1
|
Ameliorative effects of clonidine on ethanol induced kidney injury in rats: Potential role for imidazoline-1 receptor. Eur J Pharmacol 2018; 824:148-156. [DOI: 10.1016/j.ejphar.2018.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 01/18/2018] [Accepted: 02/06/2018] [Indexed: 02/06/2023]
|
2
|
Fasick V, Spengler RN, Samankan S, Nader ND, Ignatowski TA. The hippocampus and TNF: Common links between chronic pain and depression. Neurosci Biobehav Rev 2015; 53:139-59. [PMID: 25857253 DOI: 10.1016/j.neubiorev.2015.03.014] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 02/02/2015] [Accepted: 03/28/2015] [Indexed: 02/07/2023]
Abstract
Major depression and chronic pain are significant health problems that seriously impact the quality of life of affected individuals. These diseases that individually are difficult to treat often co-exist, thereby compounding the patient's disability and impairment as well as the challenge of successful treatment. The development of efficacious treatments for these comorbid disorders requires a more comprehensive understanding of their linked associations through common neuromodulators, such as tumor necrosis factor-α (TNFα), and various neurotransmitters, as well as common neuroanatomical pathways and structures, including the hippocampal brain region. This review discusses the interaction between depression and chronic pain, emphasizing the fundamental role of the hippocampus in the development and maintenance of both disorders. The focus of this review addresses the hypothesis that hippocampal expressed TNFα serves as a therapeutic target for management of chronic pain and major depressive disorder (MDD).
Collapse
Affiliation(s)
- Victoria Fasick
- Department of Pathology and Anatomical Sciences, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, United States
| | | | - Shabnam Samankan
- Department of Pathology and Anatomical Sciences, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, United States
| | - Nader D Nader
- Department of Pathology and Anatomical Sciences, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, United States; Department of Anesthesiology, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, United States
| | - Tracey A Ignatowski
- Department of Pathology and Anatomical Sciences, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, United States; NanoAxis, LLC, Clarence, NY 14031, United States; Program for Neuroscience, School of Medicine and Biomedical Science, University at Buffalo, The State University of New York, Buffalo, NY 14214, United States.
| |
Collapse
|
3
|
Abelaira HM, Réus GZ, Petronilho F, Barichello T, Quevedo J. Neuroimmunomodulation in depression: a review of inflammatory cytokines involved in this process. Neurochem Res 2014; 39:1634-9. [PMID: 24996933 DOI: 10.1007/s11064-014-1372-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 06/02/2014] [Accepted: 06/25/2014] [Indexed: 01/26/2023]
Abstract
Depression is a debilitating mental disease that affects a large number of people globally; however the pathophysiological mechanisms of this disease remain incompletely understood. Some studies have shown that depression is associated with inflammatory activity, and the mode of action of several antidepressants appears to involve immunomodulation. In this case, the induction of a pro-inflammatory state in healthy or depressive subjects induces a 'sickness behaviour' resembling depressive symptomatology. Potential mechanisms of pro-inflammatory cytokines are effects on monoamine levels, disruption of the hypothalamic-pituitary-adrenal axis, activation of the pathological microglial cells, such as the macrophages and alterations in neuroplasticity and brain functions. Thus, this review will highlight the role of inflammation in depression, the possible mechanisms involved, and also explore effective treatments that act on the immune system.
Collapse
Affiliation(s)
- Helena M Abelaira
- Laboratório de Neurociências, Unidade Acadêmica de Ciências da Saúde, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | | | | | | | | |
Collapse
|
4
|
Krügel U, Fischer J, Radicke S, Sack U, Himmerich H. Antidepressant effects of TNF-α blockade in an animal model of depression. J Psychiatr Res 2013; 47:611-6. [PMID: 23394815 DOI: 10.1016/j.jpsychires.2013.01.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/26/2012] [Accepted: 01/11/2013] [Indexed: 12/31/2022]
Abstract
Pro-inflammatory cytokines such as tumour necrosis factor-alpha (TNF-α) have repeatedly been shown to play a pivotal role in the pathophysiology of depression. Therefore, we tested the possible antidepressant-like effect of the anti-TNF-α drug etanercept in an animal model of chronic mild stress. Male Wistar rats were assigned to a non-restrained and a restrained protocol for 5 weeks. From beginning of the third week the animals were treated either with Ringer solution daily or with etanercept twice a week (0.3 mg/kg, i.p.) instead of Ringer solution (n = 12 each). As reference, imipramine (10 mg/kg, i.p.) was administered in a third restraint group daily. Naïve non-treated non-restrained rats served as healthy controls (n = 12). In the forced swim test (FST) depression-like behaviour induced by restraint was recorded as enhanced immobile time and reduced climbing activity of the vehicle-treated group in comparison to the naïve and the non-restrained vehicle treated group. The treatment with etanercept significantly reduced the depression-like effects resulting in reduced immobile time in the FST and intensified climbing behaviour (p < 0.01, p < 0.05), both similar to the antidepressive-like effect of imipramine (p < 0.01 both). The repeated restraint induced a loss of body weight gain in the Ringer-treated group which was not reversed, neither by imipramine nor by etanercept. The antidepressant effects of blocking TNF-α using etanercept may be caused by enhancement of serotonergic or noradrenergic neurotransmission or normalization of stress hormone secretion which has to be substantiated in further studies.
Collapse
Affiliation(s)
- Ute Krügel
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Härtelstrasse 16-18, D-04107 Leipzig, Germany.
| | | | | | | | | |
Collapse
|
5
|
Kim JE, Ryu HJ, Kang TC. P2X7 receptor activation ameliorates CA3 neuronal damage via a tumor necrosis factor-α-mediated pathway in the rat hippocampus following status epilepticus. J Neuroinflammation 2011; 8:62. [PMID: 21631954 PMCID: PMC3123566 DOI: 10.1186/1742-2094-8-62] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Accepted: 06/02/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The release of tumor necrosis factor-α (TNF-α) appears depend on the P2X7 receptor, a purinergic receptor. In the present study, we addressed the question of whether P2X7 receptor-mediated TNF-α regulation is involved in pathogenesis and outcome of status epilepticus (SE). METHODS SE was induced by pilocarpine in rats that were intracerebroventricularly infused with saline-, 2',3'-O-(4-benzoylbenzoyl)-adenosine 5'-triphosphate (BzATP), adenosine 5'-triphosphate-2',3'-dialdehyde (OxATP), A-438079, or A-740003 prior to SE induction. Thereafter, we performed Fluoro-Jade B staining and immunohistochemical studies for TNF-α and NF-κB subunit phosphorylations. RESULTS Following SE, P2X7 receptor agonist (BzATP) infusion increased TNF-α immunoreactivity in dentate granule cells as compared with that in saline-infused animals. In addition, TNF-α immunoreactivity was readily apparent in the mossy fibers, while TNF-α immunoreactivity in CA1-3 pyramidal cells was unaltered. However, P2X7 receptor antagonist (OxATP-, A-438079, and A-740003) infusion reduced SE-induced TNF-α expression in dentate granule cells. In the CA3 region, BzATP infusion attenuated SE-induced neuronal damage, accompanied by enhancement of p65-Ser276 and p65-Ser311 NF-κB subunit phosphorylations. In contrast, OxATP-, A-438079, and A-740003 infusions increased SE-induced neuronal death. Soluble TNF p55 receptor (sTNFp55R), and cotreatment with BzATP and sTNFp55R infusion also increased SE-induced neuronal damage in CA3 region. However, OxATP-, sTNFp55R or BzATP+sTNFp55R infusions could not exacerbate SE-induced neuronal damages in the dentate gyrus and the CA1 region, as compared to BzATP infusion. CONCLUSIONS These findings suggest that TNF-α induction by P2X7 receptor activation may ameliorate SE-induced CA3 neuronal damage via enhancing NF-κB p65-Ser276 and p65-Ser311 phosphorylations.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do 200-702, South Korea
