51
|
Aida-Yasuoka K, Nishimura N, Fujisawa N, Endo N, Narumiya S, Tohyama C. The role of prostaglandin E 2 receptor EP1 in 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced neonatal hydronephrosis in mice. Toxicology 2019; 415:10-17. [PMID: 30641090 DOI: 10.1016/j.tox.2019.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 10/27/2022]
Abstract
Prostaglandin E2 (PGE2) is a critical factor in the pathogenesis of dioxin-induced neonatal hydronephrosis. Since the PGE2 receptor has four subtypes, EP1 - EP4, this study was aimed to challenge the hypothesis that at least one of the four subtypes is responsible for the pathogenesis of dioxin-induced hydronephrosis. To this end, we used mouse pups, with a C57BL/6 J background, genetically lacking EP1, EP2, or EP3, and wild-type pups in whom EP4 was suppressed by administering ONO-AE3-208 (ONO), an EP4 antagonist, from postnatal day 1 (PND 1) to PND 13. To expose the pups to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) via lactation, the dams were administered TCDD at an oral dose of 20 μg/kg on PND 1. The pups' urine and kidneys were collected on PND 14 for urinalysis and histological examination, respectively. We found that the incidence of hydronephrosis was 80% in the EP1+/+ group, but was markedly reduced to 28.6% in the EP1-/- group despite the fact that PGE2 concentration in the urine was similarly increased in the both groups. In contrast, the incidence of hydronephrosis was 80% and 100% in the EP2+/+ and EP2-/-groups, respectively, and 88.9% and 100% in the EP3+/+ and EP3-/- groups, respectively. With regard to EP4, the incidence of hydronephrosis in vehicle (saline)-treated groups and ONO-treated was 88.9% and 100%, respectively. Therefore, we concluded that among PGE2 receptor subtypes, EP1 plays a predominant role in the onset of TCDD-induced neonatal hydronephrosis in mouse pups.
Collapse
Affiliation(s)
- Keiko Aida-Yasuoka
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Noriko Nishimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Nozomi Fujisawa
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Nozomi Endo
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan.
| |
Collapse
|
52
|
Chang RB. Body thermal responses and the vagus nerve. Neurosci Lett 2019; 698:209-216. [PMID: 30634012 PMCID: PMC7061531 DOI: 10.1016/j.neulet.2019.01.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 12/17/2022]
Abstract
While thermosensation from external environment has been extensively studied, physiological responses to temperature changes inside the body and the underlying regulatory mechanisms are less understood. As a critical link between body and brain that relays visceral organ information and regulates numerous physiological functions, the vagus nerve has been proposed to mediate diverse visceral thermal reflexes and indirectly regulate body temperature. However, the precise role of the vagus nerve in body thermal responses or visceral organ-related thermoregulation is still under debate due to extensive contradictory results. This data discrepancy is likely due to the high cell heterogeneity in the vagus nerve, as diverse vagal neuron types mediate numerous and sometimes opposite physiological functions. Here, we will review evidences that support and against the role of the vagus nerve in body thermosensation and thermoregulation and discuss potential future approaches for better understanding of this critical issue.
Collapse
Affiliation(s)
- Rui B Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06520, United States.
| |
Collapse
|
53
|
Maurer SF, Dieckmann S, Kleigrewe K, Colson C, Amri EZ, Klingenspor M. Fatty Acid Metabolites as Novel Regulators of Non-shivering Thermogenesis. Handb Exp Pharmacol 2019; 251:183-214. [PMID: 30141101 DOI: 10.1007/164_2018_150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Fatty acids are essential contributors to adipocyte-based non-shivering thermogenesis by acting as activators of uncoupling protein 1 and serving as fuel for mitochondrial heat production. Novel evidence suggests a contribution to this thermogenic mechanism by their conversion to bioactive compounds. Mammalian cells produce a plethora of oxylipins and endocannabinoids, some of which have been identified to affect the abundance or thermogenic activity of brown and brite adipocytes. These effectors are produced locally or at distant sites and signal toward thermogenic adipocytes via a direct interaction with these cells or indirectly via secondary mechanisms. These interactions are evoked by the activation of receptor-mediated pathways. The endogenous production of these compounds is prone to modulation by the dietary intake of the respective precursor fatty acids. The effect of nutritional interventions on uncoupling protein 1-derived thermogenesis may thus at least in part be conferred by the production of a supportive oxylipin and endocannabinoid profile. The manipulation of this system in future studies will help to elucidate the physiological potential of these compounds as novel, endogenous regulators of non-shivering thermogenesis.
Collapse
Affiliation(s)
- Stefanie F Maurer
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany.
- ZIEL Institute for Food and Health, TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
| | - Sebastian Dieckmann
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL Institute for Food and Health, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich, Freising, Germany
| | | | | | - Martin Klingenspor
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL Institute for Food and Health, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| |
Collapse
|
54
|
Abstract
Maintenance of a homeostatic body core temperature is a critical brain function accomplished by a central neural network. This orchestrates a complex behavioral and autonomic repertoire in response to environmental temperature challenges or declining energy homeostasis and in support of immune responses and many behavioral states. This review summarizes the anatomical, neurotransmitter, and functional relationships within the central neural network that controls the principal thermoeffectors: cutaneous vasoconstriction regulating heat loss and shivering and brown adipose tissue for heat production. The core thermoregulatory network regulating these thermoeffectors consists of parallel but distinct central efferent pathways that share a common peripheral thermal sensory input. Delineating the neural circuit mechanism underlying central thermoregulation provides a useful platform for exploring its functional organization, elucidating the molecular underpinnings of its neuronal interactions, and discovering novel therapeutic approaches to modulating body temperature and energy homeostasis.
Collapse
Affiliation(s)
- S F Morrison
- Department of Neurological Surgery, Oregon Health and Science University, Portland, Oregon 97239, USA;
| | - K Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
55
|
Attiq A, Jalil J, Husain K, Ahmad W. Raging the War Against Inflammation With Natural Products. Front Pharmacol 2018; 9:976. [PMID: 30245627 PMCID: PMC6137277 DOI: 10.3389/fphar.2018.00976] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/08/2018] [Indexed: 12/31/2022] Open
Abstract
Over the last few decade Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) are the drugs of choice for treating numerous inflammatory diseases including rheumatoid arthritis. The NSAIDs produces anti-inflammatory activity via inhibiting cyclooxygenase enzyme, responsible for the conversation of arachidonic acid to prostaglandins. Likewise, cyclooxegenase-2 inhibitors (COX-2) selectively inhibit the COX-2 enzyme and produces significant anti-inflammatory, analgesic, and anti-pyretic activity without producing COX-1 associated gastrointestinal and renal side effects. In last two decades numerous selective COX-2 inhibitors (COXIBs) have been developed and approved for various inflammatory conditions. However, data from clinical trials have suggested that the prolong use of COX-2 inhibitors are also associated with life threatening cardiovascular side effects including ischemic heart failure and myocardial infection. In these scenario secondary metabolites from natural product offers a great hope for the development of novel anti-inflammatory compounds. Although majority of the natural product based compounds exhibit more selectively toward COX-1. However, the data suggest that slight structural modification can be helpful in developing COX-2 selective secondary metabolites with comparative efficacy and limited side effects. This review is an effort to highlight the secondary metabolites from terrestrial and marine source with significant COX-2 and COX-2 mediated PGE2 inhibitory activity, since it is anticipated that isolates with ability to inhibit COX-2 mediated PGE2 production would be useful in suppressing the inflammation and its classical sign and symptoms. Moreover, this review has highlighted the potential lead compounds including berberine, kaurenoic acid, α-cyperone, curcumin, and zedoarondiol for further development with the help of structure-activity relationship (SAR) studies and their current status.
Collapse
Affiliation(s)
- Ali Attiq
- Drug and Herbal Research Centre, Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Juriyati Jalil
- Drug and Herbal Research Centre, Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Khairana Husain
- Drug and Herbal Research Centre, Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Waqas Ahmad
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Malaysia
| |
Collapse
|
56
|
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
|
57
|
Firmino M, Weis SN, Souza JMF, Gomes BRB, Mól AR, Mortari MR, Souza GEP, Coca GC, Williams TCR, Fontes W, Ricart CAO, de Sousa MV, Veiga-Souza FH. Label-free quantitative proteomics of rat hypothalamus under fever induced by LPS and PGE 2. J Proteomics 2018; 187:182-199. [PMID: 30056254 DOI: 10.1016/j.jprot.2018.07.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/13/2018] [Accepted: 07/24/2018] [Indexed: 12/21/2022]
Abstract
Fever is a brain-mediated increase in body temperature mainly during inflammatory or infectious challenges. Although there is considerable data regarding the inflammation pathways involved in fever, metabolic alterations necessary to orchestrate the complex inflammatory response are not totally understood. We performed proteomic analysis of rat hypothalamus using label-free LC-MS/MS in a model of fever induced by lipopolysaccharide (LPS) or prostaglandin E2 (PGE2). In total, 7021 proteins were identified. As far as we know, this is the largest rat hypothalamus proteome dataset available to date. Pathway analysis showed proteins from both stimuli associated with inflammatory and metabolic pathways. Concerning metabolic pathways, rats exposed to LPS or PGE2 presented lower relative abundance of proteins involved in glycolysis, pentose phosphate pathway and tricarboxylic acid cycle. Mitochondrial function may also be altered by both stimuli because significant downregulation of several proteins was found, mainly in complexes I and IV. LPS was able to induce downregulation of important proteins in the enzymatic antioxidant system, thereby contributing to oxidative stress. The results offered comprehensive information about fever responses and helped to reveal new insights into proteins potentially involved in inflammatory signaling and metabolic changes in the hypothalamus during systemic LPS and central PGE2 administration. SIGNIFICANCE The evolutionary persistence of fever, despite the elevated cost for maintenance of this response, suggests that elevation in core temperature may represent an interesting strategy for survival. Fever response is achieved through the integrated behavioral, physiological, immunological and biochemical processes that determine the balance between heat generation and elimination. The development of such complex response arouses interest in studying how the cell metabolism responds or even contributes to promote fever. Our results offered comprehensive information about fever responses, including metabolic and inflammatory pathways, providing new insights into candidate proteins potentially involved in inflammatory signaling and metabolic changes in the hypothalamus during fever induced by systemic LPS and central PGE2 perturbation.
