1
|
Hong Y, Liu Q, Peng M, Bai M, Li J, Sun R, Guo H, Xu P, Xie Y, Li Y, Liu L, Du J, Liu X, Yang B, Xu G. High-frequency repetitive transcranial magnetic stimulation improves functional recovery by inhibiting neurotoxic polarization of astrocytes in ischemic rats. J Neuroinflammation 2020; 17:150. [PMID: 32375835 PMCID: PMC7203826 DOI: 10.1186/s12974-020-01747-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/13/2020] [Indexed: 12/15/2022] Open
Abstract
Background Repetitive transcranial magnetic stimulation (rTMS) is a noninvasive treatment for ischemic stroke. Astrocytes regulation has been suggested as one mechanism for rTMS effectiveness. But how rTMS regulates astrocytes remains largely undetermined. There were neurotoxic and neuroprotective phenotypes of astrocytes (also denoted as classically and alternatively activated astrocytes or A1 and A2 astrocytes) pertaining to pro- or anti-inflammatory gene expression. Pro-inflammatory or neurotoxic polarized astrocytes were induced during cerebral ischemic stroke. The present study aimed to investigate the effects of rTMS on astrocytic polarization during cerebral ischemic/reperfusion injury. Methods Three rTMS protocols were applied to primary astrocytes under normal and oxygen-glucose deprivation/reoxygenation (OGD/R) conditions. Cell survival, proliferation, and phenotypic changes were assessed after 2-day treatment. Astrocytes culture medium (ACM) from control, OGD/R, and OGD/R + rTMS groups were mixed with neuronal medium to culture neurons for 48 h and 7 days, in order to explore the influence on neuronal survival and synaptic plasticity. In vivo, rats were subjected to middle cerebral artery occlusion (MCAO), and received posterior orbital intravenous injection of ACM collected from different groups at reperfusion, and at 3 days post reperfusion. The apoptosis in the ischemic penumbra, infarct volumes, and the modified Neurological Severity Score (mNSS) were evaluated at 1 week after reperfusion, and cognitive functions were evaluated using the Morris Water Maze (MWM) tests. Finally, the 10 Hz rTMS was directly applied to MCAO rats to verify the rTMS effects on astrocytic polarization. Results Among these three frequencies, the 10 Hz protocol exerted the greatest potential to modulate astrocytic polarization after OGD/R injury. Classically activated and A1 markers were significantly inhibited by rTMS treatment. In OGD/R model, the concentration of pro-inflammatory mediator TNF-α decreased from 57.7 to 23.0 рg/mL, while anti-inflammatory mediator IL-10 increased from 99.0 to 555.1 рg/mL in the ACM after rTMS treatment. The ACM collected from rTMS-treated astrocytes significantly alleviated neuronal apoptosis induced by OGD/R injury, and promoted neuronal plasticity. In MCAO rat model, the ACM collected from rTMS treatment decreased neuronal apoptosis and infarct volumes, and improved cognitive functions. The neurotoxic astrocytes were simultaneously inhibited after rTMS treatment. Conclusion Inhibition of neurotoxic astrocytic polarization is a potential mechanism for the effectiveness of high-frequency rTMS in cerebral ischemic stroke.
Collapse
Affiliation(s)
- Ye Hong
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Qian Liu
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Mengna Peng
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Maosheng Bai
- Department of Orthopedics, Nanjing Tongren Hospital, Nanjing, 210002, Jiangsu, China.,Department of Orthopedics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210002, Jiangsu, China
| | - Juanji Li
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Rui Sun
- Department of Neurology, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, Jiangsu, China.,Department of Neurology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, 210000, China
| | - Hongquan Guo
- Department of Neurology, Jinling Hospital, Southern Medical University, Nanjing, 210002, Jiangsu, China
| | - Pengfei Xu
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China.,Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Yi Xie
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Yunzi Li
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Ling Liu
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Juan Du
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Xinfeng Liu
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Bin Yang
- Department of Ultrasonography, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| | - Gelin Xu
- Department of Neurology, Jingling Hospital, Nanjing University School of Medicine, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
2
|
Yao M, Gao W, Tao H, Yang J, Huang T. The regulation effects of danofloxacin on pig immune stress induced by LPS. Res Vet Sci 2016; 110:65-71. [PMID: 28159239 DOI: 10.1016/j.rvsc.2016.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 11/03/2016] [Accepted: 11/05/2016] [Indexed: 01/01/2023]
Abstract
Danofloxacin (DAN) is one of the Fluoroquinolone drugs (FQs) that has been widely used in the control and prevention of bacterial infectious disease in animal production. Most of the FQs have an obvious protective effect against lipopolysaccharide (LPS) induced Immune stress. However, the effect of DAN on the host immune system of animals remains unknown. In this study, a fever piglet model was built and a systematic survey of the response of inflammatory genes and mediators to DAN treatment and LPS induction was performed in the pig. The results indicated that the IL-1β, TNF-α, IL-6, NO (nitric oxide), and PGE2 (prostaglandin E2) levels were significantly suppressed in plasma and porcine alveolar macrophage 3D4/2 cells compared with the LPS treatment group. Interestingly, the IL-10 production was further stimulated by DAN treatment in the LPS challenged piglet. Our results suggested that DAN could relieve acute inflammation through inhibiting the activation of inflammatory genes introduced by LPS.
Collapse
Affiliation(s)
- Min Yao
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China
| | - Weihua Gao
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China
| | - Hengxun Tao
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China
| | - Jun Yang
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China
| | - Tinghua Huang
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China.
| |
Collapse
|
3
|
Bacterial endotoxin modifies heat shock factor-1 activity in RAW 264.7 cells: implications for TNF-α regulation during exposure to febrile range temperatures. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519040100030401] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recent studies have identified heat shock factor (HSF)-1, the predominant heat/stress-stimulated transcriptional activator of heat shock protein genes as a repressor of certain cytokine genes, including TNF-α and IL-1β. We previously showed that exposing macrophages to febrile-range temperature (FRT; 39.5°C) activates HSF-1 to a DNA binding form that does not activate heat shock protein gene transcription, but apparently represses TNF-α and IL-1β transcription. Prewarming macrophages to 39.5°C for 30 min prior to stimulation with bacterial lipopolysaccharide (LPS) does not change the induction of TNF-α transcription, but markedly reduces its duration. This raised the question of how TNF-α transcription could occur at all in the presence of activated HSF-1. We used RAW 264.7 cells to test the hypothesis that macrophage activation triggers a transient reversal of HSF-1-mediated repression, thereby allowing induction of TNF-α transcription. Electrophoretic mobility shift assays revealed that LPS triggers a transient inactivation of HSF-1 that temporally correlates with TNF-α transcription and was associated with a transient increase in HSF-1 molecular weight, a decrease in its pI, and appearance of HSF-1 phosphorylating activity. The serine/threonine phosphatase inhibitor, calyculin A, blocked the inhibitory affect of FRT on LPS-induced TNF-α generation and prevented the re-activation of HSF-1. We propose that LPS stimulation of FRT-exposed macrophages stimulates a sequential phosphorylation and dephosphorylation of HSF-1, causing a cycle of inactivation and re-activation of HSF-1 repressor activity that allows a temporally-limited period of gene transcription.
Collapse
|
4
|
|
5
|
Gottschall PE, Tatsuno I, Arimura A. Increased sensitivity of glioblastoma cells to interleukin 1 after long-term incubation with dexamethasone. Mol Cell Neurosci 2012; 3:49-55. [PMID: 19912845 DOI: 10.1016/1044-7431(92)90008-p] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/1991] [Indexed: 11/26/2022] Open
Abstract
Cytokines may act as chemical messengers in the central nervous system and affect trophic, immune, and neuroendocrine functions. However, little is known about the regulation of cytokine production in nervous system tissue, although it has been demonstrated recently that glucocorticoids augment interleukin 1 (IL 1) receptor expression in the glioblastoma cell line, U87MG. Here, the effects of glucocorticoids on IL 1-induced interleukin 6 (IL 6) production are determined in the same cell line. During short-term incubations of U87MG cells (6 h) in the presence of IL 1beta and dexamethasone (DEX), DEX inhibited IL 1beta-stimulated IL 6 production over the entire range of the dose-response curve. However, when cells were preincubated in the presence of DEX for 15 h and then challenged with IL 1beta or IL 1beta and DEX, there was a left-shift in the IL 1,B dose-response curve, suggesting an increased sensitivity of the cells to respond to IL 1 and produce IL 6. In fact, the ED(50) for IL 1beta-stimulated IL 6 production was about 1.3 pM in cells not preincubated with DEX, but was reduced to 0.25 pM in cells that were preincubated with DEX. However, maximum IL 6 production at high doses of IL 1beta was inhibited in cells cultured in the presence of DEX during the IL 1 challenge. Thus, the results suggest the possibility that when glucocorticoids are elevated for extended periods, and concentrations of IL 1 are low because of steroid suppression of IL 1 production, actions of IL 1 may be maintained or even augmented due to up-regulated IL 1 receptor expression in particular cell types. This would allow for glucocorticoid-targeted actions of IL-1 to be maintained in particular cell types that up-regulate IL 1 receptors in response to glucocorticoid, whereas in those cell types that do not up-regulate IL 1 receptor number, IL 1 actions are suppressed.
