 (IL-1), and its primary signaling partner, interleukin-1 receptor type 1 (IL-1R1), have been associated with neuroprotection in the CNS. However, it is still unknown how IL-1/IL-1R1 signaling impacts the processes associated with neuroprotection. Methods Using our IL-1R1 restore genetic mouse model, mouse lines were generated to restrict IL-1R1 expression either to endothelia (Tie2-Cre-Il1r1r/r) or microglia (Cx3Cr1-Cre-Il1r1 r/r), in addition to either global ablation (Il1r1 r/r) or global restoration of IL-1R1 (Il1r1 GR/GR). The LPS preconditioning paradigm consisted of four daily i.p. injections of LPS at 1 mg/kg (4d LPS). 24 hrs following the final i.p. LPS injection, tissue was collected for qPCR analysis, immunohistochemistry, or FAC sorting. Results Following 4d LPS, we found multiple phenotypes that are dependent on IL-1R1 signaling such as microglia morphology alterations, increased microglial M2-like gene expression, and clustering of microglia onto the brain vasculature. We determined that 4d LPS induces microglial morphological changes, clustering at the vasculature, and gene expression changes are dependent on endothelial IL-1R1, but not microglial IL-1R1. A novel observation was the induction of microglial IL-1R1 (mIL-1R1) following 4d LPS. The induced mIL-1R1 permits a unique response to central IL-1β: the mIL-1R1 dependent induction of IL-1R1 antagonist (IL-1RA) and IL-1β gene expression. Analysis of RNA sequencing datasets revealed that mIL-1R1 is also induced in neurodegenerative diseases. Discussion Here, we have identified cell type-specific IL-1R1 mediated mechanisms, which may contribute to the neuroprotection observed in LPS preconditioning. These findings identify key cellular and molecular contributors in LPS-induced neuroprotection.
Collapse
Affiliation(s)
- Daniel P Nemeth
- College of Dentistry, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA,Correspondence: Daniel P Nemeth; Ning Quan, 5353 Parkside Drive, Jupiter, FL, 33458, USA, Email ;
| | - Xiaoyu Liu
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Daniel B McKim
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Damon J DiSabato
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Braedan Oliver
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Anu Herd
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Asish Katta
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Christina E Negray
- College of Dentistry, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - James Floyd
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Samantha McGovern
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Paige S Pruden
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Feiyang Zhutang
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Maria Smirnova
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Jonathan P Godbout
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - John Sheridan
- College of Dentistry, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Ning Quan
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
11
|
Li F, Lu X, Ma Y, Gu Y, Ye T, Huang C. Monophosphoryl Lipid A Tolerance Against Chronic Stress-Induced Depression-Like Behaviors in Mice. Int J Neuropsychopharmacol 2022; 25:399-411. [PMID: 35015863 PMCID: PMC9154281 DOI: 10.1093/ijnp/pyab097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/29/2021] [Accepted: 01/06/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUNDS Our recent studies reported that a single injection with lipopolysaccharide (LPS) before stress exposure prevents depression-like behaviors in stressed mice. Monophosphoryl lipid A (MPL) is a derivative of LPS that lacks the undesirable properties of LPS. We hypothesize that MPL can exert a prophylactic effect on depression. METHODS The experimental mice were pre-injected with MPL before stress exposure. Depression in mice was induced through chronic social defeat stress (CSDS). Behavioral tests were conducted to identify depression-like behaviors. Real-time polymerase chain reaction and biochemical assays were performed to examine the gene and protein expression levels of pro-inflammatory cytokines. RESULTS A single MPL injection 1 day before stress exposure at the dosages of 400, 800, and 1600 μg/kg but not 200 μg/kg prevented CSDS-induced depression-like behaviors in mice. This effect of MPL, however, vanished with the extension of the interval time between drug injection and stress exposure from 1 day or 5 days to 10 days, which was rescued by a second MPL injection 10 days after the first MPL injection or by a 4× MPL injection 10 days before stress exposure. A single MPL injection (800 μg/kg) before stress exposure prevented CSDS-induced increases in the gene expression levels of pro-inflammatory cytokines in the hippocampus and prefrontal cortex. Pre-inhibiting the innate immune stimulation by minocycline pretreatment (40 mg/kg) abrogated the preventive effect of MPL on CSDS-induced depression-like behaviors and neuroinflammatory responses in animal brains. CONCLUSIONS MPL, through innate immune stimulation, prevents stress-induced depression-like behaviors in mice by preventing neuroinflammatory responses.