Collapse
Affiliation(s)
- Marina Firmino
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Simone N Weis
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Jaques M F Souza
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Bruna R B Gomes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Alan R Mól
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Márcia R Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Gloria E P Souza
- Laboratory of Pharmacology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-903, Brazil
| | - Guilherme C Coca
- Laboratory of Plant Biochemistry, Department of Botany, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Thomas C R Williams
- Laboratory of Plant Biochemistry, Department of Botany, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Carlos André O Ricart
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, DF 70910-900, Brazil
| | - Marcelo V de Sousa
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, DF 70910-900, Brazil.
| | - Fabiane H Veiga-Souza
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, DF 70910-900, Brazil; School of Ceilandia, University of Brasilia, Brasília, DF 72220-275, Brazil.
| |
Collapse
|
58
|
Yamagishi-Kimura R, Honjo M, Aihara M. Contribution of prostanoid FP receptor and prostaglandins in transient inflammatory ocular hypertension. Sci Rep 2018; 8:11098. [PMID: 30038227 PMCID: PMC6056481 DOI: 10.1038/s41598-018-29273-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/07/2018] [Indexed: 11/30/2022] Open
Abstract
We explored the involvement of FP receptor and endogenous prostaglandins (PGs) in transient ocular hypertension (OH) induced by PGE2 or PGF2α in mouse eyes. PGE2 and PGF2α were topically applied to induce transient OH in Wild-type (WT) and FP-, EP1-, EP2-, and EP3-deficient (knockout [KO]) mice. To suppress endogenous PG production, the non-steroidal anti-inflammatory drug nepafenac was applied topically before treatment. PGE2 and PGF2α induced significant OH in the WT, FPKO, and EP1–3KO mice compared to the control 30 min after instillation, and the increase in IOP at 30 or 60 min after instillation in FPKO mice was significantly higher than that in the WT mice. The effects of PGF2α on the increase in IOP were significantly weaker than those of PGE2, especially in EP1KO and EP3KO mice. Transient OH induced by PGE2 and PGF2α was significantly attenuated by nepafenac treatment in FPKO mice. Transient OH was induced by PGE2 and PGF2α in WT, FPKO, and EP1–3KO mice, which was enhanced in FPKO mice. This OH was significantly diminished by nepafenac treatment in FPKO mice, suggesting that FP receptor may have an important naïve physiological role in the eye, and could regulate IOP elevation during PG-associated ocular inflammation.
Collapse
Affiliation(s)
| | - Megumi Honjo
- Department of Ophthalmology, The University of Tokyo School of Medicine, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, The University of Tokyo School of Medicine, Tokyo, Japan.
| |
Collapse
|
59
|
Pelšs A, Gandhamsetty N, Smith JR, Mailhol D, Silvi M, Watson AJA, Perez-Powell I, Prévost S, Schützenmeister N, Moore PR, Aggarwal VK. Reoptimization of the Organocatalyzed Double Aldol Domino Process to a Key Enal Intermediate and Its Application to the Total Synthesis of Δ 12 -Prostaglandin J 3. Chemistry 2018; 24:9542-9545. [PMID: 29774967 PMCID: PMC6055629 DOI: 10.1002/chem.201802498] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Indexed: 12/15/2022]
Abstract
Re-investigation of the l-proline catalyzed double aldol cascade dimerization of succinaldehyde for the synthesis of a key bicyclic enal intermediate, pertinent in the field of stereoselective prostaglandin synthesis, is reported. The yield of this process has been more than doubled, from 14 % to a 29 % isolated yield on a multi-gram scale (32 % NMR yield), through conducting a detailed study of the reaction solvent, temperature, and concentration, as well as a catalyst screen. The synthetic utility of this enal intermediate has been further demonstrated through the total synthesis of Δ12 -prostaglandin J3 , a compound with known anti-leukemic properties.
Collapse
Affiliation(s)
- Andrejs Pelšs
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, U.K
| | | | - James R Smith
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, U.K
| | - Damien Mailhol
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, U.K
| | - Mattia Silvi
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, U.K
| | - Andrew J A Watson
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, U.K
| | - Isabel Perez-Powell
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, U.K
| | - Sébastien Prévost
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, U.K
| | - Nina Schützenmeister
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, U.K
| | - Peter R Moore
- Pharmaceutical Technology and Development, AstraZeneca, Silk Road Business Park, Charter Way, Macclesfield, SK10 2NA, U.K
| | - Varinder K Aggarwal
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, U.K
| |
Collapse
|
60
|
Bastos-Pereira AL, Fraga D, Dreifuss AA, Zampronio AR. Central mediators of the zymosan-induced febrile response. J Basic Clin Physiol Pharmacol 2018; 28:555-562. [PMID: 28981444 DOI: 10.1515/jbcpp-2017-0061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/15/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Zymosan is a fungal cell wall protein-carbohydrate complex that is known to activate inflammatory pathways through the Toll-like receptors and is commonly used to induce fever. Nevertheless, the central mediators that are involved in the zymosan-induced febrile response are only partially known. METHODS The present study evaluated the participation of prostaglandins, substance P, endothelin-1 (ET-1), and endogenous opioids (eOPs) in the zymosan-induced febrile response by using inhibitors and antagonists in male Wistar rats. RESULTS Both nonselective (indomethacin) and selective (celecoxib) cyclooxygenase inhibitors reduced the febrile response induced by an intraperitoneal (i.p.) injection of zymosan. Indomethacin also blocked the increase in the prostaglandin E2 levels in the cerebrospinal fluid. An intracerebroventricular injection of the neurokinin-1, ETB, and μ-opioid receptor antagonists also reduced the febrile response induced by the i.p. injected zymosan. Moreover, the μ-opioid receptor antagonist CTAP also reduced the febrile response induced by intra-articular injection of zymosan. CONCLUSIONS These results demonstrate that prostaglandins, substance P, ET-1, and eOPs are central mediators of the zymosan-induced febrile response.
Collapse
|
61
|
Screening for the anti-inflammation quality markers of Xiaojin Pills based on HPLC-MS/MS method, COX-2 inhibition test and protein interaction network. Sci Rep 2018; 8:7454. [PMID: 29748583 PMCID: PMC5945850 DOI: 10.1038/s41598-018-25582-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/18/2018] [Indexed: 11/18/2022] Open
Abstract
Nowadays, breast disorders seriously affect women’s health in an increasing number. In China, Xiaojin Pills are commonly used in the treatment of breast diseases. Doctors have concluded that the combined use of Xiaojin Pills with conventional therapy can significantly improve the efficacy with fewer side effects. However, the prescription of Xiaojin Pills is complicated and their quality control methods cannot completely ensure the quality of Xiaojin Pills. On the basis of its mechanism, our study combined chemical evaluation and biological evaluation to identify the anti-inflammatory markers of Xiaojin Pills. In this manuscript, 13 compounds in Xiaojin Pills were quantified. At the same time, the cyclooxygenase-2 inhibition rates of different Xiaojin Pills were measured and the possible markers were screened by spectrum-effect relationship. Further, anti-inflammatory activities of markers were verified and protein interaction network was analyzed, identifying the components of Protocatechuate, Beta-Boswellic acid and Levistilide A as the anti-inflammatory quality markers of Xiaojin Pills. We hope our studies can provide a scientific theoretical basis for accurately quality control of Xiaojin Pills and reasonable suggestions for pharmaceutical companies and new ideas for the quality control of other medicines.
Collapse
|
62
|
Selectively targeting prostanoid E (EP) receptor-mediated cell signalling pathways: Implications for lung health and disease. Pulm Pharmacol Ther 2018; 49:75-87. [DOI: 10.1016/j.pupt.2018.01.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/05/2018] [Accepted: 01/25/2018] [Indexed: 12/18/2022]
|
63
|
Garami A, Steiner AA, Romanovsky AA. Fever and hypothermia in systemic inflammation. HANDBOOK OF CLINICAL NEUROLOGY 2018; 157:565-597. [PMID: 30459026 DOI: 10.1016/b978-0-444-64074-1.00034-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Systemic inflammation-associated syndromes (e.g., sepsis and septic shock) often have high mortality and remain a challenge in emergency medicine. Systemic inflammation is usually accompanied by changes in body temperature: fever or hypothermia. In animal studies, systemic inflammation is often modeled by administering bacterial lipopolysaccharide, which triggers autonomic and behavioral thermoeffector responses and causes either fever or hypothermia, depending on the dose and ambient temperature. Fever and hypothermia are regulated changes of body temperature, which correspond to mild and severe forms of systemic inflammation, respectively. Mediators of fever and hypothermia are called endogenous pyrogens and cryogens; they are produced when the innate immune system recognizes an infectious pathogen. Upon an inflammatory challenge, hepatic and pulmonary macrophages (and later brain endothelial cells) start to release lipid mediators, of which prostaglandin (PG) E2 plays the key role, and cytokines. Blood PGE2 enters the brain and triggers fever. At later stages of fever, PGE2 synthesized within the blood-brain barrier maintains fever. In both cases, PGE2 is synthesized by cyclooxygenase-2 and microsomal PGE2synthase-1. Mediators of hypothermia are not well established. Both fever and hypothermia are beneficial host defense responses. Based on evidence from studies in laboratory animals and clinical trials in humans, fever is beneficial for fighting mild infection. Based mainly on animal studies, hypothermia is beneficial in severe systemic inflammation and infection.
Collapse
Affiliation(s)
- Andras Garami
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary.
| | - Alexandre A Steiner
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Andrej A Romanovsky
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| |
Collapse
|
64
|
ANDO A, SATOMI Y. A Simple and Highly Sensitive Quantitation of Eicosanoids in Biological Samples Using Nano-flow Liquid Chromatography/Mass Spectrometry. ANAL SCI 2018; 34:177-182. [DOI: 10.2116/analsci.34.177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Ayumi ANDO
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited
| | - Yoshinori SATOMI
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited
| |
Collapse
|
65
|
Abstract
Stress affects core body temperature (Tc). Many kinds of stress induce transient, monophasic hyperthermia, which diminishes gradually if the stressor is terminated. Stronger stressors produce a longer-lasting effect. Repeated/chronic stress induces anticipatory hyperthermia, reduces diurnal changes in Tc, or slightly increases Tc throughout the day. Animals that are exposed to chronic stress or a cold environment exhibit an enhanced hyperthermic response to a novel stress. These changes persist for several days after cessation of stress exposure. In contrast, long-lasting inescapable stress sometimes induces hypothermia. In healthy humans, psychologic stress induces slight increases in Tc, which are within the normal range of Tc or just above it. Some individuals, however, develop extremely high Tc (up to 41°C) when they are exposed to emotional events or show persistent low-grade high Tc (37-38°C) during or after chronic stress situations. In addition to the nature of the stressor itself, such stress-induced thermal responses are modulated by sex, age, ambient temperature, cage mates, past stressful experiences and cold exposure, and coping. Stress-induced hyperthermia is driven by mechanisms distinct from infectious fever, which requires inflammatory mediators. However, both stress and infection activate the dorsomedial hypothalamus-rostral medullary raphe region-sympathetic nerve axis to increase Tc.