Collapse
Affiliation(s)
- P E Gottschall
- U.S.-Japan Biomedical Research Laboratories, Tulane University Hebert Center, Belle Chasse, Louisiana 70037, USA; Departments of Medicine, Anatomy and hysiology, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | | | | |
Collapse
|
6
|
Tada M, de Tribolet N. Immunobiology of malignant gliomas. J Clin Neurosci 2012; 3:102-13. [PMID: 18638850 DOI: 10.1016/s0967-5868(96)90001-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/1995] [Accepted: 05/25/1995] [Indexed: 12/19/2022]
Abstract
The immune system of patients with malignant gliomas is profoundly suppressed. The suppression involves both the cellular and humoral immunity and it is mainly attributable to selective depletion and malfunction of helper T cells. Malignant glioma cells express potent immunosuppressive factors such as transforming growth factor-beta(2), inteleukin-10 and prostaglandin E(2). Malignant glioma cells also produce chemoattractants and immunostimulatory cytokines which may activate the immune cells. However, the production of these stimulatory cytokines is not self-destructive to glioma cells because of the immunosuppression. Rather, the tumour cells use them to gain a growth advantage. Indeed the cytokines may act as a growth stimulator of the tumour cells themselves (autocrine mechanism), they may act as angiogenic factors to endothelial cells (paracrine mechanism) or induce the attracted immune cells to secrete angiogenic factors. Some cytokines produced by malignant glioma cells are known to be growth inhibitory to normal astrocytes. Recent studies on tumour suppressor genes suggest a close link between the aberrant genes and the immunobiologic features of malignant glioma cells.
Collapse
Affiliation(s)
- M Tada
- Department of Neurosurgery, University Hospital, Lausanne, Switzerland
| | | |
Collapse
|
7
|
Dexamethasone therapy for bacterial meningitis: Better never than late? Can J Infect Dis 2012; 5:210-5. [PMID: 22346503 DOI: 10.1155/1994/257198] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/1994] [Accepted: 05/25/1994] [Indexed: 11/18/2022] Open
Abstract
A multicentre randomized controlled trial was conducted in children with bacterial meningitis using dexamethasone or placebo for four days within 24 h of starting antibiotics. Primary outcomes were hearing loss and neurological abnormalities at 12 months after meningitis. The dexamethasone (n=50) and placebo (n=51) groups were similar in age, severity of illness and etiological agent. Hearing loss occurred in 10% and 11% of the dexamethasone and placebo groups and neurological deficits occurred in 20% and 18% of patients, respectively. Duodenal perforation occurred in one dexamethasone-treated child. In conclusion, there was no significant benefit in those receiving dexamethasone. The lack of benefit may have been due to the delay in administration of dexamethasone (median delay of 11 h after antibiotics). Therefore, if dexamethasone is used for meningitis it should be given immediately with the antibiotic.
Collapse
|
8
|
Miljković D, Timotijević G, Stojković MM. Astrocytes in the tempest of multiple sclerosis. FEBS Lett 2011; 585:3781-8. [DOI: 10.1016/j.febslet.2011.03.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Revised: 03/21/2011] [Accepted: 03/23/2011] [Indexed: 12/11/2022]
|
9
|
Ryan KJ, Griffin ÉW, Connor TJ. Complementary anti-inflammatory actions of the β2-adrenoceptor agonist clenbuterol and the glucocorticoid dexamethasone in rat brain. J Neuroimmunol 2011; 232:209-16. [DOI: 10.1016/j.jneuroim.2010.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Revised: 09/14/2010] [Accepted: 10/04/2010] [Indexed: 10/18/2022]
|
10
|
Sirsat S, Burkholder K, Muthaiyan A, Dowd S, Bhunia A, Ricke S. Effect of sublethal heat stress on Salmonella Typhimurium virulence. J Appl Microbiol 2011; 110:813-22. [DOI: 10.1111/j.1365-2672.2011.04941.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
11
|
Abstract
Heatstroke is the most severe form of heat-related disorders that include mild heat intolerance, heat exhaustion and heat stress. The incidence of heat-related disorders is increasing due to several factors that include climate change, co-morbidities and drug usage. Patients with heatstroke present with a core body temperature above 40°C, multiorgan dysfunction and central nervous system disorder. The pathogenesis of heatstroke is not fully understood; however, heat-shock proteins, inflammatory cytokines and their modulators have been implicated. The clinical biochemistry laboratory plays an important role in the management of patients with heatstroke. Biochemical findings in patients with heatstroke include elevated urea, creatinine, cardiac and skeletal muscle enzymes, myoglobin and troponin. There is also biochemical evidence of metabolic acidosis, respiratory alkalosis, hepatic injury with elevated enzyme levels as well as abnormal hematological and coagulation indices. This review article aims at increasing awareness of the biochemical changes seen in patients with heatstroke and their possible role in prognosis and in elucidating the pathogenesis of heatstroke.
Collapse
Affiliation(s)
- Ibrahim A Hashim
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9073, USA
| |
Collapse
|
12
|
Levin TC, Wickliffe KE, Leppla SH, Moayeri M. Heat shock inhibits caspase-1 activity while also preventing its inflammasome-mediated activation by anthrax lethal toxin. Cell Microbiol 2008; 10:2434-46. [PMID: 18671821 DOI: 10.1111/j.1462-5822.2008.01220.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Anthrax lethal toxin (LT) rapidly kills macrophages from certain mouse strains in a mechanism dependent on the breakdown of unknown protein(s) by the proteasome, formation of the Nalp1b (NLRP1b) inflammasome and subsequent activation of caspase-1. We report that heat-shocking LT-sensitive macrophages rapidly protects them against cytolysis by inhibiting caspase-1 activation without upstream effects on LT endocytosis or cleavage of the toxin's known cytosolic substrates (mitogen-activated protein kinases). Heat shock protection against LT occurred through a mechanism independent of de novo protein synthesis, HSP90 activity, p38 activation or proteasome inhibition and was downstream of mitogen-activated protein kinase cleavage and degradation of an unknown substrate by the proteasome. The heat shock inhibition of LT-mediated caspase-1 activation was not specific to the Nalp1b (NLRP1b) inflammasome, as heat shock also inhibited Nalp3 (NLRP3) inflammasome-mediated caspase-1 activation in macrophages. We found that heat shock induced pro-caspase-1 association with a large cellular complex that could prevent its activation. Additionally, while heat-shocking recombinant caspase-1 did not affect its activity in vitro, lysates from heat-shocked cells completely inhibited recombinant active caspase-1 activity. Our results suggest that heat shock inhibition of active caspase-1 can occur independently of an inflammasome platform, through a titratable factor present within intact, functioning heat-shocked cells.
Collapse
Affiliation(s)
- Tera C Levin
- Bacterial Toxins and Therapeutics Section, Laboratory of Bacterial Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
13
|
Liu X, Han Q, Sun R, Li Z. Dexamethasone regulation of matrix metalloproteinase expression in experimental pneumococcal meningitis. Brain Res 2008; 1207:237-43. [DOI: 10.1016/j.brainres.2008.01.106] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 01/17/2008] [Accepted: 01/21/2008] [Indexed: 10/22/2022]
|
14
|
Abstract
Heat stroke is a life-threatening illness that affects all segments of society, including the young, aged, sick, and healthy. The recent high death toll in France (Dorozynski, 2003) and the death of high-profile athletes has increased public awareness of the adverse effects of heat injury. However, the etiology of the long-term consequences of this syndrome remains poorly understood such that preventive/treatment strategies are needed to mitigate its debilitating effects. Cytokines are important modulators of the acute phase response (APR) to stress, infection, and inflammation. Current data implicating cytokines in heat stroke responses are mainly from correlation studies showing elevated plasma levels in heat stroke patients and experimental animal models. Correlation data fall far short of revealing the mechanisms of cytokine actions such that additional research to determine the role of these endogenous substances in the heat stroke syndrome is required. Furthermore, cytokine determinations have occurred mainly at end-stage heat stroke, such that the role of these substances in progression and long-term recovery is poorly understood. Despite several studies implicating cytokines in heat stroke pathophysiology, few studies have examined the protective effect(s) of cytokine antagonism on the morbidity and mortality of heat stroke. This is particularly surprising since heat stroke responses resemble those observed in the endotoxemic syndrome, for which a role for endogenous cytokines has been strongly implicated. The implication of cytokines as mediators of endotoxemia and the presence of circulating endotoxin in heat stroke patients suggests that much knowledge can be gained from applying our current understanding of endotoxemic pathophysiology to the study of heat stroke. Heat shock proteins (HSPs) are highly conserved proteins that function as molecular chaperones for denatured proteins and reciprocally modulate cytokine production in response to stressful stimuli. HSPs have been shown repeatedly to confer protection in heat stroke and injury models. Interactions between HSPs and cytokines have received considerable attention in the literature within the last decade such that a complex pathway of interactions between cytokines, HSPs, and endotoxin is thought to be occurring in vivo in the orchestration of the APR to heat injury. These data suggest that much of the pathophysiologic changes observed with heat stroke are not a consequence of heat exposure, per se, but are representative of interactions among these three (and presumably additional) components of the innate immune response. This chapter will provide an overview of current knowledge regarding cytokine, HSP, and endotoxin interactions in heat stroke pathophysiology. Insight is provided into the potential therapeutic benefit of cytokine neutralization for mitigation of heat stroke morbidity and mortality based on our current understanding of their role in this syndrome.
Collapse
Affiliation(s)
- Lisa R Leon
- US Army Research Institute of Environmental Medicine, Thermal and Mountain Medicine Division, Natick, MA 01760-5007, USA.
| |
Collapse
|
15
|
Hashioka S, Klegeris A, Schwab C, McGeer PL. Interferon-gamma-dependent cytotoxic activation of human astrocytes and astrocytoma cells. Neurobiol Aging 2008; 30:1924-35. [PMID: 18375019 DOI: 10.1016/j.neurobiolaging.2008.02.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2008] [Revised: 02/21/2008] [Accepted: 02/26/2008] [Indexed: 10/22/2022]
Abstract
Astrocytes and microglia become activated in a broad spectrum of inflammatory neurodegenerative diseases. Activated microglia are widely believed to be the principal source of inflammation-induced neuronal degeneration in these disorders. To investigate the neurotoxic potential of human astrocytes, we exposed them and human astrocytic U-373 MG cells to a variety of inflammatory stimulants. We then assessed the effects of their supernatants on human SH-SY5 cells. When astrocytes and U-373 MG cells were stimulated with interferon (IFN)-gamma (150U/ml), their supernatants significantly reduced SH-SY5Y cell viability. Other powerful inflammatory stimulants such as lipopolysaccharide (0.5mug/ml), tumor necrosis factor-alpha (10ng/ml) and interleukin-1beta (10ng/ml), alone or in combination, were without effect. These combinations were also unable to enhance the IFN-gamma effect. The induced cytotoxicities were reversed by JAK inhibitor I, a potent and specific inhibitor of JAKs. This result indicates that the neurotoxic effect was proceeding through the IFN-gamma receptor (IFNGR)-JAK-STAT intracellular pathway. To establish that the IFNGR is expressed on both cultured astrocytes and U-373 MG cells, we performed RT-PCR on total RNA extracts to identify a specific IFNGR product. We showed the protein product on these cultured cells by immunocytochemistry using an antibody to IFNGR. Finally, using human postmortem material, we showed sharp upregulation of the IFNGR on activated astrocytes in affected areas in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. These findings suggest that activated astrocytes may become neurotoxic when stimulated by IFN-gamma and may therefore exacerbate the pathology in a spectrum of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sadayuki Hashioka
- Kinsmen Laboratory of Neurological Research, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1W5 Canada
| | | | | | | |
Collapse
|
16
|
Kim H, Moon C, Ahn M, Lee Y, Kim S, Matsumoto Y, Koh CS, Kim MD, Shin T. Increased phosphorylation of cyclic AMP response element-binding protein in the spinal cord of Lewis rats with experimental autoimmune encephalomyelitis. Brain Res 2007; 1162:113-20. [PMID: 17617386 DOI: 10.1016/j.brainres.2007.05.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Revised: 05/29/2007] [Accepted: 05/31/2007] [Indexed: 12/29/2022]
Abstract
To investigate whether the phosphorylation of cyclic AMP response element-binding protein (CREB) is implicated in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), the change in the level of CREB phosphorylation was analyzed in the spinal cord of Lewis rats with EAE. Western blot analysis showed that the phosphorylation of CREB in the spinal cord of rats increased significantly at the peak stage of EAE compared with the controls (p<0.05) and declined significantly in the recovery stage (p<0.05). Immunohistochemistry showed that the phosphorylated form of CREB (p-CREB) was constitutively immunostained in few astrocytes and dorsal horn neurons in the spinal cord of normal rats. In the EAE-affected spinal cord, p-CREB was mainly found in ED1-positive macrophages at the peak stage of EAE, and the number of p-CREB-immunopositive astrocytes was markedly increased in the spinal cord with EAE compared with the controls. Moreover, p-CREB immunoreactivity of sensory neurons, which are closely associated with neuropathic pain, was significantly increased in the dorsal horns at the peak stage of EAE. Based on these results, we suggest that the increased phosphorylation of CREB in EAE lesions was mainly attributable to the infiltration of inflammatory cells and astrogliosis, possibly activating gene transcription, and that its increase in the sensory neurons in the dorsal horns is involved in the generation of neuropathic pain in the rat EAE model.
Collapse
MESH Headings
- Activating Transcription Factor 1/metabolism
- Animals
- Cyclic AMP Response Element-Binding Protein/metabolism
- Ectodysplasins/metabolism
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Fetal Blood/metabolism
- Gene Expression Regulation/physiology
- Glial Fibrillary Acidic Protein/metabolism
- Male
- Myelin Basic Protein
- Phosphorylation
- Rats
- Rats, Inbred Lew
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
Collapse
Affiliation(s)
- Heechul Kim
- Department of Veterinary Medicine and Applied Radiological Science Research Institute, Cheju National University, Jeju 690-756, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Mori K, Kaneko YS, Nakashima A, Nagatsu I, Takahashi H, Ota A. Peripheral lipopolysaccharide induces apoptosis in the murine olfactory bulb. Brain Res 2005; 1039:116-29. [PMID: 15781053 DOI: 10.1016/j.brainres.2005.01.078] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2004] [Revised: 01/14/2005] [Accepted: 01/19/2005] [Indexed: 12/24/2022]
Abstract
The olfactory bulb (OB) is one of the few structures in the adult mammalian CNS that contains a continuous supply of newly generated neurons in the subventricular zone. Therefore, the balance between the supply of new cells and apoptosis in the OB might determine olfactory function. Lipopolysaccharide-induced tumor necrosis factor (TNF)-alpha triggers the apoptotic cascade mediated by the TNF/TNF receptor (TNFR) pathway. The present study therefore examines the effect of the propagated innate immune reaction triggered by peripheral lipopolysaccharide on the OB of C3H/HeN mice. Within 2 h of an intraperitoneal injection of lipopolysaccharide, mRNA expression levels of the genes encoding IkappaB, TNF-alpha, and TNFR type 1 in the mouse OB were significantly enhanced. Double immunofluorescence microscopy confirmed that almost all TNF-alpha-immunopositive cells in the OB of the TNF-injected mice were located in the subependymal zone and that they overlapped cells immunostained with antibody against glial fibrillary acidic protein, but not with the antibody against F4/80, an antigenic marker of microglia. The number of TUNEL-positive cells identified exclusively in the granule cell layer was significantly increased in mice injected with lipopolysaccharide and sacrificed at 24 h thereafter. These results suggest that peripheral lipopolysaccharide causes disequilibrium between the supply and disappearance of the cells in the OB, which might lead to olfactory dysfunction.
Collapse
Affiliation(s)
- Keiji Mori
- Department of Physiology, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
| | | | | | | | | | | |
Collapse
|
18
|
Waterman WR, Xu LL, Tetradis S, Motyckova G, Tsukada J, Saito K, Webb AC, Robinson DR, Auron PE. Glucocorticoid inhibits the human pro-interleukin lbeta gene (ILIB) by decreasing DNA binding of transactivators to the signal-responsive enhancer. Mol Immunol 2005; 43:773-82. [PMID: 16095699 DOI: 10.1016/j.molimm.2005.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2005] [Accepted: 07/06/2005] [Indexed: 11/20/2022]
Abstract
Elucidating the role of glucocorticoid in regulating gene expression is crucial to developing effective strategies against inflammatory diseases such as arthritis. In this report we demonstrate that glucocorticoid inhibits transcription directed by the IL-lbeta gene (IL1B) upstream induction sequence (UIS) enhancer, and to a much lesser extent by the tissue-specific basal promoter. Within the enhancer, three transcription factor binding sites, previously demonstrated by us to be important for the induction of IL1B by lipopolysaccharide, are now shown to be directly inhibited by the synthetic glucocorticoid, dexamethasone. We also previously showed that one of these sites could bind a novel STAT-like factor, while the other two bound heterodimers containing NF-IL6(C/EBPbeta). Although it has been reported by others that NF-IL6 homodimers can interact with glucocorticoid receptor (GR) to enhance transcription of the alpha1-acid glycoprotein gene, it now appears that glucocorticoid represses DNA binding of NF-IL6 heterodimers as well as the novel STAT-like factor to the critical sites within the IL1B UIS. Thus, GR likely disrupts the DNA binding capability of critical IL1B factors via transrepression.
Collapse
Affiliation(s)
- Wayne R Waterman
- Philadelphia College of Osteopathic Medicine, Department of Neurosurgery, Philadelphia, PA 19131, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kim YS, Honkaniemi J, Sharp FR, Täuber MG. Expression of proinflammatory cytokines tumor necrosis factor-α and interleukin-1β in the brain during experimental Group B streptococcal meningitis. ACTA ACUST UNITED AC 2004; 128:95-102. [PMID: 15337322 DOI: 10.1016/j.molbrainres.2004.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2004] [Indexed: 10/26/2022]
Abstract
We performed mRNA in situ hybridization for TNF-alpha and IL-1beta from infant rats with group B streptococcal meningitis. Induction of both cytokines was seen in the ependyma and the meninges at 4 h. Both cytokines were expressed in the brain parenchyma at 12 h. Induction of IL-1beta mRNA was seen in vessels within the brain cortex. Neutrophilic infiltrate at all time points examined was minimal and could not account for the observed cytokine expression.
