1
|
Bellon M, Nicot C. Increased H19/miR-675 Expression in Adult T-Cell Leukemia Is Associated with a Unique Notch Signature Pathway. Int J Mol Sci 2024; 25:5130. [PMID: 38791169 PMCID: PMC11120950 DOI: 10.3390/ijms25105130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/21/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
The Notch pathway is a key cancer driver and is important in tumor progression. Early research suggested that Notch activity was highly dependent on the expression of the intracellular cleaved domain of Notch-1 (NICD). However, recent insights into Notch signaling reveal the presence of Notch pathway signatures, which may vary depending on different cancer types and tumor microenvironments. Herein, we perform a comprehensive investigation of the Notch signaling pathway in adult T-cell leukemia (ATL) primary patient samples. Using gene arrays, we demonstrate that the Notch pathway is constitutively activated in ATL patient samples. Furthermore, the activation of Notch in ATL cells remains elevated irrespective of the presence of activating mutations in Notch itself or its repressor, FBXW7, and that ATL cells are dependent upon Notch-1 expression for proliferation and survival. We demonstrate that ATL cells exhibit the expression of pivotal Notch-related genes, including notch-1, hes1, c-myc, H19, and hes4, thereby defining a critical Notch signature associated with ATL disease. Finally, we demonstrate that lncRNA H19 is highly expressed in ATL patient samples and ATL cells and contributes to Notch signaling activation. Collectively, our results shed further light on the Notch pathway in ATL leukemia and reveal new therapeutic approaches to inhibit Notch activation in ATL cells.
Collapse
MESH Headings
- Humans
- Leukemia-Lymphoma, Adult T-Cell/genetics
- Leukemia-Lymphoma, Adult T-Cell/metabolism
- Leukemia-Lymphoma, Adult T-Cell/pathology
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Signal Transduction
- Cell Line, Tumor
- Receptor, Notch1/metabolism
- Receptor, Notch1/genetics
- Gene Expression Regulation, Leukemic
- Receptors, Notch/metabolism
- Receptors, Notch/genetics
- Cell Proliferation/genetics
- F-Box-WD Repeat-Containing Protein 7/metabolism
- F-Box-WD Repeat-Containing Protein 7/genetics
- Gene Expression Regulation, Neoplastic
- Adult
Collapse
Affiliation(s)
| | - Christophe Nicot
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| |
Collapse
|
2
|
Losada A, Izquierdo-Useros N, Aviles P, Vergara-Alert J, Latino I, Segalés J, Gonzalez SF, Cuevas C, Raïch-Regué D, Muñoz-Alonso MJ, Perez-Zsolt D, Muñoz-Basagoiti J, Rodon J, Chang LA, Warang P, Singh G, Brustolin M, Cantero G, Roca N, Pérez M, Bustos-Morán E, White K, Schotsaert M, García-Sastre A. Plitidepsin as an Immunomodulator against Respiratory Viral Infections. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1307-1318. [PMID: 38416036 PMCID: PMC10984758 DOI: 10.4049/jimmunol.2300426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/26/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
Plitidepsin is a host-targeted compound known for inducing a strong anti-SARS-CoV-2 activity, as well as for having the capacity of reducing lung inflammation. Because IL-6 is one of the main cytokines involved in acute respiratory distress syndrome, the effect of plitidepsin in IL-6 secretion in different in vitro and in vivo experimental models was studied. A strong plitidepsin-mediated reduction of IL-6 was found in human monocyte-derived macrophages exposed to nonproductive SARS-CoV-2. In resiquimod (a ligand of TLR7/8)-stimulated THP1 human monocytes, plitidepsin-mediated reductions of IL-6 mRNA and IL-6 levels were also noticed. Additionally, although resiquimod-induced binding to DNA of NF-κB family members was unaffected by plitidepsin, a decrease in the regulated transcription by NF-κB (a key transcription factor involved in the inflammatory cascade) was observed. Furthermore, the phosphorylation of p65 that is required for full transcriptional NF-κB activity was significantly reduced by plitidepsin. Moreover, decreases of IL-6 levels and other proinflammatory cytokines were also seen in either SARS-CoV-2 or H1N1 influenza virus-infected mice, which were treated at low enough plitidepsin doses to not induce antiviral effects. In summary, plitidepsin is a promising therapeutic agent for the treatment of viral infections, not only because of its host-targeted antiviral effect, but also for its immunomodulatory effect, both of which were evidenced in vitro and in vivo by the decrease of proinflammatory cytokines.
Collapse
Affiliation(s)
- Alejandro Losada
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute, Can Ruti Campus, Badalona, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo Aviles
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | - Júlia Vergara-Alert
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Irene Latino
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Bellinzona, Switzerland
| | - Joaquim Segalés
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Bellinzona, Switzerland
| | - Carmen Cuevas
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | | | - María J Muñoz-Alonso
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | | | | | - Jordi Rodon
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Lauren A Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Marco Brustolin
- Unit of Entomology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Guillermo Cantero
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Núria Roca
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Mònica Pérez
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal, Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Eugenio Bustos-Morán
- Department of Research and Development, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | - Kris White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
3
|
Haselager MV, Thijssen R, Bax D, Both D, De Boer F, Mackay S, Dubois J, Mellink C, Kater AP, Eldering E. JAK-STAT signalling shapes the NF-κB response in CLL towards venetoclax sensitivity or resistance via Bcl-XL. Mol Oncol 2022. [PMID: 36550750 DOI: 10.1002/1878-0261.13364] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/22/2022] [Revised: 12/02/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
Preventing or overcoming resistance to the Bcl-2 inhibitor venetoclax is an emerging unmet clinical need in patients with chronic lymphocytic leukaemia (CLL). The upregulation of anti-apoptotic Bcl-2 members through signalling pathways within the tumor microenvironment appears as a major factor leading to resistance to venetoclax. Previously, we reported that T cells can drive resistance through CD40 and non-canonical NF-κB activation and subsequent Bcl-XL induction. Moreover, the T cell-derived cytokines IL-21 and IL-4 differentially affect Bcl-XL expression and sensitivity to venetoclax via unknown mechanisms. Here, we mechanistically dissected how Bcl-XL is regulated in the context of JAK-STAT signalling in primary CLL. First, we demonstrated a clear antagonistic role of IL-21/STAT3 signalling in the NF-κB-mediated expression of Bcl-XL, whereas IL-4/STAT6 further promoted the expression of Bcl-XL. In comparison, Bfl-1, another NF-κB target, was not differentially affected by either cytokine. Second, STAT3 and STAT6 affected Bcl-XL transcription by binding to its promoter without disrupting the DNA-binding activity of NF-κB. Third, in situ proximity ligation assays (isPLAs) indicated crosstalk between JAK-STAT signalling and NF-κB, in which STAT3 inhibited canonical NF-κB by accelerating nuclear export, and STAT6 promoted non-canonical NF-κB. Finally, NF-κB inducing kinase (NIK) inhibition interrupted the NF-κB/STAT crosstalk and resensitized CLL cells to venetoclax. In conclusion, we uncovered distinct crosstalk mechanisms that shape the NF-κB response in CLL towards venetoclax sensitivity or resistance via Bcl-XL, thereby revealing new potential therapeutic targets.
Collapse
Affiliation(s)
- Marco V Haselager
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, The Netherlands.,Amsterdam institute for Infection & Immunity, The Netherlands.,Cancer Immunology, Cancer Center Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, The Netherlands
| | - Rachel Thijssen
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC location University of Amsterdam, The Netherlands
| | - Danique Bax
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, The Netherlands.,Amsterdam institute for Infection & Immunity, The Netherlands.,Cancer Immunology, Cancer Center Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, The Netherlands
| | - Demi Both
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, The Netherlands.,Amsterdam institute for Infection & Immunity, The Netherlands.,Cancer Immunology, Cancer Center Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, The Netherlands
| | | | - Simon Mackay
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Julie Dubois
- Department of Hematology, Amsterdam UMC location University of Amsterdam, The Netherlands
| | - Clemens Mellink
- Department of Clinical Genetics, Amsterdam UMC location University of Amsterdam, The Netherlands
| | - Arnon P Kater
- Amsterdam institute for Infection & Immunity, The Netherlands.,Cancer Immunology, Cancer Center Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC location University of Amsterdam, The Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, The Netherlands.,Amsterdam institute for Infection & Immunity, The Netherlands.,Cancer Immunology, Cancer Center Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, The Netherlands
| |
Collapse
|
4
|
IKKα plays a major role in canonical NF-kB signalling in colorectal cells. Biochem J 2022; 479:305-325. [PMID: 35029639 PMCID: PMC8883499 DOI: 10.1042/bcj20210783] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/23/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/17/2022]
Abstract
Inhibitor of kappa B (IκB) kinase β (IKKβ) has long been viewed as the dominant IKK in the canonical nuclear factor-κB (NF-κB) signalling pathway, with IKKα being more important in non-canonical NF-κB activation. Here we have investigated the role of IKKα and IKKβ in canonical NF-κB activation in colorectal cells using CRISPR–Cas9 knock-out cell lines, siRNA and selective IKKβ inhibitors. IKKα and IKKβ were redundant for IκBα phosphorylation and turnover since loss of IKKα or IKKβ alone had little (SW620 cells) or no (HCT116 cells) effect. However, in HCT116 cells IKKα was the dominant IKK required for basal phosphorylation of p65 at S536, stimulated phosphorylation of p65 at S468, nuclear translocation of p65 and the NF-κB-dependent transcriptional response to both TNFα and IL-1α. In these cells, IKKβ was far less efficient at compensating for the loss of IKKα than IKKα was able to compensate for the loss of IKKβ. This was confirmed when siRNA was used to knock-down the non-targeted kinase in single KO cells. Critically, the selective IKKβ inhibitor BIX02514 confirmed these observations in WT cells and similar results were seen in SW620 cells. Notably, whilst IKKα loss strongly inhibited TNFα-dependent p65 nuclear translocation, IKKα and IKKβ contributed equally to c-Rel nuclear translocation indicating that different NF-κB subunits exhibit different dependencies on these IKKs. These results demonstrate a major role for IKKα in canonical NF-κB signalling in colorectal cells and may be relevant to efforts to design IKK inhibitors, which have focused largely on IKKβ to date.
Collapse
|
5
|
Feng J, Ge C, Li W, Li R. 3-(3-Hydroxyphenyl)propionic acid, a microbial metabolite of quercetin, inhibits monocyte binding to endothelial cells via modulating E-selectin expression. Fitoterapia 2021; 156:105071. [PMID: 34743931 DOI: 10.1016/j.fitote.2021.105071] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/27/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 11/18/2022]
Abstract
Adhesion of monocytes to endothelial cells is an important initiating step in atherogenesis. One of the most abundant flavonoids in the diet, quercetin has been reported to inhibit monocyte adhesion to endothelial cells. However, it is poorly absorbed in the upper gastrointestinal tract during oral intake but rather is metabolized by the intestinal microbiota into various phenolic acids. Since the biological properties of the microbial metabolites of quercetin remain largely unknown, herein, we investigated how the microbial metabolite of quercetin, 3-(3-hydroxyphenyl)propionic acid (3HPPA) impact monocyte adhesion to endothelial cells. Direct treatment with 3HPPA for 24 h was not cytotoxic to human aortic endothelial cells (HAECs). Cotreatment with 3HPPA inhibited tumor necrosis factor α (TNFα)-induced adhesion of THP-1 monocytes to HAECs, and suppressed the upregulation of cell adhesion molecule E-selectin but not intercellular adhesion molecule 1 or vascular cell adhesion molecule 1. Furthermore, 3HPPA was found to inhibit TNFα-induced nuclear translocation and phosphorylation of the p65 subunit of nuclear factor κB (NF-κB). We conclude that 3HPPA mitigates the adhesion of monocytes to endothelial cells by suppressing the expression of the cell adhesion molecule E-selectin in HAECs via inhibition of the NF-κB pathway, providing additional evidence for the health benefits of dietary flavonoids and their microbial metabolites as therapeutic agents in atherosclerosis.
Collapse
Affiliation(s)
- Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, Guangdong, People's Republic of China
| | - Chenchen Ge
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, Guangdong, People's Republic of China
| | - Weixi Li
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, Guangdong, People's Republic of China
| | - Rongsong Li
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
6
|
Aliena-Valero A, Rius-Pérez S, Baixauli-Martín J, Torregrosa G, Chamorro Á, Pérez S, Salom JB. Uric Acid Neuroprotection Associated to IL-6/STAT3 Signaling Pathway Activation in Rat Ischemic Stroke. Mol Neurobiol 2021; 58:408-423. [PMID: 32959172 DOI: 10.1007/s12035-020-02115-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/29/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022]
Abstract
Despite the promising neuroprotective effects of uric acid (UA) in acute ischemic stroke, the seemingly pleiotropic underlying mechanisms are not completely understood. Recent evidence points to transcription factors as UA targets. To gain insight into the UA mechanism of action, we investigated its effects on pertinent biomarkers for the most relevant features of ischemic stroke pathophysiology: (1) oxidative stress (antioxidant enzyme mRNAs and MDA), (2) neuroinflammation (cytokine and Socs3 mRNAs, STAT3, NF-κB p65, and reactive microglia), (3) brain swelling (Vegfa, Mmp9, and Timp1 mRNAs), and (4) apoptotic cell death (Bcl-2, Bax, caspase-3, and TUNEL-positive cells). Adult male Wistar rats underwent intraluminal filament transient middle cerebral artery occlusion (tMCAO) and received UA (16 mg/kg) or vehicle (Locke's buffer) i.v. at 20 min reperfusion. The outcome measures were neurofunctional deficit, infarct, and edema. UA treatment reduced cortical infarct and brain edema, as well as neurofunctional impairment. In brain cortex, increased UA: (1) reduced tMCAO-induced increases in Vegfa and Mmp9/Timp1 ratio expressions; (2) induced Sod2 and Cat expressions and reduced MDA levels; (3) induced Il6 expression, upregulated STAT3 and NF-κB p65 phosphorylation, induced Socs3 expression, and inhibited microglia activation; and (4) ameliorated the Bax/Bcl-2 ratio and induced a reduction in caspase-3 cleavage as well as in TUNEL-positive cell counts. In conclusion, the mechanism for morphological and functional neuroprotection by UA in ischemic stroke is multifaceted, since it is associated to activation of the IL-6/STAT3 pathway, attenuation of edematogenic VEGF-A/MMP-9 signaling, and modulation of relevant mediators of oxidative stress, neuroinflammation, and apoptotic cell death.
Collapse
Affiliation(s)
- Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universitat de València, Torre A, Lab 5.05, Ave Fernando Abril Martorell 106, 46026, Valencia, Spain
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain
| | - Sergio Rius-Pérez
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain
| | - Júlia Baixauli-Martín
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain
| | - Germán Torregrosa
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universitat de València, Torre A, Lab 5.05, Ave Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Ángel Chamorro
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Comprehensive Stroke Center, Department of Neuroscience, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Departamento de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Salvador Pérez
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain.
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universitat de València, Torre A, Lab 5.05, Ave Fernando Abril Martorell 106, 46026, Valencia, Spain.
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain.
| |
Collapse
|
7
|
Darbinian N, Darbinyan A, Merabova N, Gomberg R, Chabriere E, Simm M, Selzer ME, Amini S. DING Protein Inhibits Transcription of HIV-1 Gene through Suppression of Phosphorylation of NF-κB p65. ACTA ACUST UNITED AC 2020; 6. [PMID: 34307877 PMCID: PMC8296972 DOI: 10.16966/2380-5536.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/23/2022]
Abstract
Introduction: Novel plant DING proteins (full-length 38 kDa p38SJ, and 27 kDa p27SJ) exhibit phosphatase activity and modulate HIV-1 gene transcription. Previously, we demonstrated that DING regulates HIV-1 gene transcription by dephosphorylation and inactivation of CTD RNA polymerase II, the major elongating factor of HIV-1 Long Terminal Repeats (LTR). Because the transcription of HIV-1 is controlled by several viral and cellular factors, including p65/p50 subunits of NF-κB, we hypothesized that DING phosphatase can also affect the phosphorylation and activity of p65 NF-κB, in addition to C-terminal Domain (CTD) of RNA Polymerase II (RNAPII), to suppress HIV-1 gene transcription and inhibit HIV-1 infection. Methods: Here, we describe the inhibition of HIV-1 infection and the p65/p50 NF-κB phosphorylation by DING protein, analyzed by ELISA and northern-blot assays, western-blot assays, cell fractionation, and promoter-reporter assays in DING-expressing cells, using a pTet-on inducible system. Results: Results from HIV-1 infection assays demonstrate a strong inhibition of HIV-1 and HIV-LTR RNA expression by DING protein, determined by p24 ELISA and by northern blot assay. Results from the western blot assays and cell fractionation assays show that there is an increase in the level of hypo-phosphorylated form of p65 NF-κB in DING-expressing cells. Both fractions of p65/p50, nuclear or cytoplasmic, are affected by DING phosphatase, but more cytoplasmic accumulation of p65 NF-κB was found in the presence of DING, suggesting that subsequent activation and nuclear import of active NF-κB is affected by DING. The major portion of nuclear p65 was dephosphorylated in DING-expressing cells. The promoter-reporter assay demonstrated that DING-mediated dephosphorylation and dysregulation of NF-κB p65 lead to the suppression of its binding to HIV-1 LTR, and resulted in the inhibition of p65-mediated activation of LTR transcription. Mapping of the region within LTR that was affected by DING revealed that both, NF-κB and CTD RNA Polymerase II binding sites were important, and cooperativity of these cellular factors was diminished by DING. In addition, mapping of the region within DING-p38SJ that affected LTR transcription, revealed that phosphate-binding domain is essential for this inhibitory activity. Conclusion: We have demonstrated the effect of DING phosphatases on HIV-1 infection, phosphorylation of p65 NF-κB, and transcription of HIV-1 LTR. Our studies suggest that one possible mechanism by which DING can regulate the expression of HIV-1 LTR can be through dysregulation of the transcription factor NF-κB p65 by preventing its phosphorylation and translocation to the nucleus and binding to the HIV-1 LTR, an action that could contribute to the utility of DING p38SJ as an antiviral agent. Importantly, DING not only inhibits HIV-1 LTR gene transcription in the presence of increased p65 NF-κB, but also suppresses HIV-1 infection. DING protein improved inhibitory effects of the known anti-retroviral drugs, Tenofovir (TFV) and Emtricitabine (FTS) on HIV-1, since in the combination with these drugs; the suppression of HIV-1 by DNG was significantly higher when it was in combination with these drugs, compared to controls or cases without DING. Thus, our data support the use of neuroprotective DING proteins as novel therapeutic antiviral drugs that suppress HIV-1 LTR transcription by interfering with the function of NF-κB.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, USA
| | - Armine Darbinyan
- Department of Pathology, Yale University School of Medicine, USA
| | - Nana Merabova
- Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, USA
| | - Rebeccah Gomberg
- Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, USA
| | - Erik Chabriere
- Aix-Marseille Université, Institut Universitaire de France, IHU Mediterranée Infection, France
| | - Malgorzata Simm
- University of Pikeville, Kentucky College of Osteopathic Medicine, USA
| | - Michael E Selzer
- Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, USA
| | - Shohreh Amini
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, USA
| |
Collapse
|
8
|
Sevilla-Movilla S, Arellano-Sánchez N, Martínez-Moreno M, Gajate C, Sánchez-Vencells A, Valcárcel LV, Agirre X, Valeri A, Martínez-López J, Prósper F, Mollinedo F, Teixidó J. Upregulated expression and function of the α4β1 integrin in multiple myeloma cells resistant to bortezomib. J Pathol 2020; 252:29-40. [PMID: 32501543 DOI: 10.1002/path.5480] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/06/2020] [Revised: 04/29/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022]
Abstract
The interaction of multiple myeloma (MM) cells with the bone marrow (BM) microenvironment promotes MM cell retention, survival, and resistance to different anti-MM agents, including proteasome inhibitors (PIs) such as bortezomib (BTZ). The α4β1 integrin is a main adhesion receptor mediating MM cell-stroma interactions and MM cell survival, and its expression and function are downregulated by BTZ, leading to inhibition of cell adhesion-mediated drug resistance (CAM-DR) and MM cell apoptosis. Whether decreased α4β1 expression and activity are maintained or recovered upon development of resistance to BTZ represents an important question, as a potential rescue of α4β1 function could boost MM cell survival and disease progression. Using BTZ-resistant MM cells, we found that they not only rescue their α4β1 expression, but its levels were higher than in parental cells. Increased α4β1 expression in resistant cells correlated with enhanced α4β1-mediated cell lodging in the BM, and with disease progression. BTZ-resistant MM cells displayed enhanced NF-κB pathway activation relative to parental counterparts, which contributed to upregulated α4 expression and to α4β1-dependent MM cell adhesion. These data emphasize the upregulation of α4β1 expression and function as a key event during resistance to BTZ in MM, which might indirectly contribute to stabilize this resistance, as stronger MM cell attachment to BM stroma will regain CAM-DR and MM cell growth and survival. Finally, we found a strong correlation between high ITGB1 (integrin β1) expression in MM and poor progression-free survival (PFS) and overall survival (OS) during treatment of MM patients with BTZ and IMIDs, and combination of high ITGB1 levels and presence of the high-risk genetic factor amp1q causes low PFS and OS. These results unravel a novel prognostic value for ITGB1 in myeloma. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Silvia Sevilla-Movilla
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Nohemí Arellano-Sánchez
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Mónica Martínez-Moreno
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Consuelo Gajate
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Anna Sánchez-Vencells
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Luis V Valcárcel
- Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona, Spain
| | - Xabier Agirre
- Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona, Spain
| | - Antonio Valeri
- Department of Translational Hematology, Hospital Universitario 12 de Octubre, Centro Nacional de Investigaciones Oncológicas, CIBERONC, Madrid, Spain
| | - Joaquin Martínez-López
- Department of Translational Hematology, Hospital Universitario 12 de Octubre, Centro Nacional de Investigaciones Oncológicas, CIBERONC, Madrid, Spain
| | - Felipe Prósper
- Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona, Spain.,Department of Hematology, Clínica Universidad de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Faustino Mollinedo
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Joaquin Teixidó
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| |
Collapse
|
9
|
Cai M, Liao Z, Zou X, Xu Z, Wang Y, Li T, Li Y, Ou X, Deng Y, Guo Y, Peng T, Li M. Herpes Simplex Virus 1 UL2 Inhibits the TNF-α-Mediated NF-κB Activity by Interacting With p65/p50. Front Immunol 2020; 11:549. [PMID: 32477319 PMCID: PMC7237644 DOI: 10.3389/fimmu.2020.00549] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/07/2019] [Accepted: 03/10/2020] [Indexed: 12/31/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) is a large double-stranded DNA virus that encodes at least 80 viral proteins, many of which are involved in the virus-host interaction and are beneficial to the viral survival and reproduction. However, the biological functions of some HSV-1-encoded proteins are not fully understood. Nuclear factor κB (NF-κB) activation is the major antiviral innate response, which can be triggered by various signals induced by cellular receptors from different pathways. Here, we demonstrated that HSV-1 UL2 protein could antagonize the tumor necrosis factor α (TNF-α)-mediated NF-κB activation. Co-immunoprecipitation assays showed that UL2 could interact with the NF-κB subunits p65 and p50, which also revealed the region of amino acids 9 to 17 of UL2 could suppress the NF-κB activation and interact with p65 and p50, and UL2 bound to the immunoglobulin-like plexin transcription factor functional domain of p65. However, UL2 did not affect the formation of p65/p50 dimerization and their nuclear localizations. Yet, UL2 was demonstrated to inhibit the NF-κB activity by attenuating TNF-α-induced p65 phosphorylation at Ser536 and therefore decreasing the expression of downstream inflammatory chemokine interleukin 8. Taken together, the attenuation of NF-κB activation by UL2 may contribute to the escape of host's antiviral innate immunity for HSV-1 during its infection.