Collapse
Affiliation(s)
| | | | | | | | - Ting Ye
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chao Huang
- Correspondence: Chao Huang, Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China ()
| |
Collapse
|
12
|
Eskilsson A, Shionoya K, Engblom D, Blomqvist A. Fever During Localized Inflammation in Mice Is Elicited by a Humoral Pathway and Depends on Brain Endothelial Interleukin-1 and Interleukin-6 Signaling and Central EP 3 Receptors. J Neurosci 2021; 41:5206-5218. [PMID: 33941650 PMCID: PMC8211540 DOI: 10.1523/jneurosci.0313-21.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/01/2021] [Accepted: 04/26/2021] [Indexed: 02/02/2023] Open
Abstract
We examined the signaling route for fever during localized inflammation in male and female mice, elicited by casein injection into a preformed air pouch. The localized inflammation gave rise to high concentrations of prostaglandins of the E species (PGE2) and cytokines in the air pouch and elevated levels of these inflammatory mediators in plasma. There were also elevated levels of PGE2 in the cerebrospinal fluid, although there was little evidence for PGE2 synthesis in the brain. Global deletion of the PGE2 prostaglandin E receptor 3 (EP3) abolished the febrile response as did deletion of the EP3 receptor in neural cells, whereas its deletion on peripheral nerves had no effect, implying that PGE2 action on this receptor in the CNS elicited the fever. Global deletion of the interleukin-1 receptor type 1 (IL-1R1) also abolished the febrile response, whereas its deletion on neural cells or peripheral nerves had no effect. However, deletion of the IL-1R1 on brain endothelial cells, as well as deletion of the interleukin-6 receptor α on these cells, attenuated the febrile response. In contrast, deletion of the PGE2 synthesizing enzymes cyclooxygenase-2 and microsomal prostaglandin synthase-1 in brain endothelial cells, known to attenuate fever evoked by systemic inflammation, had no effect. We conclude that fever during localized inflammation is not mediated by neural signaling from the inflamed site, as previously suggested, but is dependent on humoral signaling that involves interleukin actions on brain endothelial cells, probably facilitating PGE2 entry into the brain from the circulation and hence representing a mechanism distinct from that at work during systemic inflammation.
Collapse
Affiliation(s)
- Anna Eskilsson
- Division of Neurobiology and Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, S-58185 Linköping, Sweden
| | - Kiseko Shionoya
- Division of Neurobiology and Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, S-58185 Linköping, Sweden
| | - David Engblom
- Division of Neurobiology and Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, S-58185 Linköping, Sweden
| | - Anders Blomqvist
- Division of Neurobiology and Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, S-58185 Linköping, Sweden
| |
Collapse
|
13
|
Jin K, Lu J, Yu Z, Shen Z, Li H, Mou T, Xu Y, Huang M. Linking peripheral IL-6, IL-1β and hypocretin-1 with cognitive impairment from major depression. J Affect Disord 2020; 277:204-211. [PMID: 32829196 DOI: 10.1016/j.jad.2020.08.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 07/06/2020] [Accepted: 08/09/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cognitive impairment has long challenged the patients with major depressive disorder (MDD), hypocretins and inflammation have recently been implicated in cognitive function. However, limited studies have compressively assessed their associations with cognitive impairment in MDD. METHODS A total of 100 MDD patients and 100 healthy controls (HC) were recruited for this study. They were tested with HAMD, HAMA, and MCCB scales. The plasma level of selected inflammatory factors (IL-1β, IL-6, and TNF-α) and hypocretin-1 were determined using enzyme-linked immunosorbent assay (ELISA). Correlation analysis was performed to explore the relationship between the plasma level of the factors and clinical performances. RESULTS Patients with MDD showed cognitive impairment in each MCCB subdomain except working memory compared with HC. The levels of IL-6, IL-1β and hypocretin-1 in MDD patients were higher than HC. Besides, IL-1β levels was negatively correlated with overall cognitive function in the combined group. Hypocretin-1 was positively correlated with socially cognitive impairment in MDD patients. A negative correlation between plasma hypocretin-1 levels and HAMA scales was also observed in MDD patients. LIMITATION The study was cross-sectional, thereby limiting causal inference, and had a relatively small sample size. There are no subcategories for MDD based on characteristics. CONCLUSION IL-1β, IL-6 and Hypocretin-1 were reported as potential factors involved in MDD pathology. Hypocretin-1 could contribute to the biological mechanisms of anxiety relief. Hypocretin-1, therefore, may be important in exploring the pathological mechanisms of social cognitive impairment in MDD patients. Conclusively, this study provides new insights for exploring cognitive impairment in depression.
Collapse
Affiliation(s)
- Kangyu Jin
- Department of Psychiatry, the First Affiliated Hospital, Zhejjiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Jing Lu
- Department of Psychiatry, the First Affiliated Hospital, Zhejjiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Zhebin Yu
- Department of Epidemiology and Health Statistics, Zhejiang University School of Public Health
| | - Zhe Shen
- Department of Psychiatry, the First Affiliated Hospital, Zhejjiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Haimei Li
- Department of Psychiatry, the First Affiliated Hospital, Zhejjiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Tingting Mou
- Department of Psychiatry, the First Affiliated Hospital, Zhejjiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Yi Xu
- Department of Psychiatry, the First Affiliated Hospital, Zhejjiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China.
| | - Manli Huang
- Department of Psychiatry, the First Affiliated Hospital, Zhejjiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China.
| |
Collapse
|
14
|
Böttcher M, Müller-Fielitz H, Sundaram SM, Gallet S, Neve V, Shionoya K, Zager A, Quan N, Liu X, Schmidt-Ullrich R, Haenold R, Wenzel J, Blomqvist A, Engblom D, Prevot V, Schwaninger M. NF-κB signaling in tanycytes mediates inflammation-induced anorexia. Mol Metab 2020; 39:101022. [PMID: 32446877 PMCID: PMC7292913 DOI: 10.1016/j.molmet.2020.101022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Infections, cancer, and systemic inflammation elicit anorexia. Despite the medical significance of this phenomenon, the question of how peripheral inflammatory mediators affect the central regulation of food intake is incompletely understood. Therefore, we have investigated the sickness behavior induced by the prototypical inflammatory mediator IL-1β. METHODS IL-1β was injected intravenously. To interfere with IL-1β signaling, we deleted the essential modulator of NF-κB signaling (Nemo) in astrocytes and tanycytes. RESULTS Systemic IL-1β increased the activity of the transcription factor NF-κB in tanycytes of the mediobasal hypothalamus (MBH). By activating NF-κB signaling, IL-1β induced the expression of cyclooxygenase-2 (Cox-2) and stimulated the release of the anorexigenic prostaglandin E2 (PGE2) from tanycytes. When we deleted Nemo in astrocytes and tanycytes, the IL-1β-induced anorexia was alleviated whereas the fever response and lethargy response were unchanged. Similar results were obtained after the selective deletion of Nemo exclusively in tanycytes. CONCLUSIONS Tanycytes form the brain barrier that mediates the anorexic effect of systemic inflammation in the hypothalamus.
Collapse
Affiliation(s)
- Mareike Böttcher
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany
| | - Helge Müller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Sivaraj M Sundaram
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany
| | - Sarah Gallet
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Lille, France; University of Lille, FHU 1000 days for Health, School of Medicine, U1172, Lille, France
| | - Vanessa Neve
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany
| | - Kiseko Shionoya
- Department of Clinical and Experimental Medicine, Linköping University, S-581 85, Linköping, Sweden
| | - Adriano Zager
- Department of Clinical and Experimental Medicine, Linköping University, S-581 85, Linköping, Sweden
| | - Ning Quan
- Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Xiaoyu Liu
- Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Ruth Schmidt-Ullrich
- Department of Signal Transduction in Tumor Cells, Max-Delbrück-Center (MDC) for Molecular Medicine, 13125, Berlin, Germany
| | - Ronny Haenold
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), 07745, Jena, Germany; Matthias Schleiden Institute of Genetics, Bioinformatics and Molecular Botany, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Jan Wenzel
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Anders Blomqvist
- Department of Clinical and Experimental Medicine, Linköping University, S-581 85, Linköping, Sweden
| | - David Engblom
- Department of Clinical and Experimental Medicine, Linköping University, S-581 85, Linköping, Sweden
| | - Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Lille, France; University of Lille, FHU 1000 days for Health, School of Medicine, U1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany.
| |
Collapse
|
15
|
Oliveira MK, dos Santos RS, Cabral LD, Vilela FC, Giusti-Paiva A. Simvastatin attenuated sickness behavior and fever in a murine model of endotoxemia. Life Sci 2020; 254:117701. [DOI: 10.1016/j.lfs.2020.117701] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 01/01/2023]
|
16
|
Pinteaux E, Abdulaal WH, Mufazalov IA, Humphreys NE, Simonsen-Jackson M, Francis S, Müller W, Waisman A. Cell-specific conditional deletion of interleukin-1 (IL-1) ligands and its receptors: a new toolbox to study the role of IL-1 in health and disease. J Mol Med (Berl) 2020; 98:923-930. [PMID: 32468079 PMCID: PMC7343756 DOI: 10.1007/s00109-020-01928-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 01/08/2023]
Abstract
The pro-inflammatory cytokine interleukin-1 (IL-1) plays a key role in many physiological processes and during the inflammatory and immune response to most common diseases. IL-1 exists as two agonists, IL-1α and IL-1β that bind to the only signaling IL-1 type 1 receptor (IL-1R1), while a second decoy IL-1 type 2 receptor (IL-1R2) binds both forms of IL-1 without inducing cell signaling. The field of immunology and inflammation research has, over the past 35 years, unraveled many mechanisms of IL-1 actions, through in vitro manipulation of the IL-1 system or by using genetically engineered mouse models that lack either member of the IL-1 family in ubiquitous constitutive manner. However, the limitation of global mouse knockout technology has significantly hampered our understanding of the precise mechanisms of IL-1 actions in animal models of disease. Here we report and review the recent generation of new conditional mouse mutants in which exons of Il1a, Il1b, Il1r1, and Il1r2 genes flanked by loxP sites (fl/fl) can be deleted in cell-/tissue-specific constitutive or inducible manner by Cre recombinase expression. Hence, IL-1αfl/fl, IL-1βfl/fl, IL-1R1fl/fl, and IL-1R2fl/fl mice constitute a new toolbox that will provide a step change in our understanding of the cell-specific role of IL-1 and its receptor in health and disease and the potential development of targeted IL-1 therapies.
Collapse
Affiliation(s)
- Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom.
| | - Wesam H Abdulaal
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, P.O.BOX 80203, Jeddah, 21589, Kingdom of Saudi Arabia
| | - Ilgiz A Mufazalov
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Langenbeckstrasse 1, Building 308A, 55131, Mainz, Germany
| | - Neil E Humphreys
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
- Epigenetics and Neurobiology Unit, Adriano Buzzati-Traverso Campus, EMBL-Rome, Via Ramarini, 3200015, Monterotondo, RM, Italy
| | - Maj Simonsen-Jackson
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Sheila Francis
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, University of Sheffield, S10 2RX, Sheffield, United Kingdom
| | - Werner Müller
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Langenbeckstrasse 1, Building 308A, 55131, Mainz, Germany
| |
Collapse
|
17
|
Acute Inflammation Alters Brain Energy Metabolism in Mice and Humans: Role in Suppressed Spontaneous Activity, Impaired Cognition, and Delirium. J Neurosci 2020; 40:5681-5696. [PMID: 32513828 PMCID: PMC7363463 DOI: 10.1523/jneurosci.2876-19.2020] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 01/09/2023] Open
Abstract
Systemic infection triggers a spectrum of metabolic and behavioral changes, collectively termed sickness behavior, which while adaptive, can affect mood and cognition. In vulnerable individuals, acute illness can also produce profound, maladaptive, cognitive dysfunction including delirium, but our understanding of delirium pathophysiology remains limited. Here, we used bacterial lipopolysaccharide (LPS) in female C57BL/6J mice and acute hip fracture in humans to address whether disrupted energy metabolism contributes to inflammation-induced behavioral and cognitive changes. LPS (250 µg/kg) induced hypoglycemia, which was mimicked by interleukin (IL)-1β (25 µg/kg) but not prevented in IL-1RI−/− mice, nor by IL-1 receptor antagonist (IL-1RA; 10 mg/kg). LPS suppression of locomotor activity correlated with blood glucose concentrations, was mitigated by exogenous glucose (2 g/kg), and was exacerbated by 2-deoxyglucose (2-DG) glycolytic inhibition, despite preventing IL-1β synthesis. Using the ME7 model of chronic neurodegeneration in female mice, to examine vulnerability of the diseased brain to acute stressors, we showed that LPS (100 µg/kg) produced acute cognitive dysfunction, selectively in those animals. These acute cognitive impairments were mimicked by insulin (11.5 IU/kg) and mitigated by glucose, demonstrating that acutely reduced glucose metabolism impairs cognition selectively in the vulnerable brain. To test whether these acute changes might predict altered carbohydrate metabolism during delirium, we assessed glycolytic metabolite levels in CSF in humans during inflammatory trauma-induced delirium. Hip fracture patients showed elevated CSF lactate and pyruvate during delirium, consistent with acutely altered brain energy metabolism. Collectively, the data suggest that disruption of energy metabolism drives behavioral and cognitive consequences of acute systemic inflammation. SIGNIFICANCE STATEMENT Acute systemic inflammation alters behavior and produces disproportionate effects, such as delirium, in vulnerable individuals. Delirium has serious short and long-term sequelae but mechanisms remain unclear. Here, we show that both LPS and interleukin (IL)-1β trigger hypoglycemia, reduce CSF glucose, and suppress spontaneous activity. Exogenous glucose mitigates these outcomes. Equivalent hypoglycemia, induced by lipopolysaccharide (LPS) or insulin, was sufficient to trigger cognitive impairment selectively in animals with existing neurodegeneration and glucose also mitigated those impairments. Patient CSF from inflammatory trauma-induced delirium also shows altered brain carbohydrate metabolism. The data suggest that the degenerating brain is exquisitely sensitive to acute behavioral and cognitive consequences of disrupted energy metabolism. Thus “bioenergetic stress” drives systemic inflammation-induced dysfunction. Elucidating this may offer routes to mitigating delirium.
Collapse
|
18
|
Nutritional psychoneuroimmunology: Is the inflammasome a critical convergence point for stress and nutritional dysregulation? Curr Opin Behav Sci 2019; 28:20-24. [PMID: 31667204 DOI: 10.1016/j.cobeha.2019.01.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Psychoneuroimmunology (PNI) aims to elucidate mechanisms by which the immune system can influence behavior. Given the complexity of the brain, studies using inbred rodents have shed critical insight into the presumed vagaries of the human condition. This is particularly true for stress modeling where adverse stimuli, conditions and/or interactions elicit patterned behavioral reactions that can translate across species. As example, sickness behaviors are as easily recognized in mice as they are in humans, and a family pet. Recently, nutrition has gained prominence as a regulator of brain function. Once perceived as mostly a peripheral player, except when manifest at extremes like starvation or gluttony, nutritional and/or metabolic stress is now recognized as a worrisome contributor to poor mental health especially in those who suffer from food insecurity or overnutrition. In this review, we will explore emerging areas of rodent research that demonstrate the impact of nutritional status on the stressed brain. Our overall goal is to implicate inflammasome activation as a critical convergence point for stress and nutritional dysregulation. In doing so, we will present results from studies focused on macronutrient, micronutrient and dietary bioactives so as to encourage innovative investigation into the emerging field of nutritional PNI.
