1
|
Khan H, Rihal V, Kaur A, Singh TG. Proposed Hypothesis of TWEAK/Fn14 Receptor Modulation in Autism Spectrum Disorder. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:257-262. [PMID: 39473248 DOI: 10.2174/0118715273330549241015073953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 05/13/2025]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with a complex, multiple etiology that is marked by impaired social interaction, communication, and repetitive behaviour. There is presently no pharmaceutical treatment for the core symptoms of ASD, even though the prevalence of ASD is increasing worldwide. Treatment of autism spectrum disorder involves the interaction of numerous signalling pathways, such as the Wnt/beta-catenin pathway, probiotics and kynurenine pathway, PPAR pathway, PI3K-AKT-mTOR pathway, Hedgehog signaling pathway, etc. The scientific literature has revealed TWEAK/Fn14 to not be explored in the autism spectrum disorder. In vitro and in vivo, TWEAK can control a wide range of cellular responses. Recent research has revealed that TWEAK and Fn14 are expressed in the Central Nervous System (CNS) and upregulated in perivascular endothelial cells, astrocytes, neurons, and microglia in response to various stimuli, including cerebral ischemia. This upregulation is followed by cell death and an increase in Blood-brain Barrier (BBB) permeability. The study has revealed that Aurintricarboxylic Acid (ATA) acts as an agent that suppresses TWEAK/Fn14 signaling. Similarly, from the discussion, it has been emphasized that the proposed molecular TWEAK/Fn14 signalling pathway can be considered as a therapeutic approach in the management of autism spectrum disorder.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Vivek Rihal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
2
|
Balasubramaniam M, Mokhtar AMA. Past and present discovery of the BAFF/APRIL system - A bibliometric study from 1999 to 2023. Cell Signal 2024; 120:111201. [PMID: 38714287 DOI: 10.1016/j.cellsig.2024.111201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/09/2024]
Abstract
Cytokines from the Tumour Necrosis Factor (TNF) family are important regulators of both physiological and pathological processes. The discovery of novel TNF ligands and receptors, BAFF and APRIL, have opened up new possibilities for scientists to explore the effect of these cytokines on the human immune system. The role of BAFF/APRIL system in B lymphocytes is particularly important for survival and maintenance of homeostasis. Aberrant expression of the system is associated with various immunological disorders. Hence, this study provides a comprehensive overview of the past and present BAFF/APRIL system research development in a bibliometric perspective. To our best knowledge, this is the first ever bibliometric analysis conducted focusing on the BAFF/APRIL system. A total of 1055 relevant documents were retrieved from WoSCC. Microsoft Excel, VOSviewer, and Biblioshiny of R studio were bibliometric tools used to analyse the scientific literature. From 1999, the annual publications showed an upward trend, with Journal of Immunology being the most productive journal. USA leads the race for BAFF/APRIL system research developments. Pascal Schneider, a senior researcher affiliated with University of Lausanne, Switzerland was recognised as the most productive author and institution in the BAFF/APRIL system research field. The research focus transitioned from focusing on the role of the system in B cell biology, to immunological disorders and finally to development of BAFF/APRIL targeting drugs. Despite several studies elucidating briefly the pathway mechanism of BAFF/APRIL system in B-cell selection, substantial research on the mechanism of action in disease models and T cell activation and development of immunomodulating drugs from natural origins remains largely unexplored. Therefore, future research focusing on these areas are crucial for the deeper understanding of the system in disease manifestations and progression allowing a better treatment management for various immunological disorders.
Collapse
Affiliation(s)
- Muggunna Balasubramaniam
- Small G protein Research Group, Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia; Green Biopolymer Coating and Packaging Centre, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
| | - Ana Masara Ahmad Mokhtar
- Small G protein Research Group, Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia; Green Biopolymer Coating and Packaging Centre, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia.
| |
Collapse
|
3
|
Abstract
TWEAK (tumor necrosis factor-like weak inducer of apoptosis) is a member of the TNF superfamily that controls a multitude of cellular events including proliferation, migration, differentiation, apoptosis, angiogenesis, and inflammation. TWEAK control of these events is via an expanding list of intracellular signalling pathways which include NF-κB, ERK/MAPK, Notch, EGFR and AP-1. Two receptors have been identified for TWEAK - Fn14, which targets the membrane bound form of TWEAK, and CD163, which scavenges the soluble form of TWEAK. TWEAK appears to elicit specific events based on the receptor to which it binds, tissue type in which it is expressed, specific extrinsic conditions, and the presence of other cytokines. TWEAK signalling is protective in healthy tissues, but in chronic inflammatory states become detrimental to the tissue. Consistent data show a role for the TWEAK/FN14/CD163 axis in metabolic disease, chronic autoimmune diseases, and acute ischaemic stroke. Low circulating concentrations of soluble TWEAK are predictive of poor cardiovascular outcomes in those with and without diabetes. This review details the current understanding of the TWEAK/Fn14/CD163 axis as one of the chief regulators of immune signalling and its cell-specific role in metabolic disease development and progression.
Collapse
Affiliation(s)
- Wiktoria Ratajczak
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, Ulster University, Altnagelvin Hospital Campus, C-TRIC Building Glenshane Road, Derry/Londonderry, Northern Ireland, UK
| | - Sarah D Atkinson
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, Ulster University, Altnagelvin Hospital Campus, C-TRIC Building Glenshane Road, Derry/Londonderry, Northern Ireland, UK
| | - Catriona Kelly
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, Ulster University, Altnagelvin Hospital Campus, C-TRIC Building Glenshane Road, Derry/Londonderry, Northern Ireland, UK.
| |
Collapse
|
4
|
Wang Y, Li C, Gong X, Chen X, Liu C, Zhang H, Li S, Luo Y. Single-Cell Transcriptomics Reveals Splicing Features of Adult Neural Stem Cells in the Subventricular Zone. Front Cell Dev Biol 2022; 10:822934. [PMID: 35300421 PMCID: PMC8921602 DOI: 10.3389/fcell.2022.822934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/03/2022] [Indexed: 11/30/2022] Open
Abstract
The central nervous system has enormously complex cellular diversity with hundreds of distinct cell types, yet alternative splicing features in single cells of important cell types at neurogenic regions are not well understood. By employing in silico analysis, we systematically identified 3,611 alternative splicing events from 1,908 genes in 28 single-cell transcriptomic data of adult mouse ependymal and subependymal regions, and found that single-cell RNA-seq has the advantage in uncovering rare splicing isoforms compared to bulk RNA-seq at the population level. We uncovered that the simultaneous presence of multiple isoforms from the same gene in a single cell is prevalent, and quiescent stem cells, activated stem cells, and neuroblast cells exhibit high heterogeneity of splicing variants. Furthermore, we also demonstrated the existence of novel bicistronic transcripts in quiescent stem cells.
Collapse
Affiliation(s)
- Yanlu Wang
- Human Aging Research Institute and School of Life Science, Nanchang University, Nanchang, China
| | - Chun Li
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.,Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xi Gong
- Human Aging Research Institute and School of Life Science, Nanchang University, Nanchang, China
| | - Xiao Chen
- College of Architectural Engineering, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Chenming Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hailei Zhang
- Novogene Bioinformatics Technology Co., Ltd., Beijing, China
| | - Siguang Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuping Luo
- Human Aging Research Institute and School of Life Science, Nanchang University, Nanchang, China.,Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Aberrant Expression of a Proliferation-Inducing Ligand Underlies Autoimmune Mechanisms in Immune Thrombocytopenia. J Immunol Res 2021; 2021:3676942. [PMID: 33564689 PMCID: PMC7867467 DOI: 10.1155/2021/3676942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/12/2020] [Accepted: 01/09/2021] [Indexed: 01/05/2023] Open
Abstract
Purpose To study the relationship between surface membrane-bound APRIL and ITP. Methods The peripheral blood of all subjects, 50 patients diagnosed with ITP and 25 healthy controls, was collected. Flow cytometry was used to detect the expression of membrane-bound APRIL on immune cells and platelets. ELISA was used to detect the content of soluble APRIL in plasma. Results Membrane-bound APRIL was only expressed on the surface of platelets in both ITP patients and controls. APRIL expression on the platelet surface was significantly lower in newly diagnosed (P < 0.001) and chronic (P < 0.001) ITP patients than in controls. Platelet surface APRIL level was significantly enhanced in patients with complete remission after treatment (P = 0.02) but not in those with no response after treatment. Platelet surface APRIL level in ITP patients was negatively correlated with serum APRIL level (r = −0.09765, P = 0.0424). Conclusions Platelet surface APRIL may play a key immunoregulative role. Platelet surface APRIL is likely to be one source of the excessive serum APRIL in ITP patients. The effectiveness of treatment may be measured by determining the platelet surface APRIL levels in ITP patients.
Collapse
|
6
|
Zhang Y, Zeng W, Xia Y. TWEAK/Fn14 axis is an important player in fibrosis. J Cell Physiol 2020; 236:3304-3316. [PMID: 33000480 DOI: 10.1002/jcp.30089] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022]
Abstract
Fibrosis is a common pathological condition associated with abnormal repair after tissue injury. However, the etiology and molecular mechanisms of fibrosis are still not well-understood. Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) belongs to the TNF superfamily and acts by binding to its receptor, fibroblast growth factor-inducible 14 (Fn14), thereby activating a variety of intracellular signal transduction pathways in various types of cells. Besides promoting the expression of growth factors, activation of TWEAK/Fn14 signaling after tissue injury can promote the expression of pro-inflammatory cytokines, which trigger the immune response, thereby exacerbating the injury. Severe or repetitive injury leads to a dysregulated tissue repair process, in which the TWEAK/Fn14 axis promotes the activation and proliferation of myofibroblasts, induces the secretion of the extracellular matrix, and regulates profibrotic mediators to further perpetuate and sustain the fibrotic process. In this review, we summarize the available experimental evidence on the underlying molecular mechanisms by which the TWEAK/Fn14 pathway mediates the development and progression of fibrosis. In addition, we discuss the therapeutic potential of the TWEAK/Fn14 pathway in fibrosis-associated diseases based on evidence derived from multiple models and cells from injured tissue and fibrotic tissue.
Collapse
Affiliation(s)
- Yitian Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
7
|
Magliozzi R, Marastoni D, Calabrese M. The BAFF / APRIL system as therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2020; 24:1135-1145. [PMID: 32900236 DOI: 10.1080/14728222.2020.1821647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The complex system of BAFF (B-cell-activating factor of the TNF family) and APRIL (A proliferation-inducing ligand) has been studied in animal models of autoimmune diseases such as those resembling human systemic lupus erythematosus and Sjogren's syndrome and multiple sclerosis (MS). Accumulating evidence suggests that BAFF and APRIL have a physiological role in B cell immunity regulation, however inappropriate production of these factors may represent a key event which disrupts immune tolerance which is associated with systemic autoimmune diseases. AREAS COVERED We provide an update on the latest studies of the BAFF/APRIL system in multiple sclerosis, as well as on related clinical trials. EXPERT OPINION Experimental and clinical evidence suggests that increased BAFF levels may interfere directly and indirectly with B cell immunity; this can lead to breakdown of immune tolerance, the production of autoantibodies and continuous local intracerebral inflammation and brain tissue destruction. A more comprehensive understanding of the cell/molecular mechanism immune reactions specifically regulated by BAFF/APRIL in MS would better elucidate the specific cell phenotype targeted by actual anti-BAFF/APRIL therapies; this may enable the identification of either specific biomarkers of MS subgroups that would benefit of anti-BAFF/APRIL treatments or new targets of MS-specific anti-BAFF/APRIL therapies.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| | - Damiano Marastoni
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| | - Massimiliano Calabrese
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| |
Collapse
|
8
|
Wang M, Xie Z, Xu J, Feng Z. TWEAK/Fn14 axis in respiratory diseases. Clin Chim Acta 2020; 509:139-148. [PMID: 32526219 DOI: 10.1016/j.cca.2020.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/31/2020] [Accepted: 06/03/2020] [Indexed: 02/08/2023]
Abstract
Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a well known multifunctional cytokine extensively distributed in cell types and tissues. Accumulating evidence has shown that TWEAK binding to the receptor factor-inducible 14 (Fn14) participates in diverse pathologic processes including cell proliferation and death, angiogenesis, carcinogenesis and inflammation. Interestingly, alterations of intracellular signaling cascades are correlated to the development of respiratory disease. Recently, a several lines of evidence suggests that TWEAK in lung tissues are closely associated with these signaling pathways. In this review, we explore if TWEAK could provide a novel therapeutic strategy for managing respiratory disease in general and pulmonary arterial hypertension (PAH), obstructive sleep apnea syndrome (OSAS), asthma, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC), specifically.
Collapse
Affiliation(s)
- Min Wang
- Department of Otorhinolaryngology, University of South China Affiliated Nanhua Hospital, Hengyang 421002, China
| | - Zhijuan Xie
- Department of Nephrology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Jin Xu
- School of Pharmaceutical Sciences, Changsha Medical University, Changsha 410219, Hunan, China.
| | - Zhuyu Feng
- Department of Critical Care Medicine, University of South China Affiliated Nanhua Hospital, Hengyang 421002, China.
| |
Collapse
|
9
|
McCartney AM, Hyland EM, Cormican P, Moran RJ, Webb AE, Lee KD, Hernandez-Rodriguez J, Prado-Martinez J, Creevey CJ, Aspden JL, McInerney JO, Marques-Bonet T, O'Connell MJ. Gene Fusions Derived by Transcriptional Readthrough are Driven by Segmental Duplication in Human. Genome Biol Evol 2020; 11:2678-2690. [PMID: 31400206 PMCID: PMC6764479 DOI: 10.1093/gbe/evz163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2019] [Indexed: 12/14/2022] Open
Abstract
Gene fusion occurs when two or more individual genes with independent open reading frames becoming juxtaposed under the same open reading frame creating a new fused gene. A small number of gene fusions described in detail have been associated with novel functions, for example, the hominid-specific PIPSL gene, TNFSF12, and the TWE-PRIL gene family. We use Sequence Similarity Networks and species level comparisons of great ape genomes to identify 45 new genes that have emerged by transcriptional readthrough, that is, transcription-derived gene fusion. For 35 of these putative gene fusions, we have been able to assess available RNAseq data to determine whether there are reads that map to each breakpoint. A total of 29 of the putative gene fusions had annotated transcripts (9/29 of which are human-specific). We carried out RT-qPCR in a range of human tissues (placenta, lung, liver, brain, and testes) and found that 23 of the putative gene fusion events were expressed in at least one tissue. Examining the available ribosome foot-printing data, we find evidence for translation of three of the fused genes in human. Finally, we find enrichment for transcription-derived gene fusions in regions of known segmental duplication in human. Together, our results implicate chromosomal structural variation brought about by segmental duplication with the emergence of novel transcripts and translated protein products.
Collapse
Affiliation(s)
- Ann M McCartney
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Ireland.,Computational and Molecular Evolutionary Biology Group, School of Biology, Faculty of Biological Sciences, The University of Leeds, United Kingdom
| | - Edel M Hyland
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Ireland.,Institute for Global Food Security, Queens University Belfast, United Kingdom
| | - Paul Cormican
- Teagasc Animal and Bioscience Research Department, Animal & Grassland Research and Innovation Centre, Teagasc, Grange, Dunsany, County Meath, Ireland
| | - Raymond J Moran
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Ireland.,Computational and Molecular Evolutionary Biology Group, School of Biology, Faculty of Biological Sciences, The University of Leeds, United Kingdom
| | - Andrew E Webb
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Ireland
| | - Kate D Lee
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Ireland.,School of Biological Sciences, University of Auckland, New Zealand.,School of Fundamental Sciences, Massey University, New Zealand
| | | | - Javier Prado-Martinez
- Institute of Evolutionary Biology (UPF-CSIC), PRBB, Dr. Aiguader 88, 08003 Barcelona, Spain.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Christopher J Creevey
- Institute for Global Food Security, Queens University Belfast, United Kingdom.,Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, United Kingdom
| | - Julie L Aspden
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, The University of Leeds, United Kingdom
| | - James O McInerney
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, M13 9PL, United Kingdom.,School of Life Sciences, Faculty of Medicine and Health Sciences, The University of Nottingham, NG7 2RD, United Kingdom
| | - Tomas Marques-Bonet
- Institute of Evolutionary Biology (UPF-CSIC), PRBB, Dr. Aiguader 88, 08003 Barcelona, Spain.,Catalan Institution of Research and Advanced Studies (ICREA), Passeig de Lluís Companys, 23, 08010, Barcelona, Spain.,NAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 4, 08028 Barcelona, Spain.,Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, Edifici ICTA-ICP, c/ Columnes s/n, 08193 Cerdanyola del Vallés, Barcelona, Spain
| | - Mary J O'Connell
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Ireland.,Computational and Molecular Evolutionary Biology Group, School of Biology, Faculty of Biological Sciences, The University of Leeds, United Kingdom.,School of Life Sciences, Faculty of Medicine and Health Sciences, The University of Nottingham, NG7 2RD, United Kingdom
| |
Collapse
|
10
|
Jackson SW, Davidson A. BAFF inhibition in SLE-Is tolerance restored? Immunol Rev 2019; 292:102-119. [PMID: 31562657 PMCID: PMC6935406 DOI: 10.1111/imr.12810] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023]
Abstract
The B cell activating factor (BAFF) inhibitor, belimumab, is the first biologic drug approved for the treatment of SLE, and exhibits modest, but durable, efficacy in decreasing disease flares and organ damage. BAFF and its homolog APRIL are TNF-like cytokines that support the survival and differentiation of B cells at distinct developmental stages. BAFF is a crucial survival factor for transitional and mature B cells that acts as rheostat for the maturation of low-affinity autoreactive cells. In addition, BAFF augments innate B cell responses via complex interactions with the B cell receptor (BCR) and Toll like receptor (TLR) pathways. In this manner, BAFF impacts autoreactive B cell activation via extrafollicular pathways and fine tunes affinity selection within germinal centers (GC). Finally, BAFF and APRIL support plasma cell survival, with differential impacts on IgM- and IgG-producing populations. Therapeutically, BAFF and combined BAFF/APRIL inhibition delays disease onset in diverse murine lupus strains, although responsiveness to BAFF inhibition is model dependent, in keeping with heterogeneity in clinical responses to belimumab treatment in humans. In this review, we discuss the mechanisms whereby BAFF/APRIL signals promote autoreactive B cell activation, discuss whether altered selection accounts for therapeutic benefits of BAFF inhibition, and address whether new insights into BAFF/APRIL family complexity can be exploited to improve human lupus treatments.
Collapse
Affiliation(s)
- Shaun W Jackson
- Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Anne Davidson
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| |
Collapse
|
11
|
Barresi V, Cosentini I, Scuderi C, Napoli S, Di Bella V, Spampinato G, Condorelli DF. Fusion Transcripts of Adjacent Genes: New Insights into the World of Human Complex Transcripts in Cancer. Int J Mol Sci 2019; 20:ijms20215252. [PMID: 31652751 PMCID: PMC6862657 DOI: 10.3390/ijms20215252] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/18/2019] [Accepted: 10/20/2019] [Indexed: 12/12/2022] Open
Abstract
The awareness of genome complexity brought a radical approach to the study of transcriptome, opening eyes to single RNAs generated from two or more adjacent genes according to the present consensus. This kind of transcript was thought to originate only from chromosomal rearrangements, but the discovery of readthrough transcription opens the doors to a new world of fusion RNAs. In the last years many possible intergenic cis-splicing mechanisms have been proposed, unveiling the origins of transcripts that contain some exons of both the upstream and downstream genes. In some cases, alternative mechanisms, such as trans-splicing and transcriptional slippage, have been proposed. Five databases, containing validated and predicted Fusion Transcripts of Adjacent Genes (FuTAGs), are available for the scientific community. A comparative analysis revealed that two of them contain the majority of the results. A complete analysis of the more widely characterized FuTAGs is provided in this review, including their expression pattern in normal tissues and in cancer. Gene structure, intergenic splicing patterns and exon junction sequences have been determined and here reported for well-characterized FuTAGs. The available functional data and the possible roles in cancer progression are discussed.
Collapse
Affiliation(s)
- Vincenza Barresi
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy.
| | - Ilaria Cosentini
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy.
| | - Chiara Scuderi
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy.
| | - Salvatore Napoli
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy.
| | - Virginia Di Bella
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy.
| | - Giorgia Spampinato
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy.
| | - Daniele Filippo Condorelli
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
12
|
Shabgah AG, Shariati-Sarabi Z, Tavakkol-Afshari J, Mohammadi M. The role of BAFF and APRIL in rheumatoid arthritis. J Cell Physiol 2019; 234:17050-17063. [PMID: 30941763 DOI: 10.1002/jcp.28445] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/10/2019] [Accepted: 02/14/2019] [Indexed: 12/17/2022]
Abstract
Development and activation of B cells quickly became clear after identifying new ligands and receptors in the tumor necrosis factor superfamily. B cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL) are the members of membrane proteins Type 2 family released by proteolytic cleavage of furin to form active, soluble homotrimers. Except for B cells, ligands are expressed by all such immune cells like T cells, dendritic cells, monocytes, and macrophages. BAFF and APRIL have two common receptors, namely TNFR homolog transmembrane activator and Ca2+ modulator and CAML interactor (TACI) and B cell-maturation antigen. BAFF alone can also be coupled with a third receptor called BAFFR (also called BR3 or BLyS Receptor). These receptors are often expressed by immune cells in the B-cell lineage. The binding of BAFF or APRIL to their receptors supports B cells differentiation and proliferation, immunoglobulin production and the upregulation of B cell-effector molecules expression. It is possible that the overexpression of BAFF and APRIL contributes to the pathogenesis of autoimmune diseases. In BAFF transgenic mice, there is a pseudo-autoimmune manifestation, which is associated with an increase in B-lymphocytes, hyperglobulinemia, anti-single stranded DNA, and anti-double-stranded DNA antibodies, and immune complexes in their peripheral blood. Furthermore, overexpressing BAFF augments the number of peripheral B220+ B cells with a normal proliferation rate, high levels of Bcl2, and prolonged survival and hyperactivity. Therefore, in this review article, we studied BAFF and APRIL as important mediators in B-cell and discussed their role in rheumatoid arthritis.
Collapse
Affiliation(s)
- Arezoo G Shabgah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zhaleh Shariati-Sarabi
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mojgan Mohammadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Lee WH, Seo D, Lim SG, Suk K. Reverse Signaling of Tumor Necrosis Factor Superfamily Proteins in Macrophages and Microglia: Superfamily Portrait in the Neuroimmune Interface. Front Immunol 2019; 10:262. [PMID: 30838001 PMCID: PMC6389649 DOI: 10.3389/fimmu.2019.00262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor necrosis factor (TNF) superfamily (TNFSF) is a protein superfamily of type II transmembrane proteins commonly containing the TNF homology domain. The superfamily contains more than 20 protein members, which can be released from the cell membrane by proteolytic cleavage. Members of the TNFSF function as cytokines and regulate diverse biological processes, including immune responses, proliferation, differentiation, apoptosis, and embryogenesis, by binding to TNFSF receptors. Many TNFSF proteins are also known to be responsible for the regulation of innate immunity and inflammation. Both receptor-mediated forward signaling and ligand-mediated reverse signaling play important roles in these processes. In this review, we discuss the functional expression and roles of various reverse signaling molecules and pathways of TNFSF members in macrophages and microglia in the central nervous system (CNS). A thorough understanding of the roles of TNFSF ligands and receptors in the activation of macrophages and microglia may improve the treatment of inflammatory diseases in the brain and periphery. In particular, TNFSF reverse signaling in microglia can be exploited to gain further insights into the functions of the neuroimmune interface in physiological and pathological processes in the CNS.
Collapse
Affiliation(s)
- Won-Ha Lee
- BK21 Plus KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, South Korea
| | - Donggun Seo
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, School of Medicine, Brain Science & Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Su-Geun Lim
- BK21 Plus KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, South Korea
| | - Kyoungho Suk
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, School of Medicine, Brain Science & Engineering Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
14
|
The role of APRIL - A proliferation inducing ligand - In autoimmune diseases and expectations from its targeting. J Autoimmun 2018; 95:179-190. [DOI: 10.1016/j.jaut.2018.10.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
|
15
|
Howard L, Wosnitzka E, Okakpu D, White MA, Wyatt S, Davies AM. TWE-PRIL reverse signalling suppresses sympathetic axon growth and tissue innervation. Development 2018; 145:dev.165936. [PMID: 30337376 PMCID: PMC6262789 DOI: 10.1242/dev.165936] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022]
Abstract
TWE-PRIL is a naturally occurring fusion protein of components of two TNF superfamily members: the extracellular domain of APRIL; and the intracellular and transmembrane domains of TWEAK with no known function. Here, we show that April−/− mice (which lack APRIL and TWE-PRIL) exhibited overgrowth of sympathetic fibres in vivo, and sympathetic neurons cultured from these mice had significantly longer axons than neurons cultured from wild-type littermates. Enhanced axon growth from sympathetic neurons cultured from April−/− mice was prevented by expressing full-length TWE-PRIL in these neurons but not by treating them with soluble APRIL. Soluble APRIL, however, enhanced axon growth from the sympathetic neurons of wild-type mice. siRNA knockdown of TWE-PRIL but not siRNA knockdown of APRIL alