1
|
Paciello F, Pisani A, Rolesi R, Montuoro R, Mohamed-Hizam V, Boni G, Ripoli C, Galli J, Sisto R, Fetoni AR, Grassi C. Oxidative stress and inflammation cause auditory system damage via glial cell activation and dysregulated expression of gap junction proteins in an experimental model of styrene-induced oto/neurotoxicity. J Neuroinflammation 2024; 21:4. [PMID: 38178142 PMCID: PMC10765700 DOI: 10.1186/s12974-023-02996-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/13/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Redox imbalance and inflammation have been proposed as the principal mechanisms of damage in the auditory system, resulting in functional alterations and hearing loss. Microglia and astrocytes play a crucial role in mediating oxidative/inflammatory injury in the central nervous system; however, the role of glial cells in the auditory damage is still elusive. OBJECTIVES Here we investigated glial-mediated responses to toxic injury in peripheral and central structures of the auditory pathway, i.e., the cochlea and the auditory cortex (ACx), in rats exposed to styrene, a volatile compound with well-known oto/neurotoxic properties. METHODS Male adult Wistar rats were treated with styrene (400 mg/kg daily for 3 weeks, 5/days a week). Electrophysiological, morphological, immunofluorescence and molecular analyses were performed in both the cochlea and the ACx to evaluate the mechanisms underlying styrene-induced oto/neurotoxicity in the auditory system. RESULTS We showed that the oto/neurotoxic insult induced by styrene increases oxidative stress in both cochlea and ACx. This was associated with macrophages and glial cell activation, increased expression of inflammatory markers (i.e., pro-inflammatory cytokines and chemokine receptors) and alterations in connexin (Cxs) and pannexin (Panx) expression, likely responsible for dysregulation of the microglia/astrocyte network. Specifically, we found downregulation of Cx26 and Cx30 in the cochlea, and high level of Cx43 and Panx1 in the ACx. CONCLUSIONS Collectively, our results provide novel evidence on the role of immune and glial cell activation in the oxidative/inflammatory damage induced by styrene in the auditory system at both peripheral and central levels, also involving alterations of gap junction networks. Our data suggest that targeting glial cells and connexin/pannexin expression might be useful to attenuate oxidative/inflammatory damage in the auditory system.
Collapse
Affiliation(s)
- Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Anna Pisani
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rolando Rolesi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Raffaele Montuoro
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Giammarco Boni
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Jacopo Galli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Renata Sisto
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers' Compensation Authority (INAIL), Monte Porzio Catone, Rome, Italy
| | - Anna Rita Fetoni
- Department of Neuroscience, Unit of Audiology, Università Degli Studi di Napoli Federico II, Naples, Italy.
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| |
Collapse
|
2
|
Xu Y, Liu Y, Li K, Miao S, Lv C, Wang C, Zhao J. Regulation of PGE 2 Pathway During Cerebral Ischemia Reperfusion Injury in Rat. Cell Mol Neurobiol 2021; 41:1483-1496. [PMID: 32621176 DOI: 10.1007/s10571-020-00911-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 06/22/2020] [Indexed: 01/14/2023]
Abstract
Stroke is an acute central nervous system disease with high morbidity and mortality rate. Cerebral ischemia reperfusion (I/R) injury is easily induced during the development or treatment of stroke and subsequently leads to more serious brain damage. Prostaglandin E2 (PGE2) is one of the most important inflammatory mediators in the brain and contributes to both physiological and pathophysiological functions. It may be upregulated and subsequently plays a key role in cerebral ischemia reperfusion injury. The synthesis and degradation of PGE2 is an extremely complex process, with multiple key stages and molecules. However, there are few comprehensive and systematic studies conducted to investigate the synthesis and degradation of PGE2 during cerebral I/R injury, which is what we want to demonstrate. In this study, qRT-PCR and immunoblotting demonstrated that the key enzymes in PGE2 synthesis, including COX-1, COX-2, mPGES-1 and mPGES-2, were upregulated during cerebral I/R injury, but 15-PGDH, the main PGE2 degradation enzyme, was downregulated. In addition, two of PGE2 receptors, EP3 and EP4, were also increased. Meanwhile, immunohistochemistry demonstrated the localization of these molecules in ischemic areas, including cortex, striatum and hippocampus, and reflected their expression patterns in different regions. Combining the results of PCR, Western blotting and immunohistochemistry, we can determine where the increase or decrease of these molecules occurs. Overall, these results further indicate a possible pathway that mediates enhanced production of PGE2, and thus that may impact production of inflammatory cytokines including IL-1β and TNF-α during cerebral I/R injury.
Collapse
Affiliation(s)
- Yunfei Xu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China
| | - Ying Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China.
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China.
| | - Kexin Li
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China
| | - Shuying Miao
- Department of Pathology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Caihong Lv
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China
| | - Chunjiang Wang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
3
|
Neuroinflammatory responses in Parkinson's disease: relevance of Ibuprofen in therapeutics. Inflammopharmacology 2020; 29:5-14. [PMID: 33052479 DOI: 10.1007/s10787-020-00764-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) pathogenesis inevitably involves neuroinflammatory responses attained through contribution of both neuron and glial cells. Investigation done in both experimental models of PD and in samples of PD patients suggested the involvement of both central and peripheral inflammatory responses during PD pathogenesis. Such neuroinflammatory responses could be regulated by neuron-glia interaction which is one of the recently focused areas in the field of disease diagnosis, pathogenesis and therapeutics. Such aggravated neuroinflammatory responses during PD are very well associated with augmented levels of cyclooxygenase (COX). An increased expression of cyclooxygenase (COX) with a concomitant increase in the prostaglandin E2 (PGE2) levels has been observed during PD pathology. Ibuprofen is one of the non-steroidal anti-inflammatory drugs (NSAID) and clinically being used for PD patients. This review focuses on the neuroinflammatory responses during PD pathology as well as the effect of ibuprofen on various disease related signaling factors and mechanisms involving nitrosative stress, neurotransmission, neuronal communication and peroxisome proliferator-activated receptor-γ. Such mechanistic effect of ibuprofen has been mostly reported in experimental models of PD and clinical investigations are still required. Since oxidative neuronal death is one of the major neurodegenerative mechanisms in PD, the antioxidant capacity of ibuprofen along with its antidepressant effects have also been discussed. This review will direct the readers towards fulfilling the existing gaps in the mechanistic aspect of ibuprofen and enhance its clinical relevance in PD therapeutics and probably in other age-related neurodegenerative diseases.
Collapse
|
4
|
Jesus LB, Santos AB, Jesus EEV, Santos RGD, Grangeiro MS, Bispo-da-Silva A, Arruda MR, Argolo DS, Pinheiro AM, El-Bachá RS, Costa SL, Costa MFD. IDO, COX and iNOS have an important role in the proliferation of Neospora caninum in neuron/glia co-cultures. Vet Parasitol 2019; 266:96-102. [PMID: 30736955 DOI: 10.1016/j.vetpar.2019.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 01/03/2019] [Accepted: 01/05/2019] [Indexed: 12/23/2022]
Abstract
Central nervous system (CNS) is the main site for encystment of Neospora caninum in different animal species. In this tissue, glial cells (astrocytes and microglia) modulate responses to aggression in order to preserve homeostasis and neuronal function. Previous data showed that when primary cultures of glial cells are infected with N. caninum, they develop gliosis and the immune response is characterized by the release of TNF and IL-10, followed by the control of parasite proliferation. In order to elucidate this control, three enzymatic systems involved in parasite-versus-host interactions were observed on a model of neuron/glia co/cultures obtained from rat brains. Indoleamine 2,3-dioxygenase (IDO), induced nitric oxide synthase (iNOS) responsible for the catabolism of tryptophan and arginine, respectively, and cycloxigenase (COX) were studied comparing their modulation by respective inhibitors with the number of tachyzoites or the immune response measured by the release of IL-10 and TNF. Cells were treated with the inhibitors of iNOS (1.5 mM L-NAME), IDO (1 mM 1-methyl tryptophan), COX-1 (1 μM indomethacin) and COX-2 (1 μM nimesulide) before infection with tachyzoites of N. caninum (1:1 cell: parasite). After 72 h of infection, immunocytochemistry showed astrogliosis and a significant increase in the number and length of neurites, compared with uninfected co-cultures, while an increase of IL-10 and TNF was verified. N. caninum did not change iNOS activity, but the inhibition of the basal levels of this enzyme stimulated parasite proliferation. Additionally, a significant increase of about 40% was verified in the IDO activity, whose inhibition caused 1.2-fold increase in parasitic growth. For COX-2 activity, infection of cultures stimulated a significant increase in release of PGE2 and its inhibition by nimesulide allowed the parasitic growth. These data indicate that iNOS, IDO and COX-2 control the proliferation of N. caninum in this in vitro model. On the other hand, the release of IL-10 by glia besides modulating the inflammation also allow the continuity of parasitism.
Collapse
Affiliation(s)
- L B Jesus
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - A B Santos
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - E E V Jesus
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - R G D Santos
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - M S Grangeiro
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - A Bispo-da-Silva
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - M R Arruda
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - D S Argolo
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - A M Pinheiro
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; Centro de Ciências Agrárias Ambientais e Biológica, Universidade do Recôncavo da Bahia - URBA, R. Ruy Barbosa 710 Centro, CEP 44380-000, Cruz das Almas, Bahia, Brazil
| | - R S El-Bachá
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; INCT de Neurociência Translacional (INNT)- CNPq, Brazil
| | - S L Costa
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; INCT de Neurociência Translacional (INNT)- CNPq, Brazil.
| | - M F D Costa
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; INCT de Neurociência Translacional (INNT)- CNPq, Brazil.
| |
Collapse
|
5
|
Mohan S, Koller EJ, Fazal JA, De Oliveria G, Pawlowicz AI, Doré S. Genetic Deletion of PGF 2α-FP Receptor Exacerbates Brain Injury Following Experimental Intracerebral Hemorrhage. Front Neurosci 2018; 12:556. [PMID: 30233287 PMCID: PMC6134069 DOI: 10.3389/fnins.2018.00556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/23/2018] [Indexed: 01/17/2023] Open
Abstract
Background: The release of inflammatory molecules such as prostaglandins (e.g., PGF2α) is associated with brain damage following an intracerebral hemorrhagic (ICH) stroke; however, the role of PGF2α and its cognate FP receptor in ICH remains unclear. This study focused on investigating the role of the FP receptor as a target for novel neuroprotective drugs in a preclinical model of ICH, aiming to investigate the contribution of the PGF2α-FP axis in modulating functional recovery and anatomical outcomes following ICH. Results: Neurological deficit scores in FP−/− mice were significantly higher compared to WT mice 72 h after ICH (6.1 ± 0.7 vs. 3.1 ± 0.8; P < 0.05). Assessing motor skills, the total time mice stayed on the rotating rod was significantly less in FP−/−mice compared to WT mice 24 h after ICH (27.0 ± 7.5 vs. 52.4 ± 11.2 s; P < 0.05). Using grip strength to quantify forepaw strength, results showed that the FP−/− mice had significantly less strength compared to WT mice 72 h after ICH (96.4 ± 17.0 vs. 129.6 ± 5.9 g; P < 0.01). In addition to the behavioral outcomes, histopathological measurements were made. In Cresyl violet stained brain sections, the FP−/− mice showed a significantly larger lesion volume compared to the WT (15.0 ± 2.2 vs. 3.2 ± 1.7 mm3; P < 0.05 mice.) To estimate the presence of ferric iron in the peri-hematoma area, Perls' staining was performed, which revealed that FP−/− mice had significantly greater staining than the WT mice (186.3 ± 34.4% vs. 86.9 ± 13.0% total positive pixel counts, P < 0.05). Immunoreactivity experiments on brain sections from FP−/− and WT mice post-ICH were performed to monitor changes in microgliosis and astrogliosis using antibodies against Iba1 and GFAP respectively. These experiments showed that FP−/− mice had a trend toward greater astrogliosis than WT mice post-ICH. Conclusion: We showed that deletion of the PGF2α FP receptor exacerbates behavioral impairments and increases lesion volumes following ICH compared to WT-matched controls.Detailed mechanisms responsible for these novel results are actively being pursued.
