1
|
Sharfe N, Dalal I, Naghdi Z, Lefaudeux D, Vong L, Dadi H, Navarro H, Tasher D, Ovadia A, Zangen T, Ater D, Ngan B, Hoffmann A, Roifman CM. NFκB pathway dysregulation due to reduced RelB expression leads to severe autoimmune disorders and declining immunity. J Autoimmun 2023; 137:102946. [PMID: 36402602 DOI: 10.1016/j.jaut.2022.102946] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Genetic aberrations in the NFκB pathway lead to primary immunodeficiencies with various degrees of severity. We previously demonstrated that complete ablation of the RelB transcription factor, a key component of the alternative pathway, results in an early manifested combined immunodeficiency requiring stem cell transplantation. OBJECTIVE To study the molecular basis of a progressive severe autoimmunity and immunodeficiency in three patients. METHODS Whole exome sequencing was performed to identify the genetic defect. Molecular and cellular techniques were utilized to assess the variant impact on NFκB signaling, canonical and alternative pathway crosstalk, as well as the resultant effects on immune function. RESULTS Patients presented with multiple autoimmune progressive severe manifestations encompassing the liver, gut, lung, and skin, becoming debilitating in the second decade of life. This was accompanied by a deterioration of the immune system, demonstrating an age-related decline in naïve T cells and responses to mitogens, accompanied by a gradual loss of all circulating CD19+ cells. Whole exome sequencing identified a novel homozygous c. C1091T (P364L) transition in RELB. The P364L RelB protein was unstable, with extremely low expression, but retained some function and could be transiently and partially upregulated following Toll-like receptor stimulation. Stimulation of P364L patient fibroblasts resulted in a marked rise in a cluster of pro-inflammatory hyper-expressed transcripts consistent with the removal of RelB inhibitory effect on RelA function. This is likely the main driver of autoimmune manifestations in these patients. CONCLUSION Incomplete loss of RelB provided a unique opportunity to gain insights into NFκB's pathway interactions as well as the pathogenesis of autoimmunity. The P364L RelB mutation leads to gradual decline in immune function with progression of severe debilitating autoimmunity.
Collapse
Affiliation(s)
- Nigel Sharfe
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada
| | - Ilan Dalal
- Pediatric Department, E. Wolfson Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Zahra Naghdi
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada
| | - Diane Lefaudeux
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Linda Vong
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada
| | - Harjit Dadi
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada
| | - Hector Navarro
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Diana Tasher
- Pediatric Department, E. Wolfson Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Ovadia
- Pediatric Department, E. Wolfson Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tzili Zangen
- Pediatric Department, E. Wolfson Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Ater
- Pediatric Pulmonology Unit, Assuta Medical Center, Tel Aviv, Israel
| | - Bo Ngan
- Department of Laboratory Medicine and Pathobiology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Chaim M Roifman
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Deka K, Li Y. Transcriptional Regulation during Aberrant Activation of NF-κB Signalling in Cancer. Cells 2023; 12:788. [PMID: 36899924 PMCID: PMC10001244 DOI: 10.3390/cells12050788] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/16/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
The NF-κB signalling pathway is a major signalling cascade involved in the regulation of inflammation and innate immunity. It is also increasingly recognised as a crucial player in many steps of cancer initiation and progression. The five members of the NF-κB family of transcription factors are activated through two major signalling pathways, the canonical and non-canonical pathways. The canonical NF-κB pathway is prevalently activated in various human malignancies as well as inflammation-related disease conditions. Meanwhile, the significance of non-canonical NF-κB pathway in disease pathogenesis is also increasingly recognized in recent studies. In this review, we discuss the double-edged role of the NF-κB pathway in inflammation and cancer, which depends on the severity and extent of the inflammatory response. We also discuss the intrinsic factors, including selected driver mutations, and extrinsic factors, such as tumour microenvironment and epigenetic modifiers, driving aberrant activation of NF-κB in multiple cancer types. We further provide insights into the importance of the interaction of NF-κB pathway components with various macromolecules to its role in transcriptional regulation in cancer. Finally, we provide a perspective on the potential role of aberrant NF-κB activation in altering the chromatin landscape to support oncogenic development.
Collapse
Affiliation(s)
- Kamalakshi Deka
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore 637551, Singapore
| | - Yinghui Li
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore 637551, Singapore
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore 138673, Singapore
| |
Collapse
|
3
|
Ghosh G, Wang VYF. Origin of the Functional Distinctiveness of NF-κB/p52. Front Cell Dev Biol 2021; 9:764164. [PMID: 34888310 PMCID: PMC8650618 DOI: 10.3389/fcell.2021.764164] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
The transcription regulators of the NF-κB family have emerged as a critical factor affecting the function of various adult tissues. The NF-κB family transcription factors are homo- and heterodimers made up of five monomers (p50, p52, RelA, cRel and RelB). The family is distinguished by sequence homology in their DNA binding and dimerization domains, which enables them to bind similar DNA response elements and participate in similar biological programs through transcriptional activation and repression of hundreds of genes. Even though the family members are closely related in terms of sequence and function, they all display distinct activities. In this review, we discuss the sequence characteristics, protein and DNA interactions, and pathogenic involvement of one member of family, NF-κB/p52, relative to that of other members. We pinpoint the small sequence variations within the conserved region that are mostly responsible for its distinct functional properties.
Collapse
Affiliation(s)
- Gourisankar Ghosh
- Department of Biochemistry, University of California, San Diego, San Diego, CA, United States
| | - Vivien Ya-Fan Wang
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.,Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| |
Collapse
|
4
|
Mockenhaupt K, Gonsiewski A, Kordula T. RelB and Neuroinflammation. Cells 2021; 10:1609. [PMID: 34198987 PMCID: PMC8307460 DOI: 10.3390/cells10071609] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation within the central nervous system involves multiple cell types that coordinate their responses by secreting and responding to a plethora of inflammatory mediators. These factors activate multiple signaling cascades to orchestrate initial inflammatory response and subsequent resolution. Activation of NF-κB pathways in several cell types is critical during neuroinflammation. In contrast to the well-studied role of p65 NF-κB during neuroinflammation, the mechanisms of RelB activation in specific cell types and its roles during neuroinflammatory response are less understood. In this review, we summarize the mechanisms of RelB activation in specific cell types of the CNS and the specialized effects this transcription factor exerts during neuroinflammation.
Collapse
Affiliation(s)
| | | | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VI 23298, USA; (K.M.); (A.G.)
| |
Collapse
|
5
|
Barbu MG, Condrat CE, Thompson DC, Bugnar OL, Cretoiu D, Toader OD, Suciu N, Voinea SC. MicroRNA Involvement in Signaling Pathways During Viral Infection. Front Cell Dev Biol 2020; 8:143. [PMID: 32211411 PMCID: PMC7075948 DOI: 10.3389/fcell.2020.00143] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
The study of miRNAs started in 1993, when Lee et al. observed their involvement in the downregulation of a crucial protein known as LIN-14 in the nematode Caenorhabditis elegans. Since then, great progress has been made regarding research on microRNAs, which are now known to be involved in the regulation of various physiological and pathological processes in both animals and humans. One such example is represented by their interaction with various signaling pathways during viral infections. It has been observed that these pathogens can induce the up-/downregulation of various host miRNAs in order to elude the host's immune system. In contrast, some miRNAs studied could have an antiviral effect, enabling the defense mechanisms to fight the infection or, at the very least, they could induce the pathogen to enter a latent state. At the same time, some viruses encode their own miRNAs, which could further modulate the host's signaling pathways, thus favoring the survival and replication of the virus. The goal of this extensive literature review was to present how miRNAs are involved in the regulation of various signaling pathways in some of the most important and well-studied human viral infections. Further on, knowing which miRNAs are involved in various viral infections and what role they play could aid in the development of antiviral therapeutic agents for certain diseases that do not have a definitive cure in the present. The clinical applications of miRNAs are extremely important, as miRNAs targeted inhibition may have substantial therapeutic impact. Inhibition of miRNAs can be achieved through many different methods, but chemically modified antisense oligonucleotides have shown the most prominent effects. Though scientists are far from completely understanding all the molecular mechanisms behind the complex cross-talks between miRNA pathways and viral infections, the general knowledge is increasing on the different roles played by miRNAs during viral infections.
Collapse
Affiliation(s)
- Madalina Gabriela Barbu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
| | - Carmen Elena Condrat
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
| | - Dana Claudia Thompson
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
| | - Oana Larisa Bugnar
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
| | - Dragos Cretoiu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Oana Daniela Toader
- Division of Obstetrics, Gynecology and Neonatology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Obstetrics and Gynecology, Alessandrescu-Rusescu National Institute for Mother and Child Health, Polizu Clinical Hospital, Bucharest, Romania
| | - Nicolae Suciu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
- Division of Obstetrics, Gynecology and Neonatology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Obstetrics and Gynecology, Alessandrescu-Rusescu National Institute for Mother and Child Health, Polizu Clinical Hospital, Bucharest, Romania
| | - Silviu Cristian Voinea
- Department of Surgical Oncology, Institute of Oncology Prof. Dr. Alexandru Trestioreanu, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
6
|
Mulero MC, Wang VYF, Huxford T, Ghosh G. Genome reading by the NF-κB transcription factors. Nucleic Acids Res 2019; 47:9967-9989. [PMID: 31501881 PMCID: PMC6821244 DOI: 10.1093/nar/gkz739] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/05/2019] [Accepted: 08/21/2019] [Indexed: 12/25/2022] Open
Abstract
The NF-κB family of dimeric transcription factors regulates transcription by selectively binding to DNA response elements present within promoters or enhancers of target genes. The DNA response elements, collectively known as κB sites or κB DNA, share the consensus 5'-GGGRNNNYCC-3' (where R, Y and N are purine, pyrimidine and any nucleotide base, respectively). In addition, several DNA sequences that deviate significantly from the consensus have been shown to accommodate binding by NF-κB dimers. X-ray crystal structures of NF-κB in complex with diverse κB DNA have helped elucidate the chemical principles that underlie target selection in vitro. However, NF-κB dimers encounter additional impediments to selective DNA binding in vivo. Work carried out during the past decades has identified some of the barriers to sequence selective DNA target binding within the context of chromatin and suggests possible mechanisms by which NF-κB might overcome these obstacles. In this review, we first highlight structural features of NF-κB:DNA complexes and how distinctive features of NF-κB proteins and DNA sequences contribute to specific complex formation. We then discuss how native NF-κB dimers identify DNA binding targets in the nucleus with support from additional factors and how post-translational modifications enable NF-κB to selectively bind κB sites in vivo.
Collapse
Affiliation(s)
- Maria Carmen Mulero
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Vivien Ya-Fan Wang
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Tom Huxford
- Structural Biochemistry Laboratory, Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Gourisankar Ghosh
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
MicroRNA-mediated regulation of BM-MSCs differentiation into sweat gland-like cells: targeting NF-κB. J Mol Histol 2019; 50:155-166. [PMID: 30783857 DOI: 10.1007/s10735-019-09814-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/14/2019] [Indexed: 12/27/2022]
Abstract
Sweat gland regeneration is important for patients with an extensive deep burn injury. In previous study, we reported that bone marrow-mesenchymal stem cells (BM-MSCs) could differentiate into sweat gland-like cells (SGLCs), but the underlying molecular mechanism remains unclear. Recently, microRNAs (miRNAs or miRs) are reported to manipulate many biological processes. However, whether the process of MSCs differentiation into sweat gland cells (SGCs) is regulated by miRNAs has not been reported. In this study, BM-MSCs were induced into SGLCs by co-culturing with SGCs. Differential expressions of miRNAs between BM-MSC and SGLCs were determined through miRNAs microarray and 68 miRNAs were found significantly changed in miRNA profile including hsa-miR-138-5p. Bioinformatics analysis showed that hsa-miR-138-5p targeted a group of nuclear factor-κB (NF-κB) related genes which play an important role in skin appendage development. As expected, hsa-miR-138-5p inhibitor transfected into BM-MSCs partly mimicked the effects of co-culture and increased the number of SGLCs by increasing the expression of NF-κB related genes. These results suggest that hsa-miR-138-5p and NF-κB are involved in the regulation of BM-MSCs differentiation into SGLCs. This study may also offer a new approach to yield SGCs for burn patients.
Collapse
|
8
|
Shu Y, Liu XB, Ma XH, Gao J, He W, Cao XY, Chen J. Immune response mechanism of mouse monocytes/macrophages treated with κ-carrageenan polysaccharide. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 53:191-198. [PMID: 28654831 DOI: 10.1016/j.etap.2017.06.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/10/2017] [Accepted: 06/19/2017] [Indexed: 06/07/2023]
Abstract
This study investigated the function of κ-carrageenan polysaccharide in immune regulation. The immune response of RAW 264.7 cells treated with κ-carrageenan polysaccharide was explored by MTT assay, general morphological observation, neutral red phagocytosis assay, Griess method, fluorescence method, and enzyme-linked immunosorbent assay. In addition, TLR4 blocking experiment and double-fluorescence immunostaining were performed on cells to demonstrate their immune response mechanism. Results show that κ-carrageenan polysaccharide not only promotes cell proliferation but also activates RAW 264.7 cells, thereby improving the cells' phagocytic capability, NO production, and tumor necrosis factor-α (TNF-α) secretion. In addition, the use of TLR4-specific inhibitors can significantly mediate the increased TNF-α secretion induced by κ-carrageenan polysaccharide. The RAW 264.7 cells treated with κ-carrageenan polysaccharide show upregulated TLR4 expression, and the main subunit of NF-κB (p65) is translocated. These results support the immunomodulatory function of κ-carrageenan polysaccharide in RAW 264.7 cells.
Collapse
Affiliation(s)
- Yang Shu
- Department of Food Science, College of Food Science, Hainan University, Haikou 570100, China
| | - Xue-Bing Liu
- Department of Food Science, College of Food Science, Hainan University, Haikou 570100, China
| | - Xin-Hui Ma
- Department of Food Science, College of Food Science, Hainan University, Haikou 570100, China
| | - Jian Gao
- Department of Chemical Engineering and Technology, College of Materials and Chemical Engineering, Hainan University, Haikou 570100, China
| | - Wei He
- Department of Food Science, College of Food Science, Hainan University, Haikou 570100, China
| | - Xian-Ying Cao
- Department of Food Science, College of Food Science, Hainan University, Haikou 570100, China.
| | - Jian Chen
- Department of Food Science, College of Food Science, Hainan University, Haikou 570100, China.
| |
Collapse
|
9
|
Toubiana J, Courtine E, Tores F, Asfar P, Daubin C, Rousseau C, Ouaaz F, Marin N, Cariou A, Chiche JD, Mira JP. Association of REL polymorphisms and outcome of patients with septic shock. Ann Intensive Care 2016; 6:28. [PMID: 27059500 PMCID: PMC4826362 DOI: 10.1186/s13613-016-0130-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 03/21/2016] [Indexed: 01/15/2023] Open
Abstract
Background cRel, a subunit of NF-κB, is implicated in the inflammatory response observed in autoimmune disease. Hence, knocked-out mice for cRel had a significantly higher mortality, providing new and important functions of cRel in the physiopathology of septic shock. Whether genetic variants in the human REL gene are associated with severity of septic shock is unknown. Methods We genotyped a population of 1040 ICU patients with septic shock and 855 ICU controls for two known polymorphisms of REL; REL rs842647 and REL rs13031237. Outcome of patients according to the presence of REL variant alleles was compared. Results The distribution of REL variant alleles was not significantly different between patients and controls. Among the septic shock group, REL rs13031237*T minor allele was not associated with worse outcome. In contrast, REL rs842647*G minor allele was significantly associated with more multi-organ failure and early death [OR 1.4; 95 % CI (1.02–1.8)]. Conclusion In a large ICU population, we report a significant clinical association between a variation in the human REL gene and severity and mortality of septic shock, suggesting for the first time a new insight into the role of cRel in response to infection in humans.
Collapse
Affiliation(s)
- Julie Toubiana
- Medical School, Paris Descartes University, Paris, France. .,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France. .,Department of Pediatric and Infectious Diseases, Necker University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Emilie Courtine
- Medical School, Paris Descartes University, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France
| | - Frederic Tores
- Bioinformatics Platform, Institut Imagine, Paris Descartes University- Sorbonne Paris Cité, 75015, Paris, France
| | - Pierre Asfar
- Medical Intensive Care Unit, Angers University Hospital, Angers, France
| | - Cédric Daubin
- Medical Intensive Care, Caen University Hospital, Caen, France
| | | | - Fatah Ouaaz
- INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France
| | - Nathalie Marin
- Intensive Care Unit, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alain Cariou
- Medical School, Paris Descartes University, Paris, France.,Intensive Care Unit, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Daniel Chiche
- Medical School, Paris Descartes University, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France.,Intensive Care Unit, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Paul Mira
- Medical School, Paris Descartes University, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France.,Intensive Care Unit, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
10
|
Facultative heterochromatin formation at the IL-1 beta promoter in LPS tolerance and sepsis. Cytokine 2011; 53:145-52. [PMID: 21078560 DOI: 10.1016/j.cyto.2010.10.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/23/2010] [Accepted: 10/20/2010] [Indexed: 12/27/2022]
Abstract
The clinical phenotype in sepsis that is observed as LPS tolerance is determined by silencing of pro-inflammatory genes like IL-1 beta (IL-1β). This study shows that facultative heterochromatin (fHC) silences IL-1β expression during sepsis, where we find dephosphorylated histone H3 serine 10 and increased binding of heterochromatin protein-1 (HP-1) to the promoter. In both human sepsis blood leukocytes and an LPS tolerant human THP-1 cell model, we show that IκBα and v-rel reticuloendotheliosis viral oncogene homolog B (RelB) function as dominant labile mediators of fHC formation at the IL-1β promoter. Protein synthesis inhibition decreases levels of IκBα and RelB, converts silent fHC to euchromatin, and restores IL-1β transcription. We further show TLR dependent NFκB p65 and histone H3 serine 10 phosphorylation binding at the promoter. We conclude that the resolution phase of sepsis, which correlates with survival in humans, may depend on the plasticity of chromatin structure as found in fHC.
Collapse
|
11
|
Ma X, Becker Buscaglia LE, Barker JR, Li Y. MicroRNAs in NF-kappaB signaling. J Mol Cell Biol 2011; 3:159-66. [PMID: 21502305 DOI: 10.1093/jmcb/mjr007] [Citation(s) in RCA: 480] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nuclear factor κB (NF-κB) is a transcriptional factor that regulates a battery of genes that are critical to innate and adaptive immunity, cell proliferation, inflammation, and tumor development. MicroRNAs (miRNAs) are short RNA molecules of 20-25 nucleotides in length that negatively regulate gene expression in animals and plants primarily by targeting 3' untranslated regions of mRNAs. In this work, we review the convergence of miRNAs and NF-κB signaling and dysregulation of miRNAs and NF-κB activation in human diseases, particularly in cancer. The function of miR-146, miR-155, miR-181b, miR-21, and miR-301a in NF-κB activation and their impact on tumorigenesis are discussed. Given that over 1000 human miRNAs have been identified, rendering miRNAs one of the most abundant classes of regulatory molecules, deciphering their biological function and pathological contribution in NF-κB dysregulation is essential to appreciate the complexity of immune systems and to develop therapeutics against cancer.
Collapse
Affiliation(s)
- Xiaodong Ma
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, 319 Abraham Flexner Way, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
12
|
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: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar 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
|
13
|
Chen X, El Gazzar M, Yoza BK, McCall CE. The NF-kappaB factor RelB and histone H3 lysine methyltransferase G9a directly interact to generate epigenetic silencing in endotoxin tolerance. J Biol Chem 2009; 284:27857-27865. [PMID: 19690169 DOI: 10.1074/jbc.m109.000950] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The interplay of transcription factors, histone modifiers, and DNA modification can alter chromatin structure that epigenetically controls gene transcription. During severe systemic inflammatory (SSI), the generation of facultative heterochromatin from euchromatin reversibly silences transcription of a set of acute proinflammatory genes. This gene-specific silencing is a salient feature of the endotoxin tolerant phenotype that is found in blood leukocytes of SSI patients and in a human THP-1 cell model of SSI. We previously reported that de novo induction of the NF-kappaB transcription factor RelB by endotoxin activation is necessary and sufficient for silencing transcription of acute proinflammatory genes in the endotoxin tolerant SSI phenotype. Here, we examined how RelB silences gene expression and found that RelB induces facultative heterochromatin formation by directly interacting with the histone H3 lysine 9 methyltransferase G9a. We found that heterochromatin protein 1 (HP1) and G9a formed a complex at the interleukin-1beta promoter that is dependent on the Rel homology domain (RHD) of RelB. RelB knockdown disassociated the complex and reversed transcription silencing. We also observed that whereas RelB chromatin binding was independent of G9a, RelB transcriptional silencing required G9a accumulation at the silenced promoter. Binding between RelB and G9a was confirmed by glutathione S-transferase pulldown in vitro and coimmunoprecipitation in vivo. These data provide novel insight into how RelB is required to initiate silencing in the phenotype associated with severe systemic inflammation in humans, a disease with major morbidity and mortality.
Collapse
Affiliation(s)
- Xiaoping Chen
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Mohamed El Gazzar
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Barbara K Yoza
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157; Department of General Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Charles E McCall
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157.
| |
Collapse
|
14
|
Ridder D, Schwaninger M. NF-κB signaling in cerebral ischemia. Neuroscience 2009; 158:995-1006. [DOI: 10.1016/j.neuroscience.2008.07.007] [Citation(s) in RCA: 294] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 06/27/2008] [Accepted: 07/03/2008] [Indexed: 01/04/2023]
|
15
|
Fagerlund R, Melén K, Cao X, Julkunen I. NF-kappaB p52, RelB and c-Rel are transported into the nucleus via a subset of importin alpha molecules. Cell Signal 2008; 20:1442-51. [PMID: 18462924 DOI: 10.1016/j.cellsig.2008.03.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Revised: 03/25/2008] [Accepted: 03/25/2008] [Indexed: 12/22/2022]
Abstract
In resting cells NF-kappaB transcription factors are retained in the cytoplasm as latent inactive complexes, until they are activated and rapidly transported into the nucleus. We show that all NF-kappaB proteins are imported into the nucleus via a subset of importin alpha isoforms. Our data indicate that the NF-kappaB components of the classical and alternative pathways have somewhat different specifities to importin alpha molecules. Based on the results from binding experiments of in vitro-translated and Sendai virus infection-induced or TNF-alpha-stimulated endogenous NF-kappaB proteins, it can be predicted that the specifity of NF-kappaB proteins to importin alpha molecules is different and changes upon the composition of the imported dimer. p52 protein binds directly to importin alpha3, alpha4, alpha5 and alpha6 and c-Rel binds to importin alpha5, alpha6 and alpha7 via a previously described monopartite nuclear localization signals (NLSs). Here we show that RelB, instead, has a bipartite arginine/lysine-rich NLS that mediates the binding of RelB to importin alpha5 and alpha6 and subsequent nuclear translocation of the protein. Moreover, we show that the nuclear import of p52/RelB heterodimers is mediated exclusively by the NLS of RelB. In addition, we found that the NLS of p52 mediates the nuclear import of p52/p65 heterodimers.
Collapse
Affiliation(s)
- Riku Fagerlund
- Department of Viral Diseases and Immunology, National Public Health Institute, FIN-00300, Helsinki, Finland.
| | | | | | | |
Collapse
|
16
|
Kim BJ, Kim SY, Lee H. Identification and characterization of human cdc7 nuclear retention and export sequences in the context of chromatin binding. J Biol Chem 2007; 282:30029-38. [PMID: 17711849 DOI: 10.1074/jbc.m703705200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The Cdc7 serine/threonine kinase activates the initiation of DNA replication by phosphorylating MCM proteins that are bound to the origins of DNA replication. We reported previously that human Cdc7 nuclear import is mediated directly by importin-beta through its binding to the Cdc7 nuclear localization sequence (NLS). Here, we report that human Cdc7 nuclear localization is regulated by two additional elements: nuclear retention (NRS) and export sequences (NES). Cdc7 proteins imported into the nucleus are retained in the nucleus by associating with chromatin, for which NRS-(306-326) is essential. Importantly, this binding appears to be specific to the origin of DNA replication, because the binding of wild-type Cdc7 to origin is 2.4-fold higher than to non-origin DNA. Furthermore, an NRS-defective Cdc7 mutant could not be retained in the nucleus, although it was imported into the nucleus normally. Together, our data suggest that NRS plays an important role in the activation of DNA replication by Cdc7. The Cdc7 proteins unassociated with chromatin are bound by CRM1 via two NES elements: NES1 at 458-467 within kinase insert III, and NES2 at 545-554 within the kinase IX domain. The primary function of the Cdc7-CRM1 association may be to translocate nuclear Cdc7 to the cytoplasm. However, the binding of CRM1 with Cdc7 at NES2 raises an interesting possibility that CRM1 may also down-regulate Cdc7 by masking its kinase domain.