Collapse
Affiliation(s)
- Mingsheng Cai
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Zongmin Liao
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China.,Department of Scientific Research and Education, Yuebei People's Hospital, Shaoguan, China
| | - Xingmei Zou
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Zuo Xu
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yuanfang Wang
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Tong Li
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yiwen Li
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Xiaowen Ou
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yangxi Deng
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yingjie Guo
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Tao Peng
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.,South China Vaccine Corporation Limited, Guangzhou Science Park, Guangzhou, China
| | - Meili Li
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
The human T-cell leukemia virus type-1 tax oncoprotein dissociates NF-κB p65 RelA-Stathmin complexes and causes catastrophic mitotic spindle damage and genomic instability. Virology 2019; 535:83-101. [PMID: 31299491 DOI: 10.1016/j.virol.2019.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/01/2019] [Revised: 06/21/2019] [Accepted: 07/02/2019] [Indexed: 12/23/2022]
Abstract
Genomic instability is a hallmark of many cancers; however, the molecular etiology of chromosomal dysregulation is not well understood. The human T-cell leukemia virus type-1 (HTLV-1) oncoprotein Tax activates NF-κB-signaling and induces DNA-damage and aberrant chromosomal segregation through diverse mechanisms which contribute to viral carcinogenesis. Intriguingly, Stathmin/oncoprotein-18 (Op-18) depolymerizes tubulin and interacts with the p65RelA subunit and functions as a cofactor for NF-κB-dependent transactivation. We thus hypothesized that the dissociation of p65RelA-Stathmin/Op-18 complexes by Tax could lead to the catastrophic destabilization of microtubule (MT) spindle fibers during mitosis and provide a novel mechanistic link between NF-κB-signaling and genomic instability. Here we report that the inhibition of Stathmin expression by the retroviral latency protein, p30II, or knockdown with siRNA-stathmin, dampens Tax-mediated NF-κB transactivation and counters Tax-induced genomic instability and cytotoxicity. The Tax-G148V mutant, defective for NF-κB activation, exhibited reduced p65RelA-Stathmin binding and diminished genomic instability and cytotoxicity. Dominant-negative inhibitors of NF-κB also prevented Tax-induced multinucleation and apoptosis. Moreover, cell clones containing the infectious HTLV-1 ACH. p30II mutant provirus, impaired for p30II production, exhibited increased multinucleation and the accumulation of cytoplasmic tubulin aggregates following nocodozole-treatment. These findings allude to a mechanism whereby NF-κB-signaling regulates tubulin dynamics and mitotic instability through the modulation of p65RelA-Stathmin/Op-18 interactions, and support the notion that p30II enhances the survival of Tax-expressing HTLV-1-transformed cells.
Collapse
|
11
|
Dreier RF, Ahrné E, Broz P, Schmidt A. Global Ion Suppression Limits the Potential of Mass Spectrometry Based Phosphoproteomics. J Proteome Res 2018; 18:493-507. [DOI: 10.1021/acs.jproteome.8b00812] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Affiliation(s)
- Roland Felix Dreier
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Erik Ahrné
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Petr Broz
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Alexander Schmidt
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| |
Collapse
|
12
|
Takakura K, Takatou S, Tomiyama R, Le TM, Nguyen DT, Nakamura Y, Konishi T, Matsugo S, Hori O. Inhibition of nuclear factor-κB p65 phosphorylation by 3,4-dihydroxybenzalacetone and caffeic acid phenethyl ester. J Pharmacol Sci 2018; 137:248-255. [PMID: 30037569 DOI: 10.1016/j.jphs.2018.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/15/2018] [Revised: 06/05/2018] [Accepted: 06/23/2018] [Indexed: 01/14/2023] Open
Abstract
3,4-Dihydroxybenzalacetone (DBL) and caffeic acid phenethyl ester (CAPE) are both catechol-containing phenylpropanoid derivatives with various bioactivities. In the present study, we compared the effects of these compounds and other phenylpropanoid derivatives on the activation of nuclear factor-κB (NF-κB) signaling, a major pathway in the inflammatory response, using RAW 264.7 cells. Lipopolysaccharide (LPS)- and interferon γ-induced production of nitrite was strongly suppressed by CAPE and, to a lesser extent, by DBL and caffeic acid ethyl ester. Consistent with these results, induction of NF-κB downstream genes, such as Nitric oxide synthase, interleukin 1 beta, and interleukin 6, and translocation of NF-κB p65 to the nucleus were reduced after LPS stimulation, to a greater extent with CAPE than with DBL. Interestingly, the phosphorylation of p65 was reduced by both compounds, especially by CAPE, even when the level of IκB was not altered. Furthermore, the thiol groups of p65 were modified by CAPE, and the inhibitory effects of CAPE and DBL on the p65 phosphorylation and nitrite production were reversed by pretreatment with thiol-containing reagents. These results suggest that CAPE has strong inhibitory effects on the NF-κB activation that are associated with the modification of thiol groups and phosphorylation of p65.
Collapse
Affiliation(s)
- Ken Takakura
- Graduate School of Natural Science and Technology, Division of Natural System, Kanazawa University, Kanazawa, Ishikawa, Japan; Graduate School of Medical Sciences, Department of Neuroanatomy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Shouhei Takatou
- Graduate School of Natural Science and Technology, Division of Natural System, Kanazawa University, Kanazawa, Ishikawa, Japan; Graduate School of Medical Sciences, Department of Neuroanatomy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Ryoichi Tomiyama
- Graduate School of Natural Science and Technology, Division of Natural System, Kanazawa University, Kanazawa, Ishikawa, Japan; Graduate School of Medical Sciences, Department of Neuroanatomy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Thuong Manh Le
- Graduate School of Medical Sciences, Department of Neuroanatomy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Dinh Thi Nguyen
- Graduate School of Medical Sciences, Department of Neuroanatomy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yutaka Nakamura
- Faculty of Applied Life Sciences, Niigata University of Pharmacy & Applied Life Sciences (NUPALS), Niigata, Niigata, Japan
| | - Tetsuya Konishi
- Niigata University of Pharmacy & Applied Life Sciences (NUPALS), LIAISON R/D Center, Niigata, Niigata, Japan
| | - Seiichi Matsugo
- Graduate School of Natural Science and Technology, Division of Natural System, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Osamu Hori
- Graduate School of Medical Sciences, Department of Neuroanatomy, Kanazawa University, Kanazawa, Ishikawa, Japan.
| |
Collapse
|
13
|
Li R, Zhang Z, Wang J, Huang Y, Sun W, Xie R, Hu F, Lei T. Triptolide suppresses growth and hormone secretion in murine pituitary corticotroph tumor cells via NF-kappaB signaling pathway. Biomed Pharmacother 2017; 95:771-779. [PMID: 28892788 DOI: 10.1016/j.biopha.2017.08.127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/27/2017] [Revised: 08/24/2017] [Accepted: 08/29/2017] [Indexed: 01/06/2023] Open
Abstract
Triptolide is a principal diterpene triepoxide from the Chinese medical plant Tripterygium wilfordii Hook. f., whose extracts have been utilized in dealing with diverse diseases in traditional Chinese medicine for centuries. Recently, the antitumor effect of triptolide has been found in several pre-clinical neoplasm models, but its effect on pituitary corticotroph adenomas has not been investigated so far. In this study, we are aiming to figure out the antitumor effect of triptolide and address the underlying molecular mechanism in AtT20 murine corticotroph cell line. Our results demonstrated that triptolide inhibited cell viability and colony number of AtT20 cells in a dose- and time-dependent pattern. Triptolide also suppressed proopiomelanocortin (Pomc) mRNA expression and extracellular adrenocorticotropic hormone (ACTH) secretion in AtT20 cells. Flow cytometry prompted that triptolide leaded to G2/M phase arrest, apoptosis program and mitochondrial membrane depolarization in AtT20 cells. Moreover, dose-dependent activation of caspase-3 and decreased Bcl2/Bax proportion were observed after triptolide treatment. By western blot analysis we found that triptolide impeded phosphorylation of NF-κB p65 subunit and extracellular signal-regulated kinase (ERK), along with reduction of cyclin D1, without any impact on other NF-κB related protein expression like total p65, p50, IκB-α, p-IκB-α. Furthermore, the mouse xenograft model revealed the inhibition of tumor growth and hormone secretion after triptolide administration. Altogether this compound might be a potential pharmaceutical choice in managing Cushing's disease.
Collapse
Affiliation(s)
- Ran Li
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Zhuo Zhang
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Junwen Wang
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Yiming Huang
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Wei Sun
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Ruifan Xie
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China; Research Group Experimental Neurooncology, Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Feng Hu
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Ting Lei
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China.
| |
Collapse
|
14
|
Lim SG, Kim JK, Suk K, Lee WH. Crosstalk between signals initiated from TLR4 and cell surface BAFF results in synergistic induction of proinflammatory mediators in THP-1 cells. Sci Rep 2017; 7:45826. [PMID: 28374824 PMCID: PMC5379196 DOI: 10.1038/srep45826] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/20/2016] [Accepted: 03/06/2017] [Indexed: 12/21/2022] Open
Abstract
Cellular response to stimulation is mediated by meshwork of signaling pathways that may share common signaling adaptors. Here, we present data demonstrating that signaling pathways initiated from the membrane-bound form of B-cell activating factor (BAFF) can crosstalk with lipopolysaccharide (LPS)-induced signaling for synergistic expression of proinflammatory mediators in the human macrophage-like cell line THP-1. Co-treatment of the cells with BAFF-specific monoclonal antibody and LPS resulted in enhanced mitogen-activated protein kinase (MAPK)/mitogen- and stress-activated protein kinase (MSK)-mediated phosphorylation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 subunit (Ser276), which then interacts with CREB binding protein (CBP) for subsequent acetylation. Simultaneously, the phosphorylation of cyclic AMP-response element binding protein (CREB) was enhanced through the combined action of phosphatidylinositol-3-kinase (PI3K)/AKT and MAPK/MSK pathways, and the resulting phospho-CREB interacted with the NF-κB/CBP complex. Transfection of CREB-specific siRNA inhibited the BAFF-mediated enhancing effect indicating that the formation of the CREB/NF-κB/CBP complex is required for the synergistic induction of the proinflammatory genes. These findings indicate that BAFF-mediated reverse signaling can modulate LPS-induced inflammatory activation through regulation of NF-κB and CREB activity and point out the necessity to re-evaluate the role of BAFF in diseases where its expression is high in macrophages.
Collapse
Affiliation(s)
- Su-Geun Lim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jae-Kwan Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science &Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu 41944, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
15
|
Protein Kinase R Mediates the Inflammatory Response Induced by Hyperosmotic Stress. Mol Cell Biol 2017; 37:MCB.00521-16. [PMID: 27920257 DOI: 10.1128/mcb.00521-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/21/2016] [Accepted: 12/01/2016] [Indexed: 12/25/2022] Open
Abstract
High extracellular osmolarity results in a switch from an adaptive to an inflammatory gene expression program. We show that hyperosmotic stress activates the protein kinase R (PKR) independently of its RNA-binding domain. In turn, PKR stimulates nuclear accumulation of nuclear factor κB (NF-κB) p65 species phosphorylated at serine-536, which is paralleled by the induction of a subset of inflammatory NF-κB p65-responsive genes, including inducible nitric oxide synthase (iNOS), interleukin-6 (IL-6), and IL-1β. The PKR-mediated hyperinduction of iNOS decreases cell survival in mouse embryonic fibroblasts via mechanisms involving nitric oxide (NO) synthesis and posttranslational modification of proteins. Moreover, we demonstrate that the PKR inhibitor C16 ameliorates both iNOS amplification and disease-induced phenotypic breakdown of the intestinal epithelial barrier caused by an increase in extracellular osmolarity induced by dextran sodium sulfate (DSS) in vivo Collectively, these findings indicate that PKR activation is an essential part of the molecular switch from adaptation to inflammation in response to hyperosmotic stress.
Collapse
|
16
|
Barreca MM, Spinello W, Cavalieri V, Turturici G, Sconzo G, Kaur P, Tinnirello R, Asea AAA, Geraci F. Extracellular Hsp70 Enhances Mesoangioblast Migration via an Autocrine Signaling Pathway. J Cell Physiol 2017; 232:1845-1861. [PMID: 27925208 DOI: 10.1002/jcp.25722] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/11/2016] [Accepted: 11/29/2016] [Indexed: 12/20/2022]
Abstract
Mouse mesoangioblasts are vessel-associated progenitor stem cells endowed with the ability of multipotent mesoderm differentiation. Therefore, they represent a promising tool in the regeneration of injured tissues. Several studies have demonstrated that homing of mesoangioblasts into blood and injured tissues are mainly controlled by cytokines/chemokines and other inflammatory factors. However, little is known about the molecular mechanisms regulating their ability to traverse the extracellular matrix (ECM). Here, we demonstrate that membrane vesicles released by mesoangioblasts contain Hsp70, and that the released Hsp70 is able to interact by an autocrine mechanism with Toll-like receptor 4 (TLR4) and CD91 to stimulate migration. We further demonstrate that Hsp70 has a positive role in regulating matrix metalloproteinase 2 (MMP2) and MMP9 expression and that MMP2 has a more pronounced effect on cell migration, as compared to MMP9. In addition, the analysis of the intracellular pathways implicated in Hsp70 regulated signal transduction showed the involvement of both PI3K/AKT and NF-κB. Taken together, our findings present a paradigm shift in our understanding of the molecular mechanisms that regulate mesoangioblast stem cells ability to traverse the extracellular matrix (ECM). J. Cell. Physiol. 232: 1845-1861, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Maria M Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Walter Spinello
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Vincenzo Cavalieri
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Giuseppina Turturici
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Gabriella Sconzo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Punit Kaur
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia
| | - Rosaria Tinnirello
- Biomedicine and Molecular Immunology Institute, National Center of Research, Palermo, Italy
| | - Alexzander A A Asea
- Department of Neurology and the Deanship for Scientific Research, University of Dammam, Dammam, Saudi Arabia
| | - Fabiana Geraci
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy.,Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| |
Collapse
|
17
|
Triptolide Combined with Radiotherapy for the Treatment of Nasopharyngeal Carcinoma via NF-κB-Related Mechanism. Int J Mol Sci 2016; 17:ijms17122139. [PMID: 27999372 PMCID: PMC5187939 DOI: 10.3390/ijms17122139] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2016] [Revised: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 11/17/2022] Open
Abstract
Advanced nasopharyngeal carcinoma (NPC) has a poor prognosis because of the lack of an effective treatment. Here we explored the efficiency and the molecular mechanisms of combined treatment with triptolide and ionizing radiation for treating NPC. Human nasopharyngeal carcinoma (CNE) cells were treated with triptolide, ionizing radiation, or triptolide plus ionizing radiation in vitro. Tumor potency was examined in an in vivo CNE cell xenograft mouse model, which was treated as above. Our results demonstrated that triptolide caused a significant reduction in cell growth and colony number, and induced a marked apoptosis that was further enhanced with increasing doses of ionizing radiation. Combination treatment synergistically reduced tumor weight and volume without obvious toxicity. Western blot analysis in vitro and in vivo showed that triptolide induced apoptotic protein Bax expression and inhibited phosph-NF-κB p65, Bcl-2 and VEGF proteins without affecting other NF-κB related protein expression. In conclusion, our findings revealed that triptolide plus ionizing radiation had synergistic anti-tumor and anti-angiogenesis effects in NPC via down-regulating NF-κB p65 phosphorylation. The combination therapy may provide novel mechanism insights into inhibit NPC.
Collapse
|
18
|
Sun L, Pham TT, Cornell TT, McDonough KL, McHugh WM, Blatt NB, Dahmer MK, Shanley TP. Myeloid-Specific Gene Deletion of Protein Phosphatase 2A Magnifies MyD88- and TRIF-Dependent Inflammation following Endotoxin Challenge. THE JOURNAL OF IMMUNOLOGY 2016; 198:404-416. [PMID: 27872207 DOI: 10.4049/jimmunol.1600221] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/05/2016] [Accepted: 10/21/2016] [Indexed: 12/23/2022]
Abstract
Protein phosphatase 2A (PP2A) is a member of the intracellular serine/threonine phosphatases. Innate immune cell activation triggered by pathogen-associated molecular patterns is mediated by various protein kinases, and PP2A plays a counter-regulatory role by deactivating these kinases. In this study, we generated a conditional knockout of the α isoform of the catalytic subunit of PP2A (PP2ACα). After crossing with myeloid-specific cre-expressing mice, effective gene knockout was achieved in various myeloid cells. The myeloid-specific knockout mice (lyM-PP2Afl/fl) showed higher mortality in response to endotoxin challenge and bacterial infection. Upon LPS challenge, serum levels of TNF-α, KC, IL-6, and IL-10 were significantly increased in lyM-PP2Afl/fl mice, and increased phosphorylation was observed in MAPK pathways (p38, ERK, JNK) and the NF-κB pathway (IKKα/β, NF-κB p65) in bone marrow-derived macrophages (BMDMs) from knockout mice. Heightened NF-κB activation was not associated with degradation of IκBα; instead, enhanced phosphorylation of the NF-κB p65 subunit and p38 phosphorylation-mediated TNF-α mRNA stabilization appear to contribute to the increased TNF-α expression. In addition, increased IL-10 expression appears to be due to PP2ACα-knockout-induced IKKα/β hyperactivation. Microarray experiments indicated that the Toll/IL-1R domain-containing adaptor inducing IFN-β/ TNFR-associated factor 3 pathway was highly upregulated in LPS-treated PP2ACα-knockout BMDMs, and knockout BMDMs had elevated IFN-α/β production compared with control BMDMs. Serum IFN-β levels from PP2ACα-knockout mice treated with LPS were also greater than those in controls. Thus, we demonstrate that PP2A plays an important role in regulating inflammation and survival in the setting of septic insult by targeting MyD88- and Toll/IL-1R domain-containing adaptor inducing IFN-β-dependent pathways.
Collapse
Affiliation(s)
- Lei Sun
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109;
| | - Tiffany T Pham
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Timothy T Cornell
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Kelli L McDonough
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Walker M McHugh
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Neal B Blatt
- Division of Pediatric Nephrology, Department of Pediatrics and Communicable Diseases, C.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Mary K Dahmer
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Thomas P Shanley
- Department of Pediatrics, Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611
| |
Collapse
|
19
|
Pradère JP, Hernandez C, Koppe C, Friedman RA, Luedde T, Schwabe RF. Negative regulation of NF-κB p65 activity by serine 536 phosphorylation. Sci Signal 2016; 9:ra85. [PMID: 27555662 DOI: 10.1126/scisignal.aab2820] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/28/2022]
Abstract
Nuclear factor κB (NF-κB) is a master regulator of inflammation and cell death. Whereas most of the activity of NF-κB is regulated through the inhibitor of κB (IκB) kinase (IKK)-dependent degradation of IκB, IKK also phosphorylates subunits of NF-κB. We investigated the contribution of the phosphorylation of the NF-κB subunit p65 at the IKK phosphorylation site serine 536 (Ser(536)) in humans, which is thought to be required for the activation and nuclear translocation of NF-κB. Through experiments with knock-in mice (S534A mice) expressing a mutant p65 with an alanine-to-serine substitution at position 534 (the murine homolog of human Ser(536)), we observed increased expression of NF-κB-dependent genes after injection of mice with the inflammatory stimulus lipopolysaccharide (LPS) or exposure to gamma irradiation, and the enhanced gene expression was most pronounced at late time points. Compared to wild-type mice, S534A mice displayed increased mortality after injection with LPS. Increased NF-κB signaling in the S534A mice was at least in part explained by the increased stability of the S534A p65 protein compared to that of the Ser(534)-phosphorylated wild-type protein. Together, our results suggest that Ser(534) phosphorylation of p65 in mice (and, by extension, Ser(536) phosphorylation of human p65) is not required for its nuclear translocation, but instead inhibits NF-κB signaling to prevent deleterious inflammation.
Collapse
Affiliation(s)
| | - Céline Hernandez
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Christiane Koppe
- Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center and Department of Biomedical Informatics, Columbia University, New York, NY 10032, USA
| | - Tom Luedde
- Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
20
|
Yan Y, Xin A, Liu Q, Huang H, Shao Z, Zang Y, Chen L, Sun Y, Gao H. Induction of ROS generation and NF-κB activation in MARC-145 cells by a novel porcine reproductive and respiratory syndrome virus in Southwest of China isolate. BMC Vet Res 2015; 11:232. [PMID: 26358082 PMCID: PMC4565009 DOI: 10.1186/s12917-015-0480-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/05/2015] [Accepted: 07/13/2015] [Indexed: 01/06/2023] Open
Abstract
Background Porcine reproductive and respiratory syndrome virus (PRRSV) is the cause of an economically important swine disease that has devastated the swine industry since the late 1980s. The aim of the present study was to investigate the interaction between reactive oxygen species (ROS) and NF-κB by PRRSV infection. Results We isolated the local strain of PRRSV from southwest China, designated YN-2011, then sequenced and analyzed the genome. YN-2011 was then used to evaluate the interaction of ROS and NF-κB. In PRRSV infected MARC-145 cells, there was a time-dependent increase in ROS and Maleic Dialdehyde (MDA). Accordingly, NF-κB activation was also increased as PRRSV infection progressed. Degradation of IκB mRNA was detected late in PRRSV infection, and overexpression of the dominant negative form of IκBα significantly suppressed NF-κB induced by PRRSV. Conclusions The results indicate that the generation of ROS is involved in PRRSV replication and this progression is associated with the alteration in NF-κB activity induced by ROS. These results should extend our better understanding the interaction between PRRSV and host MARC-145 cells. Electronic supplementary material The online version of this article (doi:10.1186/s12917-015-0480-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yulin Yan
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, People's Republic of China. .,State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People's Republic of China.
| | - Aiguo Xin
- Yunnan Animal Science and Veterinary Institute, Kunming, Yunnan, People's Republic of China.