Collapse
Affiliation(s)
- Shekher Mohan
- Department of Pharmaceutical Sciences, Manchester University, College of Pharmacy, Natural and Health Sciences, Fort Wayne, IN, United States
| | - Emily J Koller
- Department of Anesthesiology, University of Florida, College of Medicine, Gainesville, FL, United States
| | - Jawad A Fazal
- Department of Anesthesiology, University of Florida, College of Medicine, Gainesville, FL, United States
| | - Gabriela De Oliveria
- Department of Anesthesiology, University of Florida, College of Medicine, Gainesville, FL, United States
| | - Anna I Pawlowicz
- Department of Anesthesiology, University of Florida, College of Medicine, Gainesville, FL, United States
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida, College of Medicine, Gainesville, FL, United States.,Departments of Neurology, Psychiatry, Psychology, Pharmaceutics and Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, College of Medicine, Gainesville, FL, United States
| |
Collapse
|
6
|
Domínguez-González M, Puigpinós M, Jové M, Naudi A, Portero-Otín M, Pamplona R, Ferrer I. Regional vulnerability to lipoxidative damage and inflammation in normal human brain aging. Exp Gerontol 2018; 111:218-228. [PMID: 30077575 DOI: 10.1016/j.exger.2018.07.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 06/29/2018] [Accepted: 07/30/2018] [Indexed: 01/08/2023]
Abstract
Oxidative damage and inflammation coexist in healthy human brain aging. The present study analyzes levels of protein adduction by lipid peroxidation (LP) end-products neuroketal (NKT) and malondialdehyde (MDA), as markers of protein oxidative damage, cycloxygenase-2 (COX-2) levels, as a marker of inflammation, and cytochrome P450 2J2 (CYP2J2), responsible of generation of neuroprotective products, in twelve brain regions in normal middle-aged individuals (MA) and old-aged (OA) individuals. In addition, levels of these markers were evaluated as a function of age as a continuous variable and correction for multiple comparisons. Selection of regions was based on their different vulnerability to prevalent neurodegenerative diseases in aging. Our findings show region-dependent LP end-products, COX-2 and CYP2J2 changes in the aging human brain. However, no clear relationship can be established between NKT, MDA, COX-2 and CYP2J2 levels, and regional vulnerability to neurodegeneration in old age.
Collapse
Affiliation(s)
- Mayelín Domínguez-González
- Institute of Neuropathology, Bellvitge University Hospital, Biomedical Research Institute of Bellvitge (IDIBELL), Hospitalet de Llobregat, Spain
| | - Meritxell Puigpinós
- Institute of Neuropathology, Bellvitge University Hospital, Biomedical Research Institute of Bellvitge (IDIBELL), Hospitalet de Llobregat, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Alba Naudi
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Manuel Portero-Otín
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Isidro Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, Biomedical Research Institute of Bellvitge (IDIBELL), Hospitalet de Llobregat, Spain; Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain; Biomedical Research Network Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Spanish Ministry of Science and Innovation, Madrid, Spain; Institute of Neurosciences, University of Barcelona, Hospitalet de Llobregat, Spain.
| |
Collapse
|
7
|
Chen SH, Sung YF, Oyarzabal EA, Tan YM, Leonard J, Guo M, Li S, Wang Q, Chu CH, Chen SL, Lu RB, Hong JS. Physiological Concentration of Prostaglandin E 2 Exerts Anti-inflammatory Effects by Inhibiting Microglial Production of Superoxide Through a Novel Pathway. Mol Neurobiol 2018; 55:8001-8013. [PMID: 29492849 DOI: 10.1007/s12035-018-0965-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/16/2018] [Indexed: 01/21/2023]
Abstract
This study investigated the physiological regulation of brain immune homeostasis in rat primary neuron-glial cultures by sub-nanomolar concentrations of prostaglandin E2 (PGE2). We demonstrated that 0.01 to 10 nM PGE2 protected dopaminergic neurons against LPS-induced neurotoxicity through a reduction of microglial release of pro-inflammatory factors in a dose-dependent manner. Mechanistically, neuroprotective effects elicited by PGE2 were mediated by the inhibition of microglial NOX2, a major superoxide-producing enzyme. This conclusion was supported by (1) the close relationship between inhibition of superoxide and PGE2-induced neuroprotective effects; (2) the mediation of PGE2-induced reduction of superoxide and neuroprotection via direct inhibition of the catalytic subunit of NOX2, gp91phox, rather than through the inhibition of conventional prostaglandin E2 receptors; and (3) abolishment of the neuroprotective effect of PGE2 in NOX2-deficient cultures. In summary, this study revealed a potential physiological role of PGE2 in maintaining brain immune homeostasis and protecting neurons via an EP receptor-independent mechanism.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.
| | - Yueh-Feng Sung
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.,Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Esteban A Oyarzabal
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA
| | - Yu-Mei Tan
- U.S. Environmental Protection Agency, National Exposure Research Lab, Research Triangle Park, NC, USA
| | - Jeremy Leonard
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Mingri Guo
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.,Department of Laboratory Medicine, Tianjin Haihe Hospital/Haihe Clinical Institute of Tianjin Medical University, Tianjin, China
| | - Shuo Li
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.,Department of Respiratory, Tianjin Medical University General Hospital, Tianjin, China
| | - Qingshan Wang
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA
| | - Chun-Hsien Chu
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA
| | - Shiou-Lan Chen
- Department of Neurology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ru-Band Lu
- Institute of Behavioral Medicine, College of Medicine & Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Psychiatry, National Cheng Kung University, Tainan, Taiwan
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
8
|
Al Alam N, Kreydiyyeh SI. Signaling pathway involved in the inhibitory effect of FTY720P on the Na +/K + ATPase in HepG2 cells. J Cell Commun Signal 2017; 11:309-316. [PMID: 28197966 PMCID: PMC5704036 DOI: 10.1007/s12079-016-0369-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 11/28/2016] [Indexed: 02/05/2023] Open
Abstract
The Na+/K+ ATPase modulates the activity of many transporters in the liver, and maintains the ionic constancy of the intracellular milieu, preserving thus normal functioning of hepatocytes. Previous work showed that FTY720P, a sphingosine one phosphate receptor agonist used in the treatment of multiple sclerosis, exerts in HepG2 cells, an inhibitory effect on the activity of the ATPase, mediated via PGE2. This study is an attempt to identify the signaling molecules involved downstream of the prostaglandin. The activity of the ATPase was assayed by measuring the amount of inorganic phosphate liberated in presence and absence of ouabain, a specific inhibitor of the enzyme. The effect of FTY720P and PGE2 disappeared completely in presence of PF-04418948, a blocker of EP2 receptors, RpcAMP, an inhibitor of PKA, PD98059, an inhibitor of ERK, as well as in presence of PTIO, a nitric oxide synthase inhibitor, but was mimicked by butaprost, an EP2 agonist, dbcAMP, a cell permeable cAMP analogue, and SNAP1,a nitric oxide generator. PGE2 and dbcAMP increased the expression of phosphorylated ERK but not total ERK. This increase did not appear however in presence of PTIO, indicating that PKA is upstream of NO. It was concluded that FTY 720P induces PGE2 release which activates NOS leading to NO production and ERK activation. ERK then inhibits directly or indirectly the Na+/K+ ATPase.
Collapse
Affiliation(s)
- Nadine Al Alam
- Department of Biology, Faculty of Arts & Sciences, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
9
|
Stucky EC, Erndt-Marino J, Schloss RS, Yarmush ML, Shreiber DI. Prostaglandin E 2 Produced by Alginate-Encapsulated Mesenchymal Stromal Cells Modulates the Astrocyte Inflammatory Response. NANO LIFE 2017; 7:1750005. [PMID: 29682085 PMCID: PMC5903452 DOI: 10.1142/s1793984417500052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Astroglia are well known for their role in propagating secondary injury following brain trauma. Modulation of this injury cascade, including inflammation, is essential to repair and recovery. Mesenchymal stromal cells (MSCs) have been demonstrated as trophic mediators in several models of secondary CNS injury, however, there has been varied success with the use of direct implantation due to a failure to persist at the injury site. To achieve sustained therapeutic benefit, we have encapsulated MSCs in alginate microspheres and evaluated the ability of these encapsulated MSCs to attenuate neuro-inflammation. In this study, astroglial cultures were administered lipopolysaccharide (LPS) to induce inflammation and immediately co-cultured with encapsulated or monolayer human MSCs. Cultures were assayed for the pro-inflammatory cytokine tumor necrosis factor alpha (TNF-α) produced by astroglia, MSC-produced prostaglandin E2, and expression of neurotrophin-associated genes. We found that encapsulated MSCs significantly reduced TNF-α produced by LPS-stimulated astrocytes, more effectively than monolayer MSCs, and this enhanced benefit commences earlier than that of monolayer MSCs. Furthermore, in support of previous findings, encapsulated MSCs constitutively produced high levels of PGE2, while monolayer MSCs required the presence of inflammatory stimuli to induce PGE2 production. The early, constitutive presence of PGE2 significantly reduced astrocyte-produced TNF-α, while delayed administration had no effect. Finally, MSC-produced PGE2 was not only capable of modulating inflammation, but appears to have an additional role in stimulating astrocyte neurotrophin production. Overall, these results support the enhanced benefit of encapsulated MSC treatment, both in modulating the inflammatory response and providing neuroprotection.
Collapse
Affiliation(s)
- Elizabeth C Stucky
- Department of Chemical and Biochemical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, USA
| | - Joshua Erndt-Marino
- Department of Biomedical Engineering, The College of New Jersey, 2000 Pennington Road, Ewing, New Jersey 08628, USA
| | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, USA
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, USA
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, USA
| |
Collapse
|
10
|
Nagano T, Nishiyama R, Sanada A, Mutaguchi Y, Ioku A, Umeki H, Kishimoto S, Yamanaka D, Kimura SH, Takemura M. Prostaglandin E 2 potentiates interferon-γ-induced nitric oxide production in cultured rat microglia. J Neurochem 2017; 140:605-612. [PMID: 27973680 DOI: 10.1111/jnc.13926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/30/2016] [Accepted: 12/06/2016] [Indexed: 12/26/2022]
Abstract
Prostaglandin E2 (PGE2 ) plays crucial roles in managing microglial activation through the prostanoid EP2 receptor, a PGE2 receptor subtype. In this study, we report that PGE2 enhances interferon-γ (IFN-γ)-induced nitric oxide production in microglia. IFN-γ increased the release of nitrite, a metabolite of nitric oxide, which was augmented by PGE2 , although PGE2 by itself slightly affects nitrite release. The potentiating effect of PGE2 was positively associated with increased expression of inducible nitric oxide synthase. In contrast to nitrite release induced by IFN-γ, lipopolysaccharide-induced nitrite release was not affected by PGE2 . An EP2 agonist, ONO-AE1-259-01 also augmented IFN-γ-induced nitrite release, while an EP1 agonist, ONO-DI-004, an EP3 agonist, ONO-AE-248, or an EP4 agonist, ONO-AE1-329, did not. In addition, the potentiating effect of PGE2 was inhibited by an EP2 antagonist, PF-04418948, but not by an EP1 antagonist, ONO-8713, an EP3 antagonist, ONO-AE3-240, or an EP4 antagonist, ONO-AE3-208, at 10-6 M. Among the EP agonists, ONO-AE1-259-01 alone was able to accumulate cyclic adenosine monophosphate (AMP), and among the EP antagonists, PF-04418948 was the only one able to inhibit PGE2 -increased intracellular cyclic AMP accumulation. On the other hand, IFN-γ promoted phosphorylation of signal transducer and activator of transcription 1, which was not affected by PGE2 . Furthermore, other prostanoid receptor agonists, PGD2 , PGF2α , iloprost, and U-46119, slightly affected IFN-γ-induced nitrite release. These results indicate that PGE2 potentiates IFN-γ-induced nitric oxide production in microglia through the EP2 receptor, which may shed light on one of the pro-inflammatory aspects of PGE2 .
