1
|
Yamada A, Wake K, Imaoka S, Motoyoshi M, Yamamoto T, Asano M. Analysis of the effects of importin α1 on the nuclear translocation of IL-1α in HeLa cells. Sci Rep 2024; 14:1322. [PMID: 38225348 PMCID: PMC10789739 DOI: 10.1038/s41598-024-51521-w] [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] [Scholar Register] [Received: 03/13/2023] [Accepted: 01/06/2024] [Indexed: 01/17/2024] Open
Abstract
Interleukin-1α (IL-1α), a cytokine released by necrotic cells, causes sterile inflammation. On the other hand, IL-1α is present in the nucleus and also regulates the expression of many proteins. A protein substrate containing a classical nuclear localization signal (cNLS) typically forms a substrate/importin α/β complex, which is subsequently transported to the nucleus. To the best of our knowledge, no study has directly investigated whether IL-1α-which includes cNLS-is imported into the nucleus in an importin α/β-dependent manner. In this study, we noted that all detected importin α subtypes interacted with IL-1α. In HeLa cells, importin α1-mediated nuclear translocation of IL-1α occurred at steady state and was independent of importin β1. Importin α1 not only was engaged in IL-1α nuclear transport but also concurrently functioned as a molecule that regulated IL-1α protein level in the cell. Furthermore, we discussed the underlying mechanism of IL-1α nuclear translocation by importin α1 based on our findings.
Collapse
Affiliation(s)
- Akiko Yamada
- Department of Pathology, Nihon University School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
- Division of Immunology and Pathobiology, Dental Research Center, Nihon University School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| | - Kiyotaka Wake
- Department of Orthodontics, Nihon University School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Oral Structural and Functional Biology, Nihon University Graduate School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Saya Imaoka
- Department of Pathology, Nihon University School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Immunology and Pathobiology, Dental Research Center, Nihon University School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Mitsuru Motoyoshi
- Department of Orthodontics, Nihon University School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Clinical Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Takenori Yamamoto
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-9501, Japan
- Institute for Genome Research, Tokushima University, Kuramotocho-3, Tokushima, 770-8503, Japan
| | - Masatake Asano
- Department of Pathology, Nihon University School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Immunology and Pathobiology, Dental Research Center, Nihon University School of Dentistry, 1-8-13, Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| |
Collapse
|
2
|
Niklander SE, Murdoch C, Hunter KD. IL-1/IL-1R Signaling in Head and Neck Cancer. FRONTIERS IN ORAL HEALTH 2021; 2:722676. [PMID: 35048046 PMCID: PMC8757896 DOI: 10.3389/froh.2021.722676] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/04/2021] [Indexed: 01/22/2023] Open
Abstract
Decades ago, the study of cancer biology was mainly focused on the tumor itself, paying little attention to the tumor microenvironment (TME). Currently, it is well recognized that the TME plays a vital role in cancer development and progression, with emerging treatment strategies focusing on different components of the TME, including tumoral cells, blood vessels, fibroblasts, senescent cells, inflammatory cells, inflammatory factors, among others. There is a well-accepted relationship between chronic inflammation and cancer development. Interleukin-1 (IL-1), a potent pro-inflammatory cytokine commonly found at tumor sites, is considered one of the most important inflammatory factors in cancer, and has been related with carcinogenesis, tumor growth and metastasis. Increasing evidence has linked development of head and neck squamous cell carcinoma (HNSCC) with chronic inflammation, and particularly, with IL-1 signaling. This review focuses on the most important members of the IL-1 family, with emphasis on how their aberrant expression can promote HNSCC development and metastasis, highlighting possible clinical applications.
Collapse
Affiliation(s)
- Sven E. Niklander
- Unidad de Patología y Medicina Oral, Facultad de Odontologia, Universidad Andres Bello, Viña del Mar, Chile
| | - Craig Murdoch
- Unit of Oral and Maxillofacial Medicine, Pathology and Surgery, School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Keith D. Hunter
- Unit of Oral and Maxillofacial Medicine, Pathology and Surgery, School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
- Oral Biology and Pathology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
3
|
Dagvadorj J, Mikulska-Ruminska K, Tumurkhuu G, Ratsimandresy RA, Carriere J, Andres AM, Marek-Iannucci S, Song Y, Chen S, Lane M, Dorfleutner A, Gottlieb RA, Stehlik C, Cassel S, Sutterwala FS, Bahar I, Crother TR, Arditi M. Recruitment of pro-IL-1α to mitochondrial cardiolipin, via shared LC3 binding domain, inhibits mitophagy and drives maximal NLRP3 activation. Proc Natl Acad Sci U S A 2021; 118:e2015632118. [PMID: 33361152 PMCID: PMC7817159 DOI: 10.1073/pnas.2015632118] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The balance between NLRP3 inflammasome activation and mitophagy is essential for homeostasis and cellular health, but this relationship remains poorly understood. Here we found that interleukin-1α (IL-1α)-deficient macrophages have reduced caspase-1 activity and diminished IL-1β release, concurrent with reduced mitochondrial damage, suggesting a role for IL-1α in regulating this balance. LPS priming of macrophages induced pro-IL-1α translocation to mitochondria, where it directly interacted with mitochondrial cardiolipin (CL). Computational modeling revealed a likely CL binding motif in pro-IL-1α, similar to that found in LC3b. Thus, binding of pro-IL-1α to CL in activated macrophages may interrupt CL-LC3b-dependent mitophagy, leading to enhanced Nlrp3 inflammasome activation and more robust IL-1β production. Mutation of pro-IL-1α residues predicted to be involved in CL binding resulted in reduced pro-IL-1α-CL interaction, a reduction in NLRP3 inflammasome activity, and increased mitophagy. These data identify a function for pro-IL-1α in regulating mitophagy and the potency of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Jargalsaikhan Dagvadorj
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Karolina Mikulska-Ruminska
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213
- Institute of Physics, Faculty of Physics Astronomy and Informatics, Nicolaus Copernicus University in Toruń, 87-100 Torun, Poland
| | - Gantsetseg Tumurkhuu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | | | - Jessica Carriere
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Allen M Andres
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Stefanie Marek-Iannucci
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Yang Song
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Shuang Chen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics. David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Malcolm Lane
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Andrea Dorfleutner
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Roberta A Gottlieb
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Christian Stehlik
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Suzanne Cassel
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Fayyaz S Sutterwala
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213;
| | - Timothy R Crother
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048;
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics. David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Moshe Arditi
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048;
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics. David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| |
Collapse
|
4
|
CRISPR/Cas9 mediated mutation of mouse IL-1α nuclear localisation sequence abolishes expression. Sci Rep 2017; 7:17077. [PMID: 29213066 PMCID: PMC5719027 DOI: 10.1038/s41598-017-17387-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023] Open
Abstract
Inflammation is a host defense process against infection. Inflammatory mediators include cytokines of the interleukin-1 family, such as IL-1α and IL-1β. Unlike IL-1β, IL-1α carries an N-terminal nuclear localisation sequence (NLS) and is trafficked to the nucleus. The importance of IL-1α nuclear localisation is poorly understood. Here, we used CRISPR/Cas9 to make inactivating mutations to the NLS on the Il1a gene. A colony of NLS mutant mice was successfully generated with precise knock-in mutations to incapacitate NLS function. NLS mutant mice had no gross changes in immunophenotype or inflammatory response but, surprisingly, failed to express IL-1α. We deduced that, in making specific mutations in the Il1a gene, we also mutated a long-noncoding (lnc)RNA in the complementary strand which has cis-regulatory transcriptional control of the Il1a gene itself. The mutations generated in the Il1a gene also result in mutation of the lncRNA sequence and a predicted alteration of its secondary structure, potentially explaining a subsequent failure to function as a transcriptional activator of Il1a expression. Thus, lncRNA secondary structure may regulate IL-1α expression. Our results serve as a cautionary note that CRISPR -mediated genome editing without full knowledge of genomic context can result in unexpected, yet potentially informative observations.
Collapse
|
5
|
Rider P, Voronov E, Dinarello CA, Apte RN, Cohen I. Alarmins: Feel the Stress. THE JOURNAL OF IMMUNOLOGY 2017; 198:1395-1402. [PMID: 28167650 DOI: 10.4049/jimmunol.1601342] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/27/2016] [Indexed: 12/20/2022]
Abstract
Over the last decade, danger-associated molecular pattern molecules, or alarmins, have been recognized as signaling mediators of sterile inflammatory responses after trauma and injury. In contrast with the accepted passive release models suggested by the "danger hypothesis," it was recently shown that alarmins can also directly sense and report damage by signaling to the environment when released from live cells undergoing physiological stress, even without loss of subcellular compartmentalization. In this article, we review the involvement of alarmins such as IL-1α, IL-33, IL-16, and high-mobility group box 1 in cellular and physiological stress, and suggest a novel activity of these molecules as central initiators of sterile inflammation in response to nonlethal stress, a function we denote "stressorins." We highlight the role of posttranslational modifications of stressorins as key regulators of their activity and propose that targeted inhibition of stressorins or their modifiers could serve as attractive new anti-inflammatory treatments for a broad range of diseases.