Collapse
Affiliation(s)
- Takakazu Oka
- Department of Psychosomatic Medicine, International University of Health and Welfare Hospital, Tochigi-ken, Japan.
| |
Collapse
|
66
|
Abstract
Body core temperature of mammals is regulated by the central nervous system, in which the preoptic area (POA) of the hypothalamus plays a pivotal role. The POA receives peripheral and central thermosensory neural information and provides command signals to effector organs to elicit involuntary thermoregulatory responses, including shivering thermogenesis, nonshivering brown adipose tissue thermogenesis, and cutaneous vasoconstriction. Cool-sensory and warm-sensory signals from cutaneous thermoreceptors, monitoring environmental temperature, are separately transmitted through the spinal-parabrachial-POA neural pathways, distinct from the spinothalamocortical pathway for perception of skin temperature. These cutaneous thermosensory inputs to the POA likely impinge on warm-sensitive POA neurons, which monitor body core (brain) temperature, to alter thermoregulatory command outflows from the POA. The cutaneous thermosensory afferents elicit rapid thermoregulatory responses to environmental thermal challenges before they impact body core temperature. Peripheral humoral signals also act on neurons in the POA to transmit afferent information of systemic infection and energy storage to induce fever and to regulate energy balance, respectively. This chapter describes the thermoregulatory afferent mechanisms that convey cutaneous thermosensory signals to the POA and that integrate the neural and humoral afferent inputs to the POA to provide descending command signals to thermoregulatory effectors.
Collapse
Affiliation(s)
- Kazuhiro Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
67
|
Abstract
Lipids are potent signaling molecules that regulate a multitude of cellular responses, including cell growth and death and inflammation/infection, via receptor-mediated pathways. Derived from polyunsaturated fatty acids (PUFAs), such as arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), each lipid displays unique properties, thus making their role in inflammation distinct from that of other lipids derived from the same PUFA. This diversity arises from their synthesis, which occurs via discrete enzymatic pathways and because they elicit responses via different receptors. This review will collate the bioactive lipid research to date and summarize the major pathways involved in their biosynthesis and role in inflammation. Specifically, lipids derived from AA (prostanoids, leukotrienes, 5-oxo-6,8,11,14-eicosatetraenoic acid, lipoxins, and epoxyeicosatrienoic acids), EPA (E-series resolvins), and DHA (D-series resolvins, protectins, and maresins) will be discussed herein.
Collapse
|
68
|
Krasnow SM, Knoll JG, Verghese SC, Levasseur PR, Marks DL. Amplification and propagation of interleukin-1β signaling by murine brain endothelial and glial cells. J Neuroinflammation 2017; 14:133. [PMID: 28668091 PMCID: PMC5494131 DOI: 10.1186/s12974-017-0908-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/25/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND During acute infections and chronic illnesses, the pro-inflammatory cytokine interleukin-1β (IL-1β) acts within the brain to elicit metabolic derangements and sickness behaviors. It is unknown which cells in the brain are the proximal targets for IL-1β with respect to the generation of these illness responses. We performed a series of in vitro experiments to (1) investigate which brain cell populations exhibit inflammatory responses to IL-1β and (2) examine the interactions between different IL-1β-responsive cell types in various co-culture combinations. METHODS We treated primary cultures of murine brain microvessel endothelial cells (BMEC), astrocytes, and microglia with PBS or IL-1β, and then performed qPCR to measure inflammatory gene expression or immunocytochemistry to evaluate nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. To evaluate whether astrocytes and/or BMEC propagate inflammatory signals to microglia, we exposed microglia to astrocyte-conditioned media and co-cultured endothelial cells and glia in transwells. Treatment groups were compared by Student's t tests or by ANOVA followed by Bonferroni-corrected t tests. RESULTS IL-1β increased inflammatory gene expression and NF-κB activation in primary murine-mixed glia, enriched astrocyte, and BMEC cultures. Although IL-1β elicited minimal changes in inflammatory gene expression and did not induce the nuclear translocation of NF-κB in isolated microglia, these cells were more robustly activated by IL-1β when co-cultured with astrocytes and/or BMEC. We observed a polarized endothelial response to IL-1β, because the application of IL-1β to the abluminal endothelial surface produced a more complex microglial inflammatory response than that which occurred following luminal IL-1β exposure. CONCLUSIONS Inflammatory signals are detected, amplified, and propagated through the CNS via a sequential and reverberating signaling cascade involving communication between brain endothelial cells and glia. We propose that the brain's innate immune response differs depending upon which side of the blood-brain barrier the inflammatory stimulus arises, thus allowing the brain to respond differently to central vs. peripheral inflammatory insults.
Collapse
Affiliation(s)
- Stephanie M Krasnow
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, 97239, USA
| | - J Gabriel Knoll
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, 97239, USA
| | - Santhosh Chakkaramakkil Verghese
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, 97239, USA
| | - Peter R Levasseur
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, 97239, USA
| | - Daniel L Marks
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, 97239, USA. .,Oregon Health & Science University, Mail Code L481, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA.
| |
Collapse
|
69
|
LaPorte ME, Weems YS, Arreguin-Arevalo A, Nett TM, Tsutahara N, Sy T, Haberman J, Chon M, Randel RD, Weems CW. Effects of LPA2R, LPA3R, or EP4R agonists on luteal or endometrial function in vivo or in vitro and sirtuin or EP1R, EP2R, EP3R or EP4R agonists on endometrial secretion of PGE and PGF2α in vitro. Theriogenology 2017; 95:8-17. [DOI: 10.1016/j.theriogenology.2017.02.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 01/25/2023]
|
70
|
Singh AK, Zajdel J, Mirrasekhian E, Almoosawi N, Frisch I, Klawonn AM, Jaarola M, Fritz M, Engblom D. Prostaglandin-mediated inhibition of serotonin signaling controls the affective component of inflammatory pain. J Clin Invest 2017; 127:1370-1374. [PMID: 28287401 DOI: 10.1172/jci90678] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/17/2017] [Indexed: 01/03/2023] Open
Abstract
Pain is fundamentally unpleasant and induces a negative affective state. The affective component of pain is mediated by circuits that are distinct from those mediating the sensory-discriminative component. Here, we have investigated the role of prostaglandins in the affective dimension of pain using a rodent pain assay based on conditioned place aversion to formalin injection, an inflammatory noxious stimulus. We found that place aversion induced by inflammatory pain depends on prostaglandin E2 that is synthesized by cyclooxygenase 2 in neural cells. Further, mice lacking the prostaglandin E2 receptor EP3 selectively on serotonergic cells or selectively in the area of the dorsal raphe nucleus failed to form an aversion to formalin-induced pain, as did mice lacking the serotonin transporter. Chemogenetic manipulations revealed that EP3 receptor activation elicited conditioned place aversion to pain via inhibition of serotonergic neurons. In contrast to their role in inflammatory pain aversion, EP3 receptors on serotonergic cells were dispensable for acute nociceptive behaviors and for aversion induced by thermal pain or a κ opioid receptor agonist. Collectively, our findings show that prostaglandin-mediated modulation of serotonergic transmission controls the affective component of inflammatory pain.
Collapse
|
71
|
Nilsson A, Elander L, Hallbeck M, Örtegren Kugelberg U, Engblom D, Blomqvist A. The involvement of prostaglandin E 2 in interleukin-1β evoked anorexia is strain dependent. Brain Behav Immun 2017; 60:27-31. [PMID: 27375005 DOI: 10.1016/j.bbi.2016.06.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/15/2016] [Accepted: 06/28/2016] [Indexed: 01/08/2023] Open
Abstract
From experiments in mice in which the prostaglandin E2 (PGE2) synthesizing enzyme mPGES-1 was genetically deleted, as well as from experiments in which PGE2 was injected directly into the brain, PGE2 has been implicated as a mediator of inflammatory induced anorexia. Here we aimed at examining which PGE2 receptor (EP1-4) that was critical for the anorexic response to peripherally injected interleukin-1β (IL-1β). However, deletion of neither EP receptor in mice, either globally (for EP1, EP2, and EP3) or selectively in the nervous system (EP4), had any effect on the IL-1β induced anorexia. Because these mice were all on a C57BL/6 background, whereas previous observations demonstrating a role for induced PGE2 in IL-1β evoked anorexia had been carried out on mice on a DBA/1 background, we examined the anorexic response to IL-1β in mice with deletion of mPGES-1 on a C57BL/6 background and a DBA/1 background, respectively. We confirmed previous findings that mPGES-1 knock-out mice on a DBA/1 background displayed attenuated anorexia to IL-1β; however, mice on a C57BL/6 background showed the same profound anorexia as wild type mice when carrying deletion of mPGES-1, while displaying almost normal food intake after pretreatment with a cyclooxygenase-2 inhibitor. We conclude that the involvement of induced PGE2 in IL-1β evoked anorexia is strain dependent and we suggest that different routes that probably involve distinct prostanoids exist by which inflammatory stimuli may evoke an anorexic response and that these routes may be of different importance in different strains of mice.
Collapse
Affiliation(s)
- Anna Nilsson
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, S-581 85 Linköping, Sweden
| | - Louise Elander
- Division of Anesthesiology and Intensive Care, Department of Medical and Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - Martin Hallbeck
- Division of Experimental Pathology, Department of Clinical and Experimental Medicine, Linköping University, S-581 85 Linköping, Sweden
| | - Unn Örtegren Kugelberg
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, S-581 85 Linköping, Sweden
| | - David Engblom
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, S-581 85 Linköping, Sweden
| | - Anders Blomqvist
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, S-581 85 Linköping, Sweden.
| |
Collapse
|
72
|
Bennett M, Gilroy DW. Lipid Mediators in Inflammation. MYELOID CELLS IN HEALTH AND DISEASE 2017:343-366. [DOI: 10.1128/9781555819194.ch19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Melanie Bennett
- Roche Products Limited, Shire Park; Welwyn Garden City AL7 1TW United Kingdom
| | - Derek W. Gilroy
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, University College London; London WC1 E6JJ United Kingdom
| |
Collapse
|
73
|
Rakus K, Ronsmans M, Vanderplasschen A. Behavioral fever in ectothermic vertebrates. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:84-91. [PMID: 27381718 DOI: 10.1016/j.dci.2016.06.027] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 06/06/2023]
Abstract
Fever is an evolutionary conserved defense mechanism which is present in both endothermic and ectothermic vertebrates. Ectotherms in response to infection can increase their body temperature by moving to warmer places. This process is known as behavioral fever. In this review, we summarize the current knowledge on the mechanisms of induction of fever in mammals. We further discuss the evolutionary conserved mechanisms existing between fever of mammals and behavioral fever of ectothermic vertebrates. Finally, the experimental evidences supporting an adaptive value of behavioral fever expressed by ectothermic vertebrates are summarized.
Collapse
Affiliation(s)
- Krzysztof Rakus
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland.
| | - Maygane Ronsmans
- Immunology-Vaccinology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium.
| | - Alain Vanderplasschen
- Immunology-Vaccinology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium.
| |
Collapse
|
74
|
Zhang BJ, Shao SR, Aritake K, Takeuchi A, Urade Y, Huang ZL, Lazarus M, Qu WM. Interleukin-1β induces sleep independent of prostaglandin D 2 in rats and mice. Neuroscience 2017; 340:258-267. [DOI: 10.1016/j.neuroscience.2016.09.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/01/2016] [Accepted: 09/08/2016] [Indexed: 01/28/2023]
|
75
|
Turrin NP, Rivest S. Unraveling the Molecular Details Involved in the Intimate Link between the Immune and Neuroendocrine Systems. Exp Biol Med (Maywood) 2016; 229:996-1006. [PMID: 15522835 DOI: 10.1177/153537020422901003] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
During systemic infections, the immune system can signal the brain and act on different neuronal circuits via soluble molecules, such as proinflammatory cytokines, that act on the cells forming the blood-brain barrier and the circumventricular organs. These activated cells release prostaglandin of the E2 type (PGE2), which is the endogenous ligand that triggers the pathways involved in the control of autonomic functions necessary to restore homeostasis and provide inhibitory feedback to innate immunity. Among these neurophysiological functions, activation of the circuits that control the plasma release of glucocorticoids is probably the most critical to the survival of the host in the presence of pathogens. This review revisits this issue and describes in depth the molecular details (including the emerging role of Toll-like receptors during inflammation) underlying the influence of circulating inflammatory molecules on the cerebral tissue, focusing on their contribution in the synthesis and action PGE2 in the brain. We also provide an innovative view supporting the concept of “fast and delayed response” involving the same ligands but different groups of cells, signal transduction pathways, and target genes.
Collapse
Affiliation(s)
- Nicolas P Turrin
- Laboratory of Molecular Endocrinology, CHUL Research Center and Department of Anatomy and Physiology, Laval University, 2705 Boulevard Laurier, Québec G1V 4G2, Canada
| | | |
Collapse
|
76
|
Un Lam C, Hsu CYS, Yee R, Koh D, Lee YS, Chong MFF, Cai M, Kwek K, Saw SM, Gluckman P, Chong YS. Early-life factors affect risk of pain and fever in infants during teething periods. Clin Oral Investig 2016; 20:1861-1870. [PMID: 26592811 DOI: 10.1007/s00784-015-1658-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 11/10/2015] [Indexed: 12/25/2022]
Abstract
AIM This longitudinal study aimed to investigate the prevalence of teething-related pain and fever and the early-life factors that may affect the risk of experiencing these disturbances within the first 1.5 years of life. MATERIALS AND METHODS Participants were recruited (n = 1033) through the Growing Up in Singapore Towards healthy Outcomes (GUSTO) birth cohort (n = 1237). Interviews were performed tri-monthly regarding the prevalence of teething pain and fever in children from 6 to 18 months of age. Crude and multivariable analyses were conducted using Poisson-log regression models. RESULTS Prevalence rates for teething pain and fever were 35.5 and 49.9 % respectively. Multivariable Poisson regression analysis showed maternal second-hand tobacco smoke (SHS) exposure to increase the risk of both pain (mean ratio = 1.35; p = 0.006) and fever (mean ratio = 1.22; p = 0.025), whereas SHS exposure plus active smoking further increased risk of teething pain in the children (mean ratio = 1.89; p = 0.029). Delivery via Caesarean section increased risk of teething pain (mean ratio = 1.27; p = 0.033), while prenatal plasma vitamin D insufficiency lowered such a risk (mean ratio = 0.62; p = 0.012). Compared to Chinese infants, Indian babies exhibited lower risk of teething pain and fever (both p ≤ 0.001). CONCLUSIONS Early-life factors such as tobacco smoke exposure and vitamin insufficiency during pregnancy, ethnicity and childbirth via Caesarean section may significantly affect the child's susceptibility to teething-related pain and fever. CLINICAL RELEVANCE Knowledge of prevalence and risk factors of teething disturbances may better equip primary caregivers and healthcare professionals to accurately detect teething-related local and/or systemic signs/symptoms and effectively facilitate tobacco cessation among pregnant women.
Collapse
Affiliation(s)
- Carolina Un Lam
- Discipline of Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Chin-Ying Stephen Hsu
- Discipline of Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore, Singapore.
| | - Robert Yee
- Discipline of Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - David Koh
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Yung Seng Lee
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mary Foong-Fong Chong
- Singapore Institute for Clinical Sciences, Agency for Science and Technology Research (A*STAR), Brenner Centre for Molecular Medicine, Singapore, Singapore
| | - Meijin Cai
- Discipline of Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Kenneth Kwek
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital (KKH), Singapore, Singapore
| | - Seang Mei Saw
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Peter Gluckman
- Singapore Institute for Clinical Sciences, Agency for Science and Technology Research (A*STAR), Brenner Centre for Molecular Medicine, Singapore, Singapore
| | - Yap Seng Chong
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
77
|
Hosono K, Isonaka R, Kawakami T, Narumiya S, Majima M. Signaling of Prostaglandin E Receptors, EP3 and EP4 Facilitates Wound Healing and Lymphangiogenesis with Enhanced Recruitment of M2 Macrophages in Mice. PLoS One 2016; 11:e0162532. [PMID: 27711210 PMCID: PMC5053515 DOI: 10.1371/journal.pone.0162532] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 08/24/2016] [Indexed: 01/09/2023] Open
Abstract
Lymphangiogenesis plays an important role in homeostasis, metabolism, and immunity, and also occurs during wound-healing. Here, we examined the roles of prostaglandin E2 (PGE2) receptor (EP) signaling in enhancement of lymphangiogenesis in wound healing processes. The hole-punch was made in the ears of male C57BL/6 mice using a metal ear punch. Healing process and lymphangiogenesis together with macrophage recruitment were analyzed in EP knockout mice. Lymphangiogenesis was up-regulated in the granulation tissues at the margins of punched-hole wounds in mouse ears, and this increase was accompanied by increased expression levels of COX-2 and microsomal prostaglandin E synthase-1. Administration of celecoxib, a COX-2 inhibitor, suppressed lymphangiogenesis in the granulation tissues and reduced the induction of the pro-lymphangiogenic factors, vascular endothelial growth factor (VEGF) -C and VEGF-D. Topical applications of selective EP receptor agonists enhanced the expressions of lymphatic vessel endothelial hyaluronan receptor-1 and VEGF receptor-3. The wound-healing processes and recruitment of CD11b-positive macrophages, which produced VEGF-C and VEGF-D, were suppressed under COX-2 inhibition. Mice lacking either EP3 or EP4 exhibited reduced wound-healing, lymphangiogenesis and recruitment of M2 macrophages, compared with wild type mice. Proliferation of cultured human lymphatic endothelial cells was not detected under PGE2 stimulation. Lymphangiogenesis and recruitment of M2 macrophages that produced VEGF-C/D were suppressed in mice treated with a COX-2 inhibitor or lacking either EP3 or EP4 during wound healing. COX-2 and EP3/EP4 signaling may be novel targets to control lymphangiogenesis in vivo.
Collapse
MESH Headings
- Animals
- CD11b Antigen/metabolism
- Cyclooxygenase 2/metabolism
- Cyclooxygenase 2 Inhibitors/pharmacology
- Ear/physiology
- Gene Knockout Techniques
- Lymphangiogenesis/drug effects
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Prostaglandin-E Synthases/metabolism
- Receptors, Prostaglandin E, EP3 Subtype/deficiency
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/deficiency
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Signal Transduction/drug effects
- Up-Regulation/drug effects
- Vascular Endothelial Growth Factor C/biosynthesis
- Vascular Endothelial Growth Factor D/biosynthesis
- Wound Healing/drug effects
Collapse
Affiliation(s)
- Kanako Hosono
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Risa Isonaka
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Tadashi Kawakami
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Shuh Narumiya
- Center for Innovation in Immunoregulation Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Majima
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
78
|
Affiliation(s)
| | - Takuji Imamura
- Departments of Obstetrics and Gynecology, Pediatrics, and Molecular Biology and Pharmacology, Washington University School of Medicine, and St. Louis Children's Hospital, St. Louis, Missouri
| | - Louis J. Muglia
- Departments of Obstetrics and Gynecology, Pediatrics, and Molecular Biology and Pharmacology, Washington University School of Medicine, and St. Louis Children's Hospital, St. Louis, Missouri; Washington University School of Medicine, Box 8116, One Children's Place, St. Louis, MO 63110
| |
Collapse
|
79
|
Dinarello CA. Review: Infection, fever, and exogenous and endogenous pyrogens: some concepts have changed. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519040100040301] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
For many years, it was thought that bacterial products caused fever via the intermediate production of a host-derived, fever-producing molecule, called endogenous pyrogen (EP). Bacterial products and other fever-producing substances were termed exogenous pyrogens. It was considered highly unlikely that exogenous pyrogens caused fever by acting directly on the hypothalamic thermoregulatory center since there were countless fever-producing microbial products, mostly large molecules, with no common physical structure. In vivo and in vitro, lipopolysaccharides (LPSs) and other microbial products induced EP, subsequently shown to be interleukin-1 (IL-1). The concept of the `endogenous pyrogen' cause of fever gained considerable support when pure, recombinant IL-1 produced fever in humans and in animals at subnanomolar concentrations. Subsequently, recombinant tumor necrosis factor-α (TNF-α), IL-6 and other cytokines were also shown to cause fever and EPs are now termed pyrogenic cytokines. However, the concept was challenged when specific blockade of either IL-1 or TNF activity did not diminish the febrile response to LPS, to other microbial products or to natural infections in animals and in humans. During infection, fever could occur independently of IL-1 or TNF activity. The cytokine-like property of Toll-like receptor (TLR) signal transduction provides an explanation by which any microbial product can cause fever by engaging its specific TLR on the vascular network supplying the thermoregulatory center in the anterior hypothalamus. Since fever induced by IL-1, TNF-α, IL-6 or TLR ligands requires cyclooxygenase-2, production of prostaglandin E2 (PGE 2) and activation of hypothalamic PGE2 receptors provides a unifying mechanism for fever by endogenous and exogenous pyrogens. Thus, fever is the result of either cytokine receptor or TLR triggering; in autoimmune diseases, fever is mostly cytokine mediated whereas both cytokine and TLR account for fever during infection.