Collapse
Affiliation(s)
- Byung Ju Kim
- Department of Biochemistry, Microbiology and Immunology, the Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1M 8M5, Canada
| | | | | |
Collapse
|
17
|
Abstract
Stimulus-induced nuclear factor-kappaB (NF-kappaB) activity, the central mediator of inflammatory responses and immune function, comprises a family of dimeric transcription factors that regulate diverse gene expression programs consisting of hundreds of genes. A family of inhibitor of kappaB (IkappaB) proteins controls NF-kappaB DNA-binding activity and nuclear localization. IkappaB protein metabolism is intricately regulated through stimulus-induced degradation and feedback re-synthesis, which allows for dynamic control of NF-kappaB activity. This network of interactions has been termed the NF-kappaB signaling module. Here, we summarize the current understanding of the molecular structures and biochemical mechanisms that determine NF-kappaB dimer formation and the signal-processing characteristics of the signaling module. We identify NF-kappaB-kappaB site interaction specificities and dynamic control of NF-kappaB activity as mechanisms that generate specificity in transcriptional regulation. We discuss examples of gene regulation that illustrate how these mechanisms may interface with other transcription regulators and promoter-associated events, and how these mechanisms suggest regulatory principles for NF-kappaB-mediated gene activation.
Collapse
Affiliation(s)
- A Hoffmann
- Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
18
|
Huang DB, Vu D, Ghosh G. NF-κB RelB Forms an Intertwined Homodimer. Structure 2005; 13:1365-73. [PMID: 16154093 DOI: 10.1016/j.str.2005.06.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2005] [Revised: 06/14/2005] [Accepted: 06/15/2005] [Indexed: 11/22/2022]
Abstract
The X-ray structure of the RelB dimerization domain (DD) reveals that the RelBDD assumes an unexpected intertwined fold topology atypical of other NF-kappaB dimers. All typical NF-kappaB dimers are formed by the association of two independently folded immunoglobulin (Ig) domains. In RelBDD, two polypeptides reconstruct both Ig domains in the dimer with an extra beta sheet connecting the two domains. Residues most critical to NF-kappaB dimer formation are invariant in RelB, and Y300 plays a positive role in RelBDD dimer formation. The presence of RelB-specific nonpolar residues at the surface removes several intradomain surface hydrogen bonds that may render the domain fold unstable. Intertwining may stabilize the RelBDD homodimer by forming the extra beta sheet. We show that, as in the crystal, RelB forms an intertwined homodimer in solution. We suggest that the transiently stable RelB homodimer might prevent its rapid degradation, allowing for heterodimer formation with p50 and p52.
Collapse
Affiliation(s)
- De-Bin Huang
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
19
|
Schjerven H, Tran TN, Brandtzaeg P, Johansen FE. De novo synthesized RelB mediates TNF-induced up-regulation of the human polymeric Ig receptor. THE JOURNAL OF IMMUNOLOGY 2004; 173:1849-57. [PMID: 15265917 DOI: 10.4049/jimmunol.173.3.1849] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Secretory Abs, which operate in a principally noninflammatory fashion, constitute the first line of acquired immune defense of mucosal surfaces. Such Abs are generated by polymeric Ig receptor (pIgR)-mediated export of dimeric IgA and pentameric IgM. TNF activates a proinflammatory gene repertoire in mucosal epithelial cells and also enhances pIgR expression. In this study we show that TNF-induced up-regulation of the human pIgR critically depends on an NF-kappa B site and flanking sequences within a 204-bp region of the first intron in the pIgR gene, a region largely overlapping with a recently characterized IL-4-responsive enhancer. The intronic NF-kappa B site was rapidly bound by NF-kappa B p65/p50 heterodimers present in nuclear extracts after TNF treatment of HT-29 cells, but a more delayed binding of RelB agreed better with the slow, protein synthesis-dependent, transcriptional activation of the pIgR gene. Overexpression of NF-kappa B p65 caused transient up-regulation of a pIgR-derived reporter gene, whereas overexpression of RelB showed a stronger and more sustained effect. Finally, we demonstrated that inhibition of endogenous RelB by RNA interference severely reduced the TNF responsiveness of our pIgR-derived reporter gene. Thus, TNF-induced signaling pathways required for up-regulated pIgR expression appear to differ from those of the proinflammatory gene repertoire.
Collapse
Affiliation(s)
- Hilde Schjerven
- Laboratory for Immunohistochemistry and Immunopathology, Institute and Department of Pathology, Rikshospitalet University Hospital, Oslo, Norway
| | | | | | | |
Collapse
|
20
|
Maier HJ, Marienfeld R, Wirth T, Baumann B. Critical role of RelB serine 368 for dimerization and p100 stabilization. J Biol Chem 2003; 278:39242-50. [PMID: 12874295 DOI: 10.1074/jbc.m301521200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In mature B cells RelB-containing complexes are constitutively present in the nucleus, and they are less susceptible to inhibitory kappaB proteins. In most other cell types inhibitory kappaB proteins prevent nuclear translocation and activation of NFkappaB. We reasoned that this characteristic might be because of post-translational modifications of RelB. In Drosophila, signal-dependent phosphorylation of the Rel homologue Dorsal at serine 317 has been shown to be critical for nuclear import. The evolutionary conservation of this serine prompted us to analyze the function of the corresponding site in RelB. As a model system we used the murine S107 plasmacytoma cell line, which lacks endogenous RelB expression. Analysis of S107 cells expressing wild type RelB and serine 368 mutants reveals that serine 368 is not required for nuclear import but that it is critical for RelB dimerization with other members of the NFkappaB family. Similar effects were obtained when the conserved serine in RelA was mutated. We further demonstrate that expression of functional RelB, but not of serine 368 mutants, severely reduces p52 generation and strongly increases expression of the p52 precursor, p100. Wild type RelB, but not mutant RelB, prolonged p100 half-life. We therefore suggest an inhibitory effect of RelB on p100 processing, which is possibly regulated in a signal-dependent manner.
Collapse
Affiliation(s)
- Harald Jakob Maier
- Department of Physiological Chemistry, Ulm University, Ulm 89081, Germany
| | | | | | | |
Collapse
|
21
|
Gapuzan MER, Pitoc GA, Gilmore TD. Mutations within a conserved protein kinase A recognition sequence confer temperature-sensitive and partially defective activities onto mouse c-Rel. Biochem Biophys Res Commun 2003; 307:92-9. [PMID: 12849986 DOI: 10.1016/s0006-291x(03)01123-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We have created two mutants of mouse transcription factor c-Rel (c-G29E and c-R266H) that are analogous to mutants previously shown to have temperature-sensitive (ts) functions for the homologous Drosophila protein Dorsal and the retroviral oncoprotein v-Rel. In vitro, c-R266H shows both a ts and a concentration-dependent ability to bind DNA, suggesting that the lesion affects the ability of c-Rel to form homodimers. In contrast, the ability of mouse c-G29E to bind DNA in vitro is not ts. c-Rel mutant c-R266H also shows a ts ability to activate transcription from a kappaB-site reporter plasmid, whereas c-G29E activates transcription well above control levels at both 33 and 39 degrees C. Insertion of two amino acids (Pro-Trp) between amino acids 266 and 267 in mouse c-Rel (mutant c-SPW) also creates a c-Rel protein with distinct properties: mutant c-SPW is partially defective in that it cannot form DNA-binding homodimers but can form DNA-binding heterodimers with p50. Interestingly, the mutations in c-Rel that affect homodimer formation (c-R266H and c-SPW) fall within a consensus protein kinase A recognition sequence but are not predicted to lie in the dimer interface. Conditional and partially defective mutants such as those described herein may be useful for identifying physiological responses and genes regulated by specific Rel/NF-kappaB family members.
Collapse
Affiliation(s)
- Maria-Emily R Gapuzan
- Biology Department, Boston University, 5 Cummington Street, Boston, MA 02215-2406, USA
| | | | | |
Collapse
|
22
|
Marienfeld R, May MJ, Berberich I, Serfling E, Ghosh S, Neumann M. RelB forms transcriptionally inactive complexes with RelA/p65. J Biol Chem 2003; 278:19852-60. [PMID: 12657634 DOI: 10.1074/jbc.m301945200] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RelB is an unusual member of the NF-kappaB transcription factor family that acts as both a transcriptional activator as well as a repressor of NF-kappaB-dependent gene expression. Although RelB promotes gene expression when it associates with p50/NF-kappaB1 or p52/NF-kappaB2, the precise molecular mechanisms through which it represses NF-kappaB remain unclear. To examine this inhibitory function in more detail, we employed reporter gene assays and found that RelB represses at the level of RelA. Furthermore, electrophoretic mobility shift analysis revealed that in vitro translated RelB impaired the DNA binding activity of RelA and that overexpressed RelB significantly reduced tumor necrosis factor-alpha-induced RelA activity in murine embryonic fibroblasts. Intriguingly, this inhibitory effect was due to the formation of RelA.RelB heterodimers that were unable to bind to kappaB sites in vitro strongly suggesting that these newly described NF-kappaB dimers cannot bind DNA. Expression pattern analysis revealed that RelA.RelB heterodimers appeared at relatively low levels in both lymphoid and non-lymphoid cells. However, the presence of these complexes increased following stimulation with phorbolesters or lipopolysaccharide or by overexpression of constitutively active IKKbeta. Functional characterization of RelA.RelB heterodimers in NIH3T3 murine embryonic fibroblasts revealed that they are not regulated by IkappaB proteins and are located in both the cytoplasm and the nucleus. Taken together, our findings demonstrate that sequestration of RelA in transcriptionally inactive RelA.RelB complexes provides a molecular mechanism that may explain the repressive role of RelB on NF-kappaB-dependent gene expression.
Collapse
Affiliation(s)
- Ralf Marienfeld
- Section of Immunobiology and Department of Molecular Biophysics and Biochemistry, Yale University Medical School, New Haven, Connecticut 06520, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Solan NJ, Miyoshi H, Carmona EM, Bren GD, Paya CV. RelB cellular regulation and transcriptional activity are regulated by p100. J Biol Chem 2002; 277:1405-18. [PMID: 11687592 DOI: 10.1074/jbc.m109619200] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RelB mediates the constitutive nuclear pool of NF-kappaB transcriptional activity in myeloid and lymphoid cells, which is believed to be secondary to its weak interaction with the classical NF-kappaB inhibitor proteins, the IkappaBs. In other cell types, RelB is located in the cytosol, thus suggesting that RelB is also regulated by an inhibitory protein(s). In this study, it is demonstrated that RelB is associated in the cytosol with p100 but not with IkappaBalpha, IkappaBbeta, IkappaBepsilon, nor p105. Its cytosolic control is not affected by stimuli that lead to RelA nuclear translocation, and RelB nuclear localization is prevented by p100, but not by p105 or IkappaBalpha. Structure function analysis p100-RelB interactions indicates that p100 amino acids 623-900 are required for effective interaction and repression of nuclear translocation and RelB driven NF-kappaB-dependent transcription. Moreover, this carboxyl-portion of p100 contains a nuclear export signal(s), which is required for effective retrieval of RelB from the nucleus. Finally, overexpression of NF-kappaB-inducing kinase, a kinase that has recently been shown to induce p100 processing, possibly through IKKalpha activation, causes nuclear translocation of RelB protein. Thus, these studies indicate that p100 is a bone fide inhibitor of RelB and that this transcription factor may be regulated by NF-kappaB-inducing kinase and/or IKKalpha.
Collapse
Affiliation(s)
- Nancie J Solan
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
24
|
Piffat KA, Hrdlicková R, Nehyba J, Ikeda T, Liss A, Huang S, Sif S, Gilmore TD, Bose HR. The chicken RelB transcription factor has transactivation sequences and a tissue-specific expression pattern that are distinct from mammalian RelB. MOLECULAR CELL BIOLOGY RESEARCH COMMUNICATIONS : MCBRC 2001; 4:266-75. [PMID: 11529676 DOI: 10.1006/mcbr.2001.0290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Rel/NF-kappaB proteins are eukaryotic transcription factors that control the expression of genes involved in a large variety of cellular processes. Rel proteins share a highly conserved DNA-binding/dimerization domain called the Rel Homology (RH) domain. We have constructed and characterized a composite cDNA encoding most of the chicken RelB transcription factor. The predicted chicken RelB protein has a high degree of sequence similarity to other vertebrate RelB proteins within the RH domain, but is much less conserved outside this domain. Chicken RelB does not bind DNA as a homodimer, but forms DNA-binding heterodimers with NF-kappaB p50 or p52. Overexpressed chicken RelB localizes to the nucleus in chicken embryo fibroblasts, and the nonconserved C-terminal sequences of chicken RelB contain a transactivation domain that functions in chicken and mouse fibroblasts. Thus, chicken RelB has functional properties similar to other vertebrate RelB proteins. However, Western blotting of diverse chicken tissues indicates that chicken RelB is more widely expressed than mammalian RelB.
Collapse
Affiliation(s)
- K A Piffat
- Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Hart DJ, Speight RE, Sutherland JD, Blackburn JM. Analysis of the NF-kappaB p50 dimer interface by diversity screening. J Mol Biol 2001; 310:563-75. [PMID: 11439024 DOI: 10.1006/jmbi.2001.4724] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An in vivo screen has been devised for NF-kappaB p50 activity in Escherichia coli exploiting the ability of the mammalian transcription factor to emulate a prokaryotic repressor. Active intracellular p50 was shown to repress the expression of a green fluorescent protein reporter gene allowing for visual screening of colonies expressing active p50 on agar plates. A library of mutants was constructed in which the residues Y267, L269, A308 and V310 of the dimer interface were simultaneously randomised and twenty-five novel functional interfaces were selected which repressed the reporter gene to similar levels as the wild-type protein. The leucine-269 alanine-308 core was repeatedly, but not exclusively, selected from the library whilst a diversity of predominantly non-polar residues were selected at positions 267 and 310. These results indicate that L269 and A308 may form a hot spot of interaction and allow an insight into the processes of dimer selectivity and evolution within this family of transcription factors.
Collapse
Affiliation(s)
- D J Hart
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK
| | | | | | | |
Collapse
|
26
|
López-Rodríguez C, Aramburu J, Jin L, Rakeman AS, Michino M, Rao A. Bridging the NFAT and NF-kappaB families: NFAT5 dimerization regulates cytokine gene transcription in response to osmotic stress. Immunity 2001; 15:47-58. [PMID: 11485737 DOI: 10.1016/s1074-7613(01)00165-0] [Citation(s) in RCA: 176] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The transcription factor NFAT5/TonEBP is evolutionarily the oldest member of the NFAT/Rel family of transcription factors. We show that NFAT5 is uniquely related to NF-kappaB and is the only member of the Rel/NFAT family to be activated by osmotic stress. Like Rel/NF-kappaB proteins but unlike the calcium-regulated NFAT proteins, NFAT5 is constitutively dimeric, and dimerization is essential for DNA binding and transcriptional activity. Using dominant-negative proteins that inhibit NFAT5 dimerization, we show that NFAT5 regulates expression of the TNFalpha and lymphotoxin-beta genes in osmotically stressed T cells. Chromatin immunoprecipitation experiments confirm that NFAT5 binds to the TNFalpha promoter in vivo. We suggest that NFAT5 participates in specific aspects of host defense by upregulating TNF family genes and other target genes in T cells.
Collapse
Affiliation(s)
- C López-Rodríguez
- The Center for Blood Research and, Department of Pathology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
27
|
Huxford T, Malek S, Ghosh G. Structure and mechanism in NF-kappa B/I kappa B signaling. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2001; 64:533-40. [PMID: 11232330 DOI: 10.1101/sqb.1999.64.533] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- T Huxford
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093-0359, USA
| | | | | |
Collapse
|
28
|
O'Sullivan BJ, MacDonald KP, Pettit AR, Thomas R. RelB nuclear translocation regulates B cell MHC molecule, CD40 expression, and antigen-presenting cell function. Proc Natl Acad Sci U S A 2000; 97:11421-6. [PMID: 11027342 PMCID: PMC17215 DOI: 10.1073/pnas.97.21.11421] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mice with targeted RelB mutations demonstrated an essential role for RelB in immune responses and in myeloid dendritic cell differentiation. Human studies suggested a more global transcriptional role in antigen presentation. Burkitt lymphoma cell lines were used as a model to examine the role of RelB in antigen presentation. After transient transfection of BJAB with RelB, strong nuclear expression of RelB-p50 heterodimers was associated with increased APC function and expression of CD40 and MHC class I. Antisense RelB in DG75 reduced antigen-presenting capacity and CD40-mediated up-regulation of MHC molecules. The data indicate that RelB transcriptional activity directly affects antigen presentation and CD40 synthesis. Stimulation of RelB transcriptional activity may provide a positive feedback loop for facilitating productive APC/T cell interactions.
Collapse
Affiliation(s)
- B J O'Sullivan
- Centre for Immunology and Cancer Research, University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland, 4102, Australia
| | | | | | | |
Collapse
|
29
|
Sengchanthalangsy LL, Datta S, Huang DB, Anderson E, Braswell EH, Ghosh G. Characterization of the dimer interface of transcription factor NFkappaB p50 homodimer. J Mol Biol 1999; 289:1029-40. [PMID: 10369780 DOI: 10.1006/jmbi.1999.2823] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dimers of the Rel/NFkappaB transcription factor family form with differential stabilities through the combinatorial association of five polypeptides: p50, p52, p65, cRel, and RelB. Here, we have characterized the nature of the monomer-dimer equilibrium of the p50 homodimer. Sedimentation equilibrium studies show that the equilibrium constant for p50 dimer dissociation is in the low micromolar range. Using the X-ray crystal structure of the p50 homodimer as a guide, we have created site-directed alanine mutations at ten dimer-forming residues in p50 and measured their effects on p50 homodimerization. Characterization of these alanine mutants by a series of chemical crosslinking, size-exclusion chromatography, and sedimentation equilibrium experiments shows that the most critical residue in stabilizing the p50 dimer interface is Y267. Sedimentation equilibrium experiments show that an alanine substitution at position 267 destabilizes the dimer interface by 2.0 kcal/mol. Alanine substitutions at two other positions, L269 and V310, significantly destabilize the p50 dimer interface. These two residues are observed to mediate critical interactions in the crystal structure. Together, these three residues constitute the "hot-spot" of protein-protein interaction in p50 dimerization. Of the four charged residues in the dimer interface, R252, D254, E265, and D302, only D302 contributes significantly to p50 dimer stability. D254 appears to slightly destabilize the subunit interface. Although residues H304, R305, and F307 occupy positions at the hydrophobic core of the interface and appear to be involved in multiple interactions in the X-ray crystal structure, alanine substitutions at these positions do not significantly reduce the affinity for p50 dimerization. Upon evaluating the roles of these amino acid residues at the p50 dimer interface, we propose that differential contributions of a few key residues dictate the selectivity of dimer formation within the Rel/NFkappaB family.
Collapse
Affiliation(s)
- L L Sengchanthalangsy
- Department of Chemistry and Biochemistry, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0359, USA
| | | | | | | | | | | |
Collapse
|
30
|
Huang DB, Huxford T, Chen YQ, Ghosh G. The role of DNA in the mechanism of NFkappaB dimer formation: crystal structures of the dimerization domains of the p50 and p65 subunits. Structure 1997; 5:1427-36. [PMID: 9384558 DOI: 10.1016/s0969-2126(97)00293-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Members of the rel/NFkappaB family of transcription factors play a vital role in the regulation of rapid cellular responses, such as those required to fight infection or react to cellular stress. Members of this family of proteins form homo- and heterodimers with differing affinities for dimerization. They share a structural motif known as the rel homology region (RHR), the C-terminal one third of which mediates protein dimerization. Crystal structures of the rel/NFkappaB family members p50 and p65 in their DNA-bound homodimeric form have been solved. These structures showed that the residues from the dimerization domains of both p50 and p65 participate in DNA binding and that the DNA-protein and protein dimerization surfaces form one continuous overlapping interface. We desired to investigate the contribution of DNA to NFkappaB dimerization and to identify the mechanism for the selective association of rel/NFkappaB family peptides into transcriptionally active dimers. RESULTS We report here the crystal structures of the dimerization domains of murine p50 and p65 at 2.2 A and 2.0 A resolution, respectively. A comparison of these two structures suggests that conservative amino acid changes at three positions are responsible for the differences in their dimer interfaces. The presence of the target DNA does not change the dimer interface of either protein in any significant manner. CONCLUSIONS These two structures suggest that the rel/NFkappaB family of transcription factors use only a few conservative changes in their amino acid sequences to form a host of dimers with varying affinities for dimerization. Amino acids at positions corresponding to 254, 267, and 307 of murine p50, function as primary determinants for the observed differences in dimerization affinity. The DNA-contacting charged amino acid sidechains from the dimerization domains are held in a similar conformation in both the DNA-bound and free states, therefore, no major structural rearrangement is required to bring these residues into contact with the DNA.
Collapse
Affiliation(s)
- D B Huang
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0359, USA
| | | | | | | |
Collapse
|
31
|
Betts JC, Nabel GJ. Differential regulation of NF-kappaB2(p100) processing and control by amino-terminal sequences. Mol Cell Biol 1996; 16:6363-71. [PMID: 8887665 PMCID: PMC231638 DOI: 10.1128/mcb.16.11.6363] [Citation(s) in RCA: 82] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Proteolytic degradation of the C-terminal region of NF-(kappa)B precursors to their active DNA binding forms represents an important regulatory step in the activation of NF-(kappa)B. NF-(kappa)B2(p100) is found ubiquitously in the cytoplasm; however, the site and mechanism of processing to p52 have not previously been defined. We show by deletion mapping that processing of NF-(kappa)B2(p100) terminates at alanine 405 to generate p52 and is prevented by specific inhibitors of the multicatalytic proteinase complex. Although the C-terminal I(kappa)B-like domain of NF-(kappa)B2(p100) was constitutively phosphorylated, disruption of this phosphorylation by mutagenesis demonstrated that it was not required as a signal to mediate processing. Mutational analysis further showed that cleavage of NF-(kappa)B2 is not dependent on a specific sequence motif adjacent to alanine 405, the ankyrin repeats, or other C-terminal sequences but is directed by structural determinants amino terminal to the cleavage site, within the Rel homology domain and/or the glycine hinge region. The level of processing of NF-(kappa)B2(p100) was much lower than that of NF-(kappa)B1(p105) and differed from that of I(kappa)B-alpha, suggesting differential control of processing of NF-(kappa)B/I(kappa)B family members.
Collapse
Affiliation(s)
- J C Betts
- Department of Internal Medicine, Howard Hughes Medical Institute, University of Michigan Medical Center, Ann Arbor 48109-0650, USA
| | | |
Collapse
|
32
|
Abstract
The transcription factor NF-kappa B has attracted widespread attention among researchers in many fields based on the following: its unusual and rapid regulation, the wide range of genes that it controls, its central role in immunological processes, the complexity of its subunits, and its apparent involvement in several diseases. A primary level of control for NF-kappa B is through interactions with an inhibitor protein called I kappa B. Recent evidence confirms the existence of multiple forms of I kappa B that appear to regulate NF-kappa B by distinct mechanisms. NF-kappa B can be activated by exposure of cells to LPS or inflammatory cytokines such as TNF or IL-1, viral infection or expression of certain viral gene products, UV irradiation, B or T cell activation, and by other physiological and nonphysiological stimuli. Activation of NF-kappa B to move into the nucleus is controlled by the targeted phosphorylation and subsequent degradation of I kappa B. Exciting new research has elaborated several important and unexpected findings that explain mechanisms involved in the activation of NF-kappa B. In the nucleus, NF-kappa B dimers bind to target DNA elements and activate transcription of genes encoding proteins involved with immune or inflammation responses and with cell growth control. Recent data provide evidence that NF-kappa B is constitutively active in several cell types, potentially playing unexpected roles in regulation of gene expression. In addition to advances in describing the mechanisms of NF-kappa B activation, excitement in NF-kappa B research has been generated by the first report of a crystal structure for one form of NF-kappa B, the first gene knockout studies for different forms of NF-kB and of I kappa B, and the implications for therapies of diseases thought to involve the inappropriate activation of NF-kappa B.
Collapse
Affiliation(s)
- A S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill 27599, USA
| |
Collapse
|
33
|
White DW, Pitoc GA, Gilmore TD. Interaction of the v-Rel oncoprotein with NF-kappaB and IkappaB proteins: heterodimers of a transformation-defective v-Rel mutant and NF-2 are functional in vitro and in vivo. Mol Cell Biol 1996; 16:1169-78. [PMID: 8622661 PMCID: PMC231099 DOI: 10.1128/mcb.16.3.1169] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The v-Rel oncoprotein of the avian Rev-T retrovirus is a member of the Rel/NF-kappa B family of transcription factors. The mechanism by which v-Rel malignantly transforms chicken spleen cells is not precisely known. To gain a better understanding of functions needed for transformation by v-Rel, we have now characterized the activities of mutant v-Rel proteins that are defective for specific protein-protein interactions. Mutant v-delta NLS, which has a deletion of the primary v-Rel nuclear localizing sequence, does not interact efficiently with I kappa B-alpha but still transforms chicken spleen cells approximately as well as wild-type v-Rel, indicating that interaction with I kappa B-alpha is not essential for the v-Rel transforming function. A second v-Rel mutant, v-SPW, has been shown to be defective for the formation of homodimers, DNA binding, and transformation. However, we now find that v-SPW can form functional DNA-binding heterodimers in vitro and in vivo with the cellular protein NF-kappa B p-52. Most strikingly, coexpression of v-SPW and p52 from a retroviral vector can induce the malignant transformation of chicken spleen cells, whereas expression of either protein alone cannot. Our results are most consistent with a model wherein Rel homodimers or heterodimers must bind DNA and alter gene expression in order to transform lymphoid cells.
Collapse
Affiliation(s)
- D W White
- Department of Biology, Boston Univeristy, Massachusetts 02215, USA
| | | | | |
Collapse
|