Collapse
Affiliation(s)
- Takayuki Nagano
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Ryo Nishiyama
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Ayaka Sanada
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yukiko Mutaguchi
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Anna Ioku
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hirohisa Umeki
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Satoshi Kishimoto
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Daisuke Yamanaka
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Shinya H Kimura
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Motohiko Takemura
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
11
|
Feinstein DL, Kalinin S, Braun D. Causes, consequences, and cures for neuroinflammation mediated via the locus coeruleus: noradrenergic signaling system. J Neurochem 2016; 139 Suppl 2:154-178. [PMID: 26968403 DOI: 10.1111/jnc.13447] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 12/31/2022]
Abstract
Aside from its roles in as a classical neurotransmitter involved in regulation of behavior, noradrenaline (NA) has other functions in the CNS. This includes restricting the development of neuroinflammatory activation, providing neurotrophic support to neurons, and providing neuroprotection against oxidative stress. In recent years, it has become evident that disruption of physiological NA levels or signaling is a contributing factor to a variety of neurological diseases and conditions including Alzheimer's disease (AD) and Multiple Sclerosis. The basis for dysregulation in these diseases is, in many cases, due to damage occurring to noradrenergic neurons present in the locus coeruleus (LC), the major source of NA in the CNS. LC damage is present in AD, multiple sclerosis, and a large number of other diseases and conditions. Studies using animal models have shown that experimentally induced lesion of LC neurons exacerbates neuropathology while treatments to compensate for NA depletion, or to reduce LC neuronal damage, provide benefit. In this review, we will summarize the anti-inflammatory and neuroprotective actions of NA, summarize examples of how LC damage worsens disease, and discuss several approaches taken to treat or prevent reductions in NA levels and LC neuronal damage. Further understanding of these events will be of value for the development of treatments for AD, multiple sclerosis, and other diseases and conditions having a neuroinflammatory component. The classical neurotransmitter noradrenaline (NA) has critical roles in modulating behaviors including those involved in sleep, anxiety, and depression. However, NA can also elicit anti-inflammatory responses in glial cells, can increase neuronal viability by inducing neurotrophic factor expression, and can reduce neuronal damage due to oxidative stress by scavenging free radicals. NA is primarily produced by tyrosine hydroxylase (TH) expressing neurons in the locus coeruleus (LC), a relatively small brainstem nucleus near the IVth ventricle which sends projections throughout the brain and spinal cord. It has been known for close to 50 years that LC neurons are lost during normal aging, and that loss is exacerbated in neurological diseases including Parkinson's disease and Alzheimer's disease. LC neuronal damage and glial activation has now been documented in a variety of other neurological conditions and diseases, however, the causes of LC damage and cell loss remain largely unknown. A number of approaches have been developed to address the loss of NA and increased inflammation associated with LC damage, and several methods are being explored to directly minimize the extent of LC neuronal cell loss or function. In this review, we will summarize some of the consequences of LC loss, consider several factors that likely contribute to that loss, and discuss various ways that have been used to increase NA or to reduce LC damage. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Douglas L Feinstein
- Department of Anesthesiology, University of Illinois, Chicago, IL, USA. .,Jesse Brown VA Medical Center, Chicago, IL, USA.
| | - Sergey Kalinin
- Department of Anesthesiology, University of Illinois, Chicago, IL, USA.,Jesse Brown VA Medical Center, Chicago, IL, USA
| | - David Braun
- Department of Anesthesiology, University of Illinois, Chicago, IL, USA.,Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
12
|
Rashed E, Lizano P, Dai H, Thomas A, Suzuki CK, Depre C, Qiu H. Heat shock protein 22 (Hsp22) regulates oxidative phosphorylation upon its mitochondrial translocation with the inducible nitric oxide synthase in mammalian heart. PLoS One 2015; 10:e0119537. [PMID: 25746286 PMCID: PMC4352051 DOI: 10.1371/journal.pone.0119537] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 01/18/2015] [Indexed: 11/21/2022] Open
Abstract
Objectives Stress-inducible heat shock protein 22 (Hsp22) confers protection against ischemia through induction of the inducible isoform of nitric oxide synthase (iNOS). Hsp22 overexpression in vivo stimulates cardiac mitochondrial respiration, whereas Hsp22 deletion in vivo significantly reduces respiration. We hypothesized that Hsp22-mediated regulation of mitochondrial function is dependent upon its mitochondrial translocation together with iNOS. Methods and Results Adenoviruses harboring either the full coding sequence of Hsp22 (Ad-WT-Hsp22) or a mutant lacking a N-terminal 20 amino acid putative mitochondrial localization sequence (Ad-N20-Hsp22) were generated, and infected in rat neonatal cardiomyocytes. Compared to β-Gal control, WT-Hsp22 accumulated in mitochondria by 2.5 fold (P<0.05) and increased oxygen consumption rates by 2-fold (P<0.01). This latter effect was abolished upon addition of the selective iNOS inhibitor, 1400W. Ad-WT-Hsp22 significantly increased global iNOS expression by about 2.5-fold (P<0.01), and also increased iNOS mitochondrial localization by 4.5 fold vs β-gal (P<0.05). Upon comparable overexpression, the N20-Hsp22 mutant did not show significant mitochondrial translocation or stimulation of mitochondrial respiration. Moreover, although N20-Hsp22 did increase global iNOS expression by 4.6-fold, it did not promote iNOS mitochondrial translocation. Conclusion Translocation of both Hsp22 and iNOS to the mitochondria is necessary for Hsp22-mediated stimulation of oxidative phosphorylation.
Collapse
Affiliation(s)
- Eman Rashed
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Paulo Lizano
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Huacheng Dai
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Andrew Thomas
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Carolyn K. Suzuki
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Christophe Depre
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Hongyu Qiu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
- Department of Basic Science, Division of Physiology, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
- * E-mail:
| |
Collapse
|
13
|
Tolba RH, Fet N, Yonezawa K, Taura K, Nakajima A, Hata K, Okamura Y, Uchinami H, Klinge U, Minor T, Yamaoka Y, Yamamoto Y. Role of preferential cyclooxygenase-2 inhibition by meloxicam in ischemia/reperfusion injury of the rat liver. Eur Surg Res 2014; 53:11-24. [PMID: 24854565 DOI: 10.1159/000362411] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Ischemia/reperfusion injury (IRI) is one of the major clinical problems in liver and transplant surgery. Livers subjected to warm ischemia in vivo often show a severe dysfunction and the release of numerous inflammatory cytokines and arachidonic acid metabolites. Cyclooxygenase (COX)-2 is the inducible isoform of an intracellular enzyme that converts arachidonic acid into prostaglandins. The aim of the study was to evaluate the effect of COX-2 inhibition and the role of Kupffer cells in IRI of the liver. METHODS Male Wistar rats [250- 280 g body weight (BW)] were anesthetized and subjected to 30-min warm ischemia of the liver (Pringle's maneuver) and 60-min reperfusion after median laparotomy. The I/R group received no additional treatment. In the COX-2 inhibitor (COX-2I) group, the animals received 1 mg/kg BW meloxicam prior to operation. Gadolinium chloride (GdCl3) (10 mg/kg BW) was given 24 h prior to operation in the GdCl3 and GdCl3 + COX-2I groups for the selective depletion of Kupffer cells. The GdCl3 + COX-2I group received both GdCl3 and meloxicam treatment prior to operation. Blood and liver samples were obtained at the end of the experiments for further investigations. RESULTS After 30 min of warm ischemia in vivo, severe hepatocellular damage was observed in the I/R group. These impairments could be significantly prevented by the selective COX-2 inhibition and the depletion of Kupffer cells. Alanine aminotransferase was significantly reduced upon meloxicam and GdCl3 treatment compared to the I/R group: I/R, 3,240 ± 1,262 U/l versus COX-2I, 973 ± 649 U/l, p < 0.001; I/R versus GdCl3, 1,611 ± 600 U/l, p < 0.05, and I/R versus GdCl3 + COX-2I, 1,511 ± 575 U/l, p < 0.01. Plasma levels of tumor necrosis factor alpha (TNF-α) were significantly reduced in the COX-2I treatment group compared to I/R (3.5 ± 1.5 vs. 16.3 ± 11.7 pg/ml, respectively; p < 0.05). Similarly, the amount of TxB2, a marker for COX-2 metabolism, was significantly reduced in the meloxicam treatment groups compared to the I/R group: I/R, 22,500 ± 5,210 pg/ml versus COX-2I, 1,822 ± 938 pg/ml, p < 0.001, and I/R versus GdCl3 + COX-2I, 1,530 ± 907 pg/ml, p < 0.001. All values are given as mean ± SD (n = 6). CONCLUSION These results suggest that the inhibition of COX-2 suppressed the initiation of an inflammatory cascade by attenuating the release of TNF-α, which is an initiator of the inflammatory reaction in hepatic IRI. Therefore, we conclude that preferential inhibition of COX-2 is a possible therapeutic approach against warm IRI of the liver.
Collapse
Affiliation(s)
- René H Tolba
- Division of Surgical Research, University Hospital Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Singh V, Bhatia H, Kumar A, de Oliveira A, Fiebich B. Histone deacetylase inhibitors valproic acid and sodium butyrate enhance prostaglandins release in lipopolysaccharide-activated primary microglia. Neuroscience 2014; 265:147-57. [DOI: 10.1016/j.neuroscience.2014.01.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 01/17/2014] [Accepted: 01/19/2014] [Indexed: 10/25/2022]
|
15
|
Seyedi SY, Salehi F, Payandemehr B, Hossein S, Hosseini-Zare MS, Nassireslami E, Yazdi BB, Sharifzadeh M. Dual effect of cAMP agonist on ameliorative function of PKA inhibitor in morphine-dependent mice. Fundam Clin Pharmacol 2013; 28:445-54. [PMID: 24033391 DOI: 10.1111/fcp.12045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 06/24/2013] [Accepted: 07/08/2013] [Indexed: 12/15/2022]
Abstract
The present study shows interactive effects of bucladesine (db-cAMP) as a cyclic adenosine monophosphate (cAMP) agonist and H-89 as a protein kinase A (PKA) inhibitor on naloxone-induced withdrawal signs in morphine-dependent mice. Animals were treated subcutaneously with morphine thrice daily with doses progressively increased from 50 to 125 mg/kg. A last dose of morphine (50 mg/kg) was administered on the 4th day. Several withdrawal signs were precipitated by intraperitoneal (i.p.) administration of naloxone (5 mg/kg). Different doses of bucladesine (50, 100, 200 nm/mouse) and H-89 (0.05, 0.5, 1, 5 mg/kg) were administered (i.p.) 60 min before naloxone injection. In combination groups, bucladesine was injected 15 min before H-89 injection. Single administration of H-89 (0.5, 1, 5 mg/kg) and bucladesine (50, 100 nm/mouse) significantly attenuated prominent behavioral signs of morphine withdrawal. Lower doses of bucladesine (50, 100 nm/mouse) in combination with H-89 (0.05 mg/kg) increased the inhibitory effects of H-89 on withdrawal signs while in high dose (200 nm/mouse) decreased the ameliorative function of H-89 (0.05 mg/kg) in morphine-dependent animals. It is concluded that H-89 and bucladesine could affect morphine withdrawal syndrome via possible interaction with cyclic nucleotide messengering systems, protein kinase A signaling pathways, and modified related neurotransmitters.