Collapse
Affiliation(s)
- Charles A. Dinarello
- Department of Medicine, Division of Infectious Diseases, University of Colorado Health Sciences Center, Denver, Colorado, USA,
| |
Collapse
|
80
|
Andreasson KI, Bachstetter AD, Colonna M, Ginhoux F, Holmes C, Lamb B, Landreth G, Lee DC, Low D, Lynch MA, Monsonego A, O’Banion MK, Pekny M, Puschmann T, Russek-Blum N, Sandusky LA, Selenica MLB, Takata K, Teeling J, Town T, Van Eldik LJ, Russek-Blum N, Monsonego A, Low D, Takata K, Ginhoux F, Town T, O’Banion MK, Lamb B, Colonna M, Landreth G, Andreasson KI, Sandusky LA, Selenica MLB, Lee DC, Holmes C, Teeling J, Lynch MA, Van Eldik LJ, Bachstetter AD, Pekny M, Puschmann T. Targeting innate immunity for neurodegenerative disorders of the central nervous system. J Neurochem 2016; 138:653-93. [PMID: 27248001 PMCID: PMC5433264 DOI: 10.1111/jnc.13667] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/01/2016] [Accepted: 04/30/2016] [Indexed: 12/21/2022]
Abstract
Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview of physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia and astrocyte cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article. Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer's disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview on physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Niva Russek-Blum
- The Dead Sea and Arava Science Center, Central Arava Branch, Yair Station, Hazeva, Israel
| | - Alon Monsonego
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, The Faculty of Health Sciences: The National Institute of Biotechnology in the Negev, and Zlotowski Center for Neuroscience, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kazuyuki Takata
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Terrence Town
- Departments of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089,
| | - M. Kerry O’Banion
- Departments of Neuroscience and Neurology, Del Monte Neuromedicine Institute, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642,
| | - Bruce Lamb
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH 44106
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gary Landreth
- Department of Neurosciences, Case Western Reserve University 44106
| | - Katrin I. Andreasson
- Department of Neurology and Neurological Sciences, Stanford Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leslie A. Sandusky
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Maj-Linda B. Selenica
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Daniel C. Lee
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Clive Holmes
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | - Jessica Teeling
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | | | | | | | - Milos Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| | - Till Puschmann
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
81
|
Structural features of subtype-selective EP receptor modulators. Drug Discov Today 2016; 22:57-71. [PMID: 27506873 DOI: 10.1016/j.drudis.2016.08.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/20/2016] [Accepted: 08/01/2016] [Indexed: 12/11/2022]
Abstract
Prostaglandin E2 is a potent endogenous molecule that binds to four different G-protein-coupled receptors: EP1-4. Each of these receptors is a valuable drug target, with distinct tissue localisation and signalling pathways. We review the structural features of EP modulators required for subtype-selective activity, as well as the structural requirements for improved pharmacokinetic parameters. Novel EP receptor subtype selective agonists and antagonists appear to be valuable drug candidates in the therapy of many pathophysiological states, including ulcerative colitis, glaucoma, bone healing, B cell lymphoma, neurological diseases, among others, which have been studied in vitro, in vivo and in early phase clinical trials.
Collapse
|
82
|
Ni H, Shen J, Song Y, Cao M, Liu X, Huang J, Zhang W, Xie L, Ning X, Ke K. EP3, Prostaglandin E2 Receptor Subtype 3, Associated with Neuronal Apoptosis Following Intracerebral Hemorrhage. Cell Mol Neurobiol 2016; 36:971-980. [PMID: 26718710 PMCID: PMC11482414 DOI: 10.1007/s10571-015-0287-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022]
Abstract
EP3 is prostaglandin E2 receptor subtype 3 and mediates the activation of several signaling pathways, changing in cAMP levels, calcium mobilization, and activation of phospholipase C. Previous studies demonstrated a direct role for EP3 in various neurodegenerative disorders, such as stroke and Alzheimer disease. However, the distribution and function of EP3 in ICH diseases remain unknown. Here, we demonstrate that EP3 may be involved in neuronal apoptosis in the processes of intracerebral hemorrhage (ICH). From the results of Western blot and immunohistochemistry, we obtained a significant up-regulation of EP3 in neurons adjacent to the hematoma following ICH. Up-regulation of EP3 was found to be accompanied by the increased expression of active caspase-3 and pro-apoptotic Bcl-2-associated X protein (Bax) and decreased expression of anti-apoptotic protein B cell lymphoma-2 (Bcl-2) in vivo and vitro studies. Furthermore, the expression of these three proteins reduced active caspase-3 and Bax expression, while increased Bcl-2 were changed after knocking down EP3 by RNA interference in PC12 cells, further confirmed that EP3 might exert its pro-apoptotic function on neuronal apoptosis. Thus, EP3 may play a role in promoting the neuronal apoptosis following ICH.
Collapse
Affiliation(s)
- Haidan Ni
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yan Song
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaorong Liu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jie Huang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Weidong Zhang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Lili Xie
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaojin Ning
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
83
|
Kim SO, Duffy DM. Mapping PTGERs to the Ovulatory Follicle: Regional Responses to the Ovulatory PGE2 Signal. Biol Reprod 2016; 95:33. [PMID: 27307073 PMCID: PMC5029471 DOI: 10.1095/biolreprod.116.140574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/27/2016] [Accepted: 06/03/2016] [Indexed: 12/11/2022] Open
Abstract
Prostaglandin E2 (PGE2) is a key intrafollicular mediator of ovulation in many, if not all, mammalian species. PGE2 acts at follicular cells via four distinct PGE2 receptors (PTGERs). Within the ovulatory follicle, each cell type (e.g., oocyte, cumulus granulosa cell, mural granulosa cell, theca cell, endothelial cell) expresses a different subset of the four PTGERs. Expression of a subset of PTGERs has consequences for the generation of intracellular signals and ultimately the unique functions of follicular cells that respond to PGE2. Just as the ovulatory LH surge regulates PGE2 synthesis, the LH surge also regulates expression of the four PTGERs. The pattern of expression of the four PTGERs among follicular cells before and after the LH surge forms a spatial and temporal map of PGE2 responses. Differential PTGER expression, coupled with activation of cell-specific intracellular signals, may explain how a single paracrine mediator can have pleotropic actions within the ovulatory follicle. Understanding the role of each PTGER in ovulation may point to previously unappreciated opportunities to both promote and prevent fertility.
Collapse
Affiliation(s)
- Soon Ok Kim
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Diane M Duffy
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| |
Collapse
|
84
|
Fritz M, Klawonn AM, Nilsson A, Singh AK, Zajdel J, Wilhelms DB, Lazarus M, Löfberg A, Jaarola M, Kugelberg UÖ, Billiar TR, Hackam DJ, Sodhi CP, Breyer MD, Jakobsson J, Schwaninger M, Schütz G, Parkitna JR, Saper CB, Blomqvist A, Engblom D. Prostaglandin-dependent modulation of dopaminergic neurotransmission elicits inflammation-induced aversion in mice. J Clin Invest 2016; 126:695-705. [PMID: 26690700 DOI: 10.1172/jci83844] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/12/2015] [Indexed: 01/18/2023] Open
Abstract
Systemic inflammation causes malaise and general feelings of discomfort. This fundamental aspect of the sickness response reduces the quality of life for people suffering from chronic inflammatory diseases and is a nuisance during mild infections like common colds or the flu. To investigate how inflammation is perceived as unpleasant and causes negative affect, we used a behavioral test in which mice avoid an environment that they have learned to associate with inflammation-induced discomfort. Using a combination of cell-type–specific gene deletions, pharmacology, and chemogenetics, we found that systemic inflammation triggered aversion through MyD88-dependent activation of the brain endothelium followed by COX1-mediated cerebral prostaglandin E2 (PGE2) synthesis. Further, we showed that inflammation-induced PGE2 targeted EP1 receptors on striatal dopamine D1 receptor–expressing neurons and that this signaling sequence induced aversion through GABA-mediated inhibition of dopaminergic cells. Finally, we demonstrated that inflammation-induced aversion was not an indirect consequence of fever or anorexia but that it constituted an independent inflammatory symptom triggered by a unique molecular mechanism. Collectively, these findings demonstrate that PGE2-mediated modulation of the dopaminergic motivational circuitry is a key mechanism underlying the negative affect induced by inflammation.
Collapse
|
85
|
Hadley KB, Ryan AS, Forsyth S, Gautier S, Salem N. The Essentiality of Arachidonic Acid in Infant Development. Nutrients 2016; 8:216. [PMID: 27077882 PMCID: PMC4848685 DOI: 10.3390/nu8040216] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/01/2016] [Accepted: 04/05/2016] [Indexed: 01/16/2023] Open
Abstract
Arachidonic acid (ARA, 20:4n-6) is an n-6 polyunsaturated 20-carbon fatty acid formed by the biosynthesis from linoleic acid (LA, 18:2n-6). This review considers the essential role that ARA plays in infant development. ARA is always present in human milk at a relatively fixed level and is accumulated in tissues throughout the body where it serves several important functions. Without the provision of preformed ARA in human milk or infant formula the growing infant cannot maintain ARA levels from synthetic pathways alone that are sufficient to meet metabolic demand. During late infancy and early childhood the amount of dietary ARA provided by solid foods is low. ARA serves as a precursor to leukotrienes, prostaglandins, and thromboxanes, collectively known as eicosanoids which are important for immunity and immune response. There is strong evidence based on animal and human studies that ARA is critical for infant growth, brain development, and health. These studies also demonstrate the importance of balancing the amounts of ARA and DHA as too much DHA may suppress the benefits provided by ARA. Both ARA and DHA have been added to infant formulas and follow-on formulas for more than two decades. The amounts and ratios of ARA and DHA needed in infant formula are discussed based on an in depth review of the available scientific evidence.