Collapse
Affiliation(s)
- Peleg Rider
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | | | - Ron N Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Idan Cohen
- Faculty of Medicine, Galilee Medical Center, Nahariya Hospital, 22100 Nahariya, Israel
| |
Collapse
|
6
|
Almog T, Kandel-Kfir M, Shaish A, Dissen M, Shlomai G, Voronov E, Apte RN, Harats D, Kamari Y. Knockdown of interleukin-1α does not attenuate LPS-induced production of interleukin-1β in mouse macrophages. Cytokine 2015; 73:138-43. [DOI: 10.1016/j.cyto.2015.01.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 12/15/2014] [Accepted: 01/23/2015] [Indexed: 12/25/2022]
|
7
|
Li X, Li T, You B, Shan Y, Shi S, Cao X, Qian L. Expression and Function of HAX-1 in Human Cutaneous Squamous Cell Carcinoma. J Cancer 2015; 6:351-9. [PMID: 25767605 PMCID: PMC4349875 DOI: 10.7150/jca.11093] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/01/2015] [Indexed: 01/08/2023] Open
Abstract
HAX-1 is an anti-apoptotic factor and overexpressed in many types of cancers. However, the functional role of HAX-1 in human cutaneous squamous cell carcinoma (cSCC) remains unclear. Our aim was to investigate the expression of HAX-1 in cSCC and its relationship with the development of cSCC. HAX-1 expression in cSCC tissues and in-vitro cell models were evaluated by real-time quantitative PCR (RT-qPCR), Western blot and immunohistochemistry. And the RNAi strategy was used to observe the relationship of HAX-1 and cSCC in A431 cells. The mRNA and protein level of HAX-1 were significantly higher in cSCC compared with normal tissue. There were significant differences in thickness (P=0.014), differentiation (P=0.027) and TNM stages (P=0.007). After knockdown the expression of HAX-1 by siRNA, the proliferation and the motility of A431 cell was inhibited obviously, and the apoptosis of A431 cells were induced too. HAX-1 may be a risk factor for patients with cSCC. As a potential tumor promoter in cSCC, HAX-1 may be a novel potential therapeutic target for cSCC treatment and deserves further investigation.
Collapse
Affiliation(s)
- Xingyu Li
- 1. Department of Pathological Anatomy, Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Tiejun Li
- 1. Department of Pathological Anatomy, Medical School of Nantong University, Nantong 226001, Jiangsu, China; ; 2. Small RNA Technology and Application Institute, Nantong University, E&T Development Area, Nantong 226016, Jiangsu, China
| | - Bo You
- 3. Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Yin Shan
- 3. Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Si Shi
- 3. Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Xiaolei Cao
- 1. Department of Pathological Anatomy, Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Li Qian
- 4. Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| |
Collapse
|
8
|
Lamacchia C, Rodriguez E, Palmer G, Gabay C. Endogenous IL-1α is a chromatin-associated protein in mouse macrophages. Cytokine 2013; 63:135-44. [PMID: 23684408 DOI: 10.1016/j.cyto.2013.04.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 03/19/2013] [Accepted: 04/10/2013] [Indexed: 12/21/2022]
Abstract
The cytokine interleukin-1α (IL-1α) is synthesized as a 31kDa peptide that lacks a leader peptide and is not secreted by the conventional secretory pathway. A distinctive characteristic of pro-IL-1α is the presence of a nuclear localization sequence in its amino-terminal moiety that allows its translocation to the nucleus. However no nuclear function(s) of the endogenous pro-IL-1α has been reported to date. In the present study, we used murine macrophages that produce IL-1α in response to pro-inflammatory stimuli, to gain further insight into the biology of the endogenous IL-1α protein in innate immune cells. We show that endogenous IL-1α is essentially found as a chromatin-associated nuclear protein in LPS-stimulated macrophages. In contrast to IL-1β, IL-1α was not released upon inflammasome activation unless significant cell damage occurred. IL-1β mRNA and protein levels were specifically decreased in IL-1α deficient macrophages after LPS stimulation. However, overexpression of human pro-IL-1α did not rescue this defective IL-1β production, suggesting that this finding might be related to the insertion of the targeting construct into the IL-1 locus, rather than to a specific nuclear function of pro-IL-1α. Finally, by using both genomic and proteomic approaches, we could not identify a nuclear function of IL-1α. Taken together, these observations suggest that in macrophages IL-1α primarily acts as an alarmin that is rapidly released upon cell damage to activate early mechanisms of host defense.
Collapse
Affiliation(s)
- Céline Lamacchia
- Division of Rheumatology, Department of Internal Medicine, University Hospital of Geneva, 26 Avenue Beau-Séjour, 1211 Geneva 14, Switzerland.
| | | | | | | |
Collapse
|
9
|
Mitogen-activated protein kinase-dependent interleukin-1α intracrine signaling is modulated by YopP during Yersinia enterocolitica infection. Infect Immun 2011; 80:289-97. [PMID: 22083707 DOI: 10.1128/iai.05742-11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Yersinia enterocolitica is a food-borne pathogen that preferentially infects the Peyer's patches and mesenteric lymph nodes, causing an acute inflammatory reaction. Even though Y. enterocolitica induces a robust inflammatory response during infection, the bacterium has evolved a number of virulence factors to limit the extent of this response. We previously demonstrated that interleukin-1α (IL-1α) was critical for the induction of gut inflammation characteristic of Y. enterocolitica infection. More recently, the known actions of IL-1α are becoming more complex because IL-1α can function both as a proinflammatory cytokine and as a nuclear factor. In this study, we tested the ability of Y. enterocolitica to modulate intracellular IL-1α-dependent IL-8 production in epithelial cells. Nuclear translocation of pre-IL-1α protein and IL-1α-dependent secretion of IL-8 into the culture supernatant were increased during infection with a strain lacking the 70-kDa virulence plasmid compared to the case during infection with the wild type, suggesting that Yersinia outer proteins (Yops) might be involved in modulating intracellular IL-1α signaling. Infection of HeLa cells with a strain lacking the yopP gene resulted in increased nuclear translocation of pre-IL-1α and IL-1α-dependent secretion of IL-8 similar to what is observed with bacteria lacking the virulence plasmid. YopP is a protein acetylase that inhibits mitogen-activated protein kinase (MAP kinase)- and NF-κB-dependent signal transduction pathways. Nuclear translocation of pre-IL-1α and IL-1α-dependent secretion of IL-8 in response to Yersinia enterocolitica infection were dependent on extracellular signal-regulated kinase (ERK) and p38 MAP kinase signaling but independent of NF-κB. These data suggest that Y. enterocolitica inhibits intracellular pre-IL-1α signaling and subsequent proinflammatory responses through inhibition of MAP kinase pathways.
Collapse
|
10
|
Reduced atherosclerosis and inflammatory cytokines in apolipoprotein-E-deficient mice lacking bone marrow-derived interleukin-1α. Biochem Biophys Res Commun 2011; 405:197-203. [PMID: 21219852 DOI: 10.1016/j.bbrc.2011.01.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 01/04/2011] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Interleukin (IL)-1α and IL-1β are products of macrophages, endothelial cells and vascular smooth muscle cells; moreover, each of these cell types is affected by the pro-inflammatory properties of both IL-1's. Whereas several studies demonstrate the proatherogenic properties of IL-1β, the role of IL-1α in atherogenesis remains unclear. We assessed whether IL-1α and IL-1β from tissue resident vascular cells or emigrating bone marrow-derived cells promote the development of atherosclerosis in apoE-/- mice and determined the effect of selective macrophage IL-1α or IL-1β deficiency on degradation of LDL and cytokine production. METHODS We generated strains of double knock-out (KO) mice (apoE-/-/IL-1α-/- and apoE-/-/IL-1β-/-) and created chimeras consisting of apoE-/- mice reconstituted with bone marrow-derived cells from apoE-/-/IL-1+/+, apoE-/-/IL-1α-/- and apoE-/-/IL-1β-/-. RESULTS The areas of aortic sinus lesions were lower in either double KO mice compared to solely apoE-/- mice, despite higher non-HDL cholesterol levels. Importantly, selective deficiency of IL-1α or IL-1β in bone marrow-derived cells inhibited atherogenesis to the same extent as in double KO mice without affecting plasma lipids. Aortic sinus lesions in apoE-/- mice transplanted with IL-1β-/- or IL-1α-/- cells were 32% and 52% lower, respectively, than in IL-1+/+ transplanted mice. Ex vivo, isolated IL-1α-/- macrophages from atherosclerotic mice degraded LDL and secreted IL-6, TNFα and IL-12 similarly to IL-1+/+ macrophages; however, IL-1α deficient macrophages secreted reduced levels of IL-1β (-50%) and 2-3-fold higher levels of the anti-inflammatory cytokine IL-10. CONCLUSION We show for the first time that it is IL-1α from bone marrow-derived cells that accelerates atherogenesis in apoE-deficient mice rather than constitutive IL-1α in vascular cells, possibly by increasing the inflammatory cytokine profile of macrophages.