Collapse
Affiliation(s)
- Young S Kim
- Division of Infectious Diseases, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
20
|
Li L, Shui QX, Zhao ZY. Regulation of brain-derived neurotrophic factor (BDNF) expression following antibiotic treatment of experimental bacterial meningitis. J Child Neurol 2003; 18:828-34. [PMID: 14736076 DOI: 10.1177/088307380301801202] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although more and more new potent antibiotics have been used, mortality and neurologic deficits still occur frequently following bacterial meningitis in children. In this article, the expression of brain-derived neurotrophic factor messenger ribonucleic acid (RNA) and its production in the brains of rats were investigated during the course of experimental bacterial meningitis and after treatment with an antibiotic plus dexamethasone. In the brains of Streptococcus pneumoniae-inoculated rats, brain-derived neurotrophic factor (BDNF) messenger RNA was obviously up-regulated after inoculation for 24 hours (P < .01) and then declined but was still greater than that in the brains of control rats after inoculation for 5 days (P < .05). The expression of brain-derived neurotrophic factor in the brains of infected rats treated by antibiotic was dose dependent, down-regulated, and almost undetectable (P < .01) but up-regulated after treatment with an antibiotic plus dexamethasone (P < .01). However, the expression of brain-derived neurotrophic factor messenger RNA did not change in control rats treated with an antibiotic. Brain-derived neurotrophic factor protein showed similar changes, except it declined to normal levels 5 days after inoculation. Brain-derived neurotrophic factor messenger RNA and its production were observed in some infiltrating inflammatory cells in the brain of infected rats. The results of our studies support the hypothesis that brain-derived neurotrophic factor might play a neuroprotective role in brain damage during bacterial meningitis, and the expression of brain-derived neurotrophic factor messenger RNA and its production might be inhibited after treatment with antibiotics. The findings suggest that both eradicating the bacterial pathogen with antibiotics and adjuvant administering of brain-derived neurotrophic factor might be more beneficial to prevent brain damage.
Collapse
Affiliation(s)
- Ling Li
- Department of Neurology, Affiliated Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | | | | |
Collapse
|
21
|
Buzas B, Rosenberger J, Kim KW, Cox BM. Inflammatory mediators increase the expression of nociceptin/orphanin FQ in rat astrocytes in culture. Glia 2002; 39:237-46. [PMID: 12203390 DOI: 10.1002/glia.10106] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In the central nervous system, glial cells play an important role in inflammatory and immune responses, and opioid peptides have been identified as essential mediators between the nervous and the immune systems. We report the profound upregulation of the opioid-related nociceptin/orphanin FQ (N/OFQ) by inflammatory mediators in astrocytes. The bacterial endotoxin, lipopolysaccharide (LPS), and the proinflammatory cytokines, interleukin-beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha), induced levels of N/OFQ mRNA and immunoreactivity. HPLC analysis of the immunoreactivity in astrocyte extracts revealed that a large molecular weight precursor for N/OFQ is being synthesized and released in response to LPS and astrocytes appear to lack the enzymes required to process the precursor protein. Western blot analysis showed that LPS treatment elicited the activation of ERK 1/2 and p38 MAP kinases. Blockade of the p38 or the ERK MAP kinase pathways prevented the LPS-induced increase in N/OFQ mRNA levels indicating a role for these cascades in the regulation of N/OFQ genes in response to LPS. Regulation of N/OFQ gene expression by ERK and p38 activation may be mediated through the transcription factor CREB. We observed CREB phosphorylation in response to LPS, which was also prevented by SB202190 and PD98059. The NFkappaB pathway also appears to be involved in the induction of N/OFQ transcription by LPS, since NFkappaB inhibitors antagonized the effect of LPS on N/OFQ expression. Regulation of N/OFQ by inflammatory mediators in astrocytes may suggest a role for N/OFQ in neural-glial communication and in inflammatory responses in certain neuropathophysiological conditions.
Collapse
Affiliation(s)
- Beata Buzas
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA.
| | | | | | | |
Collapse
|
22
|
Xie Y, Chen C, Stevenson MA, Auron PE, Calderwood SK. Heat shock factor 1 represses transcription of the IL-1beta gene through physical interaction with the nuclear factor of interleukin 6. J Biol Chem 2002; 277:11802-10. [PMID: 11801594 DOI: 10.1074/jbc.m109296200] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heat shock factor (HSF) 1 is the major heat shock transcription factor that regulates stress-inducible synthesis of heat shock proteins and is also essential in protection against endotoxic shock. Following our previous study, which demonstrated the transcriptional repression of the IL-1beta gene by HSF1 (Cahill, C. M., Waterman, W. R., Xie, Y., Auron, P. E., and Calderwood, S. K. (1996) J. Biol. Chem. 271, 24874-24879), we have examined the mechanisms of transcriptional repression. Our studies show that HSF1 represses the lipopolyliposaccharide-induced transcription of the IL-1beta promoter through direct interaction with the nuclear factor of interleukin 6 (NF-IL6, also known as CCAAT enhancer binding protein (C/EBPbeta), an essential regulator in IL-1beta transcription. We show for the first time that HSF1 binds directly to NF-IL6 in vivo and antagonizes its activity. The HSF1/NF-IL6 interaction involves a sequence of HSF1 containing the trimerization and regulatory domains and the bZip region of NF-IL6. HSF1 has little effect on IL-1beta promoter activity stimulated by the essential monocytic transcription factor Spi.1 but is strongly inhibitory to transcriptional activation by NF-IL6 and to the synergistic activation by NF-IL6 and Spi.1. Because of its ability to bind to specific C/EBP elements in the promoters of multiple genes and its ability to interact with other transcription factors, NF-IL6 is involved in transcriptional regulation of a wide range of genes. Interaction between HSF1 and NF-IL6 could thus be an important mechanism in HSF1 regulation of general gene transcription during endotoxin stress.
Collapse
Affiliation(s)
- Yue Xie
- Department of Radiation Oncology, Dana Farber Cancer Institute, Harvard Medical School, Beth Israel and Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
23
|
Sonna LA, Fujita J, Gaffin SL, Lilly CM. Invited review: Effects of heat and cold stress on mammalian gene expression. J Appl Physiol (1985) 2002; 92:1725-42. [PMID: 11896043 DOI: 10.1152/japplphysiol.01143.2001] [Citation(s) in RCA: 417] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This review examines the effects of thermal stress on gene expression, with special emphasis on changes in the expression of genes other than heat shock proteins (HSPs). There are approximately 50 genes not traditionally considered to be HSPs that have been shown, by conventional techniques, to change expression as a result of heat stress, and there are <20 genes (including HSPs) that have been shown to be affected by cold. These numbers will likely become much larger as gene chip array and proteomic technologies are applied to the study of the cell stress response. Several mechanisms have been identified by which gene expression may be altered by heat and cold stress. The similarities and differences between the cellular responses to heat and cold may yield key insights into how cells, and by extension tissues and organisms, survive and adapt to stress.
Collapse
Affiliation(s)
- Larry A Sonna
- Thermal and Mountain Medicine Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts 01760, USA.
| | | | | | | |
Collapse
|
24
|
Yanagawa Y, Kawakami M, Okada Y. Moderate hypothermia alters interleukin-6 and interleukin-1alpha reactions in ischemic brain in mice. Resuscitation 2002; 53:93-9. [PMID: 11947985 DOI: 10.1016/s0300-9572(01)00499-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Female C57BL/6 mice were decapitated and their brains were removed and cultured at either 37 or 33 degrees C for 48 h to investigate whether or not moderate hypothermia alters the cytokine reactions in the ischemic brain. The interleukin-6 and interleukin-1alpha levels in the culture media were significantly elevated in a time-dependent manner. The interleukin-6 levels after the incubation at 33 degrees C were significantly lower than those at 37 degrees C. The interleukin-1alpha levels at 33 degrees C were significantly higher than those at 37 degrees C. The interleukin-1alpha levels incubated with interleukin-6 antibody were significantly higher than those without IL-6 antibody. At 37 degrees C, the mRNA expression of interleukin-6 was observed from 2 to 48 h after incubation, but the same expression of interleukin-1alpha was only detected until 12 h. Accordingly, the ischemic brain incubated at 33 degrees C showed a decreased interleukin-6 production in comparison with that at 37 degrees C and the level of interleukin-6 showed negative feedback for the production of interleukin-1alpha. The temperature should, therefore, be carefully considered when evaluating the cytokine reaction for cerebral ischemia.