Collapse
Affiliation(s)
- Kevin B Hadley
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| | - Alan S Ryan
- Clinical Research Consulting, 9809 Halston Manor, Boynton Beach, FL 33473, USA.
| | - Stewart Forsyth
- School of Medicine, Dentistry & Nursing, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK.
| | - Sheila Gautier
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| | - Norman Salem
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| |
Collapse
|
86
|
Jones VC, Birrell MA, Maher SA, Griffiths M, Grace M, O'Donnell VB, Clark SR, Belvisi MG. Role of EP2 and EP4 receptors in airway microvascular leak induced by prostaglandin E2. Br J Pharmacol 2016; 173:992-1004. [PMID: 26639895 PMCID: PMC4831025 DOI: 10.1111/bph.13400] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 10/21/2015] [Accepted: 11/30/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Airway microvascular leak (MVL) involves the extravasation of proteins from post-capillary venules into surrounding tissue. MVL is a cardinal sign of inflammation and an important feature of airway inflammatory diseases such as asthma. PGE2, a product of COX-mediated metabolism of arachidonic acid, binds to four receptors, termed EP1–4. PGE2 has a wide variety of effects within the airway, including modulation of inflammation, sensory nerve activation and airway tone. However, the effect of PGE2 on airway MVL and the receptor/s that mediate this have not been described. EXPERIMENTAL APPROACH Evans Blue dye was used as a marker of airway MVL, and selective EP receptor agonists and antagonists were used alongside EP receptor-deficient mice to define the receptor subtype involved. KEY RESULTS PGE2 induced significant airway MVL in mice and guinea pigs. A significant reduction in PGE2-induced MVL was demonstrated in Ptger2−/− and Ptger4−/− mice and in wild-type mice pretreated simultaneously with EP2 (PF-04418948) and EP4 (ER-819762) receptor antagonists. In a model of allergic asthma, an increase in airway levels of PGE2 was associated with a rise in MVL; this change was absent in Ptger2−/− and Ptger4−/− mice. CONCLUSIONS AND IMPLICATIONS PGE2 is a key mediator produced by the lung and has widespread effects according to the EP receptor activated. Airway MVL represents a response to injury and under ‘disease’ conditions is a prominent feature of airway inflammation. The data presented highlight a key role for EP2 and EP4 receptors in MVL induced by PGE2.
Collapse
MESH Headings
- Allergens
- Animals
- Asthma/metabolism
- Azetidines/pharmacology
- Benzazepines/pharmacology
- Bronchi/metabolism
- Capillary Permeability
- Dinoprostone/analogs & derivatives
- Dinoprostone/metabolism
- Dinoprostone/pharmacology
- Guinea Pigs
- Imidazoles/pharmacology
- Male
- Methyl Ethers/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Ovalbumin
- Receptors, Prostaglandin E, EP2 Subtype/agonists
- Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/agonists
- Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Trachea/metabolism
Collapse
|
87
|
Tamura K, Naraba H, Hara T, Nakamura K, Yoshie M, Kogo H, Tachikawa E. A positive feedback loop between progesterone and microsomal prostaglandin E synthase-1-mediated PGE2 promotes production of both in mouse granulosa cells. Prostaglandins Other Lipid Mediat 2016; 123:56-62. [DOI: 10.1016/j.prostaglandins.2016.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 04/30/2016] [Accepted: 05/04/2016] [Indexed: 12/26/2022]
|
88
|
Conti B. Prostaglandin E2 that triggers fever is synthesized through an endocannabinoid- dependent pathway. Temperature (Austin) 2016; 3:25-7. [PMID: 27227089 PMCID: PMC4861185 DOI: 10.1080/23328940.2015.1130520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Bruno Conti
- Department of Chemical Physiology, Department of Molecular and Cellular Neuroscience The Scripps Research Institute , 10550 North Torrey Pines Road SR307 , La Jolla, 92037, CA, USA
| |
Collapse
|
89
|
Romero R, Chaemsaithong P, Docheva N, Korzeniewski SJ, Tarca AL, Bhatti G, Xu Z, Kusanovic JP, Dong Z, Ahmed AI, Yoon BH, Hassan SS, Chaiworapongsa T, Yeo L. Clinical chorioamnionitis at term IV: the maternal plasma cytokine profile. J Perinat Med 2016; 44:77-98. [PMID: 26352068 PMCID: PMC5624710 DOI: 10.1515/jpm-2015-0103] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/17/2015] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Fever is a major criterion for clinical chorioamnionitis; yet, many patients with intrapartum fever do not have demonstrable intra-amniotic infection. Some cytokines, such as interleukin (IL)-1, IL-6, interferon-gamma (IFN-γ), and tumor necrosis factor alpha (TNF-α), can induce a fever. The objective of this study was to determine whether maternal plasma concentrations of cytokines could be of value in the identification of patients with the diagnosis of clinical chorioamnionitis at term who have microbial-associated intra-amniotic inflammation. METHODS A retrospective cross-sectional study was conducted, including patients with clinical chorioamnionitis at term (n=41; cases) and women in spontaneous labor at term without clinical chorioamnionitis (n=77; controls). Women with clinical chorioamnionitis were classified into three groups according to the results of amniotic fluid culture, broad-range polymerase chain reaction coupled with electrospray ionization mass spectrometry (PCR/ESI-MS), and amniotic fluid IL-6 concentration: 1) no intra-amniotic inflammation; 2) intra-amniotic inflammation without detectable microorganisms; or 3) microbial-associated intra-amniotic inflammation. The maternal plasma concentrations of 29 cytokines were determined with sensitive and specific V-PLEX immunoassays. Nonparametric statistical methods were used for analysis, adjusting for a false discovery rate of 5%. RESULTS 1) The maternal plasma concentrations of pyrogenic cytokines (IL-1β, IL-2, IL-6, IFN-γ, and TNF-α) were significantly higher in patients with clinical chorioamnionitis at term than in those with spontaneous term labor without clinical chorioamnionitis; 2) the maternal plasma concentrations of cytokines were not significantly different among the three subgroups of patients with clinical chorioamnionitis (intra-amniotic inflammation with and without detectable bacteria and those without intra-amniotic inflammation); and 3) among women with the diagnosis of clinical chorioamnionitis, but without evidence of intra-amniotic inflammation, the maternal plasma concentrations of pyrogenic cytokines were significantly higher than in patients with spontaneous labor at term. These observations suggest that a fever can be mediated by increased circulating concentrations of these cytokines, despite the absence of a local intra-amniotic inflammatory response. CONCLUSIONS 1) The maternal plasma concentrations of pyrogenic cytokines (e.g. IL-1β, IL-2, IL-6, IFN-γ, and TNF-α) are higher in patients with intra-partum fever and the diagnosis of clinical chorioamnionitis at term than in those in spontaneous labor at term without a fever; and 2) maternal plasma cytokine concentrations have limited value in the identification of patients with bacteria in the amniotic cavity. Accurate assessment of the presence of intra-amniotic infection requires amniotic fluid analysis.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Department of Molecular Obstetrics and Genetics, Wayne State University, Detroit, MI, USA
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nikolina Docheva
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Steven J. Korzeniewski
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Zhonghui Xu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Juan P. Kusanovic
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Center for Research and Innovation in Maternal-Fetal Medicine (CIMAF). Department of Obstetrics and Gynecology, Sótero del Río Hospital, Santiago, Chile
- Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Zhong Dong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ahmed I. Ahmed
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bo Hyun Yoon
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Sonia S. Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lami Yeo
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
90
|
Ceddia RP, Lee D, Maulis MF, Carboneau BA, Threadgill DW, Poffenberger G, Milne G, Boyd KL, Powers AC, McGuinness OP, Gannon M, Breyer RM. The PGE2 EP3 Receptor Regulates Diet-Induced Adiposity in Male Mice. Endocrinology 2016; 157:220-32. [PMID: 26485614 PMCID: PMC4701878 DOI: 10.1210/en.2015-1693] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mice carrying a targeted disruption of the prostaglandin E2 (PGE2) E-prostanoid receptor 3 (EP3) gene, Ptger3, were fed a high-fat diet (HFD), or a micronutrient matched control diet, to investigate the effects of disrupted PGE2-EP3 signaling on diabetes in a setting of diet-induced obesity. Although no differences in body weight were seen in mice fed the control diet, when fed a HFD, EP3(-/-) mice gained more weight relative to EP3(+/+) mice. Overall, EP3(-/-) mice had increased epididymal fat mass and adipocyte size; paradoxically, a relative decrease in both epididymal fat pad mass and adipocyte size was observed in the heaviest EP3(-/-) mice. The EP3(-/-) mice had increased macrophage infiltration, TNF-α, monocyte chemoattractant protein-1, IL-6 expression, and necrosis in their epididymal fat pads as compared with EP3(+/+) animals. Adipocytes isolated from EP3(+/+) or EP3(-/-) mice were assayed for the effect of PGE2-evoked inhibition of lipolysis. Adipocytes isolated from EP3(-/-) mice lacked PGE2-evoked inhibition of isoproterenol stimulated lipolysis compared with EP3(+/+). EP3(-/-) mice fed HFD had exaggerated ectopic lipid accumulation in skeletal muscle and liver, with evidence of hepatic steatosis. Both blood glucose and plasma insulin levels were similar between genotypes on a control diet, but when fed HFD, EP3(-/-) mice became hyperglycemic and hyperinsulinemic when compared with EP3(+/+) fed HFD, demonstrating a more severe insulin resistance phenotype in EP3(-/-). These results demonstrate that when fed a HFD, EP3(-/-) mice have abnormal lipid distribution, developing excessive ectopic lipid accumulation and associated insulin resistance.