Collapse
|
19
|
Chaskiel L, Bristow AD, Bluthé RM, Dantzer R, Blomqvist A, Konsman JP. Interleukin-1 reduces food intake and body weight in rat by acting in the arcuate hypothalamus. Brain Behav Immun 2019; 81:560-573. [PMID: 31310797 DOI: 10.1016/j.bbi.2019.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022] Open
Abstract
A reduction in food intake is commonly observed after bacterial infection, a phenomenon that can be reproduced by peripheral administration of Gram-negative bacterial lipopolysaccharide (LPS) or interleukin-1beta (IL-1β), a pro-inflammatory cytokine released by LPS-activated macrophages. The arcuate nucleus of the hypothalamus (ARH) plays a major role in food intake regulation and expresses IL-1 type 1 receptor (IL-1R1) mRNA. In the present work, we tested the hypothesis that IL-1R1 expressing cells in the ARH mediate IL-1β and/or LPS-induced hypophagia in the rat. To do so, we developed an IL-1β-saporin conjugate, which eliminated IL-R1-expressing neurons in the hippocampus, and micro-injected it into the ARH prior to systemic IL-1β and LPS administration. ARH IL-1β-saporin injection resulted in loss of neuropeptide Y-containing cells and attenuated hypophagia and weight loss after intraperitoneal IL-1β, but not LPS, administration. In conclusion, the present study shows that ARH NPY-containing neurons express functional IL-1R1s that mediate peripheral IL-1β-, but not LPS-, induced hypophagia. Our present and previous findings indicate that the reduction of food intake after IL-1β and LPS are mediated by different neural pathways.
Collapse
Affiliation(s)
- Léa Chaskiel
- Psychoneuroimmunology, Nutrition and Genetics, UMR CNRS 5226-INRA 1286, University of Bordeaux, 33076 Bordeaux, France
| | - Adrian D Bristow
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, UK
| | - Rose-Marie Bluthé
- Psychoneuroimmunology, Nutrition and Genetics, UMR CNRS 5226-INRA 1286, University of Bordeaux, 33076 Bordeaux, France
| | - Robert Dantzer
- Department of Symptom Research, MD Anderson Cancer Center, The University of Texas, Houston, TX 770030, USA
| | - Anders Blomqvist
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - Jan Pieter Konsman
- UMR CNRS 5287 Aquitaine Institute for Integrative and Cognitive Neuroscience, University of Bordeaux, 33076 Bordeaux, France.
| |
Collapse
|
20
|
Sheerin D, O'Connor D, Dold C, Clutterbuck E, Attar M, Rollier CS, Sadarangani M, Pollard AJ. Comparative transcriptomics between species attributes reactogenicity pathways induced by the capsular group B meningococcal vaccine, 4CMenB, to the membrane-bound endotoxin of its outer membrane vesicle component. Sci Rep 2019; 9:13797. [PMID: 31551511 PMCID: PMC6760121 DOI: 10.1038/s41598-019-50310-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/02/2019] [Indexed: 12/18/2022] Open
Abstract
The capsular group B meningococcal (MenB) four component vaccine (4CMenB) has been licensed for the prevention of invasive disease caused by MenB. The vaccine causes fever in infants, particularly when given in combination (concomitant) with other routinely-administered vaccines (routine), such as the standard diphtheria, tetanus, pertussis (DTP)-containing vaccine. To assess the suitability of a mouse immunisation model to study this phenomenon, we monitored temperature in mice after a second dose of routine vaccines, with or without 4CMenB, and compared the results with those in humans. Using this mouse model, we explored the reactogenicity of 4CMenB components by measuring changes in temperature, cytokines, and gene expression induced by 4CMenB, one of its components, wild-type or attenuated endotoxin outer membrane vesicles (OMVs), or lipopolysaccharide (LPS). A significant rise (p < 0.01) in temperature was observed in mice immunised with 4CMenB, wild-type OMVs, and LPS. RNA-sequencing of mouse whole blood revealed a gene signature shared by the 4CMenB, OMV, and LPS groups consisting of bacterial pattern recognition receptors and neutrophil activation marker genes. Sequencing of neutrophils isolated after concomitant 4CMenB identified cells expressing the OMV-associated genes Plek and Lcp1. Immunisation with 4CMenB or OMVs led to increased IL-6 in serum and significant upregulation (p < 0.0001) of prostaglandin-synthesising enzymes on brain tissue. These data demonstrate the suitability of a mouse model for assessing vaccine reactogenicity and strongly indicate that the fever following vaccination with 4CMenB in human infants is induced by endotoxin contained in the OMV component of the vaccine.