Collapse
Affiliation(s)
- Youichi Yanagawa
- Department of Traumatology and Critical Care Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | | | | |
Collapse
|
25
|
Xie Y, Chen C, Stevenson MA, Hume DA, Auron PE, Calderwood SK. NF-IL6 and HSF1 have mutually antagonistic effects on transcription in monocytic cells. Biochem Biophys Res Commun 2002; 291:1071-80. [PMID: 11866474 DOI: 10.1006/bbrc.2002.6562] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have examined the functional antagonism between the regulator of the heat shock response, HSF1, and NF-IL6, which plays a major role in control of the acute phase response (APR). Agents that activate HSF1 such as heat shock and sodium salicylate inhibit NF-IL6 induced transcription while NF-IL6 activators such as lipopolysaccharide (LPS) and interleukin 6 (IL-6) repressed the stress responsive HSP70B promoter. In transfection studies, the inhibitory effects of HSF1 and NF-IL6 on the c-fms promoter were shown to be highly dose-dependent. Furthermore, heat shock is inhibitory to differentiation-linked expression of macrophage colony stimulating factor (M-CSF) receptor, product of the c-fms gene, which is transcriptionally activated by NF-IL6 but repressed by HSF1. Our studies suggest a strong mutual antagonism between the heat shock response and APR, which may influence the sensitivity and duration of inflammatory responses.
Collapse
Affiliation(s)
- Yue Xie
- Department of Radiation Oncology, Dana Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
26
|
Choi SH, Choi DH, Song KS, Shin KH, Chun BG. Zaprinast, an inhibitor of cGMP-selective phosphodiesterases, enhances the secretion of TNF-alpha and IL-1beta and the expression of iNOS and MHC class II molecules in rat microglial cells. J Neurosci Res 2002; 67:411-21. [PMID: 11813247 DOI: 10.1002/jnr.10102] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Proinflammatory cytokines produced by activated glial cells may in turn augment the immune/inflammatory reactions of glial cells through autocrine and paracrine routes. The NO/cGMP signaling represents one of the reactions of activated glial cells. We investigated whether the production of proinflammatory cytokines by glial cells is affected by NO-dependent downstream cGMP signaling. In primary cultures of mixed astrocytes and microglial cells, zaprinast (0.1 mM), an inhibitor of cGMP-selective phosphodiesterases, enhanced the basal and LPS (1.0 microg/ml)-induced secretion of TNF-alpha and IL-1beta. Zaprinast also enhanced NO production induced by LPS or IFN-gamma (100 U/ml), and in microglial cell cultures, but not in astrocyte cultures, zaprinast enhanced the basal and the IFN-gamma-induced production of the cytokines, TNF-alpha and IL-1beta, and of NO. This upregulation by zaprinast was partially inhibited by KT5823 (1.0 microM), an inhibitor of protein kinase G. The LPS-induced production of TNF-alpha, IL-1beta, and NO was inhibited by ODQ (50 microM), an inhibitor of soluble guanylyl cyclase, and by KT5823. Immunohistochemical analysis of mixed glial cell cultures showed that LPS/IFN-gamma-induced iNOS expression and the enhanced expression of iNOS by zaprinast were restricted to microglial cells. Zaprinast enhanced the IFN-gamma (200 U/ml)-induced expression of MHC Class II molecules in astrocytes and microglial cells in mixed cultures, but did not enhance this IFN-gamma-induced expression in pure astrocytes, which lacked paracrine TNF-alpha from microglial cells. Summarizing, zaprinast, which is associated with cGMP/protein kinase G signaling, may augment central immune/inflammatory reactions, possibly via the increased production of TNF-alpha and IL-1beta by activated microglial cells.
Collapse
Affiliation(s)
- Sang-Hyun Choi
- Department of Pharmacology, Korea University College of Medicine, Sungbuk-Gu, Seoul, South Korea.
| | | | | | | | | |
Collapse
|
27
|
Hasday JD, Singh IS. Fever and the heat shock response: distinct, partially overlapping processes. Cell Stress Chaperones 2001. [PMID: 11189454 DOI: 10.1379/1466-1268(2000)005<0471:fathsr>2.0.co;2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The heat shock response is an ancient and highly conserved process that is essential for surviving environmental stresses, including extremes of temperature. Fever is a more recently evolved response, during which organisms temporarily subject themselves to thermal stress in the face of infections. We review studies showing that fever is beneficial in the infected host. We show that core temperatures achieved during fever can activate the heat shock response and discuss some of the biochemical consequences of such an effect. We present data suggesting 4 possible mechanisms by which fever might confer protection: (1) directly killing or inhibiting growth of pathogens; (2) inducing cytoprotective heat shock proteins (Hsps) in host cells; (3) inducing expression of pathogen Hsps, an activator of host defenses; and (4) modifying and orchestrating host defenses. Two of these mechanisms directly involve the heat shock response. We describe how heat shock factor-1, the predominant heat-induced transcriptional enhancer not only activates transcription of Hsps but also regulates expression of pivotal cytokines and early response genes. The relationship between fever and the heat shock response is an illuminating example of how a more recently evolved response might exploit preexisting biochemical pathways for a new function.
Collapse
Affiliation(s)
- J D Hasday
- Department of Medicine, University of Maryland School of Medicine and the Medicine and Research Services of the Baltimore VA Medical Center, 21201, USA.
| | | |
Collapse
|
28
|
Abstract
Sepsis is a highly lethal clinical syndrome characterized by a systemic inflammatory response to infection. Fever, a non-specific acute-phase response, has been associated with improved survival and shortened disease duration in non-life-threatening infections. However, the influence of fever and the effects of antipyresis in patients with sepsis has not been prospectively studied in humans. This paper reviews the state of our knowledge concerning the biological effects of fever in infected hosts and the influence of fever and antipyretic therapy on survival during sepsis in experimental models and in man.
Collapse
Affiliation(s)
- J D Hasday
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, and the Medicine Services of the Baltimore Veterans Affairs Centre, MD 21201, USA.
| | | | | |
Collapse
|
29
|
Fairchild KD, Viscardi RM, Hester L, Singh IS, Hasday JD. Effects of hypothermia and hyperthermia on cytokine production by cultured human mononuclear phagocytes from adults and newborns. J Interferon Cytokine Res 2000; 20:1049-55. [PMID: 11152570 DOI: 10.1089/107999000750053708] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have shown previously that febrile range temperatures modify cytokine production by adult macrophages. In this study, we compared the effects of moderate hyperthermia and hypothermia on the kinetics of lipopolysaccharide (LPS)-induced cytokine expression in monocytes and macrophages of newborns and adults. During culture at 40 degrees C, the initial rates of tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 beta (IL-1 beta) secretion were preserved, but the duration of secretion was shorter than the duration at 37 degrees C. TNF-alpha and IL1-beta concentrations in 24-h 40 degrees C culture supernatants were reduced 18%-50%. IL-6 concentration in 24-h 40 degrees C cultures was reduced 26%-29% in all cells except adult macrophages. At 32 degrees C, changes in early (2 h) and sustained (24 h) cytokine expression were reversed compared with those caused by hyperthermia. Culturing adult macrophages at 32 degrees C blunted early secretion of TNF-alpha and IL-6 by 69% and 65%, respectively, and increased TNF-alpha concentration at 24 h by 48% compared with levels at 37 degrees C. In adult monocytes cultured at 32 degrees C, early IL-6 and IL-1 beta secretion was decreased 64% and 51%, respectively. We speculate that the burst/suppression cytokine profile at febrile temperatures might enhance early activation of host defenses and prevent prolonged exposure to potentially cytotoxic cytokines. Hypothermia, on the other hand, may worsen outcome in infections by delaying and prolonging cytokine production.
Collapse
Affiliation(s)
- K D Fairchild
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 20201, USA.
| | | | | | | | | |
Collapse
|
30
|
Ostberg JR, Taylor SL, Baumann H, Repasky EA. Regulatory effects of fever‐range whole‐body hyperthermia on the LPS‐induced acute inflammatory response. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.6.815] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Julie R. Ostberg
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | - Shannon L. Taylor
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | - Heinz Baumann
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York
| | | |
Collapse
|
31
|
Abstract
The heat shock response is an ancient and highly conserved process that is essential for surviving environmental stresses, including extremes of temperature. Fever is a more recently evolved response, during which organisms temporarily subject themselves to thermal stress in the face of infections. We review studies showing that fever is beneficial in the infected host. We show that core temperatures achieved during fever can activate the heat shock response and discuss some of the biochemical consequences of such an effect. We present data suggesting 4 possible mechanisms by which fever might confer protection: (1) directly killing or inhibiting growth of pathogens; (2) inducing cytoprotective heat shock proteins (Hsps) in host cells; (3) inducing expression of pathogen Hsps, an activator of host defenses; and (4) modifying and orchestrating host defenses. Two of these mechanisms directly involve the heat shock response. We describe how heat shock factor-1, the predominant heat-induced transcriptional enhancer not only activates transcription of Hsps but also regulates expression of pivotal cytokines and early response genes. The relationship between fever and the heat shock response is an illuminating example of how a more recently evolved response might exploit preexisting biochemical pathways for a new function.
Collapse
Affiliation(s)
- J D Hasday
- Department of Medicine, University of Maryland School of Medicine and the Medicine and Research Services of the Baltimore VA Medical Center, 21201, USA.
| | | |
Collapse
|
32
|
Abstract
Sepsis is a clinical syndrome characterized by a systemic inflammatory response to infection. Mortality rates in sepsis have remained high, despite recent advances in our understanding of the immunological mechanisms that cause sepsis. Fever, a nonspecific acute-phase response, has been associated with improved survival and shortened disease duration in some infections. This article reviews the biological effects of fever and the influence of antipyretic therapy on the outcome in sepsis in experimental models and in humans and offers clinical recommendations for antipyretic therapy in early and late stages of the disorder.