Collapse
MESH Headings
- Adipose Tissue, White/immunology
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Adiposity
- Animals
- Cell Size
- Crosses, Genetic
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/immunology
- Diet, High-Fat/adverse effects
- Insulin Resistance
- Lipid Metabolism
- Liver/immunology
- Liver/metabolism
- Liver/pathology
- Macrophage Activation
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/immunology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Necrosis
- Non-alcoholic Fatty Liver Disease/etiology
- Non-alcoholic Fatty Liver Disease/immunology
- Obesity/etiology
- Obesity/metabolism
- Obesity/pathology
- Obesity/physiopathology
- Panniculitis/etiology
- Panniculitis/immunology
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Weight Gain
Collapse
Affiliation(s)
- Ryan P Ceddia
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - DaeKee Lee
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Matthew F Maulis
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Bethany A Carboneau
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - David W Threadgill
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Greg Poffenberger
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Ginger Milne
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Kelli L Boyd
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Alvin C Powers
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Owen P McGuinness
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Maureen Gannon
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Richard M Breyer
- Department of Veterans Affairs (A.C.P., M.G., R.M.B.), Tennessee Valley Health Authority, and Department of Medicine (R.M.B.), Division of Nephrology and Hypertension; Departments of Pharmacology (R.P.C., G.M., R.M.B.) and Cell and Developmental Biology (D.L., D.W.T., M.G.); Department of Medicine (M.F.M., G.P., A.C.P., M.G.), Division of Diabetes, Endocrinology, and Metabolism; and Departments of Molecular Physiology and Biophysics (B.A.C., A.C.P., O.P.G., M.G.) and Pathology, Microbiology, and Immunology (K.L.B.), Vanderbilt University Medical Center, Nashville, Tennessee 37232
| |
Collapse
|
91
|
Simm B, Ott D, Pollatzek E, Murgott J, Gerstberger R, Rummel C, Roth J. Effects of prostaglandin E2 on cells cultured from the rat organum vasculosum laminae terminalis and median preoptic nucleus. Neuroscience 2015; 313:23-35. [PMID: 26608124 DOI: 10.1016/j.neuroscience.2015.11.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/30/2015] [Accepted: 11/16/2015] [Indexed: 02/09/2023]
Abstract
The time course of the induction of enzymes responsible for the formation of prostaglandin E2 (PGE2) after an inflammatory insult, in relation to the concomitant febrile response, suggests that peripherally generated PGE2 is involved in the induction of the early phase of fever, while centrally produced PGE2 exerts pyrogenic capacities during the later stages of fever within the hypothalamic median preoptic nucleus (MnPO). The actions of peripherally derived PGE2 on the brain might occur at the level of the organum vasculosum laminae terminalis (OVLT), which lacks a tight blood-brain barrier and is implicated in fever, while the effects of PGE2 within the MnPO might interfere with glutamatergic neurotransmission within a recently characterized central efferent pathway for the activation of cold-defence reactions. Using the fura-2 ratio imaging technique we, therefore, measured changes of the intracellular Ca(2+)-concentration in primary neuroglial microcultures of rat OVLT and MnPO stimulated with PGE2 and/or glutamate. In cultures from the OVLT, as opposed to those derived from the MnPO, substantial numbers of neurons (8% of 385), astrocytes (19% of 645) and microglial cells (28% of 43) directly responded to PGE2 with a transient increase of intracellular Ca(2+). The most pronounced effect of PGE2 on cells from MnPO microcultures was its modulatory influence on the strength of glutamate-induced Ca(2+)-signals. In 72 out of 512 neurons and in 105 out of 715 astrocytes PGE2 significantly augmented glutamate-induced Ca(2+)-signals. About 30% of these neurons were GABAergic. These observations are in agreement with putative roles of peripheral PGE2 as a directly acting circulating agent at the level of the OVLT, and of central MnPO-intrinsic PGE2 as an enhancer of glutamatergic neurotransmission, which causes disinhibition of thermogenic heat production, a crucial component for the manifestation of fever. In microcultures from both brain sites investigated incubation with PGE2 significantly reduced the lipopolysaccharide-induced release of cytokines (tumor necrosis factor-α and interleukin-6) into the supernatant. PGE2, thus, seems to be involved in a negative feed-back loop to limit the strength of the brain inflammatory process and to play a dual role with pro- as well as anti-inflammatory properties.
Collapse
Affiliation(s)
- B Simm
- Institut für Veterinär-Physiologie und -Biochemie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - D Ott
- Institut für Veterinär-Physiologie und -Biochemie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - E Pollatzek
- Institut für Veterinär-Physiologie und -Biochemie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - J Murgott
- Institut für Veterinär-Physiologie und -Biochemie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - R Gerstberger
- Institut für Veterinär-Physiologie und -Biochemie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - C Rummel
- Institut für Veterinär-Physiologie und -Biochemie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - J Roth
- Institut für Veterinär-Physiologie und -Biochemie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany.
| |
Collapse
|
92
|
Zampronio AR, Soares DM, Souza GEP. Central mediators involved in the febrile response: effects of antipyretic drugs. Temperature (Austin) 2015; 2:506-21. [PMID: 27227071 PMCID: PMC4843933 DOI: 10.1080/23328940.2015.1102802] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 11/13/2022] Open
Abstract
Fever is a complex signal of inflammatory and infectious diseases. It is generally initiated when peripherally produced endogenous pyrogens reach areas that surround the hypothalamus. These peripheral endogenous pyrogens are cytokines that are produced by leukocytes and other cells, the most known of which are interleukin-1β, tumor necrosis factor-α, and interleukin-6. Because of the capacity of these molecules to induce their own synthesis and the synthesis of other cytokines, they can also be synthesized in the central nervous system. However, these pyrogens are not the final mediators of the febrile response. These cytokines can induce the synthesis of cyclooxygenase-2, which produces prostaglandins. These prostanoids alter hypothalamic temperature control, leading to an increase in heat production, the conservation of heat, and ultimately fever. The effect of antipyretics is based on blocking prostaglandin synthesis. In this review, we discuss recent data on the importance of prostaglandins in the febrile response, and we show that some endogenous mediators can still induce the febrile response even when known antipyretics reduce the levels of prostaglandins in the central nervous system. These studies suggest that centrally produced mediators other than prostaglandins participate in the genesis of fever. Among the most studied central mediators of fever are corticotropin-releasing factor, endothelins, chemokines, endogenous opioids, and substance P, which are discussed herein. Additionally, recent evidence suggests that these different pathways of fever induction may be activated during different pathological conditions.
Collapse
Affiliation(s)
- Aleksander R Zampronio
- Department of Pharmacology; Biological Sciences Section; Federal University of Paraná ; Curitiba, PR, Brazil
| | - Denis M Soares
- Department of Medicament; Faculty of Pharmacy; Federal University of Bahia ; Salvador, BA, Brazil
| | - Glória E P Souza
- Discipline of Pharmacology; Faculty of Pharmaceutical Sciences of Ribeirão Preto; University of São Paulo ; Ribeirão Preto, SP, Brazil
| |
Collapse
|
93
|
Zampronio AR, Soares DM, Souza GEP. Central mediators involved in the febrile response: effects of antipyretic drugs. Temperature (Austin) 2015. [PMID: 27227071 DOI: 10.1080/23328940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2023] Open
Abstract
Fever is a complex signal of inflammatory and infectious diseases. It is generally initiated when peripherally produced endogenous pyrogens reach areas that surround the hypothalamus. These peripheral endogenous pyrogens are cytokines that are produced by leukocytes and other cells, the most known of which are interleukin-1β, tumor necrosis factor-α, and interleukin-6. Because of the capacity of these molecules to induce their own synthesis and the synthesis of other cytokines, they can also be synthesized in the central nervous system. However, these pyrogens are not the final mediators of the febrile response. These cytokines can induce the synthesis of cyclooxygenase-2, which produces prostaglandins. These prostanoids alter hypothalamic temperature control, leading to an increase in heat production, the conservation of heat, and ultimately fever. The effect of antipyretics is based on blocking prostaglandin synthesis. In this review, we discuss recent data on the importance of prostaglandins in the febrile response, and we show that some endogenous mediators can still induce the febrile response even when known antipyretics reduce the levels of prostaglandins in the central nervous system. These studies suggest that centrally produced mediators other than prostaglandins participate in the genesis of fever. Among the most studied central mediators of fever are corticotropin-releasing factor, endothelins, chemokines, endogenous opioids, and substance P, which are discussed herein. Additionally, recent evidence suggests that these different pathways of fever induction may be activated during different pathological conditions.
Collapse
Affiliation(s)
- Aleksander R Zampronio
- Department of Pharmacology; Biological Sciences Section; Federal University of Paraná ; Curitiba, PR, Brazil
| | - Denis M Soares
- Department of Medicament; Faculty of Pharmacy; Federal University of Bahia ; Salvador, BA, Brazil
| | - Glória E P Souza
- Discipline of Pharmacology; Faculty of Pharmaceutical Sciences of Ribeirão Preto; University of São Paulo ; Ribeirão Preto, SP, Brazil
| |
Collapse
|
94
|
Abstract
Cyclooxygenase inhibitors such as ibuprofen have been used for decades to control fever through reducing the levels of the pyrogenic lipid transmitter prostaglandin E2 (PGE2). Historically, phospholipases have been considered to be the primary generator of the arachidonic acid (AA) precursor pool for generating PGE2 and other eicosanoids. However, recent studies have demonstrated that monoacyglycerol lipase (MAGL), through hydrolysis of the endocannabinoid 2-arachidonoylglycerol, provides a major source of AA for PGE2 synthesis in the mammalian brain under basal and neuroinflammatory states. We show here that either genetic or pharmacological ablation of MAGL leads to significantly reduced fever responses in both centrally or peripherally-administered lipopolysaccharide or interleukin-1β-induced fever models in mice. We also show that a cannabinoid CB1 receptor antagonist does not attenuate these anti-pyrogenic effects of MAGL inhibitors. Thus, much like traditional nonsteroidal anti-inflammatory drugs, MAGL inhibitors can control fever, but appear to do so through restricted control over prostaglandin production in the nervous system.
Collapse
|
95
|
Johansson JU, Woodling NS, Shi J, Andreasson KI. Inflammatory Cyclooxygenase Activity and PGE 2 Signaling in Models of Alzheimer's Disease. ACTA ACUST UNITED AC 2015; 11:125-131. [PMID: 28413375 PMCID: PMC5384338 DOI: 10.2174/1573395511666150707181414] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/03/2015] [Accepted: 04/19/2015] [Indexed: 11/28/2022]
Abstract
The inflammatory response is a fundamental driving force in the pathogenesis of Alzheimer’s disease (AD). In the setting of accumulating immunogenic Aß peptide assemblies, microglia, the innate immune cells of the brain, generate a non-resolving immune response and fail to adequately clear accumulating Aß peptides, accelerating neuronal and synaptic injury. Pathological, biomarker, and imaging studies point to a prominent role of the innate immune response in AD development, and the molecular components of this response are beginning to be unraveled. The inflammatory cyclooxygenase-PGE2 pathway is implicated in pre-clinical development of AD, both in epidemiology of normal aging populations and in transgenic mouse models of Familial AD. The cyclooxygenase-PGE2 pathway modulates the inflammatory response to accumulating Aß peptides through actions of specific E-prostanoid G-protein coupled receptors.