Collapse
|
11
|
Trebinska A, Rembiszewska A, Ciosek K, Ptaszynski K, Rowinski S, Kupryjanczyk J, Siedlecki JA, Grzybowska EA. HAX-1 overexpression, splicing and cellular localization in tumors. BMC Cancer 2010; 10:76. [PMID: 20196840 PMCID: PMC2843675 DOI: 10.1186/1471-2407-10-76] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Accepted: 03/02/2010] [Indexed: 12/01/2022] Open
Abstract
Background HAX-1 has been described as a protein potentially involved in carcinogenesis and especially metastasis. Its involvement in regulation of apoptosis and cell migration along with some data indicating its overexpression in cancer cell lines and tumors suggests that HAX-1 may play a role in neoplastic transformation. Here we present the first systematic analysis of HAX-1 expression in several solid tumors. Methods Using quantitative RT-PCR, we have determined the mRNA levels of HAX1 splice variant I in several solid tumors. We have also analyzed by semiquantitative and quantitative RT-PCR the expression of five HAX-1 splice variants in breast cancer samples and in normal tissue from the same individuals. Quantitative PCR was also employed to analyze the effect of estrogen on HAX1 expression in breast cancer cell line. Immunohistochemical analysis of HAX-1 was performed on normal and breast cancer samples. Results The results reveal statistically important HAX1 up-regulation in breast cancer, lung cancer and melanoma, along with some minor variations in the splicing pattern. HAX-1 up-regulation in breast cancer samples was confirmed by immunohistochemical analysis, which also revealed an intriguing HAX-1 localization in the nuclei of the tumor cells, associated with strong ER status. Conclusion HAX-1 elevated levels in cancer tissues point to its involvement in neoplastic transformation, especially in breast cancer. The connection between HAX-1 nuclear location and ER status in breast cancer samples remains to be clarified.
Collapse
|
12
|
Luheshi NM, Rothwell NJ, Brough D. Dual functionality of interleukin-1 family cytokines: implications for anti-interleukin-1 therapy. Br J Pharmacol 2010; 157:1318-29. [PMID: 19681864 PMCID: PMC2765320 DOI: 10.1111/j.1476-5381.2009.00331.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dysregulated inflammation contributes to disease pathogenesis in both the periphery and the brain. Cytokines are coordinators of inflammation and were originally defined as secreted mediators, released from expressing cells to activate plasma membrane receptors on responsive cells. However, a group of cytokines is now recognized as having dual functionality. In addition to their extracellular effects, these cytokines act inside the nuclei of cytokine-expressing or cytokine-responsive cells. Interleukin-1 (IL-1) family cytokines are key pro-inflammatory mediators, and blockade of the IL-1 system in inflammatory diseases is an attractive therapeutic goal. All current therapies target IL-1 extracellular actions. Here we review evidence that suggests IL-1 family members have dual functionality. Several IL-1 family members have been detected inside the nuclei of IL-1-expressing or IL-1-responsive cells, and intranuclear IL-1 is reported to regulate gene transcription and mRNA splicing. However, further work is required to determine the impact of IL-1 intranuclear actions on disease pathogenesis. The intranuclear actions of IL-1 family members represent a new and potentially important area of IL-1 biology and may have implications for the future development of anti-IL-1 therapies.
Collapse
Affiliation(s)
- N M Luheshi
- Faculty of Life Sciences, University of Manchester, Manchester, UK.
| | | | | |
Collapse
|
13
|
IL-32-dependent effects of IL-1beta on endothelial cell functions. Proc Natl Acad Sci U S A 2009; 106:3883-8. [PMID: 19228941 DOI: 10.1073/pnas.0813334106] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Increasing evidence demonstrates that interleukin (IL)-32 is a pro-inflammatory cytokine, inducing IL-1alpha, IL-1beta, IL-6, tumor necrosis factor (TNF)-alpha, and chemokines via nuclear factor (NF)-kappaB, p38 mitogen-activated protein kinase (MAPK), and activating protein (AP)-1 activation. Here we report that IL-32 is expressed and is also functional in human vascular endothelial cells (EC) of various origins. Compared with primary blood monocytes, high levels of IL-32 are constitutively produced in human umbilical vein EC (HUVEC), aortic macrovascular EC, and cardiac as well as pulmonary microvascular EC. At concentrations as low as 0.1 ng/ml, IL-1beta stimulated IL-32 up to 15-fold over constitutive levels, whereas 10 ng/ml of TNFalpha or 100 ng/ml of lipopolysaccharide (LPS) were required to induce similar quantities of IL-32. IL-1beta-induced IL-32 was reduced by inhibition of the IkappaB kinase-beta/NF-kappaB and ERK pathways. In addition to IL-1beta, pro-coagulant concentrations of thrombin or fresh platelets increased IL-32 protein up to 6-fold. IL-1beta and thrombin induced an isoform-switch in steady-state mRNA levels from IL-32alpha/gamma to beta/epsilon. Adult EC responded in a similar fashion. To prove functionality, we silenced endogenous IL-32 with siRNA, decreasing intracellular IL-32 protein levels by 86%. The knockdown of IL-32 resulted in reduction of constitutive as well as IL-1beta-induced intercellular adhesion molecule-1 (ICAM-1) (of 55% and 54%, respectively), IL-1alpha (of 62% and 43%), IL-6 (of 53% and 43%), and IL-8 (of 46% and 42%). In contrast, the anti-inflammatory/anti-coagulant CD141/thrombomodulin increased markedly when IL-32 was silenced. This study introduces IL-32 as a critical regulator of endothelial function, expanding the properties of this cytokine relevant to coagulation, endothelial inflammation, and atherosclerosis.
Collapse
|
14
|
Chang SY, Su PF, Lee TC. Ectopic expression of interleukin-1 receptor type II enhances cell migration through activation of the pre-interleukin 1alpha pathway. Cytokine 2008; 45:32-8. [PMID: 19026558 DOI: 10.1016/j.cyto.2008.10.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 10/01/2008] [Accepted: 10/10/2008] [Indexed: 11/26/2022]
Abstract
Expression of interleukin-1 receptor type II (IL1R2), a decoy receptor for pro-inflammatory interleukin 1 (IL-1), is enhanced by chronic exposure of the human uroepithelial cell line HUC-1 to arsenite. To explore the function of IL1R2, we ectopically expressed IL1R2 in HUC-1 cells. IL1R2 overexpression results in changes in cell morphology, actin rearrangement, and promoted cell migration. Ectopic expression of IL1R2 specifically blocked exogenous IL-1beta signaling but increased expression of the precursor form of IL-1alpha (pIL-1alpha) and its downstream targets, including interleukin 6 (IL-6), interleukin 8 (IL-8), and type I collagen alpha1 (COL1A1). However, depleting gene expression using small RNA interference specific to either pIL-1alpha or COL1A1, but not IL-6 or IL-8, significantly attenuated the migration of IL1R2-overexpressing cells. Furthermore, IL1R2 overexpression was associated with enhanced expression of Smad-interacting protein 1 (SIP-1) and reduced expression of E-cadherin. Because SIP-1 is a repressor of COL1A1-induced E-cadherin expression, the present results suggest that IL1R2 overexpression is likely through activation of the pIL-1alpha pathway to enhance cell migration.