Collapse
Affiliation(s)
- J D Hasday
- Medical Care Clinical Center, Veterans Affairs Maryland Health Care System, Baltimore, MD 21201, USA.
| | | |
Collapse
|
33
|
Condorelli F, Sortino MA, Stella AM, Canonico PL. Relative contribution of different receptor subtypes in the response of neuroblastoma cells to tumor necrosis factor-alpha. J Neurochem 2000; 75:1172-9. [PMID: 10936200 DOI: 10.1046/j.1471-4159.2000.0751172.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The effect of tumor necrosis factor-alpha (TNF-alpha) on neuronal viability has been investigated in the SK-N-BE neuroblastoma cell line. These cells undergo differentiation upon chronic treatment with retinoic acid. Exposure of SK-N-BE cells to TNF-alpha produced a proliferative response in undifferentiated cells, whereas a reduced cell number was observed in retinoic acid (RA)-differentiated cultures. This biphasic response may be related to the different expression of TNF-alpha receptors (TNFRs); a significant increase in the density of TNFR1 was in fact observed following RA-induced differentiation. Under these conditions, a pronounced increase in the formation of ceramide-1-phosphate (which was prevented by the selective inhibitor of phosphatidylcholine-specific phospholipase C, D609) and an activation of caspase-3 upon TNF-alpha challenge were evident. Selective blockade of each TNFR subtype allowed a more detailed analysis of the effect observed. Preincubation with an anti-TNFR1 antibody prevented the cytotoxic effect of TNF-alpha in RA-differentiated SK-N-BE cells, whereas the anti-TNFR2 antibody blocked the proliferative activity of the cytokine in undifferentiated cultures.
Collapse
Affiliation(s)
- F Condorelli
- Institute of Pharmacology, University of Catania, Italy
| | | | | | | |
Collapse
|
34
|
Singh IS, Viscardi RM, Kalvakolanu I, Calderwood S, Hasday JD. Inhibition of tumor necrosis factor-alpha transcription in macrophages exposed to febrile range temperature. A possible role for heat shock factor-1 as a negative transcriptional regulator. J Biol Chem 2000; 275:9841-8. [PMID: 10734139 DOI: 10.1074/jbc.275.13.9841] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously reported that expression of tumor necrosis factor-alpha (TNFalpha) was attenuated in macrophages exposed to febrile range temperatures. In this study, we analyzed the influence of temperature on TNFalpha transcription in the Raw 264.7 macrophage cell line during incubation at 37 and 39.5 degrees C. The initial activation of TNFalpha transcription in response to endotoxin (LPS) was comparable in the 37 and 39.5 degrees C cell cultures, peaking within 10 min of LPS stimulation. However, the duration of transcriptional activation was markedly reduced in the 39.5 degrees C cells (30-60 min) compared with the 37 degrees C cells (2-4 h). Deletion mapping of the TNFalpha gene revealed that the proximal 85-nucleotide promoter sequence and the 5'-untranslated region were sufficient for temperature sensitivity. This sequence contains six heat shock response element (HRE) half-sites but no complete HREs. Electrophoretic mobility shift and immunoblot assays demonstrated that nuclear transclocation of heat shock factor (HSF) and its activation to a DNA-binding form occurred in the 39.5 degrees C cells in the absence of heat shock protein-70 gene activation. The proximal TNFalpha promoter/5'-untranslated region sequence competed for HSF binding to a classic HRE. Overexpression of HSF-1 reduced activity of the TNFalpha promoter. These data suggest that partial activation of HSF-1 during exposure to febrile, sub-heat shock temperatures may block TNFalpha transcription by binding to its proximal promoter or 5'-untranslated region.
Collapse
Affiliation(s)
- I S Singh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, the University of Maryland at Baltimore Cytokine Core Laboratory, Maryland 21201, USA
| | | | | | | | | |
Collapse
|
35
|
Freyer D, Manz R, Ziegenhorn A, Weih M, Angstwurm K, Döcke WD, Meisel A, Schumann RR, Schönfelder G, Dirnagl U, Weber JR. Cerebral Endothelial Cells Release TNF-α After Stimulation with Cell Walls of Streptococcus pneumoniae and Regulate Inducible Nitric Oxide Synthase and ICAM-1 Expression Via Autocrine Loops. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.8.4308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
TNF-α, inducible NO synthase (iNOS), and ICAM-1 are considered to be key proteins in the inflammatory response of most tissues. We tested the hypothesis that cell walls of Streptococcus pneumoniae (PCW), the most common cause of adult bacterial meningitis, induce TNF-α, iNOS, and ICAM-1 expression in rat primary brain microvascular endothelial cell cultures. We detected TNF-α mRNA by RT-PCR already 1 h after stimulation with PCW, while TNF-α protein peaked at 4 h (9.4 ± 3.6 vs 0.1 ± 0.1 pg/μg protein). PCW induced iNOS mRNA 2 h after stimulation, followed by an increase of the NO degradation product nitrite (18.1 ± 4 vs 5.8 ± 1.8 at 12 h; 18.1 ± 4 vs 5.8 ± 1.8 pmol/μg protein at 72 h). The addition of TNF-α Ab significantly reduced nitrite production to 62.2 ± 14.4% compared with PCW-stimulated brain microvascular endothelial cells (100%). PCW induced the expression of ICAM-1 (measured by FACS), which was completely blocked by TNF-α Ab (142 ± 18.6 vs 97.5 ± 12.4%; 100% unstimulated brain microvascular endothelial cells). Cerebral endothelial cells express TNF-α mRNA as well as iNOS mRNA and release the bioactive proteins in response to PCW. PCW-induced NO production is mediated in part by an autocrine pathway involving TNF-α, whereas ICAM-1 expression is completely mediated by this autocrine loop. By these mechanisms, cerebral endothelial cells may regulate critical steps in inflammatory blood-brain-barrier disruption of bacterial meningitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ralf R. Schumann
- ‡Institut für Mikrobiologie und Hygiene, Universitaetsklinikum Charité, Humboldt University Berlin, Germany; and
| | - Gilbert Schönfelder
- §Institut für Klinische Pharmakologie und Toxikologie, Free University Berlin, Berlin, Germany
| | | | | |
Collapse
|
36
|
Hauck W, Samlalsingh-Parker J, Glibetic M, Ricard G, Beaudoin MC, Noya FJ, Aranda JV. Deregulation of cyclooxygenase and nitric oxide synthase gene expression in the inflammatory cascade triggered by experimental group B streptococcal meningitis in the newborn brain and cerebral microvessels. Semin Perinatol 1999; 23:250-60. [PMID: 10405195 DOI: 10.1016/s0146-0005(99)80070-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Group B Streptococcus (GBS) is the most common cause of neonatal sepsis and meningitis. Despite antibiotics, GBS in the newborn initiates a cascade of molecular and biological events leading to altered cerebral perfusion, blood-brain barrier disruption, cerebral edema, intracranial hypertension, neurological damage, and even death. Having previously shown that GBS infection impairs cerebral blood flow autoregulation and increases prostaglandin (PG) levels, we examined the regulation of some crucial inflammatory mediators (PGs, nitric oxide (NO), tumor necrosis factor-a) in the brain and cerebral microvessels (MVs) from newborn piglets. Cyclooxygenase (COX), the key enzyme in PG biosynthesis, exists in two isoforms, COX-1 and COX-2. Both may be directly induced by NO in a model of renal inflammation. Besides its neurotransmitter role, NO is a potent vasorelaxant whose production is catalyzed by at least three distinct nitric oxide synthases (NOS) (bNOS, ecNOS, iNOS). Western blot analyses showed that the newborn (4 day old) brain expressed lower levels of COX-1 (8-fold), COX-2 (20-fold), bNOS (12-fold), and ecNOS (5-fold) than in the 1 day old. MV showed approximately equal levels of COX-2, lower levels of COX-1 (4-fold), bNOS (5-fold), and higher levels of ecNOS (20-fold) in comparison to 4-day-old cerebral MV. A 4-day-old brain expressed lower levels of bNOS (5-fold), ecNOS (10-fold), and COX-1 (2-fold) than the 6-week-old pig. COX-2 protein was undetected in a 4-day-old pig brain, but present in great excess in MV. Purified MV showed lower ecNOS (14-fold), COX-1 (2-fold), and about equal levels of bNOS and COX-2 in comparison with MV from 6-week-old pigs. Reverse transcription polymerase chain reaction analyses confirmed these results. Treatment with noo-nitro-L-arginine (LNA), a NOS inhibitor, downregulated COX-1 expression in the newborn brain and both COX-1 and COX-2 cerebral MV expression. GBS infection (10(9) colony-forming units, 0.5 mL intracerebroventricular) of sedated newborn piglets induced the expression of tumor necrosis factor-alpha in the cerebrospinal fluid after 2 hours, upregulated bNOS expression in both brain and MVs, upregulated ecNOS in MVs, and downregulated COX-1, COX-2, and ecNOS in the brain. GBS did not trigger the expression of iNOS. Our data suggest that there is a net deficiency of NOS isoforms in the immature brain and microvasculature of the 4-day-old piglet and that the differences in expression lead to the immature control of NO and PG production, rendering newborns particularly susceptible to neurological damage because of the undeveloped nature of their response mechanisms. Moreover, the GBS-induced cascade deregulates the gene expression of interacting inflammatory mediators and may cause a net vasoconstrictor/vasodilator imbalance, leading to cerebral hypertension and edema in the early stages of infection. Pharmacological manipulations of the inflammatory cascade could lead to novel therapeutic approaches for the treatment of GBS meningitis.