Collapse
Affiliation(s)
- Seyedeh Y Seyedi
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, PO Box 14155-6451, Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Porter RL, Georger MA, Bromberg O, McGrath KE, Frisch BJ, Becker MW, Calvi LM. Prostaglandin E2 increases hematopoietic stem cell survival and accelerates hematopoietic recovery after radiation injury. Stem Cells 2013; 31:372-83. [PMID: 23169593 DOI: 10.1002/stem.1286] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 10/28/2012] [Indexed: 01/02/2023]
Abstract
Hematopoietic stem and progenitor cells (HSPCs), which continuously maintain all mature blood cells, are regulated within the marrow microenvironment. We previously reported that pharmacologic treatment of naïve mice with prostaglandin E2 (PGE2) expands HSPCs. However, the cellular mechanisms mediating this expansion remain unknown. Here, we demonstrate that PGE2 treatment in naïve mice inhibits apoptosis of HSPCs without changing their proliferation rate. In a murine model of sublethal total body irradiation (TBI), in which HSPCs are rapidly lost, treatment with a long-acting PGE2 analog (dmPGE2) reversed the apoptotic program initiated by TBI. dmPGE2 treatment in vivo decreased the loss of functional HSPCs following radiation injury, as demonstrated both phenotypically and by their increased reconstitution capacity. The antiapoptotic effect of dmPGE2 on HSPCs did not impair their ability to differentiate in vivo, resulting instead in improved hematopoietic recovery after TBI. dmPGE2 also increased microenvironmental cyclooxygenase-2 expression and expanded the α-smooth muscle actin-expressing subset of marrow macrophages, thus enhancing the bone marrow microenvironmental response to TBI. Therefore, in vivo treatment with PGE2 analogs may be particularly beneficial to HSPCs in the setting of injury by targeting them both directly and also through their niche. The current data provide rationale for in vivo manipulation of the HSPC pool as a strategy to improve recovery after myelosuppression.
Collapse
Affiliation(s)
- Rebecca L Porter
- Department of Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Nisar A, Malik AH, Zargar MA. Atropa acuminata Royle Ex Lindl. blunts production of pro-inflammatory mediators eicosanoids., leukotrienes, cytokines in vitro and in vivo models of acute inflammatory responses. JOURNAL OF ETHNOPHARMACOLOGY 2013; 147:584-594. [PMID: 23528361 DOI: 10.1016/j.jep.2013.03.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/21/2013] [Accepted: 03/11/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atropa acuminata Royle Ex Lindl. has been widely used in folk medicine for several inflammatory disorders such as arthritis, asthma, conjunctivitis, encephalitis, pancreatitis, peritonitis, acute infections and neuroinflammatory disorders. AIM OF THE STUDY Our aim was to evaluate Atropa acuminata for its anti-inflammatory properties and to delineate its possible mechanism of action on the modulation of the inflammatory mediators. MATERIALS AND METHODS We investigated the inhibitory action of ethanolic extract of Atropa acuminata (AAEE) on production of NO, TNF-α and IL-1β in lipopolysaccharide (LPS)-stimulated RAW264.7 cells and also assayed it for COX 1/2 and 5-LOX inhibitory activities. Next AAEE was tested in acute inflammatory animal models., carragenean induced rat paw edema, carragenean induce pleurisy in rats and vascular permeability in mice and the effects on NO, PGE2 and LTB4 production in the pleural fluid and paw exudates were evaluated. In addition the effects on leukocyte migration and exudation and vascular permeability were also observed. RESULTS Our findings summarized novel anti-inflammatory mechanisms for Atropa acuminata based on dual in vitro cyclooxygenase 1/2/ and 5-Lipoxygenase inhibitory activities and also significant downregulation of nitric oxide (NO) and pro-inflammatory cytokin (TNF-α and Il-1 β) release in LPS-stimulated RAW 246.7 macrophage cell line. In acute inflammatory models in vivo (carragenean induced edema, carragenean induced pleurisy in rats and vascular permeability in mice), AAEE exhibited an extensive diverse mechanism for anti-inflammatory properties. This was indicated on the basis of dose dependent suppression of multi targeted inflammatory mediators., NO, TNF-α and IL-1β, eicosanoids., PGE2 and leukotrienes., LTB4 along with significantly decreased leucocyte migration, exudation and decreased vascular permeability. These effects were more potent and prolonged than traditional NSAIDS, thereby indicating fewer side effects. AAEE was found to be safe for long term administration, as confirmed by the results of acute toxicity studies and MTT assay. The complex mode of action of the herbs was attributed possibly due to the high polyphenolic, flavanol and flavonoid content present in the extracts as observed by means of quantitative screening for phytochemicals. CONCLUSION Our study provides scientific evidence to support the traditional anti-inflammatory uses of Atropa acuminata and is probably due to inhibitory effects on multiple inflammatory mediators which indicates a promising potential for the development of a strong anti-inflammatory agent from this plant.
Collapse
Affiliation(s)
- Albeena Nisar
- Department of Biochemistry, University of Kashmir, Srinagar, J&K 190006, India.
| | | | | |
Collapse
|
18
|
Enhanced prostacyclin synthesis by adenoviral gene transfer reduced glial activation and ameliorated dopaminergic dysfunction in hemiparkinsonian rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:649809. [PMID: 23691265 PMCID: PMC3649752 DOI: 10.1155/2013/649809] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 02/10/2013] [Accepted: 02/18/2013] [Indexed: 12/19/2022]
Abstract
Prostacyclin (PGI2), a potent vasodilator and platelet antiaggregatory eicosanoid, is cytoprotective in cerebral circulation. It is synthesized from arachidonic acid (AA) by the sequential action of cyclooxygenase- (COX-) 1 or 2 and prostacyclin synthase (PGIS). Because prostacyclin is unstable in vivo, PGI2 analogs have been developed and demonstrated to protect against brain ischemia. This work attempts to selectively augment PGI2 synthesis in mixed glial culture or in a model of Parkinson's disease (PD) by direct adenoviral gene transfer of prostacyclin biosynthetic enzymes and examines whether it confers protection in cultures or in vivo. Confluent mixed glial cultures actively metabolized exogenous AA into PGE2 and PGD2. These PGs were largely NS398 sensitive and considered as COX-2 products. Gene transfer of AdPGIS to the cultures effectively shunted the AA catabolism to prostacyclin synthesis and concurrently reduced cell proliferation. Furthermore, PGIS overexpression significantly reduced LPS stimulation in cultures. In vivo, adenoviral gene transfer of bicistronic COX-1/PGIS to substantia nigra protected 6-OHDA- induced dopamine depletion and ameliorated behavioral deficits. Taken together, this study shows that enhanced prostacyclin synthesis reduced glial activation and ameliorated motor dysfunction in hemiparkinsonian rats. Prostacyclin may have a neuroprotective role in modulating the inflammatory response in degenerating nigra-striatal pathway.
Collapse
|
19
|
Quan Y, Jiang J, Dingledine R. EP2 receptor signaling pathways regulate classical activation of microglia. J Biol Chem 2013; 288:9293-302. [PMID: 23404506 DOI: 10.1074/jbc.m113.455816] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of EP2 receptors by prostaglandin E2 (PGE2) promotes brain inflammation in neurodegenerative diseases, but the pathways responsible are unclear. EP2 receptors couple to Gαs and increase cAMP, which associates with protein kinase A (PKA) and cAMP-regulated guanine nucleotide exchange factors (Epacs). Here, we studied EP2 function and its signaling pathways in rat microglia in their resting state or undergoing classical activation in vitro following treatment with low concentrations of lipopolysaccharide and interferon-γ. Real time PCR showed that PGE2 had no effect on expression of CXCL10, TGF-β1, and IL-11 and exacerbated the rapid up-regulation of mRNAs encoding cyclooxygenase-2, inducible NOS, IL-6, and IL-1β but blunted the production of mRNAs encoding TNF-α, IL-10, CCL3, and CCL4. These effects were mimicked fully by the EP2 agonist butaprost but only weakly by the EP1/EP3 agonist 17-phenyl trinor PGE2 or the EP4 agonist CAY10598 and not at all by the EP3/EP1 agonist sulprostone and confirmed by protein measurements of cyclooxygenase-2, IL-6, IL-10, and TNF-α. In resting microglia, butaprost induced cAMP formation and altered the mRNA expression of inflammatory mediators, but protein expression was unchanged. The PKA inhibitor H89 had little or no effect on inflammatory mediators modulated by EP2, whereas the Epac activator 8-(4-chlorophenylthio)-2'-O-methyladenosine 3',5'-cyclic monophosphate acetoxymethyl ester mimicked all butaprost effects. These results indicate that EP2 activation plays a complex immune regulatory role during classical activation of microglia and that Epac pathways are prominent in this role.
Collapse
Affiliation(s)
- Yi Quan
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
20
|
Gavins FNE, Hughes EL, Buss NAPS, Holloway PM, Getting SJ, Buckingham JC. Leukocyte recruitment in the brain in sepsis: involvement of the annexin 1-FPR2/ALX anti-inflammatory system. FASEB J 2012; 26:4977-89. [PMID: 22964301 DOI: 10.1096/fj.12-205971] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Unregulated inflammation underlies many diseases, including sepsis. Much interest lies in targeting anti-inflammatory mechanisms to develop new treatments. One such target is the anti-inflammatory protein annexin A1 (AnxA1) and its receptor, FPR2/ALX. Using intravital videomicroscopy, we investigated the role of AnxA1 and FPR2/ALX in a murine model of endotoxin-induced cerebral inflammation [intraperitoneal injection of lipopolysaccharide (LPS)]. An inflammatory response was confirmed by elevations in proinflammatory serum cytokines, increased cerebrovascular permeability, elevation in brain myeloperoxidase, and increased leukocyte rolling and adhesion in cerebral venules of wild-type (WT) mice, which were further exacerbated in AnxA1-null mice. mRNA expression of TLR2, TLR4, MyD-88, and Ly96 was also assessed. The AnxA1-mimetic peptide, AnxA1(Ac2-26) (100 μg/mouse, ∼33 μmol) mitigated LPS-induced leukocyte adhesion in WT and AnxA1-null animals without affecting leukocyte rolling, in comparison to saline control. AnxA1(Ac2-26) effects were attenuated by Boc2 (pan-FPR antagonist, 10 μg/mouse, ∼12 nmol), and by minocycline (2.25 mg/mouse, ∼6.3 nmol). The nonselective Fpr agonists, fMLP (6 μg/mouse, ∼17 nmol) and AnxA1(Ac2-26), and the Fpr2-selective agonist ATLa (5 μg/mouse, ∼11 nmol) were without effect in Fpr2/3(-/-) mice. In summary, our novel results demonstrate that the AnxA1/FPR2 system has an important role in effecting the resolution of cerebral inflammation in sepsis and may, therefore, provide a novel therapeutic target.
Collapse
Affiliation(s)
- Felicity N E Gavins
- Division of Brain Sciences, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Burlington Danes Bldg., Du Cane Rd., London W12 0NN, UK.
| | | | | | | | | | | |
Collapse
|
21
|
Peng F, Wang G, Li X, Cao D, Yang Z, Ma L, Ye H, Liang X, Ran Y, Chen J, Qiu J, Xie C, Deng C, Xiang M, Peng A, Wei Y, Chen L. Rational design, synthesis, and pharmacological properties of pyranochalcone derivatives as potent anti-inflammatory agents. Eur J Med Chem 2012; 54:272-80. [DOI: 10.1016/j.ejmech.2012.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Revised: 04/28/2012] [Accepted: 05/03/2012] [Indexed: 11/29/2022]
|
22
|
Lima IVDA, Bastos LFS, Limborço-Filho M, Fiebich BL, de Oliveira ACP. Role of prostaglandins in neuroinflammatory and neurodegenerative diseases. Mediators Inflamm 2012; 2012:946813. [PMID: 22778499 PMCID: PMC3385693 DOI: 10.1155/2012/946813] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 04/05/2012] [Indexed: 11/17/2022] Open
Abstract
Increasing data demonstrates that inflammation participates in the pathophysiology of neurodegenerative diseases. Among the different inflammatory mediators involved, prostaglandins play an important role. The effects induced by prostaglandins might be mediated by activation of their known receptors or by nonclassical mechanisms. In the present paper, we discuss the evidences that link prostaglandins, as well as the enzymes that produce them, to some neurological diseases.
Collapse
Affiliation(s)
- Isabel Vieira de Assis Lima
- Department of Pharmacology, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Leandro Francisco Silva Bastos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, Brazil
- Department of Psychology and Neuroscience, Muenzinger Building, Colorado University of Colorado Boulder, Avenida, Boulder, CO 80309-0354, USA
| | - Marcelo Limborço-Filho
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, Brazil
| | - Bernd L. Fiebich
- Department of Psychiatry and Psychotherapy, University of Freiburg Medical School, Hauptstraße 5, 79104 Freiburg, Germany
- VivaCell Biotechnology GmbH, Ferdinand-Porsche-Straße 5, 79211 Denzlingen, Germany
| | - Antonio Carlos Pinheiro de Oliveira
- Department of Pharmacology, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
- Department of Psychiatry and Psychotherapy, University of Freiburg Medical School, Hauptstraße 5, 79104 Freiburg, Germany
| |
Collapse
|
23
|
Maity B, Yadav SK, Patro BS, Tyagi M, Bandyopadhyay SK, Chattopadhyay S. Molecular mechanism of the anti-inflammatory activity of a natural diarylnonanoid, malabaricone C. Free Radic Biol Med 2012; 52:1680-91. [PMID: 22343417 DOI: 10.1016/j.freeradbiomed.2012.02.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 02/01/2012] [Accepted: 02/04/2012] [Indexed: 11/17/2022]
Abstract
The spice-derived phenolic, malabaricone C (mal C), has recently been shown to accelerate healing of the indomethacin-induced gastric ulceration in mice. In this study, we explored its anti-inflammatory activity and investigated the underlying mechanism of the action. Mal C suppressed the microvascular permeability and the levels of tumor necrosis factor-α, interleukin-1β, and nitric oxide in the lipopolysaccharide (LPS)-administered mice. At a dose of 10 mg/kg, it showed anti-inflammatory activity comparable to that of omeprazole (5 mg/kg) and dexamethasone (50 mg/kg). It also reduced the expression and activities of inducible nitric oxide synthase, cyclooxygenase-2, as well as the pro- vs anti-inflammatory cytokine ratio in the LPS-treated RAW macrophages. Mal C was found to inhibit LPS-induced NF-kB activation in RAW 264.7 cells by blocking the MyD88-dependent pathway. Mal C suppressed NF-κB activation and iNOS promoter activity, which correlated with its inhibitory effect on IκB phosphorylation and degradation, and NF-κB nuclear translocation, in the LPS-stimulated macrophages. It also inhibited LPS-induced phosphorylation of p38 and JNK, which are also upstream activators of NF-κB, without affecting Akt phosphorylation. Mal C also effectively blocked the PKR-mediated activation of NF-κB. These findings indicate that mal C exerts an anti-inflammatory effect through NF-κB-responsive inflammatory gene expressions by inhibiting the p38 and JNK-dependent canonical NF-κB pathway as well as the PKR pathway, and is a potential therapeutic agent against acute inflammation.
Collapse
Affiliation(s)
- Biswanath Maity
- Department of Biochemistry, Dr. B.C. Roy Post Graduate Institute of Basic Medical Sciences & IPGME&R, 244B, Acharya Jagadish Chandra Bose Road, Kolkata 700 020, India
| | | | | | | | | | | |
Collapse
|
24
|
Polak PE, Kalinin S, Braun D, Sharp A, Lin SX, Feinstein DL. The vincamine derivative vindeburnol provides benefit in a mouse model of multiple sclerosis: effects on the Locus coeruleus. J Neurochem 2012; 121:206-16. [PMID: 22288774 DOI: 10.1111/j.1471-4159.2012.07673.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The endogenous neurotransmitter noradrenaline (NA) plays several roles in maintaining brain homeostasis, including exerting anti-inflammatory and neuroprotective effects. The primary source of NA in the CNS are tyrosine hydroxylase (TH)-positive neurons located in the Locus coeruleus (LC) which send projections throughout the brain and spinal cord. We recently demonstrated that dysregulation of the LC:Noradrenergic system occurs in experimental autoimmune encephalomyelitis as well as in MS patients, associated with damage occurring to LC neurons. Vindeburnol, a structural analog of the cerebral vasodilator vincamine, was previously reported to increase TH expression and activity in LC neurons. Female C57BL/6 mice were immunized with myelin oligodendrocyte glycoprotein (MOG)(35-55) peptide, and treated with vindeburnol at the first appearance of clinical signs. Clinical signs continued to increase for about 1 week, at which point mice in the vehicle group continued to worsen while vindeburnol-treated mice showed improvement. Pro-inflammatory cytokine production from splenic T cells was not reduced by vindeburnol suggesting primarily central actions of treatment. In the cerebellum, vindeburnol decreased astrocyte activation and reduced the number of demyelinated regions. Vindeburnol reduced astrocyte activation in the LC, reduced TH+ neuronal hypertrophy, increased expression of several genes involved in LC survival and maturation, and increased NA levels in the spinal cord. These results suggest that treatments with drugs such as vindeburnol which target LC survival or function could be of benefit in MS patients.
Collapse
Affiliation(s)
- Paul E Polak
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
25
|
Wang G, Deng C, Xie C, Ma L, Yang J, Qiu N, Xu Q, Chen T, Peng F, Chen J, Qiu J, Peng A, Wei Y, Chen L. Synthesis and biological evaluation of novel dimethyl[1,1′-biphenyl]-2,2′-dicarboxylate derivatives containing thiazolidine-2,4-dione for the treatment of concanavalin A-induced acute liver injury of BALB/c mice. Eur J Med Chem 2011; 46:5941-8. [DOI: 10.1016/j.ejmech.2011.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 09/26/2011] [Accepted: 10/02/2011] [Indexed: 11/28/2022]
|
26
|
Affiliation(s)
- Nephi Stella
- Department of Pharmacology, Psychiatry, and Behavioral Sciences, University of Washington, Seattle, WA 98195-7280, USA.
| |
Collapse
|
27
|
Stoffels B, Yonezawa K, Yamamoto Y, Schäfer N, Overhaus M, Klinge U, Kalff JC, Minor T, Tolba RH. Meloxicam, a COX-2 inhibitor, ameliorates ischemia/reperfusion injury in non-heart-beating donor livers. ACTA ACUST UNITED AC 2011; 47:109-17. [PMID: 21757922 DOI: 10.1159/000329414] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 05/17/2011] [Indexed: 01/14/2023]
Abstract
BACKGROUND/AIMS Chronic organ donor shortage has led to the consideration to expand the donor pool with livers from non-heart-beating donors (NHBD), although a higher risk of graft dys- or nonfunction is associated with these livers. We examined the effects of selective cyclooxygenase-2 (COX-2) inhibition on hepatic warm ischemia (WI) reperfusion (I/R) injury of NHBD. METHODS Male Wistar rats were used as donors and meloxicam (5 mg/kg body weight) was administered into the preservation solution. Livers were excised after 60 min of WI in situ, flushed and preserved for 24 h at 4°C. Reperfusion was carried out in vitro at a constant flow for 45 min. During reperfusion (5, 15, 30 and 45 min), enzyme release of alanine aminotransferase and glutamate lactate dehydrogenase were measured as well as portal venous pressure, bile production and oxygen consumption. The production of malondialdehyde was quantified and TUNEL staining was performed. Quantitative PCR analyzed COX-2 mRNA. COX-2 immunohistochemistry and TxB(2) detection completed the measurements. RESULTS Meloxicam treatment led to better functional recovery concerning liver enzyme release, vascular resistance and metabolic activity over time in all animals. Oxidative stress and apoptosis were considerably reduced. CONCLUSION Cold storage using meloxicam resulted in significantly better integrity and function of livers retrieved from NHBD. Selective COX-2 inhibition is a new therapeutic approach achieving improved preservation of grafts from NHBD.
Collapse
Affiliation(s)
- B Stoffels
- Department of Gastroenterological Surgery, Graduate School of Medicine, University of Kyoto, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Milatovic D, Montine TJ, Aschner M. Prostanoid signaling: dual role for prostaglandin E2 in neurotoxicity. Neurotoxicology 2011; 32:312-9. [PMID: 21376752 PMCID: PMC3090136 DOI: 10.1016/j.neuro.2011.02.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2010] [Revised: 02/03/2011] [Accepted: 02/21/2011] [Indexed: 10/18/2022]
Abstract
The prostanoids, a naturally occurring subclass of eicosanoids, are lipid mediators generated through oxidative pathways from arachidonic acid. These cyclooxygenase metabolites, consisting of the prostaglandins (PG), prostacyclin and tromboxane, are released in response to a variety of physiological and pathological stimuli in almost all organs, including the brain. They are produced by various cell types and act upon targeted cells via specific G protein-coupled receptors. The existence of multiple receptors, cross-reactivity and coupling to different signal transduction pathways for each prostanoid, collectively establish their diverse effects. Notably, these effects can occur in functionally opposing directions within the same cell or organ. Prostaglandin E(2) (PGE(2)) is the most versatile prostanoid because of its receptors, E Prostanoid (EP) receptor subtypes 1 through 4, its biological heterogeneity and its differential expression on neuronal and glial cells throughout the central nervous system. Since PGE(2) plays an important role in processes associated with various neurological diseases, this review focuses on its dual neuroprotective and neurotoxic role in EP receptor subtype signaling pathways in different models of brain injury.
Collapse
Affiliation(s)
- Dejan Milatovic
- Department of Pediatrics, Division of Clinical Pharmacology and Toxicology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | | | |
Collapse
|
29
|
Hsieh WT, Tsai CT, Wu JB, Hsiao HB, Yang LC, Lin WC. Kinsenoside, a high yielding constituent from Anoectochilus formosanus, inhibits carbon tetrachloride induced Kupffer cells mediated liver damage. JOURNAL OF ETHNOPHARMACOLOGY 2011; 135:440-449. [PMID: 21470577 DOI: 10.1016/j.jep.2011.03.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 03/11/2011] [Accepted: 03/18/2011] [Indexed: 05/30/2023]
Abstract
AIM In the present study, we have evaluated the hepatoprotective ability of kinsenoside, a major component of Anoectochilus formosanus, in vitro and in vivo. MATERIALS AND METHODS The inhibitory action of kinsenoside on lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells and Kupffer cells were investigated. Mice hepatic injury was produced by CCl(4) twice a week for 3 weeks. Mice in the three CCl(4) group were treated daily with water and kinsenoside throughout the experimental period. RESULTS In LPS-stimulated macrophage RAW 264.7 cells and Kupffer cells, kinsenoside inhibited nitric oxide (NO) production and also blocked LPS-induced inducible NO synthase expression. Furthermore, kinsenoside inhibited the NF-κB activation induced by LPS, and this is associated with the abrogation of IκBα degradation, with subsequent decreases in nuclear p65 and p50 protein levels. Moreover, the phosphorylations of p38, ERK and JNK in LPS-stimulated RAW 264.7 cells were suppressed by kinsenoside. In the in vivo study, kinsenoside significantly protected the liver from injury, by reducing the activities of plasma aminotransferase, and by improving the histological architecture of the liver. kinsenoside inhibited Kupffer cell activation by reducing the CD 14 mRNA and protein expressions. CONCLUSION These results indicate that kinsenoside alleviates CCl(4)-induced liver injury, and this protection is probably due to the suppression of Kupffer cell activation.
Collapse
Affiliation(s)
- Wen-Tsong Hsieh
- School of Medicine and Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
30
|
Hsieh WT, Liu YT, Lin WC. Anti-inflammatory properties of Ajuga bracteosa in vivo and in vitro study and their effects on mouse model of liver fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2011; 135:116-125. [PMID: 21382463 DOI: 10.1016/j.jep.2011.02.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 01/26/2011] [Accepted: 02/27/2011] [Indexed: 05/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The entire plant of Ajuga bracteosa Wall has been used to treat various inflammatory disorders, including hepatitis, in Taiwan. AIM This study evaluated the hepatoprotective ability of Ajuga bracteosa extract (ABE). MATERIALS AND METHODS We investigated the inhibitory action of a chloroform fraction of ABE (ABCE) on lipopolysaccharide (LPS)-stimulated RAW264.7 cells and Kupffer cells. Hepatic fibrosis was induced in mice through the administration of CCl(4) twice a week for 8 weeks. Mice in three CCl(4) groups were treated daily with water and ABE throughout the duration of the experiment. RESULTS In LPS-stimulated RAW264.7 cells and Kupffer cells, ABCE inhibited the production of NO and/or TNF-α and also blocked the LPS-induced expression of NO synthase. ABCE inhibited the activation of NF-κB induced by LPS, associated with the abrogation of IκBα degradation, with a subsequent decrease in nuclear p65 and p50 protein levels. The phosphorylation of MAPKs in LPS-stimulated RAW264.7 cells was also suppressed using ABCE. In the in vivo study, ABE protected the liver from injury by reducing the activity of plasma aminotransferase, and by improving the histological architecture of the liver. RT-PCR analysis showed that ABE inhibited the hepatic mRNA expression of LPS binding protein, CD14, TNF-α, collagen(α1)(I), and α-smooth actin. CONCLUSION These results indicate that ABE alleviated CCl(4)-induced liver fibrosis, and that this protection is probably due to the suppression of macrophage activation.
Collapse
Affiliation(s)
- Wen-Tsong Hsieh
- Department of Pharmacology, School of Medicine, Graduate Institute of Basic Medical Science and Tsuzuki Institute for Traditional Medicine, China Medical University, 91 Hsueh Shih Road, Taichung, Taiwan
| | | | | |
Collapse
|
31
|
Filipovich-Rimon T, Fleisher-Berkovich S. Glial response to lipopolysaccharide: possible role of endothelins. Peptides 2010; 31:2269-75. [PMID: 20863865 DOI: 10.1016/j.peptides.2010.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 09/08/2010] [Accepted: 09/10/2010] [Indexed: 11/30/2022]
Abstract
Glial inflammation plays a major role in the development of neurodegenerative diseases. Although endothelins (ETs) are known as modulators of inflammation in the periphery, little is known about their possible role in brain inflammation. Previously, we demonstrated that all three endothelins (ET-1, ET-2 and ET-3) enhanced unstimulated synthesis of the glial pro-inflammatory mediators, prostaglandin E₂ (PGE₂) and nitric oxide (NO). In the present study, glial cells were stimulated in an in vitro model of inflammation by incubation with the bacterial endotoxin lipopolysaccharide (LPS). Indeed, the present study shows that ETs regulate basal and LPS-induced glial inflammation in an opposite fashion. Here we demonstrate that ETs significantly inhibited the LPS-induced glial synthesis of PGE₂ and NO, and each of the selective antagonists for ETA and ETB receptors (BQ123 and BQ788 respectively), significantly inhibited the ETs effects in LPS-treated cells. Similar results were observed when expression of key enzymes namely, cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) in PG and NO synthesis respectively, was measured. ET-1 significantly enhanced the expression of both COX-2 and iNOS. Whereas, it inhibited the LPS-induced expression of both enzymes. These observations suggest a novel neuro-immune feedback pathway through which inflammatory mediators' synthesis is initially enhanced by ETs and are eventually blocked by the same neuropeptide when excessive production of inflammatory mediators occurs following an inflammatory insult.
Collapse
Affiliation(s)
- Talia Filipovich-Rimon
- Department of Clinical Pharmacology, Ben-Gurion University, P.O.B. 653, Beer-Sheva 84105, Israel
| | | |
Collapse
|
32
|
Herbein G, Gras G, Khan KA, Abbas W. Macrophage signaling in HIV-1 infection. Retrovirology 2010; 7:34. [PMID: 20380698 PMCID: PMC2865443 DOI: 10.1186/1742-4690-7-34] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Accepted: 04/09/2010] [Indexed: 02/07/2023] Open
Abstract
The human immunodeficiency virus-1 (HIV-1) is a member of the lentivirus genus. The virus does not rely exclusively on the host cell machinery, but also on viral proteins that act as molecular switches during the viral life cycle which play significant functions in viral pathogenesis, notably by modulating cell signaling. The role of HIV-1 proteins (Nef, Tat, Vpr, and gp120) in modulating macrophage signaling has been recently unveiled. Accessory, regulatory, and structural HIV-1 proteins interact with signaling pathways in infected macrophages. In addition, exogenous Nef, Tat, Vpr, and gp120 proteins have been detected in the serum of HIV-1 infected patients. Possibly, these proteins are released by infected/apoptotic cells. Exogenous accessory regulatory HIV-1 proteins are able to enter macrophages and modulate cellular machineries including those that affect viral transcription. Furthermore HIV-1 proteins, e.g., gp120, may exert their effects by interacting with cell surface membrane receptors, especially chemokine co-receptors. By activating the signaling pathways such as NF-kappaB, MAP kinase (MAPK) and JAK/STAT, HIV-1 proteins promote viral replication by stimulating transcription from the long terminal repeat (LTR) in infected macrophages; they are also involved in macrophage-mediated bystander T cell apoptosis. The role of HIV-1 proteins in the modulation of macrophage signaling will be discussed in regard to the formation of viral reservoirs and macrophage-mediated T cell apoptosis during HIV-1 infection.
Collapse
Affiliation(s)
- Georges Herbein
- Department of Virology, UPRES 4266 Pathogens and Inflammation, IFR 133 INSERM, University of Franche-Comté, CHU Besançon, F-25030 Besançon, France.
| | | | | | | |
Collapse
|
33
|
Szaingurten-Solodkin I, Hadad N, Levy R. Regulatory role of cytosolic phospholipase A2alpha in NADPH oxidase activity and in inducible nitric oxide synthase induction by aggregated Abeta1-42 in microglia. Glia 2010; 57:1727-40. [PMID: 19455582 DOI: 10.1002/glia.20886] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In Alzheimer's disease, extracellular deposits of amyloid beta(1-42) (Abeta(1-42)) may induce activation of microglial cells by releasing proinflammatory factors that contribute to the neurodegeneration process. Since the activation of cytosolic phospholipase A(2)alpha (cPLA(2)alpha) has been reported in inflammatory conditions, its role in primary rat microglial cell activated by aggregated Abeta(1-42) was elucidated. The results of the present study show that activation of microglia by 5 microM aggregated Abeta(1-42) (as evident by the amoeboid morphology and increased CD68 immunofluorescence reactivity) caused an immediate activation of cPLA(2)alpha, measured by its phosphorylated form and its specific activity, followed by a gradual elevation of its expression and activity during 24 h. Inhibition of cPLA(2)alpha expression and activity by the presence of 1 microM specific antisense resulted in a significant decrease in NADPH oxidase activity that releases superoxides, PGE(2) formation, iNOS expression, and NO production, indicating a major role for cPLA(2)alpha in the regulation of these inflammatory processes. NADPH oxidase activity, which is under cPLA(2)alpha regulation, was found to upregulate cPLA(2)alpha and COX-2 protein expression through the redox-sensitive NFkappaB activation as evident by its phosphorylation on Ser-536, resulting in increased PGE(2) formation. The secreted PGE(2) induced the synthesis of iNOS and the production of NO through the PKA-CREB pathway. Taken together, our results suggest that the response of cPLA(2)alpha to aggregated Abeta(1-42) is probably a key player in the oxidative stress present in AD, regulating potent oxidative agents: the production of superoxides by NADPH oxidase and NO formation by iNOS.
Collapse
Affiliation(s)
- I Szaingurten-Solodkin
- Infectious Diseases and Immunology Laboratory, Department of Clinical Biochemistry, Faculty of Health Sciences, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer Sheva, Israel
| | | | | |
Collapse
|
34
|
Tyagi E, Agrawal R, Nath C, Shukla R. Cholinergic protection via alpha7 nicotinic acetylcholine receptors and PI3K-Akt pathway in LPS-induced neuroinflammation. Neurochem Int 2009; 56:135-42. [PMID: 19781587 DOI: 10.1016/j.neuint.2009.09.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 09/16/2009] [Accepted: 09/17/2009] [Indexed: 12/14/2022]
Abstract
The present study was planned to investigate the effect of anti-cholinesterase drugs donepezil and neostigmine on neuroinflammation induced by intracerebroventricular administration of lipopolysaccharide (LPS, 50 microg) in rat. Proinflammatory cytokines (TNF-alpha and IL-1beta), expressions of iNOS and COX-2, acetylcholinesterase activity, malondialdehyde and reduced glutathione were studied in different brain regions at 24h of LPS injection. Donepezil was found to decrease the LPS-induced AChE activity and oxidative stress in all the brain regions. It also inhibited the LPS-induced proinflammatory cytokines and iNOS expression but did not affect the increased COX-2 expression whereas neostigmine treatment had no effect on LPS-induced proinflammatory cytokines. Methyllycaconitine (MLA), a alpha7 nicotinic acetylcholine receptor antagonist, significantly antagonized the donepezil mediated inhibition of LPS-induced proinflammatory cytokines, indicating that alpha7 nicotinic acetylcholine receptor subunit was playing a role in regulation of neuroinflammation. The phosphorylation of Akt, an effector of PI3K, increased with donepezil treatment. These results suggest that increased cholinergic activity in brain by donepezil prevents LPS-induced neuroinflammation via alpha7-nAChRs, followed by the PI3K-Akt pathway and this system may form the basis for the development of novel agents for reversing neuroinflammation or provide new indications for existing drugs.
Collapse
Affiliation(s)
- Ethika Tyagi
- Division of Pharmacology, Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | | | | | | |
Collapse
|
35
|
Jonakait GM, Ni L. Prostaglandins compromise basal forebrain cholinergic neuron differentiation and survival: action at EP1/3 receptors results in AIF-induced death. Brain Res 2009; 1285:30-41. [PMID: 19555672 DOI: 10.1016/j.brainres.2009.06.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 06/11/2009] [Accepted: 06/12/2009] [Indexed: 01/26/2023]
Abstract
Activated microglia produce a factor or cocktail of factors that promotes cholinergic neuronal differentiation of undifferentiated precursors in the embryonic basal forebrain (BF) in vitro. To determine whether microglial prostaglandins mediate this action, microglia were stimulated in the presence of the cyclooxygenase inhibitor ibuprofen, and microglial conditioned medium (CM) was used to culture rat BF precursors at embryonic day 15. Choline acetyltransferase (ChAT) activity served as a measure of cholinergic differentiation. While inhibition of prostaglandin biosynthesis did not affect the ability of microglial CM to promote ChAT activity, treatment of microglia with prostaglandin E2 (PGE2) inhibited it. Agonists of E prostanoid receptors EP2 (butaprost) and EP1/3 (sulprostone) mimicked PGE2, while misoprostol (E1-4) actually enhanced the action of CM. PGE2 added directly to BF cultures together with microglial CM also inhibited ChAT activity. While BF cultures expressed all four prostanoid receptors, direct addition of sulprostone but not butaprost mimicked PGE2, suggesting that PGE2 engaged EP1/3 receptors in the BF. Neither PKA inhibition by H89 nor cAMP induction by forskolin or dibutyrl-cAMP altered the action of sulprostone. Sulprostone severely compromised ChAT activity, dendrite number, axonal length and axonal branching, but caspase inhibition did not restore these. However, sulprostone resulted in increased staining intensity and nuclear translocation of apoptosis-inducing factor (AIF) suggesting caspase-independent cell death. We have found that PGE2 action at microglial EP2 receptors inhibits the microglial production of the cholinergic differentiating cocktail, while action at neuronal EP3 receptors has a deleterious effect on cholinergic neurons causing neurite retraction and cell death.
Collapse
Affiliation(s)
- G Miller Jonakait
- Federated Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | | |
Collapse
|
36
|
Andreasson K. Emerging roles of PGE2 receptors in models of neurological disease. Prostaglandins Other Lipid Mediat 2009; 91:104-12. [PMID: 19808012 DOI: 10.1016/j.prostaglandins.2009.04.003] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 03/25/2009] [Accepted: 04/02/2009] [Indexed: 01/08/2023]
Abstract
This review presents an overview of the emerging field of prostaglandin signaling in neurological diseases, focusing on PGE(2) signaling through its four E-prostanoid (EP) receptors. A large number of studies have demonstrated a neurotoxic function of the inducible cyclooxygenase COX-2 in a broad spectrum of neurological disease models in the central nervous system (CNS), from models of cerebral ischemia to models of neurodegeneration and inflammation. Since COX-1 and COX-2 catalyze the first committed step in prostaglandin synthesis, an effort is underway to identify the downstream prostaglandin signaling pathways that mediate the toxic effect of COX-2. Recent epidemiologic studies demonstrate that chronic COX-2 inhibition can produce adverse cerebrovascular and cardiovascular effects, indicating that some prostaglandin signaling pathways are beneficial. Consistent with this concept, recent studies demonstrate that in the CNS, specific prostaglandin receptor signaling pathways mediate toxic effects in brain but a larger number appear to mediate paradoxically protective effects. Further complexity is emerging, as exemplified by the PGE(2) EP2 receptor, where cerebroprotective or toxic effects of a particular prostaglandin signaling pathway can differ depending on the context of cerebral injury, for example, in excitotoxicity/hypoxia paradigms versus inflammatory-mediated secondary neurotoxicity. The divergent effects of prostaglandin receptor signaling will likely depend on distinct patterns and dynamics of receptor expression in neurons, endothelial cells, and glia and the specific ways in which these cell types participate in particular models of neurological injury.
Collapse
Affiliation(s)
- Katrin Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
37
|
Synthesis, anti-inflammatory, and antioxidant activities of 18β-glycyrrhetinic acid derivatives as chemical mediators and xanthine oxidase inhibitors. Bioorg Med Chem 2009; 17:2785-92. [DOI: 10.1016/j.bmc.2009.02.025] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2008] [Revised: 02/15/2009] [Accepted: 02/18/2009] [Indexed: 11/22/2022]
|
38
|
Filipovich T, Fleisher-Berkovich S. Regulation of glial inflammatory mediators synthesis: possible role of endothelins. Peptides 2008; 29:2250-6. [PMID: 18838093 DOI: 10.1016/j.peptides.2008.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 09/07/2008] [Accepted: 09/08/2008] [Indexed: 01/07/2023]
Abstract
Endothelins are well known as modulators of inflammation in the periphery, but little is known about their possible role in brain inflammation. Stimulation of astrocyte prostaglandin, an inflammatory mediator, synthesis was shown so far only by endothelin 3 (ET-3). By contrast, several studies showed no change or slight decrease of basal nitric oxide synthesis after treatment of astrocytes with endothelin 1 (ET-1) and ET-3. However, a significant increase in astrocytic and microglial nitric oxide synthase (NOS) was observed after exposure to ET-1 and ET-3 in a model of forebrain ischaemia. Here we demonstrate that all three endothelins (ET-1, ET-2, ET-3) significantly enhanced the synthesis of prostaglandin E(2) and nitric oxide in glial cells. Each of the selective antagonists for ETA and ETB receptors (BQ123 and BQ788 respectively), significantly inhibited endothelins-induced production of both nitric oxide and prostaglandin E(2). These results suggest a regulatory mechanism of endothelins, interacting with both endothelin receptors, on glial inflammation. Therefore, inhibition of endothelin receptors may have a therapeutic potential in pathological conditions of the brain, when an uncontrolled inflammatory response is involved.
Collapse
Affiliation(s)
- Talia Filipovich
- Department of Clinical Pharmacology, Ben-Gurion University, P.O.B 653, Beer-Sheva 84105, Israel
| | | |
Collapse
|
39
|
Isakovic A, Harhaji L, Dacevic M, Trajkovic V. Adenosine rescues glioma cells from cytokine-induced death by interfering with the signaling network involved in nitric oxide production. Eur J Pharmacol 2008; 591:106-13. [DOI: 10.1016/j.ejphar.2008.06.076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Revised: 06/12/2008] [Accepted: 06/22/2008] [Indexed: 01/22/2023]
|
40
|
Synthesis and cytotoxic, anti-inflammatory, and anti-oxidant activities of 2',5'-dialkoxylchalcones as cancer chemopreventive agents. Bioorg Med Chem 2008; 16:7270-6. [PMID: 18606546 DOI: 10.1016/j.bmc.2008.06.031] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 06/17/2008] [Accepted: 06/18/2008] [Indexed: 11/23/2022]
Abstract
In an effort to develop novel anti-tumor, or cancer chemopreventive agents, a series of 2',5'-dialkoxylchalcones were prepared by Claisen-Schmidt condensation of appropriate acetophenones with suitable aromatic aldehyde. In vitro screening revealed low micromolar activity (IC(50)) against several human cancer cell lines. Selective compound 10 induced an accumulation of A549 cells in the G(2)/M phase arrest which was well correlated with inhibitory activity against tubulin polymerization. Cytotoxic compounds 3 and 12 showed significant inhibitory effects on NO production in lipopolysaccharide (LPS)-activated RAW 264.7 macrophage-like cells while cytotoxic compound 10 revealed potent inhibitory effect on TNF-alpha formation in RAW 264.7 cells in response to LPS. Compounds 3 and 10 also showed significant inhibitory effects on xanthine oxidase. The present results suggested that compounds 3 and 10 were potential to be served as cancer chemopreventive agents.
Collapse
|
41
|
Tilleux S, Hermans E. Neuroinflammation and regulation of glial glutamate uptake in neurological disorders. J Neurosci Res 2007; 85:2059-70. [PMID: 17497670 DOI: 10.1002/jnr.21325] [Citation(s) in RCA: 284] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Oxidative stress, neuroinflammation, and excitotoxicity are frequently considered distinct but common hallmarks of several neurological disorders, including Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, and Alzheimer's disease. Although neuron degeneration and death are the ultimate consequences of these pathological processes, it is now widely accepted that alterations in the function of surrounding glial cells are key features in the progression of these diseases. In response to alteration in their local environment, microglia, commonly considered the resident immune cells of the nervous parenchyma, become activated and release a variety of soluble factors. Among these, proinflammatory cytokines and free radicals actively participate in the degenerative insults. In addition, excitotoxic neuronal damage resulting from excessive glutamate is frequently associated with impaired handling of extracellular glutamate by gliotic astrocytes. Although several research projects have focused on the biochemical mechanisms of the regulation of glial glutamate transporters, a relationship between activation of microglia and modulation of astrocytic glutamate uptake is now suggested. The aim of this review is to summarize and discuss the data showing an influence of inflammatory mediators and related free radicals on the expression and activity of glial glutamate transporters.
Collapse
Affiliation(s)
- Sébastien Tilleux
- Laboratoire de Pharmacologie Expérimentale, Faculté de Médecine, Université catholique de Louvain, Brussels, Belgium
| | | |
Collapse
|
42
|
Lin MW, Tsao LT, Chang LC, Chen YL, Huang LJ, Kuo SC, Tzeng CC, Lee MR, Wang JP. Inhibition of lipopolysaccharide-stimulated NO production by a novel synthetic compound CYL-4d in RAW 264.7 macrophages involving the blockade of MEK4/JNK/AP-1 pathway. Biochem Pharmacol 2007; 73:1796-806. [PMID: 17379190 DOI: 10.1016/j.bcp.2007.02.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 02/15/2007] [Accepted: 02/15/2007] [Indexed: 12/25/2022]
Abstract
In the present study, a novel synthetic compound 4-(2-(cyclohex-2-enylidene)hydrazinyl)quinolin-2(1H)-one (CYL-4d) was found to inhibit lipopolysaccharide (LPS)-induced nitric oxide (NO) production without affecting cell viability or enzyme activity of expressed inducible NO synthase (iNOS) in RAW 264.7 macrophages. CYL-4d exhibited parallel inhibition of LPS-induced expression of iNOS protein, iNOS mRNA and iNOS promoter activity in the same concentration range. LPS-induced activator protein-1 (AP-1) DNA binding, AP-1-dependent reporter gene activity and c-Jun nuclear translocation were all markedly inhibited by CYL-4d with similar efficacy, whereas CYL-4d produced a weak inhibition of nuclear factor-kappaB (NF-kappaB) DNA binding, NF-kappaB-dependent reporter gene activity and p65 nuclear translocation without affecting inhibitory factor-kappa B alpha (I kappa B alpha) degradation. CYL-4d had no effect on the LPS-induced phosphorylation of extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (MAPK) and its upstream activator MAPK kinase (MEK) 3, whereas it significantly attenuated the phosphorylation of c-Jun, c-Jun NH(2)-terminal kinase (JNK) and its upstream activator MEK4 in a parallel concentration-dependent manner. Other Toll-like receptors (TLRs) ligands (peptidoglycans, double-stranded RNA, and oligonucleotide containing unmethylated CpG motifs)-induced iNOS protein expression were also inhibited by CYL-4d. Furthermore, the NO production from BV-2 microglial cells as well as rat alveolar macrophages in response to LPS was diminished by CYL-4d. These results indicate that the blockade of NO production by CYL-4d in LPS-stimulated RAW 264.7 cells is attributed mainly to interference in the MEK4-JNK-AP-1 signaling pathway. CYL-4d inhibition of NO production is not restricted to TLR4 activation and immortalized macrophage-like cells.