Collapse
Affiliation(s)
- Jenny U Johansson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Present address: SRI International, Menlo Park, CA, USA
| | - Nathaniel S Woodling
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Present address: Institute of Healthy Ageing, University College London, London, UK
| | - Ju Shi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Present address: True North Therapeutics, South San Francisco, CA, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
96
|
Hosotani R, Inoue W, Takemiya T, Yamagata K, Kobayashi S, Matsumura K. Prostaglandin transporter in the rat brain: its localization and induction by lipopolysaccharide. Temperature (Austin) 2015; 2:425-34. [PMID: 27227056 PMCID: PMC4843910 DOI: 10.1080/23328940.2015.1062953] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 06/10/2015] [Accepted: 06/10/2015] [Indexed: 11/25/2022] Open
Abstract
Prostaglandin E2 (PGE2) is produced in the brain during infectious/inflammatory diseases, and it mediates acute-phase responses including fever. In the recovery phase of such diseases, PGE2 disappears from the brain through yet unidentified mechanisms. Rat prostaglandin transporter (PGT), which facilitates transmembrane transport of PGE2, might be involved in the clearance of PGE2 from the brain. Here, we examined the cellular localization of PGT mRNA and its protein in the brains of untreated rats and those injected intraperitoneally with a pyrogen lipopolysaccharide (LPS) or saline. PGT mRNA was weakly expressed in the arachnoid membrane of untreated rats and saline-injected ones, but was induced in blood vessels of the subarachnoidal space and choroid plexus and in arachnoid membrane at 5 h and 12 h after LPS injection. In the same type of cells, PGT-like immunoreactivity was found in the cytosol and cell membrane even under nonstimulated conditions, and its level was also elevated after LPS injection. PGT-positive cells in blood vessels were identified as endothelial cells. In most cases, PGT was not colocalized with cyclooxygenase-2, a marker of prostaglandin-producing cells. The PGE2 level in the cerebrospinal fluid reached its peak at 3 h after LPS, and then dropped over 50% by 5 h, which time point coincides with the maximum PGT mRNA expression and enhanced level of PGT protein. These results suggest that PGT is involved in the clearance of PGE2 from the brain during the recovery phase of LPS-induced acute-phase responses.
Collapse
Affiliation(s)
- Rika Hosotani
- Faculty of Education; Shiga University ; Otsu, Shiga, Japan
| | - Wataru Inoue
- Department of Physiology and Pharmacology; Robarts Research Institute; Schulich School of Medicine & Dentistry; University of Western Ontario ; London, ON, Canada
| | - Takako Takemiya
- Medical Research Institute; Tokyo Women's Medical University ; Shinjuku, Tokyo, Japan
| | - Kanato Yamagata
- Neural Plasticity Project; Tokyo Metropolitan Institute of Medical Science ; Setagaya-ku, Tokyo, Japan
| | - Shigeo Kobayashi
- Department of Intelligence Science and Technology; Graduate School of Informatics; Kyoto University ; Sakyo-ku, Kyoto, Japan
| | - Kiyoshi Matsumura
- Faculty of Biomedical Engineering; Osaka Institute of Technology ; Asahi-ku, Osaka, Japan
| |
Collapse
|
97
|
Foster J, Mauger AR, Chrismas BCR, Thomasson K, Taylor L. Is prostaglandin E2 (PGE2) involved in the thermogenic response to environmental cooling in healthy humans? Med Hypotheses 2015; 85:607-11. [PMID: 26253311 DOI: 10.1016/j.mehy.2015.07.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/20/2015] [Accepted: 07/25/2015] [Indexed: 12/12/2022]
Abstract
Prostaglandin E2 (PGE2) is an eicosanoid derived from cyclooxygenase, an enzyme responsible for the cyclisation and oxygenation of arachidonic acid. In response to bacterial infection, PGE2 binds to EP3 receptors on a population of GABAergic neurons in the pre-optic area. Activation of the EP3 receptor decreases the intracellular cyclic adenosine monophosphate (cAMP) concentrations of these neurons, and the resulting dis-inhibition activates spinal motor outputs responsible for shivering thermogenesis, tachycardia, and brown adipose tissue activation. These involuntary responses increase core body temperature to varying degrees depending on the magnitude of infection; an immune response which is crucial for the survival of the host. However, evidence in animal and human models, primarily through the use of cyclooxygenase inhibitors (which block the production of PGE2), suggests that PGE2 may also be an important molecule for the defence of core temperature against body cooling and cold stress (in the absence of fever). In this paper, evidence within human and animal models is discussed which supports the hypothesis that the eicosanoid PGE2 has a role in maintaining human core temperature during environmental cooling. Given that over-the-counter PGE2 inhibiting drugs [i.e. Non-Steroidal Anti Inflammatory Drugs (NSAIDS)] are frequently used worldwide, it is possible that the use of such medication during environmental cooling could impair one's ability to thermoregulate. Support for such findings could have major implications in the pathology of hypothermia, thus, we suggest that future researchers investigate this specific hypothesis in vivo, using healthy human models. Suggestions for the implementation of such experiments are provided in the present work.
Collapse
Affiliation(s)
- Josh Foster
- Applied Sport and Exercise Physiology (ASEP) Research Group, Institute of Sport and Physical Activity Research (ISPAR), Department of Sport and Exercise Sciences, University of Bedfordshire, Bedford, UK
| | - Alexis R Mauger
- Endurance Research Group, School of Sport and Exercise Sciences, University of Kent, Chatham Maritime, UK
| | - Bryna C R Chrismas
- Applied Sport and Exercise Physiology (ASEP) Research Group, Institute of Sport and Physical Activity Research (ISPAR), Department of Sport and Exercise Sciences, University of Bedfordshire, Bedford, UK
| | - Katie Thomasson
- Applied Sport and Exercise Physiology (ASEP) Research Group, Institute of Sport and Physical Activity Research (ISPAR), Department of Sport and Exercise Sciences, University of Bedfordshire, Bedford, UK
| | - Lee Taylor
- Applied Sport and Exercise Physiology (ASEP) Research Group, Institute of Sport and Physical Activity Research (ISPAR), Department of Sport and Exercise Sciences, University of Bedfordshire, Bedford, UK.
| |
Collapse
|
98
|
Lazarus M, Huang ZL, Urade Y. Osamu Hayaishi-from the discovery of oxygenases in soil microorganisms to unraveling the enigma of sleep in mammals. Temperature (Austin) 2015; 2:303-7. [PMID: 27227031 PMCID: PMC4843939 DOI: 10.1080/23328940.2015.1072658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 07/08/2015] [Indexed: 01/26/2023] Open
Affiliation(s)
- Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS); University of Tsukuba ; Tsukuba, Ibaraki, Japan
| | - Zhi-Li Huang
- Department of Pharmacology; State Key Laboratory of Medical Neurobiology; and Institute of Brain Sciences; Shanghai Medical College of Fudan University ; Shanghai, China
| | - Yoshihiro Urade
- International Institute for Integrative Sleep Medicine (WPI-IIIS); University of Tsukuba ; Tsukuba, Ibaraki, Japan
| |
Collapse
|
99
|
Roth J, Blatteis CM. Mechanisms of fever production and lysis: lessons from experimental LPS fever. Compr Physiol 2015; 4:1563-604. [PMID: 25428854 DOI: 10.1002/cphy.c130033] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fever is a cardinal symptom of infectious or inflammatory insults, but it can also arise from noninfectious causes. The fever-inducing agent that has been used most frequently in experimental studies designed to characterize the physiological, immunological and neuroendocrine processes and to identify the neuronal circuits that underlie the manifestation of the febrile response is lipopolysaccharide (LPS). Our knowledge of the mechanisms of fever production and lysis is largely based on this model. Fever is usually initiated in the periphery of the challenged host by the immediate activation of the innate immune system by LPS, specifically of the complement (C) cascade and Toll-like receptors. The first results in the immediate generation of the C component C5a and the subsequent rapid production of prostaglandin E2 (PGE2). The second, occurring after some delay, induces the further production of PGE2 by induction of its synthesizing enzymes and transcription and translation of proinflammatory cytokines. The Kupffer cells (Kc) of the liver seem to be essential for these initial processes. The subsequent transfer of the pyrogenic message from the periphery to the brain is achieved by neuronal and humoral mechanisms. These pathways subserve the genesis of early (neuronal signals) and late (humoral signals) phases of the characteristically biphasic febrile response to LPS. During the course of fever, counterinflammatory factors, "endogenous antipyretics," are elaborated peripherally and centrally to limit fever in strength and duration. The multiple interacting pro- and antipyretic signals and their mechanistic effects that underlie endotoxic fever are the subjects of this review.
Collapse
Affiliation(s)
- Joachim Roth
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University, Giessen, Germany; Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | | |
Collapse
|
100
|
Kita Y, Yoshida K, Tokuoka SM, Hamano F, Yamazaki M, Sakimura K, Kano M, Shimizu T. Fever Is Mediated by Conversion of Endocannabinoid 2-Arachidonoylglycerol to Prostaglandin E2. PLoS One 2015. [PMID: 26196692 PMCID: PMC4511515 DOI: 10.1371/journal.pone.0133663] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Fever is a common response to inflammation and infection. The mechanism involves prostaglandin E2 (PGE2)-EP3 receptor signaling in the hypothalamus, which raises the set point of hypothalamic thermostat for body temperature, but the lipid metabolic pathway for pyretic PGE2 production remains unknown. To reveal the molecular basis of fever initiation, we examined lipopolysaccharides (LPS)-induced fever model in monoacylglycerol lipase (MGL)-deficient (Mgll-/-) mice, CB1 receptor-MGL compound-deficient (Cnr1-/-Mgll-/-) mice, cytosolic phospholipase A2α (cPLA2α)-deficient (Pla2g4a-/-) mice, and diacylglycerol lipase α (DGLα)-deficient (Dagla-/-) mice. Febrile reactions were abolished in Mgll-/- and Cnr1-/-Mgll-/- mice, whereas Cnr1-/-Mgll+/+, Pla2g4a-/- and Dagla-/- mice responded normally, demonstrating that MGL is a critical enzyme for fever, which functions independently of endocannabinoid signals. Intracerebroventricular administration of PGE2 caused fever similarly in Mgll-/- and wild-type control mice, suggesting a lack of pyretic PGE2 production in Mgll-/- hypothalamus, which was confirmed by lipidomics analysis. Normal blood cytokine responses after LPS administration suggested that MGL-deficiency does not affect pyretic cytokine productions. Diurnal body temperature profiles were normal in Mgll-/- mice, demonstrating that MGL is unrelated to physiological thermoregulation. In conclusion, MGL-dependent hydrolysis of endocannabinoid 2-arachidonoylglycerol is necessary for pyretic PGE2 production in the hypothalamus.
Collapse
Affiliation(s)
- Yoshihiro Kita
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Life Sciences Core Facility, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| | - Kenij Yoshida
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Suzumi M. Tokuoka
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Fumie Hamano
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Life Sciences Core Facility, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Niigata, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Niigata, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takao Shimizu
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|