Collapse
Affiliation(s)
- W Hauck
- Department of Pharmacology, McGill University, Lady Davis Institute, Jewish General Hospital, Montreal, Canada
| | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Xie Y, Cahill CM, Asea A, Auron PE, Calderwood SK. Heat shock proteins and regulation of cytokine expression. Infect Dis Obstet Gynecol 1999; 7:26-30. [PMID: 10231005 PMCID: PMC1784720 DOI: 10.1155/s106474499900006x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Y Xie
- Department of Adult Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
39
|
Karlsson JO, Yarmush ML, Toner M. Interaction between heat shock and interleukin 6 stimulation in the acute-phase response of human hepatoma (HepG2) cells. Hepatology 1998; 28:994-1004. [PMID: 9755236 DOI: 10.1002/hep.510280414] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Two characteristic elements of the acute-phase response are an altered pattern of circulating hepatic proteins and fever. Whereas a fever-induced heat shock response could affect expression of acute-phase proteins in the liver, the effects of a modest temperature increase on protein secretion in interleukin-6 (IL-6)-stimulated HepG2 cells were investigated. The response of HepG2 cells to IL-6 stimulation was significantly affected by heat treatment at 40 degreesC. Albumin secretion rates, which were reduced by a factor of 2 in response to either heat shock or IL-6 stimulation alone, were down-regulated by a factor of 4 when IL-6 was administered simultaneously with a continuous 40 degrees C heat shock. IL-6-induced fibrinogen up-regulation was significantly reduced by heat treatment (P < .01), and secretion rates were indistinguishable from control levels after 2 days (P > .10). Unexpectedly, heat shock at 40 degrees C induced a fivefold up-regulation of haptoglobin production in the absence of IL-6. Simultaneous heat shock and IL-6 stimulation caused a synergistic enhancement of haptoglobin expression, with secretion rates increasing up to 30-fold compared with unstimulated control cells. For all three proteins, the interaction between temperature and IL-6 concentration was statistically significant (P < .001). Heat treatment resulted in significant alterations of both the kinetics and sensitivity of IL-6-induced protein synthesis, suggesting a major modification of the mechanism of acute-phase protein regulation at 40 degreesC. In summary, the data show that heat shock can significantly modulate the pattern of acute-phase protein expression and that fever may be an important regulatory factor in the acute-phase response.
Collapse
Affiliation(s)
- J O Karlsson
- Department of Mechanical Engineering, University of Illinois, Chicago, IL, USA
| | | | | |
Collapse
|
40
|
Schumann RR, Pfeil D, Freyer D, Buerger W, Lamping N, Kirschning CJ, Goebel UB, Weber JR. Lipopolysaccharide and pneumococcal cell wall components activate the mitogen activated protein kinases (MAPK) erk-1, erk-2, and p38 in astrocytes. Glia 1998; 22:295-305. [PMID: 9482215 DOI: 10.1002/(sici)1098-1136(199803)22:3<295::aid-glia8>3.0.co;2-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cell wall compounds of gram-positive bacteria are capable of inducing the biosynthesis of proinflammatory cytokines in CNS cells in a similar way as lipopolysaccharide (LPS) of gram-negative bacteria does. Astrocytes, which lack the CD14 LPS receptor, have also been shown to respond to LPS-stimulation by increased cytokine synthesis. However, almost nothing is known about signaling steps involved in this process. We have therefore examined signaling events in primary cultures of rat astrocytes and the human astrocytoma cell line U373MG, brought about by LPS and pneumococcal cell walls (PCW). Of particular interest to us was the tyrosine phosphorylation patterns and activation states of three members of the mitogen activated protein kinase (MAPK) family, i.e., extracellular signal-regulated protein kinase (erk)-1, erk-2, and the recently identified p38. We show that LPS and PCW initiate tyrosine phosphorylation and activation of erk-1, erk-2, and p38 in a dose-dependent fashion. Inhibitors of tyrosine phosphorylation were able to alleviate this effect and also blocked cytokine production of astrocytes. Both, LPS- and PCW-induced responses of astrocytic cells required the presence of soluble CD14 (sCD14) present in serum. Unraveling the signaling steps induced by bacterial compounds in cells of the CNS may potentially help to elucidate the pathomechanisms of meningitis and central nervous complications of sepsis and may offer options for novel treatment strategies.
Collapse
Affiliation(s)
- R R Schumann
- Molecular Sepsis Research Laboratory, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Tkachuk M, Gisler RH. The promoter of macrophage colony-stimulating factor receptor is active in astrocytes. Neurosci Lett 1997; 225:121-5. [PMID: 9147389 DOI: 10.1016/s0304-3940(97)00186-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Macrophage colony-stimulating factor (M-CSF) is a hematopoietin whose actions are essential for growth and survival of macrophages, placental development, ramification of microglia and tumor progression. The expression of the receptor for macrophage colony-stimulating factor (c-fms) is regulated by two distinct promoters: distal and proximal. The distal promoter is active in trophoblasts during embryogenesis and the proximal promoter directs expression to the cells of myeloid lineage. Here we report the generation of transgenic mice expressing beta-galactosidase under the control of the human proximal c-fms promoter and demonstrate the promoter activity in astrocytes, cells of neurological origin that partially take over the role of the macrophages in the central nervous system. Enzymatic activity of beta-galactosidase was detected in homogenated spleen, bone marrow and brain and in the cell extracts from peritoneal macrophages of transgenic mice. Immunohistochemical staining of brain showed the presence of beta-galactosidase in astrocytes. We hypothesize that M-CSF released by astrocytes, upon stimulation by lipopolysaccharide (LPS), tumor necrosis factor alpha (TNF alpha) or interleukin-1 (IL-1), regulates the expression of its own receptor.
Collapse
Affiliation(s)
- M Tkachuk
- PRPG, Hofmann-La Roche AG, Basel, Switzerland
| | | |
Collapse
|
42
|
Klosterhalfen B, Töns C, Hauptmann S, Tietze L, Offner FA, Küpper W, Kirkpatrick CJ. Influence of heat shock protein 70 and metallothionein induction by zinc-bis-(DL-hydrogenaspartate) on the release of inflammatory mediators in a porcine model of recurrent endotoxemia. Biochem Pharmacol 1996; 52:1201-10. [PMID: 8937427 DOI: 10.1016/0006-2952(96)00469-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The manipulation of stress gene expression by heavy metals provides protection against the lethal effects of endotoxemia in murine models of septic shock. Recent in vitro studies with alveolar macrophages or monocytes show that induction of the stress response in these cells is followed by a decreased liberation of major cytokines [tumor necrosis factor-alpha (TNF alpha) and interleukin-1 (IL-1)] after endotoxin challenge. These findings suggest that the increased resistance to endotoxin in vivo after stress protein induction could be explained by an altered pattern of inflammatory mediator release. Therefore, we measured the time course of thromboxane-B2 (TxB2), 6-keto-PGF1 alpha, platelet activating factor (PAF), TNF alpha, interleukin-1 beta (IL-1 beta), and interleukin-6 (IL-6) formation with and without induction of the stress response in an established porcine model of recurrent endotoxemia (Klosterhalfen et al., Biochem Pharmacol 43: 2103-2109, 1992). Induction of the stress response was done by a pretreatment with Zn2+ (25 mg/kg zinc-bis-(DL-hydrogenasparate = 5 mg/kg Zn2+). Pretreatment with Zn2+ prior to lipopolysaccharide (LPS) infusion induced an increased heat shock protein 70 and metallothionein expression in the lungs, liver, and kidneys and increased plasma levels of TNF alpha, IL-1 beta, IL-6, and TxB2 as opposed to untreated controls. After LPS infusion, however, pretreated animals showed significantly decreased peak plasma levels of all mediators as opposed to the untreated group. The time course of mediator release was identical with the decreasing and increasing three peak profiles described previously. Hemodynamic data presented significantly decreased peak pulmonary artery pressures and significantly altered hypodynamic/hyperdynamic cardiac output levels in the pretreated group. In conclusion, the data show that the induction of stress proteins by Zn2+ could be a practicable strategy to prevent sepsis.