Collapse
Affiliation(s)
- Meng-Wei Lin
- Graduate Institute of Pharmaceutical Chemistry, China Medical University, Taichung, Taiwan, ROC
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Leite DFP, Echevarria-Lima J, Calixto JB, Rumjanek VM. Multidrug resistance related protein (ABCC1) and its role on nitrite production by the murine macrophage cell line RAW 264.7. Biochem Pharmacol 2007; 73:665-74. [PMID: 17169333 DOI: 10.1016/j.bcp.2006.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 09/23/2006] [Accepted: 11/06/2006] [Indexed: 11/23/2022]
Abstract
Multidrug resistance related protein 1 (MRP1/ABCC1) is an ABC transporter protein related to the extrusion of reduced glutathione (GSH), oxidized glutathione (GSSG) and GSH-conjugates, as well as leukotriene C(4) and cyclopentane prostaglandins. Inhibition of ABCC1 activity impairs lymphocyte activation. The present work studied ABCC1 expression and activity on a murine macrophage cell line, RAW 267.4 and the effects of ABCC1 classical inhibitors, as well as GSH metabolism modulators, on LPS induced activation. Approximately, 75% of resting cells were positive for ABCC1 and the classical ABCC1 reversors (indomethacin, 0.1-2mM; probenecid, 0.1-10mM and MK571, 0.01-1mM) were able to enhance intracellular CFDA accumulation in a concentration-dependent manner, suggesting ABCC1 inhibition. After LPS (100ng/ml) activation 50% of the population was positive for ABCC1, and this protein was still active. In LPS-activated cells, ABCC1 activity was also impaired by BSO (1mM), an inhibitor of GSH synthesis. Conversely, GSH (5mM) reversed the BSO effect. ABCC1 inhibition by indomethacin, probenecid or MK571 decreased LPS induced nitrite production in a concentration-dependent manner, the same result was observed with BSO and again GSH reversed its effect. The ABCC1 reversors were also able to inhibit iNOS expression. In conclusion, LPS modulated the expression and activity of ABCC1 transporters in RAW macrophages and inhibitors of these transporters were capable of inhibiting nitrite production suggesting a role for ABCC1 transporters in the inflammatory process.
Collapse
Affiliation(s)
- Daniela F P Leite
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil
| | | | | | | |
Collapse
|
44
|
Wei BL, Teng CH, Wang JP, Won SJ, Lin CN. Synthetic 2',5'-dimethoxychalcones as G(2)/M arrest-mediated apoptosis-inducing agents and inhibitors of nitric oxide production in rat macrophages. Eur J Med Chem 2007; 42:660-8. [PMID: 17320246 DOI: 10.1016/j.ejmech.2006.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Revised: 11/28/2006] [Accepted: 12/05/2006] [Indexed: 11/30/2022]
Abstract
In an effort to develop novel antitumor or chemopreventive agents, a series of 2',5'-dimethoxychalcone derivatives were prepared by Claisen-Schmidt condensation of appropriate acetophenones with suitable aromatic aldehyde. In vitro screening revealed low micromolar activity (IC(50)) against several human cancer lines. Activity in MCF-7 cells correlated with the ability to induce G(2)/M arrest-mediated apoptosis following drug treatment by the most potent agent, 8, an observation further reinforced by fluorescence microscopy. Compounds 3, 8, and 10 showed potent inhibitory effect on NO production in lipopolysaccharide (LPS)-activated RAW 264.7 macrophage-like cells. The present results demonstrated that 3, 8, and 10 are potential anti-inflammatory and cancer chemopreventive agents.
Collapse
Affiliation(s)
- Bai-Luh Wei
- Institute of Life Science, National Taitung University, Taitung, Taiwan
| | | | | | | | | |
Collapse
|
45
|
Chew LJ, Takanohashi A, Bell M. Microglia and inflammation: impact on developmental brain injuries. ACTA ACUST UNITED AC 2006; 12:105-12. [PMID: 16807890 DOI: 10.1002/mrdd.20102] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation during the perinatal period has become a recognized risk factor for developmental brain injuries over the past decade or more. To fully understand the relationship between inflammation and brain development, a comprehensive knowledge about the immune system within the brain is essential. Microglia are resident immune cells within the central nervous system and play a critical role in the development of an inflammatory response within the brain. Microglia are critically involved with both the innate and adaptive immune system, regulating inflammation and cell damage within the brain via activation of Toll-like receptors, production of cytokines, and a myriad of other intracellular and intercellular processes. In this article, microglial physiology is reviewed along with the role of microglia in developmental brain injuries in humans and animal models. Last, microglial functions within the innate and adaptive immune system will be summarized. Understanding the processes of inflammation and microglial activation is critical for formulating effective preventative and therapeutic strategies for developmental brain injuries.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, District of Columbia 20010, USA
| | | | | |
Collapse
|
46
|
Tsai MJ, Shyue SK, Weng CF, Chung Y, Liou DY, Huang CT, Kuo HS, Lee MJ, Chang PT, Huang MC, Huang WC, Liou KD, Cheng H. Effect of enhanced prostacyclin synthesis by adenovirus-mediated transfer on lipopolysaccharide stimulation in neuron-glia cultures. Ann N Y Acad Sci 2006; 1042:338-48. [PMID: 15965079 DOI: 10.1196/annals.1338.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Prostacyclin (PGI2) is known as a short-lived, potent vasodilator and platelet anti-aggregatory eicosanoid. This work attempts to selectively augment PGI2 synthesis in neuron-glia cultures by adenoviral (Ad) gene transfer of PGI synthase (PGIS) or bicistronic cyclooxygenase 1 (COX-1)/PGIS and examines whether PGI2 confers protection against lipopolysaccharide (LPS) stimulation. Cultures released low levels of eicosanoids. Upon Ad-PGIS or Ad-COX-1/PGIS infection, cultures selectively increased prostacyclin release. Both PGIS- and COX-1/PGIS-overexpressed cultures contained fewer microglial numbers. Further, they significantly attenuated LPS-induced iNOS expression and lactate, nitric oxide, and TNF-alpha production. Taken together, enhanced prostacyclin synthesis in neuron-glial cultures reduced microglia number and suppressed LPS stimulation.
Collapse
Affiliation(s)
- May-Jywan Tsai
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Shih-pai Road, Sec. 2, Taipei, Taiwan 11217
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yang MS, Ji KA, Jeon SB, Jin BK, Kim SU, Jou I, Joe E. Interleukin-13 enhances cyclooxygenase-2 expression in activated rat brain microglia: implications for death of activated microglia. THE JOURNAL OF IMMUNOLOGY 2006; 177:1323-9. [PMID: 16818793 DOI: 10.4049/jimmunol.177.2.1323] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Brain inflammation has recently attracted widespread interest because it is a risk factor for the onset and progression of brain diseases. In this study, we report that cyclooxygenase-2 (COX-2) plays a key role in the resolution of brain inflammation by inducing the death of microglia. We previously reported that IL-13, an anti-inflammatory cytokine, induced the death of activated microglia. These results revealed that IL-13 significantly enhanced COX-2 expression and production of PGE(2) and 15-deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)) in LPS-treated microglia. Two other anti-inflammatory cytokines, IL-10 and TGF-beta, neither induced microglial death nor enhanced COX-2 expression or PGE(2) or 15d-PGJ(2) production. Therefore, we hypothesized that the effect of IL-13 on COX-2 expression may be linked to death of activated microglia. We found that COX-2 inhibitors (celecoxib and NS398) suppressed the death of microglia induced by a combination of LPS and IL-13 and that exogenous addition of PGE(2) and 15d-PGJ(2) induced microglial death. Agonists of EP2 (butaprost) and peroxisome proliferator-activated receptor gamma (ciglitazone) mimicked the effect of PGE(2) and 15d-PGJ(2), and an EP2 antagonist (AH6809) and a peroxisome proliferator-activated receptor gamma antagonist (GW9662) suppressed microglial death induced by LPS in combination with IL-13. In addition, IL-13 potentiated LPS-induced activation of JNK, and the JNK inhibitor SP600125 suppressed the enhancement of COX-2 expression and attenuated microglial death. Taken together, these results suggest that IL-13 enhanced COX-2 expression in LPS-treated microglia through the enhancement of JNK activation. Furthermore, COX-2 products, PGE(2) and 15d-PGJ(2), caused microglial death, which terminates brain inflammation.
Collapse
Affiliation(s)
- Myung-Soon Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon 442-721, Korea
| | | | | | | | | | | | | |
Collapse
|
48
|
Reese J, Anderson JD, Brown N, Roman C, Clyman RI. Inhibition of cyclooxygenase isoforms in late- but not midgestation decreases contractility of the ductus arteriosus and prevents postnatal closure in mice. Am J Physiol Regul Integr Comp Physiol 2006; 291:R1717-1723. [PMID: 16857891 PMCID: PMC2819844 DOI: 10.1152/ajpregu.00259.2006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Use of cyclooxygenase (COX) inhibitors to delay preterm birth is complicated by in utero constriction of the ductus arteriosus and delayed postnatal closure. Delayed postnatal closure has been attributed to loss of vasa vasorum flow and ductus wall ischemia resulting from constriction in utero. We used the murine ductus (which does not depend on vasa vasorum flow) to determine whether delayed postnatal closure may be because of mechanisms independent of in utero constriction. Acute inhibition of both COX isoforms constricted the fetal ductus on days 18 and 19 (term) but not earlier in gestation; COX-2 inhibition constricted the fetal ductus more than COX-1 inhibition. In contrast, mice exposed to prolonged inhibition of COX-1, COX-2, or both COX isoforms (starting on day 15, when the ductus does not respond to the inhibitors) had no contractile response to the inhibitors on days 18 or 19. Newborn mice closed their ductus within 4 h of birth. Prolonged COX inhibition on days 11-14 of gestation had no effect on newborn ductal closure; however, prolonged COX inhibition on days 15-19 resulted in delayed ductus closure despite exposure to 80% oxygen after birth. Similarly, targeted deletion of COX-2 alone, or COX-1/COX-2 together, impaired postnatal ductus closure. Nitric oxide inhibition did not prevent the delay in ductus closure. These data show that impaired postnatal ductus closure is not the result of in utero ductus constriction or upregulation of nitric oxide synthesis. They are consistent with a novel role for prostaglandins in ductus arteriosus contractile development.
Collapse
Affiliation(s)
- Jeff Reese
- Department of Pediatrics, Vanderbilt University Medical Center, 1125 MRB IV Bldg., 2215 B Garland Avenue, Nashville, TN 37232-0656, USA.
| | | | | | | | | |
Collapse
|
49
|
Abstract
Elevated levels of NO produced within the central nervous system (CNS) are associated with the pathogenesis of neuroinflammatory and neurodegenerative human diseases such as multiple sclerosis, HIV dementia, brain ischemia, trauma, Parkinson's disease, and Alzheimer's disease. Resident glial cells in the CNS (astroglia and microglia) express inducible nitric oxide synthase (iNOS) and produce high levels of NO in response to a wide variety of proinflammatory and degenerative stimuli. Although pathways resulting in the expression of iNOS may vary in two different glial cells of different species, the intracellular signaling events required for the expression of iNOS in these cells are slowly becoming clear. Various signaling cascades converge to activate several transcription factors that control the transcription of iNOS in glial cells. The present review summarizes different results and discusses current understandings about signaling mechanisms for the induction of iNOS expression in activated glial cells. A complete understanding of the regulation of iNOS expression in glial cells is expected to identify novel targets for therapeutic intervention in NO-mediated neurological disorders.
Collapse
Affiliation(s)
- Ramendra N Saha
- Department of Oral Biology, Section of Neuroscience, University of Nebraska Medical Center, Lincoln, 68583, USA
| | | |
Collapse
|
50
|
Montine TJ. Symposium. Brain Pathol 2006. [DOI: 10.1111/j.1750-3639.2005.tb00508.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|