Collapse
Affiliation(s)
- Shih-Yu Chang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
| | | | | |
Collapse
|
15
|
Abstract
Pro-inflammatory members of the interleukin-1 (IL-1) family of cytokines (IL-1α and β) are important mediators of host defense responses to infection but can also exacerbate the damaging inflammation that contributes to major human diseases. IL-1α and β are produced by cells of the innate immune system, such as macrophages, and act largely after their secretion by binding to the type I IL-1 receptor on responsive cells. There is evidence that IL-1α is also a nuclear protein that can act intracellularly. In this study, we report that both IL-1α and IL-1β produced by microglia (central nervous system macrophages) in response to an inflammatory challenge are distributed between the cytosol and the nucleus. Using IL-1-β-galactosidase and IL-1-green fluorescent protein chimeras (analyzed by fluorescence recovery after photobleaching), we demonstrate that nuclear import of IL-1α is exclusively active, requiring a nuclear localization sequence and Ran, while IL-1β nuclear import is entirely passive. These data provide valuable insights into the dynamic regulation of intracellular cytokine trafficking.
Collapse
Affiliation(s)
- Nadia M Luheshi
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, UK
| | | | | |
Collapse
|
16
|
Intracellular interleukin-1alpha mediates interleukin-8 production induced by Chlamydia trachomatis infection via a mechanism independent of type I interleukin-1 receptor. Infect Immun 2007; 76:942-51. [PMID: 18086816 DOI: 10.1128/iai.01313-07] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chlamydia trachomatis infection induces a wide array of inflammatory cytokines and chemokines, which may contribute to chlamydia-induced pathologies. However, the precise mechanisms by which Chlamydia induces cytokines remain unclear. Here we demonstrate that the proinflammatory cytokine interleukin-1alpha (IL-1alpha) plays an essential role in chlamydial induction of the chemokine IL-8. Cells deficient in IL-1alpha expression or IL-1alpha-competent cells treated with IL-1alpha-specific small interfering RNA failed to produce IL-8 in response to chlamydial infection. However, neutralization of extracellular IL-1alpha or blockade of or deficiency in type I IL-1 receptor (IL-1RI) signaling did not affect chlamydial induction of IL-8 in cells capable of producing IL-1alpha. These results suggest that IL-1alpha can mediate the chlamydial induction of IL-8 via an intracellular mechanism independent of IL-1RI, especially during the early stage of the infection cycle. This conclusion is further supported by the observations that expression of a transgene-encoded full-length IL-1alpha fusion protein in the nuclei enhanced IL-8 production and that nuclear localization of chlamydia-induced precursor IL-1alpha correlated with chlamydial induction of IL-8. Thus, we have identified a novel mechanism for chlamydial induction of the chemokine IL-8.
Collapse
|
17
|
Carriere V, Roussel L, Ortega N, Lacorre DA, Americh L, Aguilar L, Bouche G, Girard JP. IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci U S A 2006; 104:282-7. [PMID: 17185418 PMCID: PMC1765450 DOI: 10.1073/pnas.0606854104] [Citation(s) in RCA: 766] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recent studies indicate that IL-1alpha functions intracellularly in pathways independent of its cell surface receptors by translocating to the nucleus and regulating transcription. Similarly, the chromatin-associated protein HMGB1 acts as both a nuclear factor and a secreted proinflammatory cytokine. Here, we show that IL-33, an IL-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines, is an endothelium-derived, chromatin-associated nuclear factor with transcriptional repressor properties. We found that IL-33 is identical to NF-HEV, a nuclear factor preferentially expressed in high endothelial venules (HEV), that we previously characterized. Accordingly, in situ hybridization demonstrated that endothelial cells constitute a major source of IL-33 mRNA in chronically inflamed tissues from patients with rheumatoid arthritis and Crohn's disease. Immunostaining with three distinct antisera, directed against the N-terminal part and IL-1-like C-terminal domain, revealed that IL-33 is a heterochromatin-associated nuclear factor in HEV endothelial cells in vivo. Association of IL-33 with heterochromatin was also observed in human and mouse cells under living conditions. In addition, colocalization of IL-33 with mitotic chromatin was noted. Nuclear localization, heterochromatin-association, and targeting to mitotic chromosomes were all found to be mediated by an evolutionarily conserved homeodomain-like helix-turn-helix motif within the IL-33 N-terminal part. Finally, IL-33 was found to possess transcriptional repressor properties, associated with the homeodomain-like helix-turn-helix motif. Together, these data suggest that, similarly to IL1alpha and HMGB1, IL-33 is a dual function protein that may function as both a proinflammatory cytokine and an intracellular nuclear factor with transcriptional regulatory properties.
Collapse
Affiliation(s)
- Virginie Carriere
- *Laboratoire de Biologie Vasculaire, Equipe Labellisée “La Ligue 2006,” Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5089, 205 Route de Narbonne, 31077 Toulouse, France; and
| | - Lucie Roussel
- *Laboratoire de Biologie Vasculaire, Equipe Labellisée “La Ligue 2006,” Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5089, 205 Route de Narbonne, 31077 Toulouse, France; and
| | - Nathalie Ortega
- *Laboratoire de Biologie Vasculaire, Equipe Labellisée “La Ligue 2006,” Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5089, 205 Route de Narbonne, 31077 Toulouse, France; and
| | - Delphine-Armelle Lacorre
- *Laboratoire de Biologie Vasculaire, Equipe Labellisée “La Ligue 2006,” Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5089, 205 Route de Narbonne, 31077 Toulouse, France; and
| | - Laure Americh
- Endocube, Prologue Biotech, BP700, Rue Pierre et Marie Curie, 31319 Labège Cedex, France
| | - Luc Aguilar
- Endocube, Prologue Biotech, BP700, Rue Pierre et Marie Curie, 31319 Labège Cedex, France
| | - Gérard Bouche
- *Laboratoire de Biologie Vasculaire, Equipe Labellisée “La Ligue 2006,” Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5089, 205 Route de Narbonne, 31077 Toulouse, France; and
| | - Jean-Philippe Girard
- *Laboratoire de Biologie Vasculaire, Equipe Labellisée “La Ligue 2006,” Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5089, 205 Route de Narbonne, 31077 Toulouse, France; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
18
|
Merhi-Soussi F, Berti M, Wehrle-Haller B, Gabay C. Intracellular interleukin-1 receptor antagonist type 1 antagonizes the stimulatory effect of interleukin-1α precursor on cell motility. Cytokine 2005; 32:163-70. [PMID: 16246569 DOI: 10.1016/j.cyto.2005.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Revised: 09/06/2005] [Accepted: 09/09/2005] [Indexed: 11/17/2022]
Abstract
Interleukin (IL)-1alpha, a proinflammatory cytokine, is produced as a 33 kDa protein precursor (preIL-1alpha) which is cleaved to generate the 17 kDa C-terminal mature IL-1alpha (mIL-1alpha) and the 16kDa N-terminal IL-1alpha propiece (NIL-1alpha). The biological effect of IL-1alpha is regulated by the IL-1 receptor antagonist (IL-1Ra), its naturally occurring inhibitor. Four different isoforms of the IL-1Ra have been described, one secreted (sIL-1Ra) and three intracellular (icIL-1Ra1, 2, 3). Whether the icIL-1Ra1 isoform can antagonize some of the biological effects of intracellular IL-1alpha is still unknown. The aim of this study is to investigate effects of preIL-1alpha and icIL-1Ra1 on cell motility in stably transfected ECV304 cells. We show that expression of preIL-1alpha in ECV304 cells significantly increases cell motility. Furthermore, transfection with NIL-1alpha propiece also increases cell motility whereas this stimulatory effect was not observed by addition of exogenous mIL-1alpha, suggesting an intracellular effect of preIL-1alpha mediated by NIL-1alpha propiece. Co-transfection of ECV304 cells with icIL-1Ra1 completely antagonizes the stimulatory effect of preIL-1alpha and NIL-1alpha propiece on cell motility. In conclusion, NIL-1alpha propiece increases ECV304 cell motility and icIL-1Ra1 exerts intracellular functions regulating this stimulatory effect.
Collapse
Affiliation(s)
- Faten Merhi-Soussi
- Division of Rheumatology, Department of Internal Medicine, University Hospital of Geneva, 26 avenue de Beau-séjour, 1211 Geneva 14, Switzerland
| | | | | | | |
Collapse
|
19
|
Böl GF, Jurrmann N, Brigelius-Flohé R. Cellular trafficking of the IL-1RI-associated kinase-1 requires intact kinase activity. Biochem Biophys Res Commun 2005; 332:279-87. [PMID: 15896328 DOI: 10.1016/j.bbrc.2005.04.121] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Accepted: 04/23/2005] [Indexed: 10/25/2022]
Abstract
Upon stimulation of cells with interleukin-1 (IL-1) the IL-1 receptor type I (IL-1RI) associated kinase-1 (IRAK-1) transiently associates to and dissociates from the IL-1RI and thereafter translocates into the nucleus. Here we show that nuclear translocation of IRAK-1 depends on its kinase activity since translocation was not observed in EL-4 cells overexpressing a kinase negative IRAK-1 mutant (EL-4(IRAK-1-K239S)). IRAK-1 itself, an endogenous substrate with an apparent molecular weight of 24kDa (p24), and exogenous substrates like histone and myelin basic protein are phosphorylated by nuclear located IRAK-1. Phosphorylation of p24 cannot be detected in EL-4(IRAK-1-K239S) cells. IL-1-dependent recruitment of IRAK-1 to the IL-1RI and subsequent phosphorylation of IRAK-1 is a prerequisite for nuclear translocation of IRAK-1. It is therefore concluded that intracellular localization of IRAK-1 depends on its kinase activity and that IRAK-1 may also function as a kinase in the nucleus as shown by a new putative endogenous substrate.