- Ji-Eun Kim, Department of Neurology, UCSF, and Veterans Affairs Medical Center, San Francisco, California 94121, USA
| | - Hea Jin Ryu
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do 200-702, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do 200-702, South Korea
| |
Collapse
|
6
|
Takei Y, Laskey R. Intracellular and Intercellular Cross Talk Between NGF and TNF. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 691:559-65. [DOI: 10.1007/978-1-4419-6612-4_58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
7
|
Reddy PH, Manczak M, Zhao W, Nakamura K, Bebbington C, Yarranton G, Mao P. Granulocyte-macrophage colony-stimulating factor antibody suppresses microglial activity: implications for anti-inflammatory effects in Alzheimer's disease and multiple sclerosis. J Neurochem 2009; 111:1514-28. [PMID: 19840215 PMCID: PMC2796704 DOI: 10.1111/j.1471-4159.2009.06432.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The objective of our study was to determine granulocyte-macrophage colony-stimulating factor (GM-CSF) activity in the brain following GM-CSF induction. We injected recombinant mouse GM-CSF into the brains of 8-month-old C57BL6 mice via intracerebroventricular injections and studied the activities of microglia, astrocytes, and neurons. We also sought to determine whether an anti-GM-CSF antibody could suppress endogenous microglial activity in the C57BL6 mice and could also suppress microglial activity induced by the recombinant mouse GM-CSF in another group of C57BL6 mice. Using quantitative real-time RT-PCR, we assessed microglial, astrocytic, and neuronal activity by measuring mRNA expression of pro-inflammatory cytokines, GFAP, and the neuronal marker NeuN in the cerebral cortex tissues from C57BL6 mice. We performed immunoblotting and immunohistochemistry of activated microglia in different regions of the brains from control (phosphate-buffered saline-injected C57BL6 mice) and experimental mice (recombinant GM-CSF-injected C57BL6 mice, GM-CSF antibody-injected C57BL6 mice, and recombinant mouse GM-CSF plus anti-GM-CSF antibody-injected C57BL6 mice). We found increased mRNA expression of CD40 (9.75-fold), tumor necrosis factor-alpha (2.1-fold), CD45 (1.73-fold), and CD11c (1.70-fold) in the cerebral cortex of C57BL6 mice that were induced with recombinant GM-CSF, compared with control mice. Further, the anti-GM-CSF antibody suppressed microglia in mice that were induced with recombinant GM-CSF. Our immunoblotting and immunohistochemistry findings of GM-CSF-associated cytokines in C57BL6 mice induced with recombinant GM-CSF, in C57BL6 mice injected with the anti-GM-CSF antibody, and in C57BL6 mice injected with recombinant mouse GM-CSF plus anti-GM-CSF antibody concurred with our real-time RT-PCR findings. These findings suggest that GM-CSF is critical for microglial activation and that anti-GM-CSF antibody suppresses microglial activity in the CNS. The findings from this study may have implications for anti-inflammatory effects of Alzheimer's disease and experimental autoimmune encephalomyelitis mice (a multiple sclerosis mouse model).
Collapse
Affiliation(s)
- P Hemachandra Reddy
- Neurogenetics Laboratory, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA.
| | | | | | | | | | | | | |
Collapse
|
8
|
Adjuvant therapy with intrathecal clonidine improves postoperative pain in patients undergoing coronary artery bypass graft. Clin J Pain 2009; 25:101-6. [PMID: 19333153 DOI: 10.1097/ajp.0b013e3181817add] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alpha2 adrenergic agonists have long been employed as analgesics and to sedate patients undergoing surgical procedures. In addition, their therapeutic response synergizes that elicited by opioids. Although this response is well known, the role of alpha2 agonists, such as clonidine, during various painful surgical procedures remains to be elucidated. The goal of our study was to evaluate the effects of the intrathecal administration of clonidine on postoperative pain control and time to extubation in patients undergoing coronary artery bypass grafting. METHODS Eighty-five patients undergoing coronary artery bypass grafting randomly received either an intrathecal injection of preservative free morphine 0.5 mg (MOR) or a combination of morphine 0.5 mg and clonidine 100 microg (CMC) before induction of anesthesia. Anesthesia was induced and maintained using a balanced anesthesia technique. Patients were transferred to the intensive care unit while intubated and weaned from mechanical ventilation following an established weaning protocol. Postoperative pain, opioid use within the first 24 hours, and time to extubation were used as primary outcome variables. Data were analyzed by a 2-tailed t test for continuous variables and Fisher exact test for nonparametric variables. RESULTS There were no demographic differences between the CMC and MOR groups. Postoperative pain, as assessed by a visual analog scale, was milder in the CMC group when compared with that of the MOR group (2.2+/-0.36 vs. 3.4+/-0.33, P<0.05). Similarly, patients in the CMC group required lower doses of morphine within 24 hours compared with the MOR group (2.02+/-0.36 vs. 6.47+/-0.49 mg, P<0.0001). Time to extubation was significantly shorter in patients receiving CMC than in those who received MOR (592+/-52 vs. 887+/-75 min, P<0.05). There was no mortality in either group. There was a trend for increased vasopressin use in the CMC group compared with the MOR group, although this was not statistically significant (P=0.07). CONCLUSIONS Addition of clonidine to neuraxial opioids improves the quality of analgesia postoperatively and expedites the process of weaning from mechanical ventilation. There were no serious adverse events in the cohort of the patients studied. However, the safety profile of this medication remains to be examined with a larger group of patients.
Collapse
|
9
|
Youn DH, Wang H, Jeong SJ. Exogenous tumor necrosis factor-alpha rapidly alters synaptic and sensory transmission in the adult rat spinal cord dorsal horn. J Neurosci Res 2008; 86:2867-75. [PMID: 18543334 DOI: 10.1002/jnr.21726] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) is involved in the generation of inflammatory and neuropathic pain. This study investigated if TNF-alpha has any effect on spinal synaptic and/or sensory transmission by using whole-cell recordings of substantia gelatinosa (SG) neurons in transverse lumbar spinal cord slices of adult rats and by using behavioral tests. After intrathecal administration of TNF-alpha in adult rats, spontaneous hind paw withdrawal behavior and thermal hyperalgesia were rapidly induced (approximately 30 min), while mechanical allodynia slowly developed. Bath application of TNF-alpha (0.1-1 nM, 8 min) depressed peak amplitude of monosynaptic Adelta and C fiber-evoked excitatory postsynaptic currents (EPSCs) without changing in holding currents and input resistances, whereas this application generally potentiated polysynaptic Adelta fiber-evoked EPSCs. Moreover, the frequencies, but not the amplitudes, of spontaneous and miniature EPSCs and spontaneous inhibitory postsynaptic currents were significantly increased by bath-applied TNF-alpha in most of the SG neurons. The effects of TNF-alpha on Adelta/C fiber-evoked monosynaptic and polysynaptic or spontaneous EPSCs were significantly blocked by 5 microM TNF-alpha antagonist that inhibits TNF-alpha binding to its type 1 receptor (TNFR1). Because this study also found high protein expression of TNFR1 in the adult dorsal root ganglion and no change of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) induced whole-cell currents by TNF-alpha, we conclude that presynaptic TNFR1 at Adelta/C primary afferent terminals contributes to the rapid alteration of synaptic transmission in the spinal SG, and the development of abnormal pain hypersensitivity by exogenous TNF-alpha.
Collapse
Affiliation(s)
- Dong-Ho Youn
- Department of Anesthesia and Critical Care, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA.
| | | | | |
Collapse
|
10
|
Takei Y, Laskey R. Interpreting crosstalk between TNF-alpha and NGF: potential implications for disease. Trends Mol Med 2008; 14:381-8. [PMID: 18693138 DOI: 10.1016/j.molmed.2008.07.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 07/04/2008] [Accepted: 07/04/2008] [Indexed: 12/19/2022]
Abstract
Tumour necrosis factor-alpha (TNF-alpha) is a proinflammatory cytokine, whereas nerve growth factor (NGF) is a neurotrophin that can promote neural cell survival, differentiation and maturation. However, recent papers indicate that TNF-alpha has a pivotal role in fate decisions of neural cells in normal noninflammatory conditions, whereas NGF contributes to maintenance of inflammation. Although these observations suggest a close relationship between NGF and TNF-alpha signalling, crosstalk between these factors is not fully understood. In this Opinion article, we review recent reports regarding possible crosstalk between NGF and TNF-alpha and we propose a positive-feedback loop of their expression. We discuss the possible mechanisms by which disturbance of the crosstalk could contribute to diseases such as cancer and Alzheimer's disease.
Collapse
Affiliation(s)
- Yoshinori Takei
- Medical Research Council (MRC) Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road Cambridge CB2 0XZ, UK.
| | | |
Collapse
|
11
|
Sud R, Spengler RN, Nader ND, Ignatowski TA. Antinociception occurs with a reversal in alpha 2-adrenoceptor regulation of TNF production by peripheral monocytes/macrophages from pro- to anti-inflammatory. Eur J Pharmacol 2008; 588:217-31. [PMID: 18514187 DOI: 10.1016/j.ejphar.2008.04.043] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 04/02/2008] [Accepted: 04/09/2008] [Indexed: 02/07/2023]
Abstract
Tumor necrosis factor-alpha (TNF) plays a role in neuropathic pain. During neuropathic pain development in the chronic constriction injury model, elevated TNF levels in the brain occur in association with enhanced alpha 2-adrenoceptor inhibition of norepinephrine release. alpha 2-Adrenoceptors are also located on peripheral macrophage where they normally function as pro-inflammatory, since they increase the production of the cytokine TNF, a proximal mediator of inflammation. How the central increase in TNF affects peripheral alpha 2-adrenoceptor function was investigated. Male, Sprague-Dawley rats had four loose ligatures placed around the right sciatic nerve. Thermal hyperalgesia was determined by comparing hind paw withdrawal latencies between chronic constriction injury and sham-operated rats. Chronic constriction injury increased TNF immunoreactivity at the lesion and the hippocampus. Amitriptyline, an antidepressant that is used as an analgesic, was intraperitoneally administered (10 mg/kg) starting simultaneous with ligature placement (day-0) or at days-4 or -6 post-surgery. Amitriptyline treatment initiated at day-0 or day-4 post-ligature placement alleviated hyperalgesia. When initiated at day-0, amitriptyline prevented increased TNF immunoreactivity in the hippocampus and at the lesion. A peripheral inflammatory response, macrophage production of TNF, was also assessed in the current study. Lipopolysaccharide (LPS)-stimulated production of TNF by whole blood cells and peritoneal macrophages was determined following activation of the alpha 2-adrenoceptor in vitro. alpha 2-Adrenoceptor regulation of TNF production from peripheral immune-effector cells reversed from potentiation in controls to inhibition in chronic constriction injured rats. This effect is accelerated with amitriptyline treatment initiated at day-0 or day-4 post-ligature placement. Amitriptyline treatment initiated day-6 post-ligature placement did not alleviate hyperalgesia and prevented the switch from potentiation to inhibition in alpha 2-adrenoceptor regulation of TNF production. Recombinant rat TNF i.c.v. microinfusion reproduces the response of peripheral macrophages from rats with chronic constriction injury. A reversal in peripheral alpha 2-adrenoceptor regulation of TNF production from pro- to anti-inflammatory is associated with effective alleviation of thermal hyperalgesia. Thus, alpha 2-adrenoceptor regulation of peripheral TNF production may serve as a potential biomarker to evaluate therapeutic regimens.
Collapse
Affiliation(s)
- Reeteka Sud
- Department of Pathology and Anatomical Sciences, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
12
|
Cytokines in Synaptic Function. ACTA ACUST UNITED AC 2008. [DOI: 10.1016/s1567-7443(07)10007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
13
|
Thomson EM, Kumarathasan P, Calderón-Garcidueñas L, Vincent R. Air pollution alters brain and pituitary endothelin-1 and inducible nitric oxide synthase gene expression. ENVIRONMENTAL RESEARCH 2007; 105:224-33. [PMID: 17662977 DOI: 10.1016/j.envres.2007.06.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2006] [Revised: 05/04/2007] [Accepted: 06/15/2007] [Indexed: 05/16/2023]
Abstract
Recent work suggests that air pollution is a risk factor for cerebrovascular and neurodegenerative disease. Effects of inhaled pollutants on the production of vasoactive factors such as endothelin (ET) and nitric oxide (NO) in the brain may be relevant to disease pathogenesis. Inhaled pollutants increase circulating levels of ET-1 and ET-3, and the pituitary is a potential source of plasma ET, but the effects of pollutants on the expression of ET and NO synthase genes in the brain and pituitary are not known. In the present study, Fischer-344 rats were exposed by nose-only inhalation to particles (0, 5, 50mg/m3 EHC-93), ozone (0, 0.4, 0.8 ppm), or combinations of particles and ozone for 4 h. Real-time reverse transcription polymerase chain reaction was used to measure mRNA levels in the cerebral hemisphere and pituitary 0 and 24 h post-exposure. Ozone inhalation significantly increased preproET-1 but decreased preproET-3 mRNAs in the cerebral hemisphere, while increasing mRNA levels of preproET-1, preproET-3, and the ET-converting enzyme (ECE)-1 in the pituitary. Inducible NO synthase (iNOS) was initially decreased in the cerebral hemisphere after ozone inhalation, but increased 24 h post-exposure. Particles decreased tumour necrosis factor (TNF)-alpha mRNA in the cerebral hemisphere, and both particles and ozone decreased TNF-alpha mRNA in the pituitary. Our results show that ozone and particulate matter rapidly modulate the expression of genes involved in key vasoregulatory pathways in the brain and pituitary, substantiating the notion that inhaled pollutants induce cerebrovascular effects.
Collapse
Affiliation(s)
- Errol M Thomson
- Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ont., Canada K1A 0K9
| | | | | | | |
Collapse
|
14
|
Dey S, Snow DM. Cocaine exposure in vitro induces apoptosis in fetal locus coeruleus neurons through TNF-alpha-mediated induction of Bax and phosphorylated c-Jun NH(2)-terminal kinase. J Neurochem 2007; 103:542-56. [PMID: 17635674 DOI: 10.1111/j.1471-4159.2007.04750.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cocaine exposure results in aberrant outgrowth and decreased survival for locus coeruleus (LC), a noradrenergic population of neurons that putatively regulates attentional function; however, the underlying mechanisms for these events are not known. We previously showed that cocaine exposure in vitro activates pro-apoptotic Bax, caspase-9, and caspase-3 in LC neurons dissected from embryonic day 14 rats, implicating that apoptosis may be orchestrated via signal transduction events. In the current study in vitro, we examined upstream events to determine the role of the pro-inflammatory cytokine, tumor necrosis factor alpha (TNF-alpha), on LC signal transduction, because cocaine exposure to LC neurons triggered TNF-alpha expression at 30 min as measured by ELISA. Exposure of LC neurons to recombinant-TNF-alpha resulted in decreased metabolic activity, an indicator of reduced neuron viability [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay], and increased apoptosis (terminal deoxynucleotidyl transferase-mediated DNA nick end labeling assay). Pro-apoptotic caspase-3 was induced by cocaine starting at 30 min. Recombinant-TNF-alpha induced caspase-3 activity earlier than cocaine (15 and 20 min). The caspase-3 levels were significantly reduced when cocaine and TNF-alpha were combined with neutralizing-TNF-alpha (nTNF-alpha), respectively. Further, cocaine alone elevated phospho-p38-mitogen-activated protein kinases that persisted when combined with nTNF-alpha. However, both cocaine and TNF-alpha independently increased phospho-c-Jun NH(2)-terminal kinase and Bax levels at concurrent time periods (30 min and 1 h), and this elevation was attenuated in the presence of nTNF-alpha. These simultaneous molecular events triggered by cocaine and TNF-alpha implicate a potential apoptotic signal transduction pathway via induction of phospho-c-Jun NH(2)-terminal kinase and Bax that may lead to caspase-3 activation and apoptosis in cocaine-exposed fetal LC neurons.
Collapse
Affiliation(s)
- Swatee Dey
- Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky 40536, USA.
| | | |
Collapse
|
15
|
Spengler RN, Sud R, Knight PR, Ignatowski TA. Antinociception mediated by alpha(2)-adrenergic activation involves increasing tumor necrosis factor alpha (TNFalpha) expression and restoring TNFalpha and alpha(2)-adrenergic inhibition of norepinephrine release. Neuropharmacology 2007; 52:576-89. [PMID: 17055005 PMCID: PMC1839855 DOI: 10.1016/j.neuropharm.2006.08.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 07/26/2006] [Accepted: 08/21/2006] [Indexed: 02/07/2023]
Abstract
The central component that establishes chronic pain from peripheral nerve injury is associated with increased tumor necrosis factor-alpha (TNFalpha) production in the brain. This study examined TNFalpha and its reciprocally permissive role with alpha(2)-adrenergic activation during peak and progressive decline of thermal hyperalgesia in sciatic nerve chronic constriction injury (CCI). Accumulation of TNFalpha mRNA (in situ hybridization) increases in the hippocampus and locus coeruleus during the onset of neuropathic pain and persists as hyperalgesia abates. Activation of alpha(2)-adrenergic receptors in control rats decreases TNFalpha mRNA accumulation in these brain regions. In contrast, during hyperalgesia, alpha(2)-adrenergic activation enhances TNFalpha mRNA accumulation. Whether this enhanced TNFalpha production is associated with changes in the regulation of norepinephrine (NE) release was tested. Hippocampal slices were electrically depolarized to evaluate alpha(2)-adrenergic and TNFalpha regulation of NE release. While inhibition of NE release by TNFalpha is maximal during peak hyperalgesia, it subsequently transforms to facilitate NE release. In addition, alpha(2)-adrenergic receptor activation with clonidine (0.2mg/kg, i.p.) in CCI rats experiencing hyperalgesia restores TNFalpha and alpha(2)-adrenergic inhibition of NE release. While TNFalpha directs the development of hyperalgesia, it also directs its resolution. Transformed sensitivity to alpha(2)-adrenergic agonists during hyperalgesia demonstrates a mechanism for therapy.
Collapse
Affiliation(s)
- Robert N. Spengler
- Department of Pathology and Anatomical Sciences, University at Buffalo, The State University of New York, 3435 Main Street, Buffalo, NY 14214
- Department of Anesthesiology, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 3435 Main Street, Buffalo, NY 14214
| | - Reeteka Sud
- Department of Pathology and Anatomical Sciences, University at Buffalo, The State University of New York, 3435 Main Street, Buffalo, NY 14214
| | - Paul R. Knight
- Department of Anesthesiology, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 3435 Main Street, Buffalo, NY 14214
| | - Tracey A. Ignatowski
- Department of Pathology and Anatomical Sciences, University at Buffalo, The State University of New York, 3435 Main Street, Buffalo, NY 14214
- Department of Anesthesiology, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 3435 Main Street, Buffalo, NY 14214
| |
Collapse
|
16
|
Sud R, Ignatowski TA, Lo CPK, Spengler RN. Uncovering molecular elements of brain-body communication during development and treatment of neuropathic pain. Brain Behav Immun 2007; 21:112-24. [PMID: 16859892 DOI: 10.1016/j.bbi.2006.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Revised: 05/22/2006] [Accepted: 06/01/2006] [Indexed: 02/07/2023] Open
Abstract
Integral to neuropathic pain is a reciprocal interaction between tumor necrosis factor-alpha (TNF) production and the alpha(2)-adrenergic receptor response, offering an attractive therapeutic target. The effects of varying levels of brain TNF on alpha(2)-adrenergic regulation of cyclic AMP (cAMP) production in the hippocampus and sciatic nerve were investigated during the development and amitriptyline treatment of chronic pain. Increased levels of TNF during the development of chronic pain transform alpha(2)-adrenergic inhibition of cAMP production in the brain to potentiation. While alpha(2)-adrenergic receptors regulate TNF production, they also affect descending noradrenergic pathways. Increases in levels of TNF in the brain deeply impact peripheral inflammation through regulating alpha(2)-adrenergic receptors, offering insight into brain-body interactions during neuropathic pain. Amitriptyline as an analgesic inhibits pain-induced increases in brain-associated TNF and transforms peripheral alpha(2)-adrenergic receptors. The dynamic equilibrium between TNF levels and alpha(2)-adrenergic functioning is uniquely altered during development and treatment of neuropathic pain. Proper manipulations of this interaction offer efficacious treatment of neuropathic pain.
Collapse
Affiliation(s)
- Reeteka Sud
- Department of Pathology and Anatomical Sciences, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 3435 Main Street, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
17
|
Vogel C, Stallforth S, Sommer C. Altered pain behavior and regeneration after nerve injury in TNF receptor deficient mice. J Peripher Nerv Syst 2006; 11:294-303. [PMID: 17117937 DOI: 10.1111/j.1529-8027.2006.00101.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The pro-inflammatory cytokine tumor necrosis factor (TNF)-alpha is an important mediator in hyperalgesia, nerve injury, and regeneration. Here, we used mice deficient of TNF receptor (TNFR) 1 or 2 to investigate the role of TNF signaling via receptor in each pain behavior and nerve de- and regeneration after chronic constriction injury (CCI) of the sciatic nerve. We found an absence of thermal hyperalgesia in mice deficient of TNFR1 and a reduction in mechanical and cold allodynia in mice deficient of TNFR1 or TNFR2 compared with wild-type mice. Nerve conduction studies and nerve pathology did not reveal major differences between genotypes in the temporal course of de- and regeneration of the nerve. We propose that the functional effects of the TNFRs on pain symptoms are independent of effects on nerve regeneration. Furthermore, the differential action of TNF via each of its receptors should be taken into account when considering clinical trials with TNF inhibitors for pain.
Collapse
Affiliation(s)
- Carola Vogel
- Department of Neurology, University of Würzburg, Würzburg, Germany
| | | | | |
Collapse
|
18
|
Laan TTJM, Bull S, Pirie RS, Fink-Gremmels J. Evaluation of cytokine production by equine alveolar macrophages exposed to lipopolysaccharide, Aspergillus fumigatus, and a suspension of hay dust. Am J Vet Res 2006; 66:1584-9. [PMID: 16261833 DOI: 10.2460/ajvr.2005.66.1584] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate cytokine production by equine alveolar macrophages after exposure to lipopolysaccharide (LPS), Aspergillus fumigatus, and hay dust, and determine the effect of clenbuterol on the cytokine response. ANIMALS 6 horses. PROCEDURE Alveolar macrophages were exposed to PBS solution (negative control), LPS, hyphae and conidia of Aspergillus fumigatus (AF), or a suspension of hay dust (HDS) and incubated for 24 hours at 37 degrees C. Concentrations of tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta were measured in the supernatant. The procedure was repeated with cells that were concurrently incubated with 0.5 microM clenbuterol. RESULTS Exposure to HDS and AF significantly increased production of TNF-alpha by equine alveolar macrophages. The increase in TNF-alpha produced in response to HDS and AF was 5 and 7 times as great, respectively, as the increase measured in response to LPS. The concentration of IL-1beta in the supernatant was significantly increased after exposure of cells to AF. Clenbuterol was effective at inhibiting TNF-alpha production by cells exposed to LPS, HDS, or AF. CONCLUSIONS AND CLINICAL RELEVANCE Increased production of TNF-alpha and IL-1 indicated that the pro-inflammatory cytokines produced by alveolar macrophages in response to allergens may play a role in recurrent airway obstruction (RAO) in horses. Equine alveolar macrophages are not only a primary pulmonary defense mechanism but may also influence the pathogenesis of equine RAO. The beta2-adrenoceptor agonist clenbuterol, a drug that is commonly used for treatment of equine RAO, promotes immediate bronchodilation and may also contribute to downward modulation of the inflammatory response.
Collapse
Affiliation(s)
- Tamarinde T J M Laan
- Department of Equine Science, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 12-16, 3508 TD Utrecht, The Netherlands
| | | | | | | |
Collapse
|
19
|
Reynolds JL, Ignatowski TA, Sud R, Spengler RN. An antidepressant mechanism of desipramine is to decrease tumor necrosis factor-alpha production culminating in increases in noradrenergic neurotransmission. Neuroscience 2005; 133:519-31. [PMID: 15878644 DOI: 10.1016/j.neuroscience.2005.02.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2004] [Revised: 02/11/2005] [Accepted: 02/12/2005] [Indexed: 02/07/2023]
Abstract
The monoamine theory of depression proposes decreased bioavailability of monoamines, such as norepinephrine (NE), as the underlying cause of depression. Thus, the antidepressant efficacy of NE-reuptake inhibitors such as desipramine is attributed to increases in synaptic concentrations of NE. The time difference between inhibition of reuptake and therapeutic efficacy, however, argues against this being the primary mechanism. If desipramine elicits its therapeutic efficacy by increasing NE release, in turn, increasing activation of the alpha(2)-adrenergic autoinhibitory receptor, then mimicking this increase with an exogenous agonist (clonidine) should support or even enhance the efficacy of the antidepressant. Intriguingly, simultaneous administration of clonidine with desipramine prevented the cellular and behavioral effects elicited by desipramine alone, in both acute and chronic administration paradigms. These results suggest the involvement of additional factor(s) in the mechanism of antidepressant action of this drug. Desipramine administration results in a virtual ablation of neuron-derived tumor necrosis factor-alpha (TNF), thus implicating an essential role of TNF in the therapeutic efficacy of this antidepressant. Additionally, following chronic administration of desipramine, TNF-regulation of NE release is transformed, from inhibition to facilitation. Here, we demonstrate that a transformation in TNF-regulation of NE release in the brain is a key element in the efficacy of this antidepressant. Interestingly, an increase in neurotransmission prior to the antidepressant's effect on TNF production prevents the efficacy of the antidepressant drug. Thus, the efficacy of desipramine is due to decreased levels of TNF in the brain induced by this drug, ultimately modifying noradrenergic neurotransmission.
Collapse
Affiliation(s)
- J L Reynolds
- Department of Pathology and Anatomical Sciences, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
20
|
Reynolds JL, Ignatowski TA, Gallant S, Spengler RN. Amitriptyline administration transforms tumor necrosis factor-alpha regulation of norepinephrine release in the brain. Brain Res 2004; 1023:112-20. [PMID: 15364025 DOI: 10.1016/j.brainres.2004.07.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2004] [Indexed: 11/23/2022]
Abstract
The present study demonstrates that the mixed action antidepressant drug amitriptyline enhances norepinephrine (NE) release by transforming the nature of the response of neurons to both tumor necrosis factor-alpha (TNF) as well as to an alpha(2)-adrenergic agonist in an area of the central nervous system (CNS) rich in adrenergic neurons. Administration of the antidepressant drug amitriptyline for 1 day or 14 days to rats significantly increases TNF bioactivity in total homogenates of the locus coeruleus (LC) and the hippocampus as assessed by the WEHI-13VAR bioassay. Superfusion and electrical field stimulation of rat hippocampal brain slices were used to study the regulation of NE release. Exposure to TNF, as well as activation of the alpha(2)-adrenergic autoreceptor inhibits stimulation-evoked norepinephrine (NE) release from adrenergic neurons of the CNS from naïve rats. Superfusion of hippocampal slices isolated from rats chronically (14 days) administered amitriptyline demonstrates that TNF inhibition of NE release is transformed, such that TNF facilitates NE release, dependent upon alpha(2)-adrenergic activation. Furthermore, chronic administration of amitriptyline increases stimulation-evoked NE release and decreases alpha(2)-adrenergic autoreceptor inhibition of NE release, an effect not observed with acute drug administration. These data support the hypothesis that chronic antidepressant drug administration, through regulation of TNF expression, transforms alpha(2)-adrenergic receptors such that they function to facilitate NE release, suggesting a mechanism of action of antidepressant drugs.
Collapse
Affiliation(s)
- Jessica L Reynolds
- Department of Pathology and Anatomical Sciences, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 206 Farber Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
21
|
Renauld AE, Ignatowski TA, Spengler RN. Alpha 2-adrenergic receptor inhibition of cAMP accumulation is transformed to facilitation by tumor necrosis factor-alpha. Brain Res 2004; 1004:212-6. [PMID: 15033439 DOI: 10.1016/j.brainres.2004.01.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2004] [Indexed: 11/26/2022]
Abstract
Activation of the alpha(2)-adrenergic receptor on neurons regulates the activity of neurons. Inhibition of forskolin-stimulated cAMP accumulation induced by alpha(2)-adrenergic receptor activation is altered following exposure of the neuron SH-SY5Y cell line to tumor necrosis factor-alpha (TNF). Acute (5 and 15 min) exposure to TNF induces a transformation in alpha(2)-adrenergic regulation of cAMP accumulation from inhibition to facilitation. These findings support an autocrine role for the regulation of TNF production from neurons.
Collapse
Affiliation(s)
- Amy E Renauld
- Department of Pathology and Anatomical Sciences, School of Medicine and Biomedical Sciences, SUNY at Buffalo, 206 Farber Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | | | | |
Collapse
|
22
|
Yoshida H, Peterfi Z, García-García F, Kirkpatrick R, Yasuda T, Krueger JM. State-specific asymmetries in EEG slow wave activity induced by local application of TNFα. Brain Res 2004; 1009:129-36. [PMID: 15120590 DOI: 10.1016/j.brainres.2004.02.055] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2004] [Indexed: 10/26/2022]
Abstract
Sleep is posited to be a fundamental property of groups of highly interconnected neurons and regulated in part by activity-dependent sleep regulatory substances such as tumor necrosis factor alpha (TNFalpha). We show that the unilateral local application of TNFalpha onto the somatosensory cortex of rats induced state- and frequency-dependent EEG asymmetries. In contrast, the unilateral injection of a TNFalpha inhibitor, a TNFalpha soluble receptor, attenuated sleep deprivation-enhanced EEG slow wave power ipsilaterally during non-rapid eye movement sleep (NREMS) but not during REMS or waking. Results are consistent with the notion that sleep begins with state changes occurring within small groups of highly interconnected neurons and is driven in part by the local production of sleep regulating substances.
Collapse
Affiliation(s)
- Hitoshi Yoshida
- Department of VCAPP, College of Veterinary Medicine, Washington State University, P.O. Box 646520, Pullman, WA 99164-6520, USA
| | | | | | | | | | | |
Collapse
|
23
|
Reynolds JL, Ignatowski TA, Sud R, Spengler RN. Brain-derived tumor necrosis factor-alpha and its involvement in noradrenergic neuron functioning involved in the mechanism of action of an antidepressant. J Pharmacol Exp Ther 2004; 310:1216-25. [PMID: 15082752 DOI: 10.1124/jpet.104.067835] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The present study documents a role for brain-derived tumor necrosis factor-alpha (TNF) in the mechanism of action of the antidepressant drug desmethylimipramine (desipramine). To establish this role, field stimulation and superfusion of rat hippocampal slices was employed to investigate the regulation of norepinephrine (NE) release by TNF. Chronic desipramine administration transforms TNF-mediated inhibition of NE release to facilitation, dependent upon alpha2-adrenergic receptor activation. Chronic i.c.v. microinfusion of polyclonal TNF antibody (pTNF-Ab) similarly transforms TNF inhibition of NE release to facilitation. To determine whether this transformation is due to desipramine-induced inhibition of TNF bioactivity in the brain, rats were i.c.v. microinfused with recombinant rat TNF (rrTNF) for 14 days, either alone or with simultaneous i.p. desipramine administration. TNF regulation of NE release in hippocampal slices isolated from these rats was compared with slices isolated from rats chronically administered desipramine alone. Although simultaneous microinfusion of rrTNF with chronic desipramine administration prevents the transformation induced by desipramine, microinfusion of rrTNF enhances TNF inhibition of NE release. These cellular events correspond to changes in immobility, analyzed by the forced swim test (FST). Intracerebroventricular microinfusion of rrTNF increases the duration of immobility of rats in the FST, compared with rats microinfused with aCSF. Desipramine administered chronically decreases immobility duration, which is mimicked by i.c.v. microinfusion of pTNF-Ab and prevented by simultaneous i.c.v. microinfusion of rrTNF. Thus, i.c.v. microinfusion of rrTNF with concomitant desipramine administration opposes decreases in neuron-associated TNF levels, required to transform presynaptic sensitivity to TNF, which is necessary for the drug to be efficacious.
Collapse
Affiliation(s)
- Jessica L Reynolds
- University at Buffalo, The State University of New York, Department of Pathology and Anatomical Sciences, 206 Farber Hall, 3435 Main Street, Buffalo, New York 14214, USA
| | | | | | | |
Collapse
|
24
|
Abstract
Individual differences in the aging process can be conceptualized as an accumulation of wear and tear of daily experiences and major life stressors that interact with the genetic constitution and predisposing early life experiences. The neuroendocrine system, autonomic nervous system, and immune system are mediators of adaptation to challenges of daily life, referred to as allostasis, meaning "maintaining stability through change." Physiological mediators such as adrenalin from the adrenal medulla, glucocorticoids from the adrenal cortex, and cytokines from cells of the immune system act upon receptors in various tissues and organs to produce effects that are adaptive in the short run but can be damaging if the mediators are not shut off when no longer needed. When release of the mediators is not efficiently terminated, their effects on target cells are prolonged, leading to other consequences that may include receptor desensitization and tissue damage. This process has been named "allostatic load," and it refers to the price the tissue or organ pays for an overactive or inefficiently managed allostatic response. Therefore, allostatic load refers to the "cost" of adaptation. This article discusses the mediators of allostasis and their contributions to allostatic load as well as their role in resilience of the aging organism to stressful experiences.
Collapse
Affiliation(s)
- Bruce S McEwen
- Laboratory of Neuroendocrinology, Rockefeller University, New York, NY 10021, USA
| |
Collapse
|
25
|
Gemma C, Imeri L, Opp MR. Serotonergic activation stimulates the pituitary-adrenal axis and alters interleukin-1 mRNA expression in rat brain. Psychoneuroendocrinology 2003; 28:875-84. [PMID: 12892655 DOI: 10.1016/s0306-4530(02)00103-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Interactions between neurotransmitters and immunomodulators within the central nervous system may be functionally relevant for communication between the immune system and the brain. Previous studies indicate that cytokines such as interleukin-1 (IL-1) alter activity of the serotonergic system at multiple levels. This study tested the hypothesis that serotonergic activation modulates cytokine mRNA expression in brain. Serotonergic activation was induced by injecting rats intraperitoneally (i.p.) prior to dark onset with the serotonin precursor L-5-hydroxytryptophan (5-HTP; 100 mg/kg). Cytokine mRNA expression in discrete brain regions at selected time points was determined by means of ribonuclease protection assay. Plasma corticosterone concentrations were also measured to determine if the hypothalamic-pituitary-adrenal axis is activated in response to this treatment, which potentially could exert feedback regulating cytokine message expression in brain. Plasma corticosterone was elevated for 4 h after 5-HTP administration. At this time IL-1alpha mRNA expression was reduced in the hippocampus, hypothalamus, and brainstem, and IL-1beta mRNA was reduced in the hippocampus. Six hours after 5-HTP injection, IL-1beta mRNA increased in the hypothalamus. These results show that activation of the serotonergic system affects cytokine message expression in rat brain, possibly by actions of corticosterone.
Collapse
Affiliation(s)
- C Gemma
- Institute of Human Physiology II, University of Milan School of Medicine, Via Mangiagalli 32, I-20133 Milan, Italy
| | | | | |
Collapse
|
26
|
Renauld AE, Spengler RN. Tumor necrosis factor expressed by primary hippocampal neurons and SH-SY5Y cells is regulated by alpha(2)-adrenergic receptor activation. J Neurosci Res 2002; 67:264-74. [PMID: 11782970 DOI: 10.1002/jnr.10101] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neuron expression of the cytokine tumor necrosis factor-alpha (TNF), and the regulation of the levels of TNF by alpha(2)-adrenergic receptor activation were investigated. Adult rat hippocampal neurons and phorbol ester (PMA)-differentiated SH-SY5Y cells were examined. Intracellular levels of TNF mRNA accumulation, as well as TNF protein and that released into the supernatant were quantified by in situ hybridization, immunocytochemistry and bioanalysis, respectively. Both neuron cultures demonstrated constitutive production of TNF. Activation of the alpha(2)-adrenergic receptor increased intracellular levels of TNF mRNA and protein in SH-SY5Y cells after addition of graded concentrations of the selective agonist, Brimonidine (UK-14304) to parallel cultures. Intracellular levels of mRNA were increased in a concentration-dependent fashion within 15 min of UK-14304 addition and were sustained during 24 hr of receptor activation. In addition, the levels of TNF in the supernatant were increased in both types of neuron cultures within 15 min of alpha(2)-adrenergic receptor activation. Furthermore, levels of TNF significantly increased in the supernatants of both neuron cultures after potassium-induced depolarization. A reduction in this depolarization-induced release occurred in hippocampal neuron cultures after exposure to the sympathomimetic tyramine with media replacement to deplete endogenous catecholamines. This finding reveals a role for endogenous catecholamines in the regulation of TNF production. Potassium-induced depolarization resulted in the release of TNF in hippocampal neuron cultures within 15 min but not until 24 hr in SH-SY5Y cultures demonstrating a temporally mediated event dependent upon cell type. Neuron expression of TNF, regulated by alpha(2)-adrenergic receptor activation demonstrates not only how a neuron controls its own production of this pleiotropic cytokine, but also displays a normal role for neurons in directing the many functions of TNF.
Collapse
Affiliation(s)
- A E Renauld
- Department of Pathology, School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | | |
Collapse
|
27
|
Nader ND, Ignatowski TA, Kurek CJ, Knight PR, Spengler RN. Clonidine suppresses plasma and cerebrospinal fluid concentrations of TNF-alpha during the perioperative period. Anesth Analg 2001. [PMID: 11473862 DOI: 10.1213/00000539-200108000-00026] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
UNLABELLED The analgesic properties of alpha(2)-agonists are well known. In experimental models, tumor necrosis factor (TNF)-alpha regulates adrenergic responses in the brain. Constitutive TNF-alpha, in brain regions involved in pain perception, is decreased after the administration of clonidine. We investigated patients undergoing lower-extremity revascularization. Seven patients were treated with clonidine 0.2 mg per os (low), and three patients received 0.4 mg per os clonidine (high) before surgery. Eight patients received placebo and served as controls. Continuous spinal anesthesia was provided by insertion of a pliable catheter into the subarachnoid space. Baseline plasma and cerebrospinal fluid (CSF) samples were obtained before injection of local anesthetic. Samples were analyzed for TNF-alpha using a biologic assay. Systemic and central release of catecholamines were assessed by high-pressure liquid chromatography measurement of norepinephrine in plasma and CSF, vanillylmandelic acid and methoxy hydroxyl phenyl glycol in 24-h urinary excretion, respectively. Clonidine 0.2 mg pretreatment decreased TNF-alpha concentrations both in plasma and CSF. Patients receiving clonidine had lower pain visual analog scale scores and required less morphine compared with the Placebo group (P < 0.01). Preoperative administration of clonidine decreased catecholamine release in the periphery, as well as in the central nervous system. A smaller norepinephrine concentration in plasma and CSF, and less secretion of vanillylmandelic acid (P < 0.01) and methoxy hydroxyl phenyl glycol in the urine, were observed. Larger dose clonidine (0.4 mg) resulted in no detectable TNF-alpha in CSF. These results suggest that an interaction between TNF-alpha and the function of adrenergic neurons in the central nervous system may contribute to the sedative and analgesic effects of adrenergic agonists. IMPLICATIONS Preoperative administration of clonidine decreases both plasma and cerebrospinal fluid concentrations of inflammatory cytokines, resulting in perioperative analgesia and decreased sympathetic tone.
Collapse
Affiliation(s)
- N D Nader
- Department of Anesthesiology, SUNY-Buffalo, Buffalo, New York, USA.
| | | | | | | | | |
Collapse
|
28
|
Nader ND, Ignatowski TA, Kurek CJ, Knight PR, Spengler RN. Clonidine suppresses plasma and cerebrospinal fluid concentrations of TNF-alpha during the perioperative period. Anesth Analg 2001; 93:363-9 , 3rd contents page. [PMID: 11473862 DOI: 10.1097/00000539-200108000-00026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
UNLABELLED The analgesic properties of alpha(2)-agonists are well known. In experimental models, tumor necrosis factor (TNF)-alpha regulates adrenergic responses in the brain. Constitutive TNF-alpha, in brain regions involved in pain perception, is decreased after the administration of clonidine. We investigated patients undergoing lower-extremity revascularization. Seven patients were treated with clonidine 0.2 mg per os (low), and three patients received 0.4 mg per os clonidine (high) before surgery. Eight patients received placebo and served as controls. Continuous spinal anesthesia was provided by insertion of a pliable catheter into the subarachnoid space. Baseline plasma and cerebrospinal fluid (CSF) samples were obtained before injection of local anesthetic. Samples were analyzed for TNF-alpha using a biologic assay. Systemic and central release of catecholamines were assessed by high-pressure liquid chromatography measurement of norepinephrine in plasma and CSF, vanillylmandelic acid and methoxy hydroxyl phenyl glycol in 24-h urinary excretion, respectively. Clonidine 0.2 mg pretreatment decreased TNF-alpha concentrations both in plasma and CSF. Patients receiving clonidine had lower pain visual analog scale scores and required less morphine compared with the Placebo group (P < 0.01). Preoperative administration of clonidine decreased catecholamine release in the periphery, as well as in the central nervous system. A smaller norepinephrine concentration in plasma and CSF, and less secretion of vanillylmandelic acid (P < 0.01) and methoxy hydroxyl phenyl glycol in the urine, were observed. Larger dose clonidine (0.4 mg) resulted in no detectable TNF-alpha in CSF. These results suggest that an interaction between TNF-alpha and the function of adrenergic neurons in the central nervous system may contribute to the sedative and analgesic effects of adrenergic agonists. IMPLICATIONS Preoperative administration of clonidine decreases both plasma and cerebrospinal fluid concentrations of inflammatory cytokines, resulting in perioperative analgesia and decreased sympathetic tone.
Collapse
Affiliation(s)
- N D Nader
- Department of Anesthesiology, SUNY-Buffalo, Buffalo, New York, USA.
| | | | | | | | | |
Collapse
|
29
|
Nickola TJ, Ignatowski TA, Spengler RN. Antidepressant drug administration modifies the interactive relationship between alpha(2)-adrenergic sensitivity and levels of TNF in the rat brain. J Neuroimmunol 2000; 107:50-8. [PMID: 10808050 DOI: 10.1016/s0165-5728(00)00244-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A reciprocally permissive interaction occurs between cellular responses elicited by the pleiotropic cytokine tumor necrosis factor-alpha (TNF) and alpha(2)-adrenergic receptor activation, such that each may adapt in response to modifications in the other's effects. Changes in presynaptic adrenergic sensitivity as well as neuronal sensitivity to TNF have been implicated in the mechanism of action of antidepressant drugs. The present study examines the influence of alpha(2)-adrenergic receptor activation on levels of TNF in regions of the brain associated with adrenergic function and the expression of mood. Additionally, the role of TNF as a neuromodulator is demonstrated by in vivo microinfusion of rrTNF proximal to the hippocampus. Administration to rats of an alpha(2)-adrenergic receptor agonist (clonidine) decreases levels of TNF in homogenates of rat locus coeruleus and hippocampus within 7.5 min. Chronic (14 days) administration of the antidepressant drugs desipramine or zimelidine transforms alpha(2)-adrenergic receptor-dependent decreases in TNF levels to increases in levels of TNF in the locus coeruleus. This transformation to an increase in total levels of TNF also occurs, although transiently, in the hippocampus following acute (1 day) antidepressant drug administration. The effect of TNF on presynaptic alpha(2)-adrenergic sensitivity was also investigated. Field stimulation of hippocampal slices from rats microinfused with rrTNF proximal to the hippocampus for 14 days demonstrates a decrease in fractional release of [3H]NE and an increase in alpha(2)-adrenergic autoreceptor sensitivity. These data demonstrate a mutual dependence between alpha(2)-adrenergic receptor activation and levels of TNF in the central nervous system that would culminate in an increase in neurotransmitter release following antidepressant drug administration.
Collapse
Affiliation(s)
- T J Nickola
- Department of Pathology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, 204 Farber Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | | | | |
Collapse
|
30
|
Covey WC, Ignatowski TA, Knight PR, Spengler RN. Brain-derived TNFalpha: involvement in neuroplastic changes implicated in the conscious perception of persistent pain. Brain Res 2000; 859:113-22. [PMID: 10720620 DOI: 10.1016/s0006-8993(00)01965-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The pleiotropic cytokine tumor necrosis factor-alpha (TNFalpha) is implicated in the development of persistent pain through its actions in the periphery and in the central nervous system (CNS). Activation of the alpha(2)-adrenergic receptor is associated with modulation of pain, possibly through its autoregulatory effect on norepinephrine (NE) release in the CNS. The present study employs a chronic constriction nerve injury (CCI) pain model to demonstrate the interactive role of presynaptic sensitivity to TNFalpha and the alpha(2)-adrenergic autoreceptor in the pathogenesis of neuropathic pain. Accumulation of TNFalpha is increased initially in a region of the brain containing the locus coeruleus (LC) at day 4 post-ligature placement, followed by an increase in TNFalpha in the hippocampus at day 8 post-ligature placement, coincident with hyperalgesia. Levels of TNFalpha in the thoraco-lumbar spinal cord are also increased at day 8 post-ligature placement. Concurrently, alpha(2)-adrenergic receptor and TNFalpha-induced inhibition of NE release are increased, and stimulated NE release is decreased in superfused hippocampal slices isolated at day 8 post-ligature placement. Stimulated NE release is also decreased in spinal cord slices (lumbar region) from animals undergoing CCI, although in contrast to that which occurs in the hippocampus, alpha(2)-adrenergic receptor inhibition of NE release is not changed. These results indicate an important role that TNFalpha plays in adrenergic neuroplastic changes in a region of the brain that, among its many functions, appears to be a crucial link in the conscious perception of pain. We predict that neuroplastic changes, involving increased functional responses of alpha(2)-adrenergic autoreceptors and increased presynaptic sensitivity to TNFalpha, culminate in decreased NE release in the CNS. These neuroplastic changes provide a mechanism for the role of CNS-derived TNFalpha in the pathogenesis of persistent pain.
Collapse
Affiliation(s)
- W C Covey
- Department of Pathology, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, 204 Farber Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
31
|
Abstract
If cytokines are constitutively expressed by and act on neurons in normal adult brain, then we may have to modify our current view that they are predominantly inflammatory mediators. We critically reviewed the literature to determine whether we could find experimental basis for such a modification. We focused on two "proinflammatory" cytokines, interleukin (IL)-1 and tumor necrosis factor-alpha (TNFalpha) because they have been most thoroughly investigated in shaping our current thinking. Evidence, although equivocal, indicates that the genes coding for these cytokines and their accessory proteins are expressed by neurons, in addition to glial cells, in normal brain. Their expression is region- and cell type-specific. Furthermore, bioactive cytokines have been extracted from various regions of normal brain. The cytokines' receptors selectively are present on all neural cell types, rendering them responsive to cytokine signaling. Blocking their action modifies multiple neural "housekeeping" functions. For example, blocking IL-1 or TNFalpha by several independent means alters regulation of sleep. This indicates that these cytokines likely modulate in the brain behavior of a normal organism. In addition, these cytokines are likely involved in synaptic plasticity, neural transmission, and Ca2+ signaling. Thus, the evidence strongly suggests that these cytokines perform neural functions in normal brain. We therefore propose that they should be thought of as neuromodulators in addition to inflammatory mediators.
Collapse
Affiliation(s)
- L Vitkovic
- CNRS-INSERM Centre de Pharmacologie-Endocrinologie, Montpellier, France
| | | | | |
Collapse
|
32
|
Ignatowski TA, Covey WC, Knight PR, Severin CM, Nickola TJ, Spengler RN. Brain-derived TNFalpha mediates neuropathic pain. Brain Res 1999; 841:70-7. [PMID: 10546989 DOI: 10.1016/s0006-8993(99)01782-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Neuropathic pain is a chronic pain state that develops a central component following acute nerve injury. However, the pathogenic mechanisms involved in the expression of this central component are not completely understood. We have investigated the role of brain-associated TNF in the evolution of hyperalgesia in the chronic constriction injury (CCI) model of neuropathic pain. Thermal nociceptive threshold has been assessed in rats (male, Sprague-Dawley) that have undergone loose, chromic gut ligature placement around the sciatic nerve. Total levels of TNF in regions of the brain, spinal cord and plasma have been assayed (WEHI-13VAR bioassay). Bioactive TNF levels are elevated in the hippocampus. During the period of injury, hippocampal noradrenergic neurotransmission demonstrates a decrease in stimulated norepinephrine (NE) release, concomitant with elevated hippocampal TNF levels. Continuous intracerebroventricular (i.c.v.) microinfusion of TNF-antibodies (Abs) starting at four days, but not six days, following ligature placement completely abolishes the hyperalgesic response characteristic of this model, as assessed by the 58 degrees C hot-plate test. Antibody infusion does not decrease spinal cord or plasma levels of TNF. Continuous i.c.v. microinfusion of rrTNF alpha exacerbates the hyperalgesic response by ligatured animals, and induces a hyperalgesic response in animals not receiving ligatures. Likewise, field-stimulated hippocampal adrenergic neurotransmission is decreased upon continuous i.c.v. microinfusion of TNF. These results indicate an important role of brain-derived TNF, both in the pathology of neuropathic pain, as well as in fundamental pain perception.
Collapse
Affiliation(s)
- T A Ignatowski
- Department of Pathology, State University of New York at Buffalo, 14214, USA
| | | | | | | | | | | |
Collapse
|
33
|
Broug-Holub E, Persoons JH, Schornagel K, Mastbergen SC, Kraal G. Effects of stress on alveolar macrophages: a role for the sympathetic nervous system. Am J Respir Cell Mol Biol 1998; 19:842-8. [PMID: 9806750 DOI: 10.1165/ajrcmb.19.5.3103] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Alveolar macrophages (AMs) play an important role in the regulation of the local immune reactivity in the lung. It was previously shown that exposure of rats to mild inescapable electrical footshock stress (20 min, 4 shocks/min, 5 s/shock, 0.8 mAmp) leads to apparent changes in the activity of AMs upon stimulation, reflected by an enhanced interleukin-1beta and tumor necrosis factor-alpha secretion and decreased nitric oxide secretion compared with the secretion by AMs isolated from nonstressed rats. Here we show that in vivo blockade of the autonomic nervous system by intraperitoneal injection of the nicotinic receptor antagonist chlorisondamine leads to complete abrogation of these stress-induced alterations in AM activity. This role for the autonomic nervous system could further be attributed to sympathetic stimulation of beta-adrenergic receptors as shown by blockade of beta-adrenoceptors. Blockade of either alpha-adrenoceptors or parasympathetic output did not result in abrogation of the stress-induced changes in AM activity. The beta-adrenergic modulation of AM activity most likely is not due to a direct effect of catecholamines on AMs because mimicking the in vivo stress effects by in vitro preincubation of AMs with various doses of catecholamines followed by lipopolysaccharide stimulation did not result in an altered cytokine secretion by AMs.
Collapse
Affiliation(s)
- E Broug-Holub
- Department of Cell Biology and Immunology and Department of Pharmacology, Faculty of Medicine, Vrije Universiteit, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
34
|
Mao L, Abdel-Rahman AA. Ethanol Counteraction of Clonidine-Evoked Inhibition of Norepinephrine Release in Rostral Ventrolateral Medulla of Rats. Alcohol Clin Exp Res 1998. [DOI: 10.1111/j.1530-0277.1998.tb03910.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
35
|
Ignatowski TA, Noble BK, Wright JR, Gorfien JL, Heffner RR, Spengler RN. Neuronal-associated tumor necrosis factor (TNF alpha): its role in noradrenergic functioning and modification of its expression following antidepressant drug administration. J Neuroimmunol 1997; 79:84-90. [PMID: 9357451 DOI: 10.1016/s0165-5728(97)00107-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Tumor necrosis factor-alpha (TNF alpha) and the alpha 2-adrenergic agonist clonidine regulate norepinephrine (NE) release from noradrenergic nerve terminals in the central nervous system (CNS). In the present study, superfusion and electrical field stimulation were applied to a series of rat hippocampal brain slices in order to investigate the regulation of [3H]-NE release. NE release had been previously determined to be decreased by TNF alpha in a concentration-dependent manner, an effect which was potentiated by the alpha 2-adrenergic antagonist idazoxan. Presently, we demonstrate that similar to alpha 2-adrenergic activation, TNF alpha regulation of NE release in a region of the brain rich in noradrenergic nerve terminals, is dependent upon the frequency of electrical stimulation applied to the hippocampal slice. Furthermore, immunoperoxidase staining has verified our previous findings of constitutive TNF alpha protein in the rat brain. Staining for TNF alpha appears to be largely localized to neurons and neuronal processes, further substantiating the proposal that TNF alpha is either synthesized de novo or is accumulated in and released by neurons. After administration of the tricyclic antidepressant desipramine, tissue sections obtained from the rat hippocampus and locus coeruleus are devoid of neuronal-associated TNF alpha immunoreactivity. TNF alpha localization in neurons and its modification of NE release comparable to alpha 2-adrenergic receptor activation, explains a functional role for the cytokine as a neuromodulator in the CNS.
Collapse
Affiliation(s)
- T A Ignatowski
- Department of Pathology, School of Medicine and Biomedical Sciences, SUNY at Buffalo 14214, USA
| | | | | | | | | | | |
Collapse
|