Collapse
Affiliation(s)
- B Klosterhalfen
- Department of Surgery, Technical University of Aachen, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Cahill CM, Waterman WR, Xie Y, Auron PE, Calderwood SK. Transcriptional Repression of the Prointerleukin 1β Gene by Heat Shock Factor 1. J Biol Chem 1996. [DOI: 10.1016/s0021-9258(18)40087-7] [Citation(s) in RCA: 147] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
44
|
Kim YS, Täuber MG. Neurotoxicity of glia activated by gram-positive bacterial products depends on nitric oxide production. Infect Immun 1996; 64:3148-53. [PMID: 8757846 PMCID: PMC174200 DOI: 10.1128/iai.64.8.3148-3153.1996] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The present study examined the mechanism by which bacterial cell walls from two gram-positive meningeal pathogens, Streptococcus pneumoniae and the group B streptococcus, induced neuronal injury in primary cultures of rat brain cells. Cell walls from both organisms produced cellular injury to similar degrees in pure astrocyte cultures but not in pure neuronal cultures. Cell walls also induced nitric oxide production in cultures of astrocytes or microglia. When neurons were cultured together with astrocytes or microglia, the cell walls of both organisms became toxic to neurons. L-NAME, a nitric oxide synthase inhibitor, protected neurons from cell wall-induced toxicity in mixed cultures with glia, as did dexamethasone. In contrast, an excitatory amino acid antagonist (MK801) had no effect. Low concentrations of cell walls from either gram-positive pathogen added together with the excitatory amino acid glutamate resulted in synergistic neurotoxicity that was inhibited by L-NAME. The induction of nitric oxide production and neurotoxicity by cell walls was independent of the presence of serum, whereas endotoxin exhibited these effects only in the presence of serum. We conclude that gram-positive cell walls can cause toxicity in neurons by inducing the production of nitric oxide in astrocytes and microglia.
Collapse
Affiliation(s)
- Y S Kim
- Infectious Diseases Laboratory, San Francisco General Hospital, California 94143, USA
| | | |
Collapse
|
45
|
Willis SA, Nisen PD. Differential induction of the mitogen-activated protein kinase pathway by bacterial lipopolysaccharide in cultured monocytes and astrocytes. Biochem J 1996; 313 ( Pt 2):519-24. [PMID: 8573086 PMCID: PMC1216937 DOI: 10.1042/bj3130519] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We recently reported that cyclic AMP (cAMP) specifically inhibits lipopolysaccharide (LPS)-induced interleukin 1 beta (IL-1 beta) transcription initiation in astrocytic cells but enhances the LPS induction of IL-1 beta in monocytic cells. The purpose of this study was to determine how cAMP differentially regulates LPS-induced IL-1 beta transcription in these two cell types. Two essential components of the mitogen-activated protein (MAP) kinase signal-transduction pathway, extracellular-signal-regulated kinase (ERK2; p41 mapk) and Raf-1, have been shown to be targets of LPS stimulation in other cell types, and therefore may be linked to the regulation of IL-1 beta transcription. In the human astrocytic cell line, U-373MG, LPS was found to strongly activate (and cAMP to inhibit) both ERK2 and Raf-1. In the human monocytic cell line, THP-1, LPS minimally activated ERK2 and did not activate Raf-1. These findings suggest that, in astrocytic cells, elevated intracellular cAMP levels may negatively regulate LPS activation of IL-1 beta via the MAP kinase signalling pathway. In contrast, this pathway is not significantly activated by LPS in monocytic cells, thus inhibition by elevated intracellular cAMP levels would not affect IL-1 beta transcription.
Collapse
Affiliation(s)
- S A Willis
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas 75235-9063, USA
| | | |
Collapse
|
46
|
Freyer D, Weih M, Weber JR, Bürger W, Scholz P, Manz R, Ziegenhorn A, Angestwurm K, Dirnagl U. Pneumococcal cell wall components induce nitric oxide synthase and TNF-alpha in astroglial-enriched cultures. Glia 1996; 16:1-6. [PMID: 8787768 DOI: 10.1002/(sici)1098-1136(199601)16:1<1::aid-glia1>3.0.co;2-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Astroglia and microglia, the most numerous cells in the central nervous system (CNS), have been shown to produce the inducible nitric oxide synthase (iNOS) and tumor necrosis factor-alpha (TNF-alpha) upon stimulation with the cytokines IFN-gamma, IL-1-beta, or bacterial lipopolysaccharides (LPS). However, it is not known whether gram-positive bacteria like Streptococcus pneumoniae cause astroglial cells to release nitric oxide (NO) and TNF-alpha. S. pneumoniae meningitis still has a high incidence and mortality in spite of antibiotic therapy. Cell wall components from S. pneumoniae (pneumococcal cell-wall components, PCW) and TNF-alpha have been shown to cause meningeal inflammation and cerebrovascular changes in experimental meningitis. Addition of PCW to cultured rat astroglial cells increased nitrite in the supernatant significantly after 24 h, from 17 +/- 21 to 133 +/- 62 nM/micrograms protein. Nitrite release was dose-dependent in a range shown to cause meningeal inflammation in vivo and was inhibited by the competitive NO synthase inhibitor NW-nitro-L-arginine (L-NA 10(-4 M)) and dexamethasone (10(-6 M)), with transcriptional and translational inhibition by actinomycin D and cycloheximide, respectively. PCW caused a significant increase in the release of TNF-alpha from astroglial cells after 4 h, from 2 +/- 3.5 pg/ml to 102 +/- 13.5 pg/ml, which was inhibited by dexamethasone (10(-6 M)). Our results suggest a role for astroglial-derived NO and TNF-alpha as mediators of vascular and inflammatory response in the early phase of experimental pneumococcal meningitis.
Collapse
Affiliation(s)
- D Freyer
- Department of Neurology, Humboldt University, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Geley S, Fiegl M, Hartmann BL, Kofler R. Genes mediating glucocorticoid effects and mechanisms of their regulation. Rev Physiol Biochem Pharmacol 1996; 128:1-97. [PMID: 8791720 DOI: 10.1007/3-540-61343-9_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- S Geley
- Institute for General and Experimental Pathology, University of Innsbruck Medical School, Austria
| | | | | | | |
Collapse
|
48
|
Abstract
Cytokines are a group of molecules with an extremely broad range of activities on a variety of target cells. This review summarizes the known cytokine and cytokine receptor expression in primary brain tumors and derived cell lines. These expression patterns are compared with those occurring in other CNS diseases, such as virus or bacterial infections, experimental allergic encephalitis, multiple sclerosis, and trauma. A variety of cytokines are expressed during CNS neoplasia; their potential involvement in tumor growth through a variety of mechanisms, such as autocrine or paracrine growth stimulation, angiogenesis, and immune surveillance evasion, are discussed. Finally, results of preliminary therapeutic approaches with cytokines are critically evaluated.
Collapse
Affiliation(s)
- E G Van Meir
- Neurosurgery Department, University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
49
|
Kimberlin DW, Velasco S, Paris MM, Hickey SM, McCracken GH, Nisen PD. Modulation of expression of genes involved in the inflammatory response by lipopolysaccharide and temperature in cultured human astroglial cells. Immunol Invest 1995; 24:775-85. [PMID: 8543341 DOI: 10.3109/08820139509060705] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In bacterial sepsis and meningitis, large concentrations of interleukin-1 beta (IL-1 beta) and tumor necrosis factor-alpha (TNF-alpha) correlate directly with morbidity and mortality. This laboratory has reported previously that elevated temperature in the physiologic range is associated with down regulation of IL-1 beta and TNF alpha expression in cultured astroglia after lipopolysaccharide (LPS) stimulation. To further investigate the role of elevated temperature in the CNS inflammatory response, the effects of LPS and elevated temperature on the expression of genes that participate in the inflammatory response were determined in cultured transformed human fetal astrocytes and in an astrocytoma cell line. The effect of physiologic temperature elevation on cytokine concentrations in cerebrospinal fluid (CSF) was also investigated in a rabbit meningitis model. The findings indicate that astrocytes express a wide variety of cytokines, growth factors, growth factor receptors, and other genes that could play important roles in CNS inflammation. Furthermore, temperature elevation in the febrile range can lead to alterations in the patterns of expression of many genes involved in the inflammatory response of these cells.
Collapse
Affiliation(s)
- D W Kimberlin
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas 75235, USA
| | | | | | | | | | | |
Collapse
|
50
|
Kimberlin DW, Willis SA, McCracken GH, Nisen PD. Protein synthesis-dependent induction of interleukin-1 beta by lipopolysaccharide is inhibited by dexamethasone via mRNA destabilization in human astroglial cells. J Clin Immunol 1995; 15:199-204. [PMID: 7593467 DOI: 10.1007/bf01541090] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Dexamethasone inhibits lipopolysaccharide-induced synthesis of the cytokine, interleukin-1 beta, in cerebrospinal fluid of patients with bacterial meningitis. Along with monocytes, astrocytes are capable of producing lipopolysaccharide-induced interleukin-1 beta in the central nervous system. The objective of this study was to investigate the induction of interleukin-1 beta mRNA by lipopolysaccharide, and the inhibition of this process by dexamethasone, in human astrocytes using the astrocytoma cell line U-373MG as a model system. Dexamethasone-mediated inhibition of induction of interleukin-1 beta mRNA by lipopolysaccharide required a functional glucocorticoid receptor. In contrast to monocytes, lipopolysaccharide induction of interleukin-1 beta mRNA in U-373MG cells required de novo protein synthesis. Dexamethasone also had no effect on lipopolysaccharide-induced interleukin-1 beta transcriptional initiation in U-373MG cells. U-373MG cells were similar to monocytes, however, with respect to the ability of dexamethasone to decrease interleukin-1 beta mRNA half-life. These findings demonstrate that the mode of lipopolysaccharide induction of interleukin-1 beta mRNA, and inhibition of this process by dexamethasone, can vary in different cell types.
Collapse
Affiliation(s)
- D W Kimberlin
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas 75235-9063, USA
| | | | | | | |
Collapse
|