Collapse
Affiliation(s)
- Gaby-Fleur Böl
- German Institute of Human Nutrition Potsdam-Rehbruecke, Biochemistry of Micronutrients, 14558 Nuthetal, Germany.
| | | | | |
Collapse
|
20
|
Prudovsky I, Mandinova A, Soldi R, Bagala C, Graziani I, Landriscina M, Tarantini F, Duarte M, Bellum S, Doherty H, Maciag T. The non-classical export routes: FGF1 and IL-1alpha point the way. J Cell Sci 2004; 116:4871-81. [PMID: 14625381 DOI: 10.1242/jcs.00872] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Non-classical protein release independent of the ER-Golgi pathway has been reported for an increasing number of proteins lacking an N-terminal signal sequence. The export of FGF1 and IL-1alpha, two pro-angiogenic polypeptides, provides two such examples. In both cases, export is based on the Cu2+-dependent formation of multiprotein complexes containing the S100A13 protein and might involve translocation of the protein across the membrane as a 'molten globule'. FGF1 and IL-1alpha are involved in pathological processes such as restenosis and tumor formation. Inhibition of their export by Cu2+ chelators is thus an effective strategy for treatment of several diseases.
Collapse
Affiliation(s)
- Igor Prudovsky
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Niki Y, Yamada H, Kikuchi T, Toyama Y, Matsumoto H, Fujikawa K, Tada N. Membrane-associated IL-1 contributes to chronic synovitis and cartilage destruction in human IL-1 alpha transgenic mice. THE JOURNAL OF IMMUNOLOGY 2004; 172:577-84. [PMID: 14688369 DOI: 10.4049/jimmunol.172.1.577] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-1 molecules are encoded by two distinct genes, IL-1alpha and IL-1beta. Both isoforms possess essentially identical activities and potencies, whereas IL-1alpha, in contrast to IL-1beta, is known to act as a membrane-associated IL-1 (MA-IL-1) and plays an important role in a variety of inflammatory situations. The transgenic (Tg) mouse line (Tg1706), which was generated in our laboratory, overexpresses human IL-1alpha (hIL-1alpha) and exhibits a severe arthritic phenotype characterized by autonomous synovial proliferation with subsequent cartilage destruction. Because the transgene encoded Lys(64) to Ala(271) of the hIL-1alpha amino acid sequence, Tg mice may overproduce MA-IL-1 as well as soluble IL-1alpha. The present study investigated whether MA-IL-1 contributes to synovial proliferation and cartilage destruction in the development of arthritis. Flow cytometric analysis revealed that both macrophage-like and fibroblast-like synoviocytes constitutively produce MA-IL-1. D10 cell proliferation assay revealed MA-IL-1 bioactivity of paraformaldehyde-fixed synoviocytes and the further induction of endogenous mouse MA-IL-1 via autocrine mechanisms. MA-IL-1 expressed on synoviocytes triggered synoviocyte self-proliferation through cell-to-cell (i.e., juxtacrine) interactions and also promoted proteoglycan release from the cartilage matrix in chondrocyte monolayer culture. Interestingly, the severity of arthritis was significantly correlated with MA-IL-1 activity rather than with soluble IL-1alpha activity or concentration of serum hIL-1alpha. Moreover, when the Tg1706 line was compared with the Tg101 line, which selectively overexpresses the 17-kDa mature hIL-1alpha, the severity of arthritis was significantly higher in the Tg1706 line than in the Tg101 line. These results suggest that MA-IL-1 contributes to synoviocyte self-proliferation and subsequent cartilage destruction in inflammatory joint disease such as rheumatoid arthritis.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Cartilage, Articular/immunology
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cell Communication/genetics
- Cell Communication/immunology
- Cell Division/genetics
- Cell Division/immunology
- Cell Membrane/genetics
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cells, Cultured
- Chronic Disease
- Extracellular Matrix/metabolism
- Flow Cytometry
- Gene Expression Regulation/immunology
- Humans
- Interleukin-1/biosynthesis
- Interleukin-1/genetics
- Interleukin-1/metabolism
- Interleukin-1/physiology
- Kinetics
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Transgenic
- Precipitin Tests
- Proteoglycans/metabolism
- Severity of Illness Index
- Solubility
- Synovial Membrane/immunology
- Synovial Membrane/metabolism
- Synovial Membrane/pathology
- Synovitis/genetics
- Synovitis/immunology
- Synovitis/pathology
Collapse
Affiliation(s)
- Yasuo Niki
- Department of Orthopedic Surgery, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | | | | | | | | | | | | |
Collapse
|
22
|
Gruber J, Vincent TL, Hermansson M, Bolton M, Wait R, Saklatvala J. Induction of interleukin-1 in articular cartilage by explantation and cutting. ACTA ACUST UNITED AC 2004; 50:2539-46. [PMID: 15334468 DOI: 10.1002/art.20369] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To investigate the effect of explantation and fine cutting of articular cartilage upon intracellular inflammatory signaling pathways and expression of interleukin-1 (IL-1). METHODS Cartilage from porcine metacarpophalangeal joints was cultured in serum-free medium. Tissue extracts were examined for ERK activation by phosphorylated-Western blotting, for JNK and p38 MAPK activity by kinase assay, and for IkappaBalpha. IL-1alpha and IL-1beta messenger RNA (mRNA) was measured by reverse transcriptase-polymerase chain reaction. IL-1 activity was measured by the induction of serum amyloid A protein in cultured chondrocytes. RESULTS All 3 MAPKs (p38, JNK, and ERK) were rapidly activated upon dissection and explantation of the cartilage. IL-1alpha and IL-1beta mRNA was also induced: the speed and magnitude of induction were increased if the explants had been finely cut. IL-1 activity that could be inhibited by IL-1 receptor antagonist or antibodies to IL-1alpha was found in extracts of explants cultured for 20 hours or lysates of cells isolated from them. This activity was likely due to intracellular proIL-1alpha that was not secreted. ProIL-1beta would not be detected because it is biologically inactive. The mechanism of inflammatory signaling pathway activation underlying the induction of IL-1 is unknown. CONCLUSION Explantation and cutting of articular cartilage activates intracellular inflammatory signaling pathways and induces expression of mRNA for IL-1alpha and IL-1beta. Biologically active IL-1alpha protein was detectable in cartilage lysates and was probably intracellular proIL-1alpha. We were unable to show that IL-1 was secreted by chondrocytes.
Collapse
|
23
|
Buryskova M, Pospisek M, Grothey A, Simmet T, Burysek L. Intracellular interleukin-1alpha functionally interacts with histone acetyltransferase complexes. J Biol Chem 2003; 279:4017-26. [PMID: 14612453 DOI: 10.1074/jbc.m306342200] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interleukin-1alpha (IL-1alpha) is an inflammatory cytokine acting extracellularly via membrane receptors. Interestingly, a significant portion of synthesized IL-1alpha is not secreted; instead, it is actively translocated into the cell nucleus. IL-1alpha was indeed shown to be involved in certain intracellular processes, such as control of proliferation, apoptosis, or migration, however, the mechanisms of such actions are not known. Here we show that intracellular IL-1alpha fused to the Gal4p DNA-binding domain (Gal4BD) possesses strong transactivation potential that can be boosted by overexpression of the transcriptional coactivator p300. We demonstrate that the IL-1alpha precursor interacts via its N-terminal peptide (IL-1NTP) with histone acetyltransferases p300, PCAF, Gcn5 and with the adaptor component Ada3, and that it integrates into the PCAF.p300 complex in a non-destructive manner. In analogy with known acidic coactivators, yeast strains expressing Gal4BD/IL-1NTP display a toxic phenotype that can be relieved by depletion of various components of the SAGA complex. Our data provide the first solid evidence for the nuclear target of the IL-1alpha precursor and suggest its novel function in transcriptional control.
Collapse
Affiliation(s)
- Miroslava Buryskova
- Department of Pharmacology of Natural Products and Clinical Pharmacology, University of Ulm, D-89081 Ulm, Germany
| | | | | | | | | |
Collapse
|
24
|
Schaub FJ, Liles WC, Ferri N, Sayson K, Seifert RA, Bowen-Pope DF. Fas and Fas-associated death domain protein regulate monocyte chemoattractant protein-1 expression by human smooth muscle cells through caspase- and calpain-dependent release of interleukin-1alpha. Circ Res 2003; 93:515-22. [PMID: 12946945 DOI: 10.1161/01.res.0000093205.42313.7c] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We previously reported that treatment of human vascular smooth muscle cells (SMCs) with proapoptotic stimuli, including Fas ligand plus cycloheximide (FasL/Chx), or overexpression of Fas-associated death domain protein (FADD) result in increased expression of monocyte chemoattractant protein-1 (MCP-1) and other proinflammatory genes. In this study, we demonstrate that Fas/FADD-induced MCP-1 upregulation is driven by an autocrine/paracrine signaling loop in which interleukin (IL)-1alpha synthesis and release are activated through caspase- and calpain-dependent processes. Untreated SMCs contain very little IL-1alpha protein or transcript. Both were increased greatly in response to Fas/FADD activation, primarily through an autocrine/paracrine pathway in which secreted IL-1alpha stimulated additional IL-1alpha synthesis and release. Caspase 8 (Csp8) activity increased in response to FasL/Chx treatment, and Csp8 inhibitors markedly reduced IL-1alpha release and MCP-1 upregulation. In contrast, Csp8 activity was not significantly increased in response to FADD overexpression and caspase inhibitors did not effect FADD-induced MCP-1 upregulation. Both FasL/Chx treatment and FADD overexpression increased the activity of calpains. Calpain inhibitors reduced IL-1alpha release and MCP-1 upregulation in both FADD-overexpressing SMCs and FasL/Chx-treated SMCs without blocking Csp8 activity. This indicates that calpains are not required for activation of caspases and that caspase activation is not sufficient for IL-1alpha release and MCP-1 upregulation. These data suggest that calpains play a dominant role in Fas/FADD-induced IL-1alpha release and MCP-1 upregulation and that caspase activation may function to amplify the effects of calpain activation.
Collapse
Affiliation(s)
- Friedemann J Schaub
- Department of Pathology, University of Washington, Box 357470, Seattle, Wash 98195-7470, USA
| | | | | | | | | | | |
Collapse
|
25
|
Bayley JP, van Rietschoten JGI, Bakker AM, van Baarsen L, Kaijzel EL, Wierenga EA, van der Pouw Kraan TCTM, Huizinga TWJ, Verweij CL. Allele-specific expression of the IL-1 alpha gene in human CD4+ T cell clones. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2349-53. [PMID: 12928381 DOI: 10.4049/jimmunol.171.5.2349] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A number of reports have described the monoallelic expression of murine cytokine genes. Here we describe the monoallelic expression of the human IL-1alpha gene in CD4+ T cells. Analysis of peripheral blood T cell clones derived from healthy individuals revealed that the IL-1alpha gene shows predominantly monoallelic expression. Monoallelic expression was observed in Th0, Th1, and Th2 cell clones. In addition, we demonstrate monoallelic expression in T cell clones from rheumatoid arthritis patients derived from synovial fluid of the knee joint, suggesting that the occurrence of this phenomenon is not different from that in clones derived from healthy individuals. The finding of monoallelic expression of a cytokine gene in human CD4+ T cell clones provides evidence for allele-specific silencing/activation as another layer of regulation of IL-1alpha gene expression.
Collapse
Affiliation(s)
- Jean-Pierre Bayley
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Murata M, Trahan C, Hirahashi J, Mankin HJ, Towle CA. Intracellular interleukin-1 receptor antagonist in osteoarthritis chondrocytes. Clin Orthop Relat Res 2003:285-95. [PMID: 12671514 DOI: 10.1097/01.blo.0000059582.08469.ac] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The objective of the current study was to determine whether the balance of interleukin-1 and intracellular interleukin-1 receptor antagonist in chondrocytes in osteoarthritic human joints favors agonist action. Chondrocytes were isolated from cartilage specimens taken at the time of joint arthroplasty. Interleukin-1alpha, interleukin-1beta, and intracellular interleukin-1 receptor antagonist messenger ribonucleic acids were assessed by reverse transcriptase-polymerase chain reaction, and chondrocyte lysates were analyzed by enzyme-linked immunosorbent assay for the respective proteins. Type I intracellular interleukin receptor antagonist transcripts were the only intracellular variant detected in osteoarthritis chondrocytes. In cartilage graded as advanced osteoarthritis both interleukin proteins in chondrocyte lysates decreased, correlating with decreased interleukin-1alpha and beta messenger ribonucleic acids. Interleukin-1 receptor antagonist exceeded interleukin-1alpha in chondrocyte lysates by one order of magnitude except that in moderate osteoarthritis, antagonist was only two- to fourfold in excess. Interleukin-1alpha and interleukin-1beta proteins were correlated closely in individual lysates, with interleukin-1beta exceeding interleukin-1beta by one order of magnitude. In moderately degenerated cartilage, intracellular antagonist may not be sufficiently abundant to block postulated intracellular functions of precursor interleukin-1alpha. Furthermore, if stored interleukin-1alpha, interleukin-1beta, and interleukin receptor antagonist are released from chondrocytes, the localized antagonist would be insufficient to prevent signaling through cell surface receptors. Chondrocyte-derived interleukin-1alpha and interleukin-1beta may locally overwhelm inhibition by interleukin receptor antagonist to promote the early degenerative changes in osteoarthritis.
Collapse
MESH Headings
- Aged
- Antirheumatic Agents/analysis
- Arthroplasty, Replacement, Hip
- Arthroplasty, Replacement, Knee
- Chondrocytes/pathology
- Female
- Gene Expression/genetics
- Humans
- Interleukin 1 Receptor Antagonist Protein
- Interleukin-1/analysis
- Interleukin-1/genetics
- Male
- Middle Aged
- Osteoarthritis, Hip/genetics
- Osteoarthritis, Hip/pathology
- Osteoarthritis, Hip/surgery
- Osteoarthritis, Knee/genetics
- Osteoarthritis, Knee/pathology
- Osteoarthritis, Knee/surgery
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Receptors, Interleukin-1/analysis
- Receptors, Interleukin-1/antagonists & inhibitors
- Receptors, Interleukin-1/genetics
- Sialoglycoproteins/analysis
- Sialoglycoproteins/genetics
Collapse
Affiliation(s)
- Minako Murata
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
27
|
Pollock AS, Turck J, Lovett DH. The prodomain of interleukin 1alpha interacts with elements of the RNA processing apparatus and induces apoptosis in malignant cells. FASEB J 2003; 17:203-13. [PMID: 12554699 DOI: 10.1096/fj.02-0602com] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Interleukin 1alpha (IL-1alpha), a 33 kDa precursor, is cleaved releasing the 17 kDa carboxyl-terminal cytokine IL-1alpha to which all of the biological properties of IL-1alpha have been attributed. We investigated the potential independent properties of the remaining 16 kDa IL-1alpha amino-terminal propiece by expression in human tumor and primary human cell lines. The IL-1alpha propiece produced apoptosis in malignant but not normal cell lines. A minimal fragment comprised of amino acids 55-108 was required for apoptosis. Deletion and mutation studies identified an extended nuclear localization sequence required for nuclear localization, induction of apoptosis and concentration of the IL-1alpha propiece in interchromatin granule clusters, concentrations of proteins in the RNA splicing and processing pathways. The IL-1alpha propiece interacted with five known components of the RNA splicing/processing pathway, suggesting that the mechanism of action may involve changes in RNA splicing or processing. Expression of the IL-1alpha propiece caused a shift in the ratio of Bcl-Xl/Bcl-Xs toward the apoptotic direction. Our findings indicate that the IL-1alpha propiece induces apoptosis in a range of tumor cells and likely operates through a mechanism involving the RNA processing apparatus and the alternate splicing of apoptosis regulatory proteins.