Collapse
Affiliation(s)
- Dylan Sheerin
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK.
| | - Daniel O'Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Elizabeth Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Moustafa Attar
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| |
Collapse
|
21
|
Hocker AD, Huxtable AG. IL-1 receptor activation undermines respiratory motor plasticity after systemic inflammation. J Appl Physiol (1985) 2018; 125:504-512. [PMID: 29565772 DOI: 10.1152/japplphysiol.01051.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inflammation undermines respiratory motor plasticity, yet we are just beginning to understand the inflammatory signaling involved. Because interleukin-1 (IL-1) signaling promotes or inhibits plasticity in other central nervous system regions, we tested the following hypotheses: 1) IL-1 receptor (IL-1R) activation after systemic inflammation is necessary to undermine phrenic long-term facilitation (pLTF), a model of respiratory motor plasticity induced by acute intermittent hypoxia (AIH), and 2) spinal IL-1β is sufficient to undermine pLTF. pLTF is significantly reduced 24 h after lipopolysaccharide (LPS; 100 μg/kg ip, 12 ± 18%, n = 5) compared with control (57 ± 25%, n = 6) and restored by peripheral IL-1R antagonism (63 ± 13%, n = 5, AF-12198, 0.5 mg/kg ip, 24 h). Furthermore, acute, spinal IL-1R antagonism (1 mM AF-12198, 15 μl it) restored pLTF (53 ± 15%, n = 4) compared with LPS-treated rats (11 ± 10%; n = 5), demonstrating IL-1R activation is necessary to undermine pLTF after systemic inflammation. However, in healthy animals, pLTF persisted after spinal, exogenous recombinant rat IL-1β (rIL-1β) (1 ng ± AIH; 66 ± 26%, n = 3, 10 ng ± AIH; 102 ± 49%, n = 4, 100 ng + AIH; 93 ± 51%, n = 3, 300 ng ± AIH; 37 ± 40%, n = 3; P < 0.05 from baseline). In the absence of AIH, spinal rIL-1β induced progressive, dose-dependent phrenic amplitude facilitation (1 ng; −3 ± 5%, n = 3, 10 ng; 8 ± 22%, n = 3, 100 ng; 31 ± 12%, P < 0.05, n = 4, 300 ng; 51 ± 17%, P < 0.01 from baseline, n = 4). In sum, IL-1R activation, both systemically and spinally, undermines pLTF after LPS-induced systemic inflammation, but IL-1R activation is not sufficient to abolish plasticity. Understanding the inflammatory signaling inhibiting respiratory plasticity is crucial to developing treatment strategies utilizing respiratory plasticity to promote breathing during ventilatory control disorders.NEW & NOTEWORTHY This study gives novel insights concerning mechanisms by which systemic inflammation undermines respiratory motor plasticity. We demonstrate that interleukin-1 signaling, both peripherally and centrally, undermines respiratory motor plasticity. However, acute, exogenous interleukin-1 signaling is not sufficient to undermine respiratory motor plasticity.
Collapse
Affiliation(s)
- Austin D. Hocker
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | | |
Collapse
|
22
|
Abstract
Fever is a common symptom of infectious and inflammatory disease. It is well-established that prostaglandin E2 is the final mediator of fever, which by binding to its EP3 receptor subtype in the preoptic hypothalamus initiates thermogenesis. Here, we review the different hypotheses on how the presence of peripherally released pyrogenic substances can be signaled to the brain to elicit fever. We conclude that there is unequivocal evidence for a humoral signaling pathway by which proinflammatory cytokines, through their binding to receptors on brain endothelial cells, evoke fever by eliciting prostaglandin E2 synthesis in these cells. The evidence for a role for other signaling routes for fever, such as signaling via circumventricular organs and peripheral nerves, as well as transfer into the brain of peripherally synthesized prostaglandin E2 are yet far from conclusive. We also review the efferent limb of the pyrogenic pathways. We conclude that it is well established that prostaglandin E2 binding in the preoptic hypothalamus produces fever by disinhibition of presympathetic neurons in the brain stem, but there is yet little understanding of the mechanisms by which factors such as nutritional status and ambient temperature shape the response to the peripheral immune challenge.
Collapse
Affiliation(s)
- Anders Blomqvist
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health, Linköping University, Linköping, Sweden
| | - David Engblom
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health, Linköping University, Linköping, Sweden
| |
Collapse
|