1
|
Cha HY, Ahn SH, Cheon JH, Park SY, Kim K. Hataedock treatment has preventive therapeutic effects for atopic dermatitis through skin barrier protection in Dermatophagoides farinae-induced NC/Nga mice. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:327-336. [PMID: 28583294 DOI: 10.1016/j.jep.2017.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hataedock treatment is traditionally used for the purpose of preventing the future skin disease by feeding herbal extracts to the newborn in traditional Chinese and Korean medicine. AIM OF THE STUDY This study investigated the preventive therapeutic effects of Hataedock (HTD) treatment for atopic dermatitis (AD) through skin barrier protection in Dermatophagoides farinae-induced NC/Nga mice. MATERIALS AND METHODS To the HTD treatment group, the extract of Coptis japonica Makino and Glycyrrhiza uralensis Fischer, which analyzed with High Performance Liquid Chromatography (HPLC)-fingerprint for quality consistency, was administered orally to the 3-week-old mice before inducing AD. After that, Dermatophagoides farinae was applied except the control group to induce AD-like skin lesions. We confirmed the effects of HTD on morphological changes, protection of skin barrier, regulation of Th2 differentiation, inflammation regulation and induction of apoptosis through histochemistry, immunohistochemistry, and Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. RESULTS HTD effectively reduced edema, angiogenesis and skin lesion. HTD also increased the levels of liver X receptor (LXR) and filaggrin but decreased the level of protein kinase C (PKC) (p<0.01). The levels of interleukin-4 (IL-4), IL-13, signal transducer and activator of transcription-6 (STAT-6) and Cluster of differentiation 40 (CD40) were significantly reduced in the HTD treated group (p<0.01). HTD also suppressed the mast cell degranulation and the level of the high-affinity IgE receptor (FcɛRI), substance P, Matrix metalloproteinases-9 (MMP-9) and 5-hydroxytryptamine (5-HT) (p<0.01). The levels of inflammatory factors such as nuclear factor-kappaB (NF-κB) p65, phosphorylated IκB (p-IκB) and inducible nitric oxide synthase (iNOS) were also decreased (p<0.01). Apoptosis of inflammatory cells was also found to increase (p<0.01). CONCLUSION Our results indicate that HTD effectively regulate the Th2 differentiation, mast cell activation and various inflammatory responses on AD-induced mice through protection of skin barrier. Therefore, HTD may have potential applications for alternative and preventive treatment in the management of AD.
Collapse
Affiliation(s)
- Ho-Yeol Cha
- Department of Korean Pediatrics, Hospital of Korean Medicine, Pusan National University, Geumo-ro 20, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 50612, Republic of Korea; Department of Korean Pediatrics, School of Korean Medicine, Pusan National University, Pusandaehak-ro 49, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 50612, Republic of Korea.
| | - Sang-Hyun Ahn
- Department of Anatomy, College of Korean Medicine, Semyung University, Semyung-ro 65, Jecheon-si, Chungbuk 27136, Republic of Korea.
| | - Jin-Hong Cheon
- Department of Korean Pediatrics, Hospital of Korean Medicine, Pusan National University, Geumo-ro 20, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 50612, Republic of Korea; Department of Korean Pediatrics, School of Korean Medicine, Pusan National University, Pusandaehak-ro 49, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 50612, Republic of Korea.
| | - Sun-Young Park
- Department of Physiology, College of Korean Medicine, Semyung University, Semyung-ro 65, Jecheon-si, Chungbuk 27136, Republic of Korea.
| | - Kibong Kim
- Department of Korean Pediatrics, Hospital of Korean Medicine, Pusan National University, Geumo-ro 20, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 50612, Republic of Korea; Department of Korean Pediatrics, School of Korean Medicine, Pusan National University, Pusandaehak-ro 49, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 50612, Republic of Korea.
| |
Collapse
|
2
|
Yuan LB, Hua CY, Gao S, Yin YL, Dai M, Meng HY, Li PP, Yang ZX, Hu QH. Astragalus Polysaccharides Attenuate Monocrotaline-Induced Pulmonary Arterial Hypertension in Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:773-789. [PMID: 28521513 DOI: 10.1142/s0192415x17500410] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Astragalus polysaccharides (APS) have been shown to possess a variety of biological activities including anti-oxidant and anti-inflammation functions in a number of diseases. However, their function in pulmonary arterial hypertension (PAH) is still unknown. Rats received APS (200[Formula: see text]mg/kg once two days) for 2 weeks after being injected with monocrotaline (MCT; 60[Formula: see text]mg/kg). The pulmonary hemodynamic index, right ventricular hypertrophy, and lung morphological features of the rat models were examined, as well as the NO/eNOS ratio of wet lung and dry lung weight and MPO. A qRT-PCR and p-I[Formula: see text]B was used to assess IL-1[Formula: see text], IL-6 and TNF-[Formula: see text] and WB was used to detect the total I[Formula: see text]B. Based on these measurements, it was found that APS reversed the MCT-induced increase in mean pulmonary arterial pressure (mPAP) (from 32.731[Formula: see text]mmHg to 26.707[Formula: see text]mmHg), decreased pulmonary vascular resistance (PVR) (from 289.021[Formula: see text]mmHg[Formula: see text][Formula: see text] min/L to 246.351[Formula: see text]mmHg[Formula: see text][Formula: see text][Formula: see text]min/L), and reduced right ventricular hypertrophy (from 289.021[Formula: see text]mmHg[Formula: see text][Formula: see text][Formula: see text]min/L to 246.351 mmHg[Formula: see text][Formula: see text][Formula: see text]min/L) ([Formula: see text]0.05). In terms of pulmonary artery remodeling, the WT% and WA% decreased with the addition of APS. In addition, it was found that APS promoted the synthesis of eNOS and the secretion of NO, promoting vasodilation and APS decreased the MCT-induced elevation of MPO, IL-1[Formula: see text], IL-6 and TNF-[Formula: see text], reducing inflammation. Furthermore, APS was able to inhibit the activation of pho-I[Formula: see text]B[Formula: see text]. In couclusion, APS ameliorates MCT-induced pulmonary artery hypertension by inhibiting pulmonary arterial remodeling partially via eNOS/NO and NF-[Formula: see text]B signaling pathways.
Collapse
Affiliation(s)
- Lin-Bo Yuan
- * Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, P. R. China.,† Key Laboratory of Pulmonary Diseases of Ministry of Health, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, P. R. China.,‡ Department of Physiology, School of Basic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China.,§ Key Laboratory of Heart Failure, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Chun-Yan Hua
- ‡ Department of Physiology, School of Basic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China.,§ Key Laboratory of Heart Failure, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Sheng Gao
- ¶ Animal Center Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Ya-Ling Yin
- †† Department of Physiology, Basic Medical College, Xinxiang Medical College, Xinxiang, Henan, P. R. China
| | - Mao Dai
- * Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, P. R. China.,† Key Laboratory of Pulmonary Diseases of Ministry of Health, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, P. R. China
| | - Han-Yan Meng
- § Key Laboratory of Heart Failure, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China.,∥ 1st Clinical College, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Piao-Piao Li
- § Key Laboratory of Heart Failure, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China.,** Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Zhong-Xin Yang
- § Key Laboratory of Heart Failure, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China.,** Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Qing-Hua Hu
- * Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, P. R. China.,† Key Laboratory of Pulmonary Diseases of Ministry of Health, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, P. R. China
| |
Collapse
|
3
|
Astragalus polysaccharide attenuates lipopolysaccharide-induced inflammatory responses in microglial cells: regulation of protein kinase B and nuclear factor-κB signaling. Inflamm Res 2015; 64:205-12. [DOI: 10.1007/s00011-015-0798-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 01/28/2015] [Accepted: 02/03/2015] [Indexed: 01/13/2023] Open
|
4
|
Mamik MK, Banerjee S, Walseth TF, Hirte R, Tang L, Borgmann K, Ghorpade A. HIV-1 and IL-1β regulate astrocytic CD38 through mitogen-activated protein kinases and nuclear factor-κB signaling mechanisms. J Neuroinflammation 2011; 8:145. [PMID: 22027397 PMCID: PMC3247131 DOI: 10.1186/1742-2094-8-145] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 10/25/2011] [Indexed: 01/06/2023] Open
Abstract
Background Infection with human immunodeficiency virus type-1 (HIV)-1 leads to some form of HIV-1-associated neurocognitive disorders (HAND) in approximately half of the cases. The mechanisms by which astrocytes contribute to HIV-1-associated dementia (HAD), the most severe form of HAND, still remain unresolved. HIV-1-encephalitis (HIVE), a pathological correlate of HAD, affects an estimated 9-11% of the HIV-1-infected population. Our laboratory has previously demonstrated that HIVE brain tissues show significant upregulation of CD38, an enzyme involved in calcium signaling, in astrocytes. We also reported an increase in CD38 expression in interleukin (IL)-1β-activated astrocytes. In the present investigation, we studied regulatory mechanisms of CD38 gene expression in astrocytes activated with HIV-1-relevant stimuli. We also investigated the role of mitogen-activated protein kinases (MAPKs) and nuclear factor (NF)-κB in astrocyte CD38 regulation. Methods Cultured human astrocytes were transfected with HIV-1YU-2 proviral clone and levels of CD38 mRNA and protein were measured by real-time PCR gene expression assay, western blot analysis and immunostaining. Astrocyte activation by viral transfection was determined by analyzing proinflammatory chemokine levels using ELISA. To evaluate the roles of MAPKs and NF-κB in CD38 regulation, astrocytes were treated with MAPK inhibitors (SB203580, SP600125, U0126), NF-κB interfering peptide (SN50) or transfected with dominant negative IκBα mutant (IκBαM) prior to IL-1β activation. CD38 gene expression and CD38 ADP-ribosyl cyclase activity assays were performed to analyze alterations in CD38 levels and function, respectively. Results HIV-1YU-2-transfection significantly increased CD38 mRNA and protein expression in astrocytes (p < 0.01) in a dose-dependent manner and induced astrocyte activation. IL-β-activation of HIV-1YU-2-transfected astrocytes significantly increased HIV-1 gene expression (p < 0.001). Treatment with MAPK inhibitors or NF-κB inhibitor SN50 abrogated IL-1β-induced CD38 expression and activity in astrocytes without altering basal CD38 levels (p < 0.001). IκBαM transfection also significantly inhibited IL-1β-mediated increases in CD38 expression and activity in astrocytes (p < 0.001). Conclusion The present findings demonstrate a direct involvement of HIV-1 and virus-induced proinflammatory stimuli in regulating astrocyte-CD38 levels. HIV-1YU-2-transfection effectively induced HIV-1p24 protein expression and activated astrocytes to upregulate CCL2, CXCL8 and CD38. In astrocytes, IL-1β-induced increases in CD38 levels were regulated through the MAPK signaling pathway and by the transcription factor NF-κB. Future studies may be directed towards understanding the role of CD38 in response to infection and thus its role in HAND.
Collapse
Affiliation(s)
- Manmeet K Mamik
- Department of Cell Biology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | | | | | | | | | | | | |
Collapse
|
5
|
Russo I, Amornphimoltham P, Weigert R, Barlati S, Bosetti F. Cyclooxygenase-1 is involved in the inhibition of hippocampal neurogenesis after lipopolysaccharide-induced neuroinflammation. Cell Cycle 2011; 10:2568-73. [PMID: 21694498 DOI: 10.4161/cc.10.15.15946] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Growing evidence indicates that neuroinflammation can alter adult neurogenesis by mechanisms as yet unclear. We have previously demonstrated that the neuroinflammatory response and neuronal damage after lipopolysaccharide (LPS) injection is reduced in cyclooxygenase-1 deficient (COX-1(-/-)) mice. In this study, we investigated the role of COX-1 on hippocampal neurogenesis during LPS-induced neuroinflammation, using COX-1(-/-) and wild type (WT) mice. We found that LPS-induced neuroinflammation resulted in the decrease of proliferation, survival and differentiation of hippocampal progenitor cells in WT but not in COX-1(-/-) mice. Thus, we demonstrate for the first time that COX-1 is involved in the inhibition of BrdU progenitor cells in proliferation and hippocampal neurogenesis after LPS. These results suggest that COX-1 may represent a viable therapeutic target to reduce neuroinflammation and promote neurogenesis in neurodegenerative diseases with a strong inflammatory component.
Collapse
Affiliation(s)
- Isabella Russo
- Molecular Neuroscience Unit, Brain Physiology and Metabolism Section, National Institute on Aging, NIH, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
6
|
Xu S, Zhi H, Hou X, Cohen RA, Jiang B. IκBβ attenuates angiotensin II-induced cardiovascular inflammation and fibrosis in mice. Hypertension 2011; 58:310-6. [PMID: 21646597 DOI: 10.1161/hypertensionaha.111.172031] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The development of cardiovascular fibrosis is associated with chronic inflammation, where activation of nuclear factor κB (NF-κB) signaling may play a critical role. NF-κB activation is tightly regulated by the cellular inhibitor of κB (IκB) family of proteins, such as IκBα and IκBβ. IκBα and IκBβ display different regulation kinetics in response to inflammatory stimulation. The present study tested the hypothesis that IκBα and IκBβ may have different roles in modulating cardiovascular inflammation and fibrosis, using a model of angiotensin II infusion-induced hypertension in wild-type mice and IκBβ knock-in mice, in which the IκBα gene is replaced by IκBβ cDNA (AKBI). In WT mice, subcutaneous angiotensin II infusion for 7 days induced increased perivascular and interstitial collagen deposition and fibrotic lesions, associated with myocardial interstitial hemosiderin accumulation and extensive macrophage infiltration. These effects of angiotensin II were dramatically limited in AKBI mice. Replacement of IκBα with IκBβ significantly attenuated angiotensin II infusion-induced expression of interleukin 1β, interleukin 6, monocyte chemotactic protein 1, collagen I and III, fibronectin, and tissue inhibitor of metalloproteinase 1 in the hearts. Furthermore, using cultured vascular smooth muscle cells, we demonstrated that interleukin 1β-induced NF-κB activation and monocyte chemotactic protein 1, vascular cell adhesion molecule 1, and tissue inhibitor of metalloproteinase 1 expressions were suppressed in the AKBI cells because of the replacement of IκBα with IκBβ. These results indicate that NF-κB has an essential role in mediating the cardiovascular inflammatory response to angiotensin II and suggest that targeting the balance of IκBα and IκBβ expression might be a novel therapeutic modality in preventing fibrosis in hypertensive cardiovascular disease.
Collapse
Affiliation(s)
- Shanqin Xu
- 77 Ave Louis Pasteur, NRB 431, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
7
|
Abstract
NF-κB (nuclear factor κB) controls diverse cellular processes and is frequently misregulated in chronic immune diseases or cancer. The activity of NF-κB is regulated by IκB (inhibitory κB) proteins which control nuclear-cytoplasmic shuttling and DNA binding of NF-κB. In the present paper, we describe a novel role for p65 as a critical regulator of the cellular localization and functions of NF-κB and its inhibitor IκBβ. In genetically modified p65-/- cells, the localization of ectopic p65 is not solely regulated by IκBα, but is largely dependent on the NLS (nuclear localization signal) and the NES (nuclear export signal) of p65. Furthermore, unlike IκBα, IκBβ does not contribute to the nuclear export of p65. In fact, the cellular localization and degradation of IκBβ is controlled by the p65-specific NLS and NES. The results of our present study also reveal that, in addition to stimulus-induced redistribution of NF-κB, changes in the constitutive localization of p65 and IκBβ specifically modulate activation of inflammatory genes. This is a consequence of differences in the DNA-binding activity and signal responsiveness between the nuclear and cytoplasmic NF-κB-IκBβ complexes. Taken together, the findings of the present study indicate that the p65 subunit controls transcriptional competence of NF-κB by regulating the NF-κB/IκBβ pathway.
Collapse
|
8
|
Atzei P, Gargan S, Curran N, Moynagh PN. Cactin targets the MHC class III protein IkappaB-like (IkappaBL) and inhibits NF-kappaB and interferon-regulatory factor signaling pathways. J Biol Chem 2010; 285:36804-17. [PMID: 20829348 PMCID: PMC2978609 DOI: 10.1074/jbc.m110.139113] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 08/10/2010] [Indexed: 11/06/2022] Open
Abstract
Toll-like receptors (TLRs) act as primary sensors of the immune system by recognizing specific microbial motifs and inducing proinflammatory genes that facilitate innate and adaptive immunity. TLRs regulate gene expression by activating transcription factors, such as NF-κB and interferon-regulatory factors. Dysregulation of these pathways can lead to inflammatory diseases, and thus they are subject to stringent control by negative regulators of innate immune signaling. Cactin (Cactus interactor) was initially discovered as a novel interactor of Drosophila Cactus, a regulator of Drosophila Toll signaling. We now describe the first functional characterization of the human ortholog of Cactin (hCactin) and show that it acts as a negative regulator of TLRs. Overexpression of hCactin suppresses TLR-induced activation of NF-κB and interferon-regulatory factor transcription factors and induction of TLR-responsive genes, whereas knockdown of endogenous hCactin augments TLR induction of these responses. hCactin also interacts with IκB-like protein and targets other proteins that are encoded by genes in the MHC Class III region of chromosome 6. We demonstrate that hCactin localizes to the nucleus, and this nuclear localization is critical for manifesting its inhibitory effects on TLR signaling. This study thus defines hCactin as a novel negative regulator of TLR signaling and reveals its capacity to target MHC Class III genes at the molecular and functional level.
Collapse
Affiliation(s)
- Paola Atzei
- From the Institute of Immunology, National University of Ireland, Maynooth, County Kildare 1, Ireland
| | - Siobhan Gargan
- From the Institute of Immunology, National University of Ireland, Maynooth, County Kildare 1, Ireland
| | - Niamh Curran
- From the Institute of Immunology, National University of Ireland, Maynooth, County Kildare 1, Ireland
| | - Paul N. Moynagh
- From the Institute of Immunology, National University of Ireland, Maynooth, County Kildare 1, Ireland
| |
Collapse
|
9
|
Scheibel M, Klein B, Merkle H, Schulz M, Fritsch R, Greten FR, Arkan MC, Schneider G, Schmid RM. IkappaBbeta is an essential co-activator for LPS-induced IL-1beta transcription in vivo. ACTA ACUST UNITED AC 2010; 207:2621-30. [PMID: 20975042 PMCID: PMC2989768 DOI: 10.1084/jem.20100864] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IkBβ forms a complex with the NF-κB subunits RelA and c-Rel that inhibits the transcription of IL-1β and other genes. Mice lacking IkBβ are protected against LPS-induced shock. Inhibitor of κB (IκB) β (IκBβ) represents one of the major primary regulators of NF-κB in mammals. In contrast to the defined regulatory interplay between NF-κB and IκBα, much less is known about the biological function of IκBβ. To elucidate the physiological role of IκBβ in NF-κB signaling in vivo, we generated IκBβ-deficient mice. These animals proved to be highly refractory to LPS-induced lethality, accompanied by a strong reduction in sepsis-associated cytokine production. In response to LPS, IκBβ is recruited to the IL-1β promoter forming a complex with the NF-κB subunits RelA/c-Rel required for IL-1β transcription. Further transcriptome analysis of LPS-stimulated wild-type and IκBβ-deficient BM-derived macrophages revealed several other genes with known regulatory functions in innate immunity arguing that a subset of NF-κB target genes is under control of IκBβ. Collectively, these findings provide an essential proinflammatory role for IκBβ in vivo, and establish a critical function for IκBβ as a transcriptional coactivator under inflammatory conditions.
Collapse
Affiliation(s)
- Melanie Scheibel
- II. Medizinische Klinik, Technische Universität München, 81675 München, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Nuclear factor κB (NF-κB) is an inducible transcription factor that tightly regulates the expression of a large cohort of genes. As a key component of the cellular machinery NF-κB is involved in a wide range of biological processes including innate and adaptive immunity, inflammation, cellular stress responses, cell adhesion, apoptosis and proliferation. Appropriate regulation of NF-κB is critical for the proper function and survival of the cell. Aberrant NF-κB activity has now been implicated in the pathogenesis of several diseases ranging from inflammatory bowel disease to autoimmune conditions such as rheumatoid arthritis. Systems governing NF-κB activity are complex and there is an increased understanding of the importance of nuclear events in regulating NF-κB's activities as a transcription factor. A number of novel nuclear regulators of NF-κB such as IκB-ζ and PDZ and LIM domain 2 (PDLIM2) have now been identified, adding another layer to the mechanics of NF-κB regulation. Further insight into the functions of these molecules raises the prospect for better understanding and rational design of therapeutics for several important diseases.
Collapse
Affiliation(s)
- Arun K Mankan
- Department of Clinical Medicine and Institute of Molecular Medicine, Trinity College, Dublin, Ireland.
| | | | | | | | | |
Collapse
|
11
|
de Souza DF, Leite MC, Quincozes-Santos A, Nardin P, Tortorelli LS, Rigo MM, Gottfried C, Leal RB, Gonçalves CA. S100B secretion is stimulated by IL-1beta in glial cultures and hippocampal slices of rats: Likely involvement of MAPK pathway. J Neuroimmunol 2008; 206:52-7. [PMID: 19042033 DOI: 10.1016/j.jneuroim.2008.10.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 10/29/2008] [Accepted: 10/29/2008] [Indexed: 12/11/2022]
Abstract
S100B is an astrocyte-derived cytokine implicated in the IL-1beta-triggered cytokine cycle in Alzheimer's disease. However, the secretion of S100B following stimulation by IL-1beta has not been directly demonstrated. We investigated S100B secretion in cortical primary astrocyte cultures, C6 glioma cells and acute hippocampal slices exposed to IL-1beta. S100B secretion was induced by IL-1beta in all preparations, involving MAPK pathway and, apparently, NF-small ka, CyrillicB signaling. Astrocytes and C6 cells exhibited different sensitivities to IL-1beta. These results suggest that IL-1beta-induced S100B secretion is a component of the neuroinflammatory response, which would support the involvement of S100B in the genesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Daniela F de Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Roach JD, Aguinaldo GT, Jonnalagadda K, Hughes FM, Spangelo BL. Gamma-aminobutyric acid inhibits synergistic interleukin-6 release but not transcriptional activation in astrocytoma cells. Neuroimmunomodulation 2008; 15:117-24. [PMID: 18679050 PMCID: PMC2859952 DOI: 10.1159/000148194] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 03/19/2008] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE A decline in the inhibitory neurotransmitter gamma-aminobutyric acid (GABA) may enhance cytokine release in Alzheimer's disease (AD) resulting in neuroinflammation. We investigated the GABA-mediated suppression of the synergistic release of interleukin (IL)-6 due to interleukin 1-beta (IL-1 beta) and tumor necrosis factor-alpha (TNF-alpha). METHODS Rat C6 astrocytoma cells were treated with IL-1 beta and TNF-alpha in the absence and presence of GABA. Activation of p38, degradation of I kappaB-alpha and total cellular IL-6 were determined by Western blot analysis. IL-6 release and gene expression were measured by ELISA and RT-PCR, respectively. RESULTS Although p38 and nuclear factor (NF)-kappaB are essential for the synergistic release of IL-6, GABA did not affect either p38 phosphorylation or I kappaB-alpha degradation. Additionally, GABA suppressed IL-6 release but did not alter cytokine-driven synergistic increases in IL-6 gene expression. Western blot analysis revealed that co-treatments with IL-1 beta and TNF-alpha resulted in an increase in intracellular IL-6 that was prevented by GABA. CONCLUSION GABA-induced inhibition of IL-6 release appears to coincide with a reduction in cellular IL-6. The GABA-induced suppression of IL-6 release may include inhibition of IL-6 gene translation.
Collapse
Affiliation(s)
- Joseph D. Roach
- Department of Chemistry, University of Nevada Las Vegas, Las Vegas, Nevada, 89154
| | - Grant T. Aguinaldo
- Department of Chemistry, University of Nevada Las Vegas, Las Vegas, Nevada, 89154
| | - Kaumudi Jonnalagadda
- Department of Biology University of North Carolina Charlotte, Charlotte, North Carolina, 28223
| | - Francis M. Hughes
- Department of Biology University of North Carolina Charlotte, Charlotte, North Carolina, 28223
| | - Bryan L. Spangelo
- Department of Chemistry, University of Nevada Las Vegas, Las Vegas, Nevada, 89154
- To whom correspondence and reprint requests should be addressed at: Department of Chemistry, University of Nevada Las Vegas, 4505 Maryland Parkway, Las Vegas, NV 89154-4003, 702.895.3797 telephone; 702.895.3171 telefax,
| |
Collapse
|
13
|
Abstract
BACKGROUND High-grade glioma remains one of the most difficult cancers to treat. Recent studies in oncology have identified a role of the ubiquitous enzyme, cyclooxygenase (Cox), especially cyclooxygenase-2 (COX-2) in cell proliferation, and its inhibition in cancer control, apoptosis, as well as synergy with other forms of therapy. The inhibitors of the Cox enzyme are well known as members of the nonsteroidal anti-inflammatory drug (NSAID) class of pharmaceuticals. METHODS In vitro and in vivo studies of different cancers expressing COX-2, including glioma studies, along with the few clinical trials that have been reported are reviewed to specifically identify the actions of these agents. RESULTS The anticancer effect of the COX-2 inhibitors may occur irrelevant of COX-2 expression, and it appears to be drug-specific, as well as dose-specific in different cancers. In combination with chemotherapeutic agents, the COX-2 inhibitors may have an additive, synergistic, or inhibitory effect on tumor growth. CONCLUSIONS As evaluations of this class of drugs begin in glioma, in vitro and in vivo data should be acquired to accurately predict which compounds will have an effect in controlling tumor growth and at which doses these should be used. The actual expression and inhibition of COX-2 may not always be relevant to the effects on tumor growth.
Collapse
Affiliation(s)
- Pamela New
- Department of Neurosurgery/Neuro-oncology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
14
|
García-Román R, Pérez-Carreón JI, Márquez-Quiñones A, Salcido-Neyoy ME, Villa-Treviño S. Persistent activation of NF-kappaB related to IkappaB's degradation profiles during early chemical hepatocarcinogenesis. J Carcinog 2007; 6:5. [PMID: 17445259 PMCID: PMC1865534 DOI: 10.1186/1477-3163-6-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2006] [Accepted: 04/19/2007] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND To define the NF-kappaB activation in early stages of hepatocarcinogenesis and its IkappaB's degradation profiles in comparison to sole liver regeneration. METHODS Western-blot and EMSA analyses were performed for the NF-kappaB activation. The transcriptional activity of NF-kappaB was determined by RT-PCR of the IkappaB-alpha mRNA. The IkappaB's degradation proteins were determined by Western-blot assay. RESULTS We demonstrated the persistent activation of NF-kappaB during early stages of hepatocarcinogenesis, which reached maximal level 30 min after partial hepatectomy. The DNA binding and transcriptional activity of NF-kappaB, were sustained during early steps of hepatocarcinogenesis in comparison to only partial hepatectomy, which displayed a transitory NF-kappaB activation. In early stages of hepatocarcinogenesis, the IkappaB-alpha degradation turned out to be acute and transitory, but the low levels of IkappaB-beta persisted even 15 days after partial hepatectomy. Interestingly, IkappaB-beta degradation is not induced after sole partial hepatectomy. CONCLUSION We propose that during liver regeneration, the transitory stimulation of the transcription factor response, assures blockade of NF-kappaB until recovery of the total mass of the liver and the persistent NF-kappaB activation in early hepatocarcinogenesis may be due to IkappaB-beta and IkappaB-alpha degradation, mainly IkappaB-beta degradation, which contributes to gene transcription related to proliferation required for neoplastic progression.
Collapse
Affiliation(s)
- Rebeca García-Román
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, México D.F., México
| | - Julio Isael Pérez-Carreón
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, México D.F., México
| | - Adriana Márquez-Quiñones
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, México D.F., México
| | - Martha Estela Salcido-Neyoy
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, México D.F., México
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, México D.F., México
| |
Collapse
|
15
|
Griffin B, Moynagh P. In vivo binding of NF-kappaB to the IkappaBbeta promoter is insufficient for transcriptional activation. Biochem J 2006; 400:115-25. [PMID: 16792530 PMCID: PMC1635438 DOI: 10.1042/bj20060786] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Despite certain structural and biochemical similarities, differences exist in the function of the NF-kappaB (nuclear factor kappaB) inhibitory proteins IkappaBalpha (inhibitory kappaBalpha) and IkappaBbeta. The functional disparity arises in part from variance at the level of gene regulation, and in particular from the substantial induction of IkappaBalpha, but not IkappaBbeta, gene expression post-NF-kappaB activation. In the present study, we probe the differential effects of IL (interleukin)-1beta on induction of IkappaBalpha and perform the first characterization of the human IkappaBbeta promoter. A consensus NF-kappaB-binding site, capable of binding NF-kappaB both in vitro and in vivo, is found in the IkappaBbeta gene 5' flanking region. However, the IkappaBbeta promoter was not substantially activated by pro-inflammatory cytokines, such as IL-1beta and tumour necrosis factor alpha, that are known to cause strong activation of NF-kappaB. Furthermore, in contrast with IkappaBalpha, NF-kappaB activation did not increase expression of endogenous IkappaBbeta as assessed by analysis of mRNA and protein levels. Unlike kappaB-responsive promoters, IkappaBbeta promoter-bound p65 inefficiently recruits RNA polymerase II, which stalls at the promoter. We present evidence that this stalling is likely due to the absence of transcription factor IIH engagement, a prerequisite for RNA polymerase II phosphorylation and transcriptional initiation. Differences in the conformation of promoter-bound NF-kappaB may underlie the variation in the ability to engage the basal transcriptional apparatus at the IkappaBbeta and kappaB-responsive promoters. This accounts for the differential expression of IkappaB family members in response to NF-kappaB activation and furthers our understanding of the mechanisms involved in transcription factor activity and IkappaBbeta gene regulation.
Collapse
Affiliation(s)
- Bryan D. Griffin
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Paul N. Moynagh
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- To whom correspondence should be addressed (email )
| |
Collapse
|
16
|
Mémet S. NF-kappaB functions in the nervous system: from development to disease. Biochem Pharmacol 2006; 72:1180-95. [PMID: 16997282 DOI: 10.1016/j.bcp.2006.09.003] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2006] [Revised: 08/31/2006] [Accepted: 09/05/2006] [Indexed: 01/01/2023]
Abstract
The transcription factor nuclear factor-kappaB (NF-kappaB) is an ubiquitously expressed dimeric molecule with post-translationally regulated activity. Its role in the immune system and host defense has been well characterized over the last two decades. In contrast, our understanding of the function of this transcription factor in the nervous system (NS) is only emerging. Given their cytoplasmic retention and nuclear translocation upon stimulus, NF-kappaB members are likely to exert an important role in transduction of signals from synaptic terminals to nucleus, to initiate transcriptional responses. This report describes recent findings deciphering the diverse functions of NF-kappaB in NS development and activity, which range from the control of cell growth, survival and inflammatory response to synaptic plasticity, behavior and cognition. Particular attention is given to the specific roles of NF-kappaB in the various cells of the NS, e.g. neurons and glia. Current knowledge of the contribution of NF-kappaB to several neurodegenerative disorders, such as Alzheimer's, Parkinson's and Huntington's diseases is also summarized.
Collapse
Affiliation(s)
- Sylvie Mémet
- Unité de Mycologie Moléculaire, FRE CNRS 2849, Department of Infection and Epidemiology, Institut Pasteur, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
17
|
Moynagh PN. The interleukin-1 signalling pathway in astrocytes: a key contributor to inflammation in the brain. J Anat 2006; 207:265-9. [PMID: 16185251 PMCID: PMC1571539 DOI: 10.1111/j.1469-7580.2005.00445.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A dysregulated inflammatory response in the central nervous system (CNS) lies at the heart of many neuropathological conditions such as multiple sclerosis and Alzheimer's disease. A key component of these inflammatory conditions is the accumulation of leukocytes in the CNS. The infiltration of leukocytes into the brain is dependent on the induction of leukocyte adhesion molecules and chemoattractant chemokines. Recent studies have suggested the astrocyte to be a key cell in mediating the inflammatory process in the brain and in expressing adhesion molecules and chemokines. Here I overview work in my laboratory and others that demonstrates interleukin-1 (IL-1) to be a key inducer of the expression of these molecules in astrocytes. The temporal expression is sustained in nature and this is due to prolonged activation of the transcription factor NFkappaB. The molecular basis to the sustained activation of NFkappaB is also discussed. The IL-1 signalling pathway thus emerges as a valuable therapeutic target in the treatment of presently incurable neuropathological conditions.
Collapse
Affiliation(s)
- Paul N Moynagh
- Department of Pharmacology, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland.
| |
Collapse
|
18
|
Griffin BD, Moynagh PN. Persistent interleukin-1beta signaling causes long term activation of NFkappaB in a promoter-specific manner in human glial cells. J Biol Chem 2006; 281:10316-26. [PMID: 16455661 DOI: 10.1074/jbc.m509973200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nuclear factor-kappaB (NFkappaB) is an inducible transcription factor that plays a key role in regulating the expression of a wide range of immune and inflammatory response genes. The activity of NFkappaB is controlled at multiple levels, including cytoplasmic retention with inhibitor of kappaB (IkappaB) proteins in the basal state. Persistent activation of the transcription factor is seen in numerous chronic inflammatory disease states, and we have previously demonstrated sustained activation of NFkappaB in human glial cells upon stimulation with interleukin (IL)-1beta. In these cells, NFkappaB retains DNA binding activity for up to 72 h despite the presence of resynthesized IkappaBalpha and in the absence of IkappaBbeta. Here we characterized the apparent inability of newly synthesized IkappaBalpha to terminate activation of NFkappaB in glial cells. We showed unexpectedly that newly synthesized IkappaBalpha can enter the nucleus, interact with the NFkappaB subunit p65, and export it to the cytoplasm. However, in vitro analysis of enzyme activity demonstrates that IL-1beta causes the long term activation of the IkappaB kinase complex leading to chronic phosphorylation of the newly synthesized IkappaBalpha signal response domain and persistent activation of NFkappaB. Such sustained activation of NFkappaB is dependent on the continuous presence and activity of IL-1beta. Interestingly, the sustained nature of NFkappaB activity is promoter type-specific. Chromatin immunoprecipitation studies revealed that p65 is detected at the promoters of both intercellular adhesion molecule-1 and IL-8 1 h following IL-1beta stimulation but is only found at the latter at 24 h. The functional significance of this finding is indicated by the transient induction of intercellular adhesion molecule-1 mRNA, but more sustained induction of IL-8 expression, by IL-1beta. These studies thus demonstrated that persistent IL-1 signaling causes sustained activation of NFkappaB in a promoter-specific manner in human glial cells, leading to prolonged induction of selective pro-inflammatory genes. This is likely to make a key contribution to chronic inflammatory conditions of the brain.
Collapse
Affiliation(s)
- Bryan D Griffin
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | |
Collapse
|
19
|
Widera D, Mikenberg I, Kaltschmidt B, Kaltschmidt C. Potential role of NF-kappaB in adult neural stem cells: the underrated steersman? Int J Dev Neurosci 2006; 24:91-102. [PMID: 16413989 DOI: 10.1016/j.ijdevneu.2005.11.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Accepted: 11/11/2005] [Indexed: 01/19/2023] Open
Abstract
Neural stem cells are precursors of neurons and glial cells. During brain development, these cells proliferate, migrate and differentiate into specific lineages. Recently neural stem cells within the adult central nervous system were identified. Informations are now emerging about regulation of stem cell proliferation, migration and differentiation by numerous soluble factors such as chemokines and cytokines. However, the signal transduction mechanisms downstream of these factors are less clear. Here, we review potential evidences for a novel central role of the transcription factor nuclear factor kappa B (NF-kappaB) in these crucial signal transduction processes. NF-kappaB is an inducible transcription factor detected in neurons, glia and neural stem cells. NF-kappaB was discovered by David Baltimore's laboratory as a transcription factor in lymphocytes. NF-kappaB is involved in many biological processes such as inflammation and innate immunity, development, apoptosis and anti-apoptosis. It has been recently shown that members of the NF-kappaB family are widely expressed by neurons, glia and neural stem cells. In the nervous system, NF-kappaB plays a crucial role in neuronal plasticity, learning, memory consolidation, neuroprotection and neurodegeneration. Recent data suggest an important role of NF-kappaB on proliferation, migration and differentiation of neural stem cells. NF-kappaB is composed of three subunits: two DNA-binding and one inhibitory subunit. Activation of NF-kappaB takes place in the cytoplasm and results in degradation of the inhibitory subunit, thus enabling the nuclear import of the DNA-binding subunits. Within the nucleus, several target genes could be activated. In this review, we suggest a model explaining the multiple action of NF-kappaB on neural stem cells. Furthermore, we discuss the potential role of NF-kappaB within the so-called brain cancer stem cells.
Collapse
Affiliation(s)
- Darius Widera
- Institut für Neurobiochemie, Universität Witten/Herdecke, Stockumer Str. 10, D-58448 Witten, Germany
| | | | | | | |
Collapse
|
20
|
Curran NM, Griffin BD, O'Toole D, Brady KJ, Fitzgerald SN, Moynagh PN. The Synthetic Cannabinoid R(+)WIN 55,212-2 Inhibits the Interleukin-1 Signaling Pathway in Human Astrocytes in a Cannabinoid Receptor-independent Manner. J Biol Chem 2005; 280:35797-806. [PMID: 16105834 DOI: 10.1074/jbc.m507959200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
R(+)WIN 55,212-2 is a synthetic cannabinoid that controls disease progression in models of multiple sclerosis. This is associated with its ability to reduce migration of leukocytes into the central nervous system. Because leukocyte migration is dependent on induction of adhesion molecules and chemokines by pro-inflammatory cytokines, we examined the effects of R(+)WIN 55,212-2 on their expression. Using 1321N1 astrocytoma and A-172 glioblastoma as cell models we show that R(+)WIN 55,212-2, but not its inactive chiral form S(-)WIN 55,212-2, strongly inhibits the interleukin-1 (IL-1) induction of the adhesion molecules intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) and the chemokine IL-8. This inhibition is not mediated via the CB1 or CB2 cannabinoid receptors, because their selective antagonists and pertussis toxin failed to affect the inhibitory effects of R(+)WIN 55,212-2. Furthermore reverse transcription-PCR analysis did not detect the expression of either receptor in 1321N1 cells. R(+)WIN 55,212-2 was shown to inhibit adhesion molecule and chemokine expression at the level of transcription, because it strongly inhibited the IL-1 induction of ICAM-1, VCAM-1, and IL-8 mRNAs and blocked the IL-1 activation of their promoters. The NFkappaB pathway was then assessed as a lead target for R(+)WIN 55,212-2. NFkappaB was measured by expression of a transfected NFkappaB-regulated reporter gene. Using this assay, R(+)WIN 55,212-2 strongly inhibited IL-1 activation of NFkappaB. Furthermore R(+)WIN 55,212-2 inhibited the ability of overexpressed Myd88, Tak-1, and IKK-2 to induce the reporter gene suggesting that R(+)WIN 55,212-2 acts at or downstream of IKK-2 in the IL-1 pathway. However R(+)WIN 55,212-2 failed to inhibit IL-1-induced degradation of IkappaBalpha, excluding IKK-2 as a direct target. In addition electrophoretic mobility shift and chromatin immunoprecipitation assays showed that R(+)WIN 55,212-2 does not regulate the IL-1-induced nuclear translocation of NFkappaB or the ability of the latter to bind to promoters regulating expression of ICAM-1 and IL-8. These data suggest that R(+)WIN 55,212-2 blocks IL-1 signaling by inhibiting the transactivation potential of NFkappaB.
Collapse
Affiliation(s)
- Niamh M Curran
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
21
|
Kaltschmidt B, Widera D, Kaltschmidt C. Signaling via NF-κB in the nervous system. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1745:287-99. [PMID: 15993497 DOI: 10.1016/j.bbamcr.2005.05.009] [Citation(s) in RCA: 224] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2005] [Revised: 05/20/2005] [Accepted: 05/23/2005] [Indexed: 12/12/2022]
Abstract
Nuclear factor kappa B (NF-kappaB) is an inducible transcription factor present in neurons and glia. Recent genetic models identified a role for NF-kappaB in neuroprotection against various neurotoxins. Furthermore, genetic evidence for a role in learning and memory is now emerging. This review highlights our current understanding of neuronal NF-kappaB in response to synaptic transmission and summarizes potential physiological functions of NF-kappaB in the nervous system. This article contains a listing of NF-kappaB activators and inhibitors in the nervous system, furthermore specific target genes are discussed. Synaptic NF-kappaB activated by glutamate and Ca2+ will be presented in the context of retrograde signaling. A controversial role of NF-kappaB in neurodegenerative diseases will be discussed. A model is proposed explaining this paradox as deregulated physiological NF-kappaB activity, where novel results are integrated, showing that p65 could be turned from an activator to a repressor of anti-apoptotic genes.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Institut für Neurobiochemie Universität Witten/Herdecke, Stockumer Street 10, D-58448 Witten, Germany
| | | | | |
Collapse
|
22
|
Hertlein E, Wang J, Ladner KJ, Bakkar N, Guttridge DC. RelA/p65 regulation of IkappaBbeta. Mol Cell Biol 2005; 25:4956-68. [PMID: 15923614 PMCID: PMC1140602 DOI: 10.1128/mcb.25.12.4956-4968.2005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Revised: 01/19/2005] [Accepted: 03/17/2005] [Indexed: 01/01/2023] Open
Abstract
IkappaB inhibitor proteins are the primary regulators of NF-kappaB. In contrast to the defined regulatory interplay between NF-kappaB and IkappaBalpha, much less is known regarding the regulation of IkappaBbeta by NF-kappaB. Here, we describe in detail the regulation of IkappaBbeta by RelA/p65. Using p65(-/-) fibroblasts, we show that IkappaBbeta is profoundly reduced in these cells, but not in other NF-kappaB subunit knockouts. This regulation prevails during embryonic and postnatal development in a tissue-specific manner. Significantly, in both p65(-/-) cells and tissues, IkappaBalpha is also reduced, but not nearly to the same extent as IkappaBbeta, thus highlighting the degree to which IkappaBbeta is dependent on p65. This dependence is based on the ability of p65 to stabilize IkappaBbeta protein from the 26S proteasome, a process mediated in large part through the p65 carboxyl terminus. Furthermore, IkappaBbeta was found to exist in both a basally phosphorylated and a hyperphosphorylated form. While the hyperphosphorylated form is less abundant, it is also more stable and less dependent on p65 and its carboxyl domain. Finally, we show that in p65(-/-) fibroblasts, expression of a proteolysis-resistant form of IkappaBbeta, but not IkappaBalpha, causes a severe growth defect associated with apoptosis. Based on these findings, we propose that tight control of IkappaBbeta protein by p65 is necessary for the maintenance of cellular homeostasis.
Collapse
Affiliation(s)
- Erin Hertlein
- Human Cancer Genetics Program, The Ohio State University, Columbus, OH, USA
| | | | | | | | | |
Collapse
|
23
|
Kiss DL, Xu W, Gopalan S, Buzanowska K, Wilczynska KM, Rydel RE, Kordula T. Duration of alpha 1-antichymotrypsin gene activation by interleukin-1 is determined by efficiency of inhibitor of nuclear factor kappa B alpha resynthesis in primary human astrocytes. J Neurochem 2005; 92:730-8. [PMID: 15686474 PMCID: PMC4558886 DOI: 10.1111/j.1471-4159.2004.02900.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Expression of alpha1antichymotrypsin (ACT) is significantly activated by interleukin-1 (IL-1) in human astrocytes; however, it is barely affected by IL-1 in hepatocytes. This tissue-specific regulation depends upon an enhancer that contains both nuclear factor kappaB (NF-kappaB) and activating protein 1 (AP-1) elements, and is also observed for an NF-kappaB reporter but not for an AP-1 reporter. We found efficient activation of NF-kappaB binding in both cell types; however, this binding was persistent in glial cells and only transient in hepatocytes. IL-1-activated NF-kappaB complexes consisted of p65 and p50, with p65 transiently phosphorylated on serine 536 in glial cells whereas more persistently in hepatic cells. Overexpression of p65 or constitutively active IKKbeta (inhibitor of NF-kappaB kinase beta) resulted in an efficient activation of the ACT reporter in hepatic cells, indicating that a specific mechanism exists in these cells terminating IL-1 signaling. IL-1 effectively induced the degradation of inhibitor of NF-kappaBalpha (IkBalpha) and IkBepsilon in both cell types but IkBbeta was not affected. However, IkBalpha was resynthesized much more rapidly in hepatic cells in comparison to glial cells. In addition, the initial levels of IkBalpha were much lower in glial cells. We propose that the tissue-specific regulation of the ACT gene expression by IL-1 is determined by different efficiencies of IkBalpha resynthesis in glial and hepatic cells.
Collapse
Affiliation(s)
- Daniel L. Kiss
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio 44115
| | - Weili Xu
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio 44115
- Department of Biochemistry, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Sunita Gopalan
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio 44115
- Department of Biochemistry, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Katarzyna Buzanowska
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio 44115
| | - Katarzyna M. Wilczynska
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio 44115
- Department of Biochemistry, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | | | - Tomasz Kordula
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio 44115
- Department of Biochemistry, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
- Corresponding author: Dr. Tomasz Kordula, Department of Biochemistry, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298, tel. (804) 828-0771, fax. (804) 828-1473,
| |
Collapse
|
24
|
Bai Y, Onuma H, Bai X, Medvedev AV, Misukonis M, Weinberg JB, Cao W, Robidoux J, Floering LM, Daniel KW, Collins S. Persistent nuclear factor-kappa B activation in Ucp2-/- mice leads to enhanced nitric oxide and inflammatory cytokine production. J Biol Chem 2005; 280:19062-9. [PMID: 15757894 PMCID: PMC1382174 DOI: 10.1074/jbc.m500566200] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
One of the phenotypes of mice with targeted disruption of the uncoupling protein-2 gene (Ucp2-/-) is greater macrophage phagocytic activity and free radical production, resulting in a striking resistance to infectious microorganisms. In this study, the molecular mechanisms of this enhanced immune response were investigated. We found that levels of nitric oxide measured in either plasma or isolated macrophages from Ucp2-/- mice are significantly elevated in response to bacterial lipopolysaccharide challenge compared with similarly treated Ucp2+/+ mice. Likewise, expression of inducible nitric-oxide synthase and inflammatory cytokines is higher in Ucp2-/- mice in vivo and in vitro. Key steps in the activation cascade of nuclear factor (NF)-kappa B, including I kappa B kinase and nuclear translocation of NF-kappa B subunits, are all remarkably enhanced in Ucp2-/- mice, most notably even under basal conditions. The elevated basal activity of I kappa B kinase in macrophages from Ucp2-/- mice can be blocked by cell-permeable inhibitors of superoxide and hydrogen peroxide generation, but not by a specific inhibitor for inducible nitric-oxide synthase. Isolated mitochondria from Ucp2-/- cells produced more superoxide/hydrogen peroxide. We conclude that mitochrondrially derived reactive oxygen from Ucp2-/- cells constitutively activates NF-kappa B, resulting in a "primed" state to both potentiate and amplify the inflammatory response upon subsequent stimulation.
Collapse
Affiliation(s)
- Yushi Bai
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Hiroki Onuma
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Xu Bai
- Department of Psychiatry and Behavioral Sciences and
| | | | - Mary Misukonis
- Division of Hematology-Oncology, Veterans Administration Medical Center/Duke University Medical Center, Durham, North Carolina 27710
| | - J. Brice Weinberg
- Division of Hematology-Oncology, Veterans Administration Medical Center/Duke University Medical Center, Durham, North Carolina 27710
| | - Wenhong Cao
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Jacques Robidoux
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Lisa M. Floering
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Kiefer W. Daniel
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Sheila Collins
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
- Department of Psychiatry and Behavioral Sciences and
- ||To whom correspondence should be addressed: Endocrine Biology Program, CIIT Centers for Health Research, 6 Davis Dr., P. O. Box 12137, Research Triangle Park, NC 27709-2137. Tel.: 919-558-1378; Fax: 919-558-1305; E-mail:
| |
Collapse
|
25
|
Chen Y, Wu J, Ghosh G. KappaB-Ras binds to the unique insert within the ankyrin repeat domain of IkappaBbeta and regulates cytoplasmic retention of IkappaBbeta x NF-kappaB complexes. J Biol Chem 2003; 278:23101-6. [PMID: 12672800 DOI: 10.1074/jbc.m301021200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The IkappaBalpha and IkappaBbeta proteins inhibit the transcriptional potential of active NF-kappaB dimers through stable complex formation. It has been shown that inactive IkappaBalpha x NF-kappaB complexes shuttle in and out of the nucleus, whereas IkappaBbeta x NF-kappaB complexes are retained exclusively in the cytoplasm of resting cells. The biochemical mechanism underlying this functional difference and its consequences are unknown. Although the two IkappaB proteins are significantly homologous, IkappaBbeta contains a unique 47-amino acid insertion of unknown function within its ankyrin repeat domain. In this study, we assess the role of the IkappaBbeta insert in regulating cytoplasmic retention of IkappaBbeta.NF-kappaB complexes. Deletion of the IkappaBbeta insert renders IkappaBbeta x NF-kappaB complexes capable of shuttling between the nucleus and cytoplasm, similar to IkappaBalpha x NF-kappaB complexes. A small Ras-like G-protein, kappaB-Ras, participates with the IkappaBbeta insert to effectively mask the NF-kappaB nuclear localization potential. Similarly, a complex between NF-kappaB and a mutant IkappaBbeta protein containing four serine to alanine mutations within its C-terminal proline, glutamic acid, serine, and threonine-rich sequence exhibits nucleocytoplasmic shuttling. This suggests a phosphorylation state-dependent role for the C-terminal proline, glutamic acid, serine, and threonine-rich sequence of IkappaBbeta in proper localization of IkappaBbeta x NF-kappaB complexes. These results are consistent with structural studies, which predicted that binary IkappaBbeta x NF-kappaB complexes should be capable of nuclear translocation, and with previous observations that hypophosphorylated IkappaBbeta.NF-kappaB complexes can reside in the nucleus.
Collapse
Affiliation(s)
- Yi Chen
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093-0359, USA
| | | | | |
Collapse
|
26
|
Maiuri MC, De Stefano D, Mele G, Fecarotta S, Greco L, Troncone R, Carnuccio R. Nuclear factor kappa B is activated in small intestinal mucosa of celiac patients. J Mol Med (Berl) 2003; 81:373-9. [PMID: 12743709 DOI: 10.1007/s00109-003-0440-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2002] [Accepted: 03/17/2003] [Indexed: 10/20/2022]
Abstract
NF-kappa B regulates inflammatory and immune response by increasing the expression of specific genes. In celiac disease proinflammatory cytokines, adhesion molecules, and enzymes whose gene expression is known to be regulated by NF-kappa B are involved. This study investigated the activation of NF-kappa B in inflamed mucosa from patients with untreated celiac disease. Biopsy specimens from control, untreated, and treated patients were subjected to molecular biology analysis. NF-kappa B activation was evaluated by electrophoretic mobility shift assay. NF-kappa B related subunit protein level, and inducible nitric oxide synthase and cyclo-oxygenase 2 protein expression was analyzed by western blot. Both NF-kappa B/DNA binding activity and p50/p65 nuclear levels were higher in biopsy specimens from untreated patients than in those from treated patients and controls. The degradation of I kappa B beta in the cytosol and the reappearance in the nucleus indicated a persistent NF-kappa B activation in celiac disease. NF-kappa B activity was maintained in cultured biopsy specimens up to 6 h and decreased at 24 h, and then the addition of peptic-tryptic digest of gliadin caused the recovery of NF-kappa B activity at 6 h. NF-kappa B/DNA binding activity was correlated with inducible nitric oxide synthase and cyclo-oxygenase-2 protein expression. These results show for the first time that NF-kappa B is activated in the inflamed mucosa of celiac patients and suggest that it may represent a molecular target for the modulation of inflammatory response in celiac disease.
Collapse
Affiliation(s)
- Maria Chiara Maiuri
- Department of Experimental Pharmacology, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Ladner KJ, Caligiuri MA, Guttridge DC. Tumor necrosis factor-regulated biphasic activation of NF-kappa B is required for cytokine-induced loss of skeletal muscle gene products. J Biol Chem 2003; 278:2294-303. [PMID: 12431991 DOI: 10.1074/jbc.m207129200] [Citation(s) in RCA: 187] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
NF-kappaB activation is classically defined as a transient response initiated by the degradation of IkappaB inhibitor proteins leading to nuclear import of NF-kappaB and culminating with the resynthesis of IkappaBalpha and subsequent inactivation of the transcription factor. Although this type of regulation is considered the paradigm for NF-kappaB activation, other regulatory profiles are known to exist. By far the most common of these is chronic or persistent activation of NF-kappaB. In comparison, regulation of NF-kappaB in a biphasic manner represents a profile that is scarcely documented and whose biological significance remains poorly understood. Here we show using differentiated skeletal muscle cells, that tumor necrosis factor (TNF) induces NF-kappaB activation in a biphasic manner. Unlike the first transient phase, which is terminated within 1 h of cytokine addition, the second phase persists for an additional 24-36 h. Biphasic activation is mediated at both the levels of NF-kappaB DNA binding and transactivation function, and both phases are dependent on the IKK/26 S proteasome pathway. We find that regulation of the first transient phase is mediated by the degradation and subsequent resynthesis of IkappaBalpha, as well as by a TNF-induced expression of A20. Second phase activity correlates with persistent down-regulation of both IkappaBalpha and IkappaBbeta proteins, derived from a continuous TNF signal. Finally, we demonstrate that inhibition of NF-kappaB prior to initiation of the second phase of activity inhibits cytokine-mediated loss of muscle proteins. We propose that the biphasic activation of NF-kappaB in response to TNF may play a key regulatory role in skeletal muscle wasting associated with cachexia.
Collapse
Affiliation(s)
- Katherine J Ladner
- Division of Human Cancer Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus 43210, USA
| | | | | |
Collapse
|
28
|
Wang JH, Manning BJ, Wu QD, Blankson S, Bouchier-Hayes D, Redmond HP. Endotoxin/lipopolysaccharide activates NF-kappa B and enhances tumor cell adhesion and invasion through a beta 1 integrin-dependent mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:795-804. [PMID: 12517943 DOI: 10.4049/jimmunol.170.2.795] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Beta(1) integrins play a crucial role in supporting tumor cell attachment to and invasion into the extracellular matrix. Endotoxin/LPS introduced by surgery has been shown to enhance tumor metastasis in a murine model. Here we show the direct effect of LPS on tumor cell adhesion and invasion in extracellular matrix proteins through a beta(1) integrin-dependent pathway. The human colorectal tumor cell lines SW480 and SW620 constitutively expressed high levels of the beta(1) subunit, whereas various low levels of alpha(1), alpha(2), alpha(4), and alpha(6) expression were detected. SW480 and SW620 did not express membrane-bound CD14; however, LPS in the presence of soluble CD14 (sCD14) significantly up-regulated beta(1) integrin expression; enhanced tumor cell attachment to fibronectin, collagen I, and laminin; and strongly promoted tumor cell invasion through the Matrigel. Anti-beta(1) blocking mAbs (4B4 and 6S6) abrogated LPS- plus sCD14-induced tumor cell adhesion and invasion. Furthermore, LPS, when combined with sCD14, resulted in NF-kappaB activation in both SW480 and SW620 cells. Inhibition of the NF-kappaB pathway significantly attenuated LPS-induced up-regulation of beta(1) integrin expression and prevented tumor cell adhesion and invasion. These results provide direct evidence that although SW480 and SW620 cells do not express membrane-bound CD14, LPS in the presence of sCD14 can activate NF-kappaB, up-regulate beta(1) integrin expression, and subsequently promote tumor cell adhesion and invasion. Moreover, LPS-induced tumor cell attachment to and invasion through extracellular matrix proteins is beta(1) subunit-dependent.
Collapse
Affiliation(s)
- Jiang Huai Wang
- Department of Academic Surgery, National University of Ireland, Cork University Hospital, Ireland.
| | | | | | | | | | | |
Collapse
|
29
|
Wang JH, Doyle M, Manning BJ, Di Wu Q, Blankson S, Redmond HP. Induction of bacterial lipoprotein tolerance is associated with suppression of toll-like receptor 2 expression. J Biol Chem 2002; 277:36068-75. [PMID: 12133836 DOI: 10.1074/jbc.m205584200] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tolerance to bacterial cell wall components including lipopolysaccharide (LPS) may represent an essential regulatory mechanism during bacterial infection. Two members of the Toll-like receptor (TLR) family, TLR2 and TLR4, recognize the specific pattern of bacterial cell wall components. TLR4 has been found to be responsible for LPS tolerance. However, the role of TLR2 in bacterial lipoprotein (BLP) tolerance and LPS tolerance is unclear. Pretreatment of human THP-1 monocytic cells with a synthetic bacterial lipopeptide induced tolerance to a second BLP challenge with diminished tumor necrosis factor-alpha and interleukin-6 production, termed BLP tolerance. Furthermore, BLP-tolerized THP-1 cells no longer responded to LPS stimulation, indicating a cross-tolerance to LPS. Induction of BLP tolerance was CD14-independent, as THP-1 cells that lack membrane-bound CD14 developed tolerance both in serum-free conditions and in the presence of a specific CD14 blocking monoclonal antibody (MEM-18). Pre-exposure of THP-1 cells to BLP suppressed mitogen-activated protein kinase phosphorylation and nuclear factor-kappaB activation in response to subsequent BLP and LPS stimulation, which is comparable with that found in LPS-tolerized cells, indicating that BLP tolerance and LPS tolerance may share similar intracellular pathways. However, BLP strongly enhanced TLR2 expression in non-tolerized THP-1 cells, whereas LPS stimulation had no effect. Furthermore, a specific TLR2 blocking monoclonal antibody (2392) attenuated BLP-induced, but not LPS-induced, tumor necrosis factor-alpha and interleukin-6 production, indicating BLP rather than LPS as a ligand for TLR2 engagement and activation. More importantly, pretreatment of THP-1 cells with BLP strongly inhibited TLR2 activation in response to subsequent BLP stimulation. In contrast, LPS tolerance did not prevent BLP-induced TLR2 overexpression. These results demonstrate that BLP tolerance develops through down-regulation of TLR2 expression.
Collapse
Affiliation(s)
- Jiang Huai Wang
- Department of Academic Surgery, National University of Ireland, Cork University Hospital, Cork, Ireland.
| | | | | | | | | | | |
Collapse
|
30
|
Haij SD, Woltman AM, Bakker AC, Daha MR, van Kooten C. Production of inflammatory mediators by renal epithelial cells is insensitive to glucocorticoids. Br J Pharmacol 2002; 137:197-204. [PMID: 12208776 PMCID: PMC1573489 DOI: 10.1038/sj.bjp.0704866] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
1. In the present study we investigated the effect of glucocorticoids on the activation of renal tubular epithelial cells, which are thought to play an important role in inflammatory processes in the kidney. 2. Activation of renal epithelial cells by IL-1, TNF-alpha or CD40L resulted in increased production of cytokines and chemokines. Both in the renal epithelial cell line HK-2 and in primary cultures of human proximal tubular epithelial cells (PTEC) production of IL-6, IL-8 and monocyte chemotactic protein 1 (MCP-1) was not inhibited by glucocorticoids, independent of the stimulus. 3. In contrast, dexamethasone strongly inhibited cytokine production by immortalized renal fibroblasts and an airway epithelial cell line (A549). 4. Stimulation of renal epithelial cells resulted in activation of NF-kappaB, a pivotal transcription factor in the regulation of cytokine genes, as was shown by IkappaB-alpha degradation and increased DNA-binding activity. In contrast to dexamethasone, addition of the NF-kappaB inhibitors pyrrolidine dithiocarbamate (PDTC) and n-tosyl-l-phenylalanine chloromethyl ketone (TPCK) completely abolished cytokine and chemokine production. 5. Renal epithelial cells express abundant levels of the functional glucocorticoid receptor alpha (GRalpha) isoform and low levels of the inhibitory beta isoform (GRbeta). 6. In conclusion, cytokine production by renal epithelial cells is insensitive to the inhibitory effects of glucocorticoids. The lack of dexamethasone-mediated inhibition was specific for renal epithelial cells and could not be explained by an increased expression of the glucocorticoid receptor beta isoform.
Collapse
Affiliation(s)
- Simone de Haij
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrea M Woltman
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Astrid C Bakker
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mohamed R Daha
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cees van Kooten
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Author for correspondence:
| |
Collapse
|
31
|
Kiemer AK, Weber NC, Vollmar AM. Induction of IkappaB: atrial natriuretic peptide as a regulator of the NF-kappaB pathway. Biochem Biophys Res Commun 2002; 295:1068-76. [PMID: 12135603 DOI: 10.1016/s0006-291x(02)00807-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Atrial natriuretic peptide (ANP) was shown to possess anti-inflammatory potential due to its potency to inhibit the production of inflammatory mediators, such as TNF-alpha. The aim of this study was to determine potential effects of ANP on endothelial cells targeted by TNF-alpha. HUVEC were treated with TNF-alpha and expression of adhesion molecules was investigated by FACS and RT-PCR. Pre-treatment of cells with ANP (30min) significantly reduced TNF-alpha-induced cell surface protein and mRNA expression of E-selectin and ICAM-1, whereas it did not influence VCAM-1. ANP reduced TNF-alpha-induced NF-kappaB activity, which was paralleled by a decreased translocation of p65 to nuclei. ANP did not alter TNF-alpha-induced phosphorylation and degradation of IkappaB-alpha, but attenuated degradation of IkappaB-epsilon. Moreover, ANP leads to a transcriptional induction of IkappaB-alpha. The induction of IkappaB by ANP is suggested as a novel mechanism for regulating inflammatory signalling in endothelial cells, leading to reduced TNF-alpha-induced expression of adhesion molecules.
Collapse
Affiliation(s)
- Alexandra K Kiemer
- Department of Pharmacy, Center of Drug Research, University of Munich, Butenandtstrasse 5-13, 81377 Munich, Germany.
| | | | | |
Collapse
|
32
|
Vancurova I, Wu R, Miskolci V, Sun S. Increased p50/p50 NF-kappaB activation in human papillomavirus type 6- or type 11-induced laryngeal papilloma tissue. J Virol 2002; 76:1533-6. [PMID: 11773428 PMCID: PMC135826 DOI: 10.1128/jvi.76.3.1533-1536.2002] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We have observed elevated NF-kappaB DNA-binding activity in nuclear extracts from human papillomavirus type 6- and 11-infected laryngeal papilloma tissues. The predominant DNA-binding species is the p50/p50 homodimer. The elevated NF-kappaB activity could be correlated with a reduced level of cytoplasmic IkappaBbeta and could be associated with the overexpression of p21(CIP1/WAF1) in papilloma cells. Increased NF-kappaB activity and cytoplasmic accumulation of p21(CIP1/WAF1) might counteract death-promoting effects elicited by overexpressed PTEN and reduced activation of Akt and STAT3 previously noted in these tissues.
Collapse
Affiliation(s)
- Ivana Vancurova
- Department of Pediatrics, Long Island Jewish Medical Center, New Hyde Park, New York, USA
| | | | | | | |
Collapse
|
33
|
Albensi BC. Potential roles for tumor necrosis factor and nuclear factor-kappaB in seizure activity. J Neurosci Res 2001; 66:151-4. [PMID: 11592109 DOI: 10.1002/jnr.1206] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- B C Albensi
- Department of Neurological Surgery, Cerebrovascular Research Center, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| |
Collapse
|