| | - Qian Liu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, People's Republic of China.
| | - Hui Huang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, People's Republic of China.
| | - Zhiyong Shao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, People's Republic of China.
| | - Yating Zang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, People's Republic of China.
| | - Ling Chen
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, People's Republic of China.
| | - Yongke Sun
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, People's Republic of China.
| | - Hong Gao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, People's Republic of China.
| |
Collapse
|
21
|
Chang Y, Lan YY, Hsiao JR, Chang NS. Strategies of oncogenic microbes to deal with WW domain-containing oxidoreductase. Exp Biol Med (Maywood) 2014; 240:329-37. [PMID: 25488911 DOI: 10.1177/1535370214561957] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022] Open
Abstract
WW domain-containing oxidoreductase (WWOX) is a well-documented tumor suppressor protein that controls growth, survival, and metastasis of malignant cells. To counteract WWOX's suppressive effects, cancer cells have developed many strategies either to downregulate WWOX expression or to functionally inactivate WWOX. Relatively unknown is, in the context of those cancers associated with certain viruses or bacteria, how the oncogenic pathogens deal with WWOX. Here we review recent studies showing different strategies utilized by three cancer-associated pathogens. Helicobactor pylori reduces WWOX expression through promoter hypermethylation, an epigenetic mechanism also occurring in many other cancer cells. WWOX has a potential to block canonical NF-κB activation and tumorigenesis induced by Tax, an oncoprotein of human T-cell leukemia virus. Tax successfully overcomes the blockage by inhibiting WWOX expression through activation of the non-canonical NF-κB pathway. On the other hand, latent membrane protein 2A of Epstein-Barr virus physically interacts with WWOX and redirects its function to trigger a signaling pathway that upregulates matrix metalloproteinase 9 and cancer cell invasion. These reports may be just "the tip of the iceberg" regarding multiple interactions between WWOX and oncogenic microbes. Further studies in this direction should expand our understanding of infection-driven oncogenesis.
Collapse
Affiliation(s)
- Yao Chang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan 70456, Taiwan Graduate Institute of Basic Medical Science, Medical College, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yu-Yan Lan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan 70456, Taiwan Graduate Institute of Basic Medical Science, Medical College, National Cheng Kung University, Tainan 70101, Taiwan
| | - Jenn-Ren Hsiao
- Department of Otolaryngology, Medical College and Hospital, National Cheng Kung University, Tainan 70101, Taiwan
| | - Nan-Shan Chang
- Institute of Molecular Medicine, Medical College, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
22
|
Shen M, Duan X, Zhou P, Zhou W, Wu X, Xu S, Chen Y, Tao Z. Lymphotoxin β receptor activation promotes bladder cancer in a nuclear factor-κB-dependent manner. Mol Med Rep 2014; 11:783-90. [PMID: 25369740 PMCID: PMC4262482 DOI: 10.3892/mmr.2014.2826] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/30/2013] [Accepted: 06/26/2014] [Indexed: 12/12/2022] Open
Abstract
Bladder cancer (BCa) is the most common tumor of the urinary system. Chronic inflammation in the papillary urothelial neoplasm of low malignant potential (PUNLMP)may contribute to carcinogenesis, including that of BCa, via poorly understood mechanisms. In this study, we show that the lymphotoxin β receptor (LTβR) is upregulated in BCa via activation of the canonical and non-canonical nuclear factor-κB (NF-κB) pathways. The mRNA expression of LTβR in 81 BCa, 10 chronic cystitis and 23 healthy bladder mucosa tissues was investigated by reverse transcription-fluorescent quantitative polymerase chain reaction (RT-FQ-PCR), and protein expression was studied in 73 BCa, 30 cystitis and 15 healthy paraffin-embedded tissue sections by immunohistochemistry. Both LTβR mRNA and protein were upregulated in BCa and cystitis compared to the healthy group (P<0.05). The mRNA level of the downstream NF-κB canonical pathway p65 gene and of the non-canonical pathway RelB gene were higher in the BCa and cystitis groups compared to the healthy one. The level of phosphorylated p65 (p-p65) protein of the canonical NF-κB pathway and that of p52, a protein of the non-canonical NF-κB pathway, were also higher in the BCa and cystitis group compared to the healthy group. The levels of these proteins significantly correlated to the pathological grade, clinical stage and lymph node metastasis of BCa patients (P<0.05). In addition, there was a positive correlation between LTβR and NF-κB pathway proteins. Thus, LTβR signaling may be involved in promoting BCa through the NF-κB pathway, and which may represent the molecular link between inflammation and BCa.
Collapse
Affiliation(s)
- Mo Shen
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang 325000, P.R. China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310009, P.R. China
| | - Ping Zhou
- Wenzhou WuMa Community Health Service Center, Wenzhou 325000, P.R. China
| | - Wu Zhou
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang 325000, P.R. China
| | - Xiuling Wu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang 325000, P.R. China
| | - Siqi Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang 325000, P.R. China
| | - Yuhua Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310009, P.R. China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310009, P.R. China
| |
Collapse
|
23
|
Regulation of HTLV-1 tax stability, cellular trafficking and NF-κB activation by the ubiquitin-proteasome pathway. Viruses 2014; 6:3925-43. [PMID: 25341660 PMCID: PMC4213571 DOI: 10.3390/v6103925] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/09/2014] [Revised: 10/15/2014] [Accepted: 10/21/2014] [Indexed: 12/22/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is a complex retrovirus that infects CD4+ T cells and causes adult T-cell leukemia/lymphoma (ATLL) in 3%–5% of infected individuals after a long latent period. HTLV-1 Tax is a trans-activating protein that regulates viral gene expression and also modulates cellular signaling pathways to enhance T-cell proliferation and cell survival. The Tax oncoprotein promotes T-cell transformation, in part via constitutive activation of the NF-κB transcription factor; however, the underlying mechanisms remain unknown. Ubiquitination is a type of post-translational modification that occurs in a three-step enzymatic cascade mediated by E1, E2 and E3 enzymes and regulates protein stability as well as signal transduction, protein trafficking and the DNA damage response. Emerging studies indicate that Tax hijacks the ubiquitin machinery to activate ubiquitin-dependent kinases and downstream NF-κB signaling. Tax interacts with the E2 conjugating enzyme Ubc13 and is conjugated on C-terminal lysine residues with lysine 63-linked polyubiquitin chains. Tax K63-linked polyubiquitination may serve as a platform for signaling complexes since this modification is critical for interactions with NEMO and IKK. In addition to NF-κB signaling, mono- and polyubiquitination of Tax also regulate its subcellular trafficking and stability. Here, we review recent advances in the diverse roles of ubiquitin in Tax function and how Tax usurps the ubiquitin-proteasome pathway to promote oncogenesis.
Collapse
|
24
|
Shanmugasundaram K, Nayak B, Shim EH, Livi CB, Block K, Sudarshan S. The oncometabolite fumarate promotes pseudohypoxia through noncanonical activation of NF-κB signaling. J Biol Chem 2014; 289:24691-9. [PMID: 25028521 DOI: 10.1074/jbc.m114.568162] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/14/2023] Open
Abstract
Inactivating mutations of the gene encoding the tricarboxylic acid cycle enzyme fumarate hydratase (FH) have been linked to an aggressive variant of hereditary kidney cancer (hereditary leiomyomatosis and renal cell cancer). These tumors accumulate markedly elevated levels of fumarate. Fumarate is among a growing list of oncometabolites identified in cancers with mutations of genes involved in intermediary metabolism. FH-deficient tumors are notable for their pronounced accumulation of the transcription factor hypoxia inducible factor-1α (HIF-1α) and aggressive behavior. To date, HIF-1α accumulation in hereditary leiomyomatosis and renal cell cancer tumors is thought to result from fumarate-dependent inhibition of prolyl hydroxylases and subsequent evasion from von Hippel-Lindau-dependent degradation. Here, we demonstrate a novel mechanism by which fumarate promotes HIF-1α mRNA and protein accumulation independent of the von Hippel-Lindau pathway. Here we demonstrate that fumarate promotes p65 phosphorylation and p65 accumulation at the HIF-1α promoter through non-canonical signaling via the upstream Tank binding kinase 1 (TBK1). Consistent with these data, inhibition of the TBK1/p65 axis blocks HIF-1α accumulation in cellular models of FH loss and markedly reduces cell invasion of FH-deficient RCC cancer cells. Collectively, our data demonstrate a novel mechanism by which pseudohypoxia is promoted in FH-deficient tumors and identifies TBK1 as a novel putative therapeutic target for the treatment of aggressive fumarate-driven tumors.
Collapse
Affiliation(s)
| | | | - Eun-Hee Shim
- the Department of Urology, University of Alabama, Birmingham, Alabama 35294, and
| | - Carolina B Livi
- Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas 78229
| | - Karen Block
- From the Departments of Medicine and the South Texas Veterans Health Care System, Audie L. Murphy Memorial Hospital Division, San Antonio, Texas 78229
| | - Sunil Sudarshan
- the Department of Urology, University of Alabama, Birmingham, Alabama 35294, and
| |
Collapse
|
25
|
Wu X, Qi J, Bradner JE, Xiao G, Chen LF. Bromodomain and extraterminal (BET) protein inhibition suppresses human T cell leukemia virus 1 (HTLV-1) Tax protein-mediated tumorigenesis by inhibiting nuclear factor κB (NF-κB) signaling. J Biol Chem 2013; 288:36094-105. [PMID: 24189064 DOI: 10.1074/jbc.m113.485029] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
The etiology of human T cell leukemia virus 1 (HTLV-1)-mediated adult T cell leukemia is associated with the ability of viral oncoprotein Tax to induce sustained NF-κB activation and the expression of many NF-κB target genes. Acetylation of the RelA subunit of NF-κB and the subsequent recruitment of bromodomain-containing factor Brd4 are important for the expression of NF-κB target genes in response to various stimuli. However, their contributions to Tax-mediated NF-κB target gene expression and tumorigenesis remain unclear. Here we report that Tax induced the acetylation of lysine 310 of RelA and the binding of Brd4 to acetylated RelA to facilitate Tax-mediated transcriptional activation of NF-κB. Depletion of Brd4 down-regulated Tax-mediated NF-κB target gene expression and cell proliferation. Inhibiting the interaction of Brd4 and acetylated RelA with the bromodomain extraterminal protein inhibitor JQ1 suppressed the proliferation of Tax-expressing rat fibroblasts and Tax-positive HTLV-1-infected cells and Tax-mediated cell transformation and tumorigenesis. Moreover, JQ1 attenuated the Tax-mediated transcriptional activation of NF-κB, triggering the polyubiquitination and proteasome-mediated degradation of constitutively active nuclear RelA. Our results identify Brd4 as a key regulator for Tax-mediated NF-κB gene expression and suggest that targeting epigenetic regulators such as Brd4 with the bromodomain extraterminal protein inhibitor might be a potential therapeutic strategy for cancers and other diseases associated with HTLV-1 infection.
Collapse
Affiliation(s)
- Xuewei Wu
- From the Department of Biochemistry and
| | | | | | | | | |
Collapse
|
26
|
Wang YP, Liu IJ, Chiang CP, Wu HC. Astrocyte elevated gene-1 is associated with metastasis in head and neck squamous cell carcinoma through p65 phosphorylation and upregulation of MMP1. Mol Cancer 2013; 12:109. [PMID: 24063540 PMCID: PMC3856534 DOI: 10.1186/1476-4598-12-109] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/03/2013] [Accepted: 09/17/2013] [Indexed: 12/21/2022] Open
Abstract
Background The survival rate of head and neck squamous cell carcinoma (HNSCC) at advanced stage is poor, despite contemporary advances in treatment modalities. Recent studies have indicated that astrocyte elevated gene-1 (AEG-1), a single transmembrane protein without any known functional domains, is overexpressed in various malignancies and is implicated in both distant metastasis and poor survival. Results High expression of AEG-1 in HNSCC was positively correlated with regional lymph node metastasis and a poor 5-year survival rate. Knockdown of AEG-1 in HNSCC cell lines reduced their capacity for colony formation, migration and invasion. Furthermore, decreased tumor volume and metastatic foci were observed after knockdown of AEG-1 in subcutaneous xenografts and pulmonary metastasis assays in vivo, respectively. We also demonstrated that AEG-1 increased phosphorylation of the p65 subunit of NF-κB, and regulated the expression of MMP1 in HNSCC cells. Moreover, compromised phosphorylation of the p65 (RelA) subunit of NF-κB at serine 536 was observed upon silencing of AEG-1 in both HNSCC cell lines and clinical specimens. Conclusion High expression of AEG-1 is associated with lymph node metastasis and its potentially associated mechanism is investigated.
Collapse
Affiliation(s)
- Yi-Ping Wang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| | | | | | | |
Collapse
|
27
|
NF-kappaB mediated transcriptional repression of acid modifying hormone gastrin. PLoS One 2013; 8:e73409. [PMID: 24009751 PMCID: PMC3751843 DOI: 10.1371/journal.pone.0073409] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/20/2012] [Accepted: 07/24/2013] [Indexed: 12/16/2022] Open
Abstract
Helicobacter pylori is a major pathogen associated with the development of gastroduodenal diseases. It has been reported that H. pylori induced pro-inflammatory cytokine IL1B is one of the various modulators of acid secretion in the gut. Earlier we reported that IL1B-activated NFkB down-regulates gastrin, the major hormonal regulator of acid secretion. In this study, the probable pathway by which IL1B induces NFkB and affects gastrin expression has been elucidated. IL1B-treated AGS cells showed nine-fold activation of MyD88 followed by phosphorylation of TAK1 within 15 min of IL1B treatment. Furthermore, it was observed that activated TAK1 significantly up-regulates the NFkB subunits p50 and p65. Ectopic expression of NFkB p65 in AGS cells resulted in about nine-fold transcriptional repression of gastrin both in the presence and absence of IL1B. The S536A mutant of NFkB p65 is significantly less effective in repressing gastrin. These observations show that a functional NFkB p65 is important for IL1B-mediated repression of gastrin. ChIP assays revealed the presence of HDAC1 and NFkB p65 along with NCoR on the gastrin promoter. Thus, the study provides mechanistic insight into the IL1B-mediated gastrin repression via NFkB.
Collapse
|
28
|
AGGF1 is a novel anti-inflammatory factor associated with TNF-α-induced endothelial activation. Cell Signal 2013; 25:1645-53. [DOI: 10.1016/j.cellsig.2013.04.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/15/2013] [Revised: 04/14/2013] [Accepted: 04/14/2013] [Indexed: 12/21/2022]
|
29
|
Lodewick J, Sampaio C, Boxus M, Rinaldi AS, Coulonval K, Willems L, Roger PP, Bex F. Acetylation at lysine 346 controls the transforming activity of the HTLV-1 Tax oncoprotein in the Rat-1 fibroblast model. Retrovirology 2013; 10:75. [PMID: 23880157 PMCID: PMC3734113 DOI: 10.1186/1742-4690-10-75] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/29/2013] [Accepted: 07/18/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Transformation by the Tax oncoprotein of the human T cell leukemia virus type 1 (HTLV-1) is governed by actions on cellular regulatory signals, including modulation of specific cellular gene expression via activation of signaling pathways, acceleration of cell cycle progression via stimulation of cyclin-dependent kinase activity leading to retinoblastoma protein (pRb) hyperphosphorylation and perturbation of survival signals. These actions control early steps in T cell transformation and development of Adult T cell leukemia (ATL), an aggressive malignancy of HTLV-1 infected T lymphocytes. Post-translational modifications of Tax by phosphorylation, ubiquitination, sumoylation and acetylation have been implicated in Tax-mediated activation of the NF-κB pathway, a key function associated with Tax transforming potential. RESULTS In this study, we demonstrate that acetylation at lysine K(346) in the carboxy-terminal domain of Tax is modulated in the Tax nuclear bodies by the acetyltransferase p300 and the deacetylases HDAC5/7 and controls phosphorylation of the tumor suppressor pRb by Tax-cyclin D3-CDK4-p21(CIP) complexes. This property correlates with the inability of the acetylation deficient K(346)R mutant, but not the acetylation mimetic K(346)Q mutant, to promote anchorage-independent growth of Rat-1 fibroblasts. By contrast, acetylation at lysine K(346) had no effects on the ability of Tax carboxy-terminal PDZ-binding domain to interact with the tumor suppressor hDLG. CONCLUSIONS The identification of the acetyltransferase p300 and the deacetylase HDAC7 as enzymes modulating Tax acetylation points to new therapeutic targets for the treatment of HTLV-1 infected patients at risk of developing ATL.
Collapse
Affiliation(s)
- Julie Lodewick
- Institute for Microbiological Research J-M Wiame (IRMW), Laboratory of Microbiology, Université Libre de Bruxelles, 1, Avenue E, Gryson, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Nakajima S, Kato H, Gu L, Takahashi S, Johno H, Umezawa K, Kitamura M. Pleiotropic Potential of Dehydroxymethylepoxyquinomicin for NF-κB Suppression via Reactive Oxygen Species and Unfolded Protein Response. THE JOURNAL OF IMMUNOLOGY 2013; 190:6559-69. [DOI: 10.4049/jimmunol.1300155] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/19/2022]
|
31
|
Van Beek M, Oravecz-Wilson KI, Delekta PC, Gu S, Li X, Jin X, Apel IJ, Konkle KS, Feng Y, Teitelbaum DH, Ruland J, McAllister-Lucas LM, Lucas PC. Bcl10 links saturated fat overnutrition with hepatocellular NF-kB activation and insulin resistance. Cell Rep 2013; 1:444-52. [PMID: 22708078 DOI: 10.1016/j.celrep.2012.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023] Open
Abstract
Excess serum free fatty acids (FFAs) are fundamental to the pathogenesis of insulin resistance. With high-fat feeding, FFAs activate NF-kB in target tissues, initiating negative crosstalk with insulin signaling. However, the mechanisms underlying FFA-dependent NF-kB activation remain unclear. Here, we demonstrate that the saturated FA, palmitate, requires Bcl10 for NF-kB activation in hepatocytes. Uptake of palmitate, metabolism to diacylglycerol, and subsequent activation of protein kinase C (PKC) appear to mechanistically link palmitate with Bcl10, known as a central component of a signaling complex that, along with CARMA3 and MALT1, activates NF-kB downstream of selected cell surface receptors. Consequently, Bcl10-deficient mice are protected from hepatic NF-kB activation and insulin resistance following brief high-fat diet, suggesting that Bcl10 plays a major role in the metabolic consequences of acute overnutrition. Surprisingly, while CARMA3 also participates in the palmitate response, MALT1 is completely dispensable, thereby revealing an apparent nonclassical role for Bcl10 in NF-kB signaling.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- B-Cell CLL-Lymphoma 10 Protein
- CARD Signaling Adaptor Proteins/metabolism
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Caspases/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Diet, High-Fat
- Fatty Acids/pharmacology
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Humans
- Insulin Resistance/physiology
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Male
- Mice
- Mice, Knockout
- Models, Animal
- Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein
- NF-kappa B/metabolism
- Neoplasm Proteins/metabolism
- Overnutrition/metabolism
- Palmitates/pharmacology
- Rats
Collapse
Affiliation(s)
- Matthew Van Beek
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bcl-3 suppresses Tax-induced NF-κB activation through p65 nuclear translocation blockage in HTLV-1-infected cells. Int J Oncol 2012; 42:269-76. [PMID: 23135533 DOI: 10.3892/ijo.2012.1685] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/24/2012] [Accepted: 10/16/2012] [Indexed: 11/05/2022] Open
Abstract
Human T cell leukemia virus type 1 (HTLV-1) Tax-induced persistent activation of the NF-κB pathway is perceived as the primary cause of adult T cell leukemia (ATL), an aggressive leukemia caused by HTLV-1. Although elevated oncoprotein Bcl-3 levels are found in many HTLV-1-infected T cell lines and ATL cells, the role of Bcl-3 in the malignant progression caused by HTLV-1 retrovirus remains poorly understood. We confirmed, in the present study, that the Tax-induced NF-κB activation involves the regulation of Bcl-3. Both knockdown and overexpression of Bcl-3 inhibit the Tax-induced NF-κB activation. Similarly, excessive Bcl-3 inhibits the NF-κB/DNA binding activity and significantly decreases Tax-induced p65 nuclear translocation. The present results demonstrate the pleiotropic roles of Bcl-3 in Tax-induced NF-κB activation and indicate that a balance in the aberrant Bcl-3 expression may be established to play an important role in the maintenance of proliferation and inhibition of apoptosis in HTLV-1-infected and ATL cells.
Collapse
|
33
|
Taylor CA, Liu Z, Tang TC, Zheng Q, Francis S, Wang TW, Ye B, Lust JA, Dondero R, Thompson JE. Modulation of eIF5A expression using SNS01 nanoparticles inhibits NF-κB activity and tumor growth in murine models of multiple myeloma. Mol Ther 2012; 20:1305-14. [PMID: 22588272 PMCID: PMC3392975 DOI: 10.1038/mt.2012.94] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/22/2011] [Accepted: 04/11/2012] [Indexed: 12/31/2022] Open
Abstract
Despite recent advances in the first-line treatment of multiple myeloma, almost all patients eventually experience relapse with drug-resistant disease. New therapeutic modalities are needed, and to this end, SNS01, a therapeutic nanoparticle, is being investigated for treatment of multiple myeloma. The antitumoral activity of SNS01 is based upon modulation of eukaryotic translation initiation factor 5A (eIF5A), a highly conserved protein that is involved in many cellular processes including proliferation, apoptosis, differentiation and inflammation. eIF5A is regulated by post-translational hypusine modification, and overexpression of hypusination-resistant mutants of eIF5A induces apoptosis in many types of cancer cells. SNS01 is a polyethylenimine (PEI)-based nanoparticle that contains both a B-cell-specific expression plasmid expressing a non-hypusinable mutant of eIF5A and a small interfering RNA (siRNA) which depletes endogenous hypusinated eIF5A. Reducing hypusine-modified eIF5A levels was found to inhibit phosphorylation and activity of ERK MAPK and nuclear factor-κB (NF-κB), and thus sensitize myeloma cells to apoptosis resulting from transfection of a plasmid expressing eIF5A(K50R). SNS01 exhibited significant antitumoral activity in both KAS-6/1 (95% inhibition; P < 0.05) and RPMI 8226 (59% inhibition; P < 0.05) multiple myeloma xenograft models following systemic administration. These results highlight the potential of using this approach as a new therapeutic strategy for multiple myeloma.
Collapse
Affiliation(s)
- Catherine A Taylor
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Zhongda Liu
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Terence C Tang
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Qifa Zheng
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Sarah Francis
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Tzann-Wei Wang
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Bin Ye
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - John A Lust
- Department of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - John E Thompson
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
- Senesco Technologies, Bridgewater, New Jersey, USA
| |
Collapse
|
34
|
HTLV-1 tax-induced rapid senescence is driven by the transcriptional activity of NF-κB and depends on chronically activated IKKα and p65/RelA. J Virol 2012; 86:9474-83. [PMID: 22740410 DOI: 10.1128/jvi.00158-12] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/08/2023] Open
Abstract
The HTLV-1 oncoprotein Tax is a potent activator of classical and alternative NF-κB pathways and is thought to promote cell proliferation and transformation via NF-κB activation. We showed recently that hyperactivation of NF-κB by Tax triggers a cellular senescence response (H. Zhi et al., PLoS Pathog. 7:e1002025, 2011). Inhibition of NF-κB activation by expression of I-κBα superrepressor or by small hairpin RNA (shRNA)-mediated knockdown of p65/RelA rescues cells from Tax-induced rapid senescence (Tax-IRS). Here we demonstrate that Tax-IRS is driven by the transcriptional activity of NF-κB. Knockdown of IKKγ, the primary Tax target, by shRNAs abrogated Tax-mediated activation of both classical and alternative NF-κB pathways and rendered knockdown cells resistant to Tax-IRS. Consistent with a critical role of IKKα in the transcriptional activity of NF-κB, IKKα deficiency drastically decreased NF-κB trans-activation by Tax, although it only modestly reduced Tax-mediated I-κBα degradation and NF-κB nuclear localization. In contrast, although IKKβ knockdown attenuated Tax-induced NF-κB transcriptional activation, the residual NF-κB activation in IKKβ-deficient cells was sufficient to trigger Tax-IRS. Importantly, the phenotypes of NIK and TAK1 knockdown were similar to those of IKKα and IKKβ knockdown, respectively. Finally, double knockdown of RelB and p100 had a minor effect on senescence induction by Tax. These data suggest that Tax, through its interaction with IKKγ, helps recruit NIK and TAK1 for IKKα and IKKβ activation, respectively. In the presence of Tax, the delineation between the classical and alternative NF-κB pathways becomes obscured. The senescence checkpoint triggered by Tax is driven by the transcriptional activity of NF-κB, which depends on activated IKKα and p65/RelA.
Collapse
|
35
|
Peng H, Guerau-de-Arellano M, Mehta VB, Yang Y, Huss DJ, Papenfuss TL, Lovett-Racke AE, Racke MK. Dimethyl fumarate inhibits dendritic cell maturation via nuclear factor κB (NF-κB) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) and mitogen stress-activated kinase 1 (MSK1) signaling. J Biol Chem 2012; 287:28017-26. [PMID: 22733812 DOI: 10.1074/jbc.m112.383380] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
Dimethyl fumarate (DMF) is an effective novel treatment for multiple sclerosis in clinical trials. A reduction of IFN-γ-producing CD4(+) T cells is observed in DMF-treated patients and may contribute to its clinical efficacy. However, the cellular and molecular mechanisms behind this clinical observation are unclear. In this study, we investigated the effects of DMF on dendritic cell (DC) maturation and subsequent DC-mediated T cell responses. We show that DMF inhibits DC maturation by reducing inflammatory cytokine production (IL-12 and IL-6) and the expression of MHC class II, CD80, and CD86. Importantly, this immature DC phenotype generated fewer activated T cells that were characterized by decreased IFN-γ and IL-17 production. Further molecular studies demonstrated that DMF impaired nuclear factor κB (NF-κB) signaling via reduced p65 nuclear translocalization and phosphorylation. NF-κB signaling was further decreased by DMF-mediated suppression of extracellular signal-regulated kinase 1 and 2 (ERK1/2) and its downstream kinase mitogen stress-activated kinase 1 (MSK1). MSK1 suppression resulted in decreased p65 phosphorylation at serine 276 and reduced histone phosphorylation at serine 10. As a consequence, DMF appears to reduce p65 transcriptional activity both directly and indirectly by promoting a silent chromatin environment. Finally, treatment of DCs with the MSK1 inhibitor H89 partially mimicked the effects of DMF on the DC signaling pathway and impaired DC maturation. Taken together, these studies indicate that by suppression of both NF-κB and ERK1/2-MSK1 signaling, DMF inhibits maturation of DCs and subsequently Th1 and Th17 cell differentiation.
Collapse
Affiliation(s)
- Haiyan Peng
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Abstract
The ability to sense and adjust to the environment is crucial to life. For multicellular organisms, the ability to respond to external changes is essential not only for survival but also for normal development and physiology. Although signaling events can directly modify cellular function, typically signaling acts to alter transcriptional responses to generate both transient and sustained changes. Rapid, but transient, changes in gene expression are mediated by inducible transcription factors such as NF-κB. For the past 25 years, NF-κB has served as a paradigm for inducible transcription factors and has provided numerous insights into how signaling events influence gene expression and physiology. Since its discovery as a regulator of expression of the κ light chain gene in B cells, research on NF-κB continues to yield new insights into fundamental cellular processes. Advances in understanding the mechanisms that regulate NF-κB have been accompanied by progress in elucidating the biological significance of this transcription factor in various physiological processes. NF-κB likely plays the most prominent role in the development and function of the immune system and, not surprisingly, when dysregulated, contributes to the pathophysiology of inflammatory disease. As our appreciation of the fundamental role of inflammation in disease pathogenesis has increased, so too has the importance of NF-κB as a key regulatory molecule gained progressively greater significance. However, despite the tremendous progress that has been made in understanding the regulation of NF-κB, there is much that remains to be understood. In this review, we highlight both the progress that has been made and the fundamental questions that remain unanswered after 25 years of study.
Collapse
Affiliation(s)
- Matthew S Hayden
- Department of Microbiology and Immunology, College of Physicians and Surgeons, New York, New York 10032, USA
| | | |
Collapse
|
38
|
Abstract
Human T-cell lymphotropic virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia/lymphoma (ATL), whereas the highly related HTLV-2 is not associated with ATL or other cancers. In addition to ATL leukemogenesis, studies of the HTLV viruses also provide an exceptional model for understanding basic pathogenic mechanisms of virus-host interactions and human oncogenesis. Accumulating evidence suggests that the viral regulatory protein Tax and host inflammatory transcription factor NF-κB are largely responsible for the different pathogenic potentials of HTLV-1 and HTLV-2. Here, we discuss the molecular mechanisms of HTLV-1 oncogenic pathogenesis with a focus on the interplay between the Tax oncoprotein and NF-κB pro-oncogenic signaling. We also outline some of the most intriguing and outstanding questions in the fields of HTLV and NF-κB. Answers to those questions will greatly advance our understanding of ATL leukemogenesis and other NF-κB-associated tumorigenesis and will help us design personalized cancer therapies.
Collapse
|
39
|
Fiume G, Vecchio E, De Laurentiis A, Trimboli F, Palmieri C, Pisano A, Falcone C, Pontoriero M, Rossi A, Scialdone A, Fasanella Masci F, Scala G, Quinto I. Human immunodeficiency virus-1 Tat activates NF-κB via physical interaction with IκB-α and p65. Nucleic Acids Res 2011; 40:3548-62. [PMID: 22187158 PMCID: PMC3333881 DOI: 10.1093/nar/gkr1224] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/19/2022] Open
Abstract
Nuclear factor (NF)-κB is a master regulator of pro-inflammatory genes and is upregulated in human immunodeficiency virus 1 (HIV-1) infection. Mechanisms underlying the NF-κB deregulation by HIV-1 are relevant for immune dysfunction in AIDS. We report that in single round HIV-1 infection, or single-pulse PMA stimulation, the HIV-1 Tat transactivator activated NF-κB by hijacking the inhibitor IκB-α and by preventing the repressor binding to the NF-κB complex. Moreover, Tat associated with the p65 subunit of NF-κB and increased the p65 DNA-binding affinity and transcriptional activity. The arginine- and cysteine-rich domains of Tat were required for IκB-α and p65 association, respectively, and for sustaining the NF-κB activity. Among an array of NF-κB-responsive genes, Tat mostly activated the MIP-1α expression in a p65-dependent manner, and bound to the MIP-1α NF-κB enhancer thus promoting the recruitment of p65 with displacement of IκB-α; similar findings were obtained for the NF-κB-responsive genes CSF3, LTA, NFKBIA and TLR2. Our results support a novel mechanism of NF-κB activation via physical interaction of Tat with IκB-α and p65, and may contribute to further insights into the deregulation of the inflammatory response by HIV-1.
Collapse
Affiliation(s)
- Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro Magna Graecia, Viale Europa-Germaneto, 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Olière S, Douville R, Sze A, Belgnaoui SM, Hiscott J. Modulation of innate immune responses during human T-cell leukemia virus (HTLV-1) pathogenesis. Cytokine Growth Factor Rev 2011; 22:197-210. [DOI: 10.1016/j.cytogfr.2011.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
|
41
|
Targeting HTLV-1 activation of NFκB in mouse models and ATLL patients. Viruses 2011; 3:886-900. [PMID: 21994759 PMCID: PMC3185776 DOI: 10.3390/v3060886] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/27/2011] [Revised: 06/07/2011] [Accepted: 06/09/2011] [Indexed: 11/17/2022] Open
Abstract
Of the millions of HTLV-1 infected carriers worldwide, 3-5% will develop an aggressive T-cell neoplasm that is highly refractory to conventional therapy. The virus carries the Tax oncogene which constitutively activates the NFκB pathway. This co-option of signaling through NFκB provides for the HTLV-1 infected cell an escape from cell cycle arrest and apoptosis, a steady source of growth factors, and a mechanism by which the virus can activate its own target cell. Therapies that target the NFκB pathway sensitize adult T-cell leukemia/lymphoma (ATLL) cells to apoptosis. A focus on translational interrogation of NFκB inhibitors in animal models and ATLL patients is needed to advance NFκB-targeted ATLL therapies to the bedside.
Collapse
|
42
|
Fiebich BL, Collado JA, Stratz C, Valina C, Hochholzer W, Muñoz E, Bellido LM. Pseudoephedrine inhibits T-cell activation by targeting NF-κB, NFAT and AP-1 signaling pathways. Immunopharmacol Immunotoxicol 2011; 34:98-106. [DOI: 10.3109/08923973.2011.582118] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/22/2023]
|
43
|
The tumor suppressor gene WWOX links the canonical and noncanonical NF-κB pathways in HTLV-I Tax-mediated tumorigenesis. Blood 2010; 117:1652-61. [PMID: 21115974 DOI: 10.1182/blood-2010-08-303073] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/10/2023] Open
Abstract
Both the canonical and noncanonical nuclear factor κB (NF-κB) pathways have been linked to tumorigenesis. However, it remains unknown whether and how the 2 signaling pathways cooperate during tumorigenesis. We report that inhibition of the noncanonical NF-κB pathway significantly delays tumorigenesis mediated by the viral oncoprotein Tax. One function of noncanonical NF-κB activation was to repress expression of the WWOX tumor suppressor gene. Notably, WWOX specifically inhibited Tax-induced activation of the canonical, but not the noncanonical NF-κB pathway. Mechanistic studies indicated that WWOX blocked Tax-induced inhibitors of κB kinaseα (IKKα) recruitment to RelA and subsequent RelA phosphorylation at S536. In contrast, WWOX Y33R, a mutant unable to block the IKKα recruitment and RelA phosphorylation, lost the ability to inhibit Tax-mediated tumorigenesis. These data provide one important mechanism by which Tax coordinates the 2 NF-κB pathways for tumorigenesis. These data also suggest a novel role of WWOX in NF-κB regulation and viral tumorigenesis.
Collapse
|
44
|
Sperone A, Dryden NH, Birdsey GM, Madden L, Johns M, Evans PC, Mason JC, Haskard DO, Boyle JJ, Paleolog EM, Randi AM. The transcription factor Erg inhibits vascular inflammation by repressing NF-kappaB activation and proinflammatory gene expression in endothelial cells. Arterioscler Thromb Vasc Biol 2010; 31:142-50. [PMID: 20966395 DOI: 10.1161/atvbaha.110.216473] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To test whether ETS-related gene (Erg) inhibits tumor necrosis factor (TNF)-α-dependent endothelial activation and inflammation. METHODS AND RESULTS Endothelial activation underlies many vascular diseases, including atherosclerosis. Endothelial activation by proinflammatory cytokines decreases expression of the ETS transcription factor Erg. By using human umbilical vein endothelial cells (HUVECs), we showed that Erg overexpression by adenovirus (AdErg) repressed basal and TNF-α-induced expression of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule (VCAM), and interleukin 8 (IL-8). Erg inhibited TNF-α-dependent activation of the ICAM-1 promoter, nuclear factor (NF)-κB activity, and NF-κB p65 phosphorylation. Basal NF-κB activity was also inhibited by Erg overexpression. Chromatin immunoprecipitation showed that Erg binds to the ICAM-1 proximal promoter region, which contains 7 putative ETS binding sites. To test the anti-inflammatory role of Erg in vivo, we used a murine model of TNF-α-dependent acute inflammation. The injection of AdErg into the paw decreased TNF-α-induced inflammation compared with control. Finally, staining of human coronary plaques showed loss of Erg expression from the endothelium overlaying active plaque shoulders. CONCLUSIONS We have identified a novel physiological anti-inflammatory pathway under the control of the transcription factor Erg; this pathway inhibits NF-κB-dependent transcription and TNF-α-induced inflammation in vivo. These results suggest a novel approach to anti-inflammatory therapies.
Collapse
Affiliation(s)
- Andrea Sperone
- Imperial College London, Hammersmith Hospital, London W12 0NN, England
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Nuclear factor-kappaB (NF-kappaB) is a pleiotropic mediator of inducible and specific gene regulation involving diverse biological activities including immune response, inflammation, cell proliferation, and death. The fine-tuning of the NF-kappaB DNA binding activity is essential for its fundamental function as a transcription factor. An increasing body of literature illustrates that this process can be elegantly and specifically controlled at multiple levels by different protein subsets. In particular, the recent identification of a non-Rel subunit of NF-kappaB itself provides a new way to understand the selective high-affinity DNA binding specificity of NF-kappaB conferred by a synergistic interaction within the whole complex. Here, we review the mechanism of the specification of DNA binding activity of NF-kappaB complexes, one of the most important aspects of NF-kappaB transcriptional control.
Collapse
Affiliation(s)
- Fengyi Wan
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
46
|
Ghosh CC, Ramaswami S, Juvekar A, Vu HY, Galdieri L, Davidson D, Vancurova I. Gene-specific repression of proinflammatory cytokines in stimulated human macrophages by nuclear IκBα. THE JOURNAL OF IMMUNOLOGY 2010; 185:3685-93. [PMID: 20696864 DOI: 10.4049/jimmunol.0902230] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
We have previously shown that increased nuclear accumulation of IkappaBalpha inhibits NF-kappaB activity and induces apoptosis in human leukocytes. In this study, we wanted to explore the possibility that the nucleocytoplasmic distribution of IkappaBalpha can be used as a therapeutic target for the regulation of NF-kappaB-dependent cytokine synthesis. Treatment of LPS-stimulated human U937 macrophages with an inhibitor of chromosome region maintenance 1-dependent nuclear export, leptomycin B, resulted in the increased nuclear accumulation of IkappaBalpha and inhibition of NF-kappaB DNA binding activity, caused by the nuclear IkappaBalpha-p65 NF-kappaB interaction. Surprisingly, however, whereas mRNA expression and cellular release of TNF-alpha, the beta form of pro-IL-1 (IL-1beta), and IL-6 were inhibited by the leptomycin B-induced nuclear IkappaBalpha, IL-8 mRNA expression and cellular release were not significantly affected. Analysis of in vivo recruitment of p65 NF-kappaB to NF-kappaB-regulated promoters by chromatin immunoprecipitation in U937 cells and human PBMCs indicated that although the p65 recruitment to TNF-alpha, IL-1beta, and IL-6 promoters was inhibited by the nuclear IkappaBalpha, p65 recruitment to IL-8 promoter was not repressed. Chromatin immunoprecipitation analyses using IkappaBalpha and S536 phosphospecific p65 NF-kappaB Abs demonstrated that although the newly synthesized IkappaBalpha induced by postinduction repression is recruited to TNF-alpha, IL-1beta, and IL-6 promoters but not to the IL-8 promoter, S536-phosphorylated p65 is recruited to IL-8 promoter, but not to TNF-alpha, IL-1beta, or IL-6 promoters. Together, these data indicate that the inhibition of NF-kappaB-dependent transcription by nuclear IkappaBalpha in LPS-stimulated macrophages is gene specific and depends on the S536 phosphorylation status of the recruited p65 NF-kappaB.
Collapse
Affiliation(s)
- Chandra C Ghosh
- Department of Biological Sciences, St. John's University, New York, NY 11439, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Regulation of IkappaBalpha function and NF-kappaB signaling: AEBP1 is a novel proinflammatory mediator in macrophages. Mediators Inflamm 2010; 2010:823821. [PMID: 20396415 PMCID: PMC2855089 DOI: 10.1155/2010/823821] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/17/2009] [Accepted: 01/12/2010] [Indexed: 02/08/2023] Open
Abstract
NF-κB comprises a family of transcription factors that are critically involved in various inflammatory processes. In this paper, the role of NF-κB in inflammation and atherosclerosis and the regulation of the NF-κB signaling pathway are summarized. The structure, function, and regulation of the NF-κB inhibitors, IκBα and IκBβ, are reviewed. The regulation of NF-κB activity by glucocorticoid receptor (GR) signaling and IκBα sumoylation is also discussed. This paper focuses on the recently reported regulatory function that adipocyte enhancer-binding protein 1 (AEBP1) exerts on NF-κB transcriptional activity in macrophages, in which AEBP1 manifests itself as a potent modulator of NF-κB via physical interaction with IκBα and a critical mediator of inflammation. Finally, we summarize the regulatory roles that recently identified IκBα-interacting proteins play in NF-κB signaling. Based on its proinflammatory roles in macrophages, AEBP1 is anticipated to serve as a therapeutic target towards the treatment of various inflammatory conditions and disorders.
Collapse
|
48
|
Abstract
Phosphorylation of the RelA (p65) NF-κB (nuclear factor κB) subunit has been previously shown to modulate its ability to induce or repress transcription. In the present study we have investigated the consequences of Thr435 phosphorylation within the C-terminal transactivation domain of RelA. We confirm that Thr435 is phosphorylated in cells and is induced by TNFα (tumour necrosis factor α) treatment. Mutational analysis of this site revealed gene-specific effects on transcription, with a T435D phosphomimetic mutant significantly enhancing Cxcl2 (CXC chemokine ligand 2) mRNA levels in reconstituted Rela−/− mouse embryonic fibroblasts. Chromatin immunoprecipitation analysis revealed that this mutation results in enhanced levels of histone acetylation associated with decreased recruitment of HDAC1 (histone deacetylase 1). Moreover, mutation of this site disrupted RelA interaction with HDAC1 in vitro. Thr435 phosphorylation of promoter-bound RelA was also detected at NF-κB target genes following TNFα treatment in wild-type mouse embryonic fibroblasts. Phosphorylation at this site therefore provides an additional mechanism through which the specificity of NF-κB transcriptional activity can be modulated in cells.
Collapse
|
49
|
Briones J, Moga E, Espinosa I, Vergara C, Alvarez E, Villa J, Bordes R, Delgado J, Prat J, Sierra J. Bcl-10 protein highly correlates with the expression of phosphorylated p65 NF-kappaB in peripheral T-cell lymphomas and is associated with clinical outcome. Histopathology 2009; 54:478-85. [PMID: 19309400 DOI: 10.1111/j.1365-2559.2009.03250.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
AIMS In T cells, protein kinase C (PKC) theta plays a major role in T-cell receptor (TCR)-mediated activation of a novel nuclear factor (NF)-kappaB pathway that involves phosphorylation of p65 at serine 536 (Pp65(Ser536)). Bcl-10 acts along the same pathway downstream of PKC theta to activate NF-kappaB. The aim was to investigate the relationship between the expression of PKC theta, Bcl-10 and P-p65(Ser536) proteins and their prognostic significance in peripheral T-cell lymphomas (PTCLs). METHODS AND RESULTS Paraffin-embedded tissues from 30 patients with PTCLs treated with curative intention were evaluated retrospectively. Expression of PKC theta, Bcl-10 and P-p65(Ser536) proteins was assessed using immunohistochemistry. Expression of PKC theta was detected in 22 of 30 cases (73%), Bcl-10 in 20 of 30 (67%) and P-p65(Ser536) in 21 of 30 (70%). Bcl-10+ tumours were associated with PKC theta (18 of 22) (P < 0.0001) and Pp65Ser536 (19 of 21) expression (P < 0.0001). Patients with Bcl-10+ or P-p65(Ser536+) tumours fared better, with a 5-year overall survival of 48 and 45%, respectively, versus 0% for negative tumours (P = 0.029 and P = 0.04, respectively). CONCLUSIONS Bcl-10 is expressed in PTCLs, correlates with PKC theta and Pp65(Ser536) expression and seems to be associated with better survival.
Collapse
Affiliation(s)
- Javier Briones
- Departments of Clinical Haematology and Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Nuclear factor kappa B signaling either stimulates or inhibits neurite growth depending on the phosphorylation status of p65/RelA. J Neurosci 2008; 28:8246-56. [PMID: 18701687 DOI: 10.1523/jneurosci.1941-08.2008] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022] Open
Abstract
Nuclear factor kappaB (NF-kappaB) signaling is known to promote neurite growth from developing sensory neurons and to enhance the size and complexity of pyramidal neuron dendritic arbors in the developing cerebral cortex. In marked contrast, here we show that NF-kappaB signaling can also exert a potent inhibitory influence on neurite growth in certain neurons, and can either promote or inhibit neurite growth in the same neurons depending on the mechanism of NF-kappaB activation. In neonatal superior cervical ganglion sympathetic neurons, enhancing NF-kappaB transcriptional activity by overexpressing either the p65 NF-kappaB subunit or the IkappaB kinase-beta (IKKbeta) subunit of the IkappaB kinase complex, or by tumor necrosis factor alpha (TNFalpha) treatment, strongly inhibits neurite growth. Paradoxically in neonatal nodose ganglion sensory neurons, enhancing NF-kappaB transcriptional activity by p65/p50 overexpression increases neurite growth, whereas enhancing NF-kappaB transcriptional activity by IKKbeta overexpression inhibits neurite growth. In addition to activating NF-kappaB, IKKbeta overexpression leads to phosphorylation of p65 on serine 536. Blockade of serine 536 phosphorylation by a S536A-p65 mutant protein prevents the growth-inhibitory effects of IKKbeta overexpression in both sensory and sympathetic neurons and the growth-inhibitory effects of TNFalpha on sympathetic neurons. Furthermore, expression of a p65 S536D phosphomimetic mutant inhibits neurite growth from sensory neurons. These results demonstrate that NF-kappaB can either stimulate or inhibit neurite growth in developing neurons depending on the phosphorylation status of p65.
Collapse
|