Collapse
Affiliation(s)
- Allan S Pollock
- The Department of Medicine, University of California, San Francisco, Northern California Institute for Research and Education, Veterans Administration Medical Center, San Francisco, California 94121, USA.
| | | | | |
Collapse
|
28
|
Abstract
IL-1 is an important mediator of inflammation and tissue damage in multiple organs, both in experimental animal models of disease and in human diseases. The IL-1 family consists of two agonists, IL-1alpha and IL-1beta, two receptors, biologically active IL-1RI and inert IL-1RII, and a specific receptor antagonist, IL-1Ra. The balance between IL-1 and IL-1Ra in local tissues plays an important role in the susceptibility to and severity of many diseases. An allelic polymorphism in the IL-1Ra gene has been associated with a variety of human diseases primarily of epithelial and endothelial cell origin. This association may be secondary to an imbalance in the IL-1 system with enhanced production of IL-1beta and reduced production of the major intracellular isoform of IL-1Ra. Treatment of RA with daily subcutaneous injections of recombinant IL-1Ra protein has been shown to be efficacious. Gene therapy approaches with IL-1Ra are being evaluated for the treatment of RA and other human diseases.
Collapse
Affiliation(s)
- William P Arend
- B115 Division of Rheumatology, Health Sciences Centre, University of Colorado, 4200 East Ninth Avenue, Denver, CO 80262, USA.
| |
Collapse
|
29
|
Yin H, Morioka H, Towle CA, Vidal M, Watanabe T, Weissbach L. Evidence that HAX-1 is an interleukin-1 alpha N-terminal binding protein. Cytokine 2001; 15:122-37. [PMID: 11554782 DOI: 10.1006/cyto.2001.0891] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During studies aimed at understanding the function of the N-terminal peptide of interleukin-1 alpha (IL-1 NTP, amino acids 1-112), which is liberated from the remainder of IL-1 alpha during intracellular processing, we identified by yeast two-hybrid analysis a putative interacting protein previously designated as HAX-1. In vitro binding studies and transient transfection experiments confirmed that HAX-1 can associate with the IL-1 NTP. HAX-1 was first identified as a protein that associates with HS1, a target of non-receptor protein tyrosine kinases within haematopoietic cells. Recent data have also revealed interactions between HAX-1 and three disparate proteins, polycystin-2 (derived from the PKD2 gene), a protein linked to polycystic kidney disease, cortactin, and Epstein-Barr virus nuclear antigen leader protein (EBNA-LP). Sequence analysis of different HAX-1 binding domains revealed a putative consensus binding motif that is present in various intracellular proteins. Overlapping peptides comprising the IL-1 NTP were synthesized, and binding experiments revealed that discrete peptides were capable of interacting with HAX-1. HAX-1 may serve to retain the IL-1 NTP in the cytoplasm, and complex formation between the IL-1 NTP and HAX-1 may play a role in motility and/or adhesion of cells.
Collapse
Affiliation(s)
- H Yin
- Orthopaedic Research Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | |
Collapse
|
30
|
Sasu S, Cooper AL, Beasley D. Juxtacrine effects of IL-1 alpha precursor promote iNOS expression in vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 2001; 280:H1615-23. [PMID: 11247772 DOI: 10.1152/ajpheart.2001.280.4.h1615] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
After injury to the blood vessel wall, vascular smooth muscle cells (SMC) synthesize interleukin (IL)-1 and inducible nitric oxide (NO) synthase (iNOS). The present study tested whether endogenous production of IL-1 alpha stimulates iNOS expression in vascular SMC, and assessed whether IL-1 alpha exerts autocrine effects on the cells producing IL-1 alpha or juxtacrine effects on cells that contact the IL-1 alpha producing cells. Rat aortic SMC were transiently transfected with expression plasmids encoding either IL-1 alpha precursor, which localizes to the plasma membrane, or mature IL-1 alpha, which remains cytosolic. iNOS mRNA levels, determined by RT-PCR, and production of nitrite, a stable oxidation product of NO, were markedly elevated in SMC overexpressing IL-1 alpha precursor, and modestly elevated in SMC overexpressing mature IL-1 alpha, relative to SMC transfected with vector alone. Exposure to exogenous IL-1 beta or TNF-alpha further stimulated iNOS gene expression in SMC producing IL-1 alpha; low levels of IL-1 beta (20 pg/ml) were effective in SMC transfected with IL-1 alpha precursor plasmid, whereas SMC transfected with mature IL-1 alpha plasmid or vector alone required higher concentrations of IL-1 beta (200 and 2,000 pg/ml, respectively). The increases in iNOS mRNA levels and NO production in SMC overexpressing IL-1 alpha precursor were prevented by exogenous IL-1 receptor antagonist, suggesting that these effects were mediated by the type I IL-1 receptor. Immunostaining studies indicated that IL-1 alpha precursor stimulates iNOS gene expression via cell-cell contact. Expression of iNOS was enhanced in cells that were in contact with a cell overexpressing IL-1 alpha precursor (identified by coexpression of green fluorescent protein), and in cells that were overexpressing IL-1 alpha themselves, but only when the cell contacted another cell. Together these results indicate that IL-1 alpha precursor acts by cell-cell contact as an autocrine and juxtacrine enhancer of iNOS gene expression, inducing moderate iNOS expression on its own, and markedly augmenting the responsiveness of rat aortic SMC to exogenous cytokines.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/enzymology
- Cells, Cultured
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/physiology
- Interleukin-1/genetics
- Interleukin-1/pharmacology
- Interleukin-1/physiology
- Kinetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/enzymology
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase Type II
- Plasmids
- Protein Precursors/genetics
- RNA, Messenger/genetics
- Rats
- Reverse Transcriptase Polymerase Chain Reaction
- Time Factors
- Transcription, Genetic
- Transfection
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- S Sasu
- Division of Nephrology, Department of Medicine, New England Medical Center Hospitals and Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | | |
Collapse
|
31
|
Tarantini F, Micucci I, Bellum S, Landriscina M, Garfinkel S, Prudovsky I, Maciag T. The precursor but not the mature form of IL1alpha blocks the release of FGF1 in response to heat shock. J Biol Chem 2001; 276:5147-51. [PMID: 11087725 DOI: 10.1074/jbc.c000714200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interleukin (IL)1alpha mediates proinflammatory events through its extracellular interaction with the IL1 type I receptor. However, IL1alpha does not contain a conventional signal peptide sequence that provides access to the endoplasmic reticulum-Golgi apparatus for secretion. Thus, we have studied the release of the precursor (p) and mature (m) forms of IL1alpha from NIH 3T3 cells. We have demonstrated that mIL1alpha but not pIL1alpha was released in response to heat shock with biochemical and pharmacological properties similar to those reported for the stress-mediated release pathway utilized by fibroblast growth factor (FGF)1. However, unlike the FGF1 release pathway, the IL1alpha release pathway appears to function independently of synaptotagmin (Syt)1 because the expression of a dominant-negative form of Syt1, which represses the release of FGF1, did not inhibit the release of mIL1alpha in response to temperature stress. Interestingly, whereas the expression of both mIL1alpha and FGF1 in NIH 3T3 cells did not impair the stress-induced release of either polypeptide, the expression of both pIL1alpha and FGF1 repressed the release of FGF1 in response to temperature stress. These data suggest that the release of mIL1alpha requires proteolytic processing of its precursor form and that mIL1alpha and FGF1 may utilize similar but distinct mechanisms for export.
Collapse
Affiliation(s)
- F Tarantini
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074
| | | | | | | | | | | | | |
Collapse
|
32
|
Blankesteijn WM, van Gijn ME, Essers-Janssen YP, Daemen MJ, Smits JF. Beta-catenin, an inducer of uncontrolled cell proliferation and migration in malignancies, is localized in the cytoplasm of vascular endothelium during neovascularization after myocardial infarction. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 157:877-83. [PMID: 10980127 PMCID: PMC1885709 DOI: 10.1016/s0002-9440(10)64601-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Beta-catenin is a protein involved in cell-cell adhesion and proliferation. In neoplastic diseases, defects in the regulation of the cellular beta-catenin content and cytoplasmic accumulation of the protein contribute to the uncontrolled cell proliferation and migration. Whether beta-catenin plays a role in the controlled proliferative and migratory responses to injury, eg, of vascular endothelial cells during neovascularization after myocardial infarction (MI), is not known. In the present study, we examined the localization of beta-catenin in the infarcted rat heart at different time points after MI. Cytoplasmic beta-catenin was observed in the endothelial cells of the newly formed and pre-existing blood vessels in the infarct area in the first week after MI, but not in the uninjured parts of the heart and not at later time points. Adenomatous polyposis coli (APC) protein was also detected; interaction of APC with beta-catenin has been reported to be critical in epithelial tube formation in vitro. Moreover, the expression of dishevelled-1, an upstream regulatory molecule of the cellular beta-catenin content, was observed in vascular endothelial cells in the infarct area. These findings suggest a role for the beta-catenin-APC complex in the proliferation and migration of vascular endothelial cells during neovascularization of the infarct area.
Collapse
Affiliation(s)
- W M Blankesteijn
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Universiteit Maastricht, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
33
|
Böl G, Kreuzer OJ, Brigelius-Flohé R. Translocation of the interleukin-1 receptor-associated kinase-1 (IRAK-1) into the nucleus. FEBS Lett 2000; 477:73-8. [PMID: 10899313 DOI: 10.1016/s0014-5793(00)01759-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Interleukin-1 (IL-1) signal transduction involves the recruitment of the IL-1 receptor-associated kinase-1 (IRAK-1). Subsequent signaling finally leads to nuclear translocation of NFkappaB. We here show that the association and autophosphorylation of IRAK-1 was already detectable 30 s after IL-1 stimulation of ECV 304 cells. Significant levels of IRAK-1 accumulated in the nucleus 30 min after IL-1 stimulation shown by Western blot analysis and confocal laser scanning microscopy. Nuclear transfer of IRAK-1 upon IL-1 stimulation was confirmed in the murine T cell line EL-4. This characterizes nuclear localization of IRAK-1 as a possibly essential event in the IL-1 signaling cascade.
Collapse
Affiliation(s)
- G Böl
- German Institute of Human Nutrition, D-14558, Potsdam-Rehbrücke, Germany.
| | | | | |
Collapse
|
34
|
Imai M, Goepfert C, Kaczmarek E, Robson SC. CD39 modulates IL-1 release from activated endothelial cells. Biochem Biophys Res Commun 2000; 270:272-8. [PMID: 10733939 DOI: 10.1006/bbrc.2000.2410] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The activation of endothelial cells (EC) and monocyte-macrophages (Mφ) by lipopolysaccharide (LPS) is considered an important element of the vascular injury observed in endotoxemia. Interleukin-1 (IL-1) beta release from Mφ in response to LPS, appears to be mediated by the autocrine/paracrine release of ATP via P2X7 receptor activation. In EC, similar nucleotide-mediated signaling pathways may be influenced by high levels of expression of CD39, the vascular nucleoside triphosphate diphosphohydrolase (NTPDase; ENTPD I). To determine whether CD39 modulates ATP-mediated release of IL-1 from EC, we stimulated human EC with LPS and measured levels of ATP secretion and IL-1 release. LPS triggered ATP secretion from EC that was soon followed by IL-1alpha release. Overexpression of CD39 following infection with recombinant CD39 adenoviral vectors (AdCD39) abrogated the initial phase of ATP secretion and inhibited IL-1alpha release; comparable results were obtained with soluble NTPDase. These data demonstrate that CD39/NTPDase modulates IL-1alpha release from LPS stimulated human EC.
Collapse
Affiliation(s)
- M Imai
- Department of Surgery, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
35
|
Mertani HC, Morel G, Lobie PE. Cytoplasmic and nuclear cytokine receptor complexes. VITAMINS AND HORMONES 1999; 57:79-121. [PMID: 10232047 DOI: 10.1016/s0083-6729(08)60641-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Much of our understanding on how hormones and cytokines transmit their message into the cell is based on the receptor activation at the plasma membrane. Many experimental in vitro models have established the paradigm for cytokine action based upon such activation of their cell surface receptor. The signaling from the plasma membrane activated cytokine receptor is driven to the nucleus by a rapid ricochet of protein phosphorylation, ultimately integrated as a differentiative, proliferative, or transcriptional message. The Janus kinase (JAK)--signal transducers and activators of transcription (STAT) pathway that was first thought to be cytokine receptor specific now appears to be activated by other noncytokine receptors. Also, evidence is accumulating showing that cytokines modulate the signal transduction machinery of the tyrosine kinase receptors and that of the heterotrimeric guanosine triphosphate (GTP)-binding protein-coupled receptors. Thus cytokine receptor signaling has become much more complex than originally hypothesized, challenging the established model of specificity of the action of a given cytokine. This review is focused on another level of complexity emerging within cytokine receptor superfamily signaling. Over the past 10 years, data from different laboratories have shown that cytokines and their receptors localize to intracellular compartments including the nucleus, and, in some cases, biological responses have been correlated with this unexpected location, raising the possibility that cytokines act as their own messenger through inter-actions with nuclear proteins. Thus, the interplay between cytokine receptor engagement and cellular signaling turns out to be more dynamic than originally suspected. The mechanisms and regulations of intracellular translocation of the cytokines, their receptors, and their signaling proteins are discussed in the context that such compartmentalization provides some of the specificity of the responses mediated by each cytokine.
Collapse
Affiliation(s)
- H C Mertani
- Institute of Molecular and Cell Biology, Singapore, Republic of Singapore
| | | | | |
Collapse
|
36
|
Beasley D, Cooper AL. Constitutive expression of interleukin-1alpha precursor promotes human vascular smooth muscle cell proliferation. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:H901-12. [PMID: 10070073 DOI: 10.1152/ajpheart.1999.276.3.h901] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vascular smooth muscle cell (VSMC) proliferation plays a critical role in the failure of vascular surgeries and contributes to the development of atherosclerotic lesions. Evidence that interleukin-1 (IL-1) is a mitogen for cultured VSMC has implicated its release by activated macrophages in the development of atherosclerosis. VSMC also produce IL-1, including the precursor form of IL-1alpha. However, it is not known whether IL-1alpha precursor is processed to mature IL-1alpha or released from VSMC, nor is it known whether either precursor or mature IL-1alpha functions as an autocrine growth factor. The goals of the present study were to establish whether proliferation is enhanced in human VSMC transfectants producing IL-1alpha constitutively at levels comparable to those produced after activation, and to determine which domains of IL-1alpha are important for its activity. Human VSMC were stably transfected with expression vectors directing constitutive expression of either full-length IL-1alpha precursor [IL-1alpha-(1-271)], its NH2-terminal domain [IL-1alpha-(1-112)], or mature IL-1alpha [IL-1alpha-(113-271)]. Both IL-1alpha-(1-271) and IL-1alpha-(113-271) stable transfectants produced moderate levels of IL-1alpha (0.2-1.0 ng/10(6) cells) and released low levels of IL-1alpha into the supernatant (<20 pg/ml). VSMC stably transfected with either IL-1alpha-(1-271) or IL-1alpha-(113-271) expression plasmids proliferated rapidly compared with nontransfected or vector-transfected VSMC and displayed a distinct morphology characterized by elongated, spindle-shaped cells. Stable transfection with IL-1alpha-(1-271) was somewhat more effective than transfection with IL-1alpha-(113-271). Interestingly, VSMC transfected with IL-1alpha-(113-271) expression plasmids also expressed IL-1alpha-(1-271) mRNA, suggesting that IL-1alpha-(113-271) activates an IL-1-induced IL-1 autocrine loop. In contrast, neither proliferation rates nor morphology was affected by stable transfection with IL-1alpha-(1-112) expression plasmids. Exogenous IL-1 receptor antagonist partially reversed the enhanced DNA synthesis in VSMC transfected with either IL-1alpha-(1-271) or IL-1alpha-(113-271) expression plasmids, suggesting that the pro-proliferative effect of VSMC-derived IL-1alpha is at least partially mediated by signaling via the type I IL-1 receptor. These results demonstrate that IL-1alpha precursor is an autocrine growth factor for human VSMC and further indicate that amino acids 113-271 play a crucial role in its actions.
Collapse
Affiliation(s)
- D Beasley
- Division of Nephrology, Department of Medicine, and Tupper Research Institute, New England Medical Center Hospitals, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | |
Collapse
|
37
|
Weissbach L, Tran K, Colquhoun SA, Champliaud MF, Towle CA. Detection of an interleukin-1 intracellular receptor antagonist mRNA variant. Biochem Biophys Res Commun 1998; 244:91-5. [PMID: 9514884 DOI: 10.1006/bbrc.1998.8217] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
At least two versions of interleukin-1 receptor antagonist generated by alternative splicing are found intracellularly, but their functions remain poorly characterized. During studies aimed at characterizing the expression of these transcripts in human articular cartilage, we detected a variant cDNA species that contained an additional 171 nucleotides within the type I interleukin-1 intracellular receptor antagonist cDNA which interrupted the coding region. This mRNA variant was also found to be expressed in keratinocytes. Translation likely initiates at an alternate methionine codon than that utilized for the previously reported interleukin-1 intracellular receptor antagonist isoforms, suggesting that this mRNA variant encodes a novel polypeptide that may play a role in interleukin-1 signaling.
Collapse
Affiliation(s)
- L Weissbach
- Orthopaedic Research Laboratories, Massachusetts General Hospital, Boston, USA.
| | | | | | | | | |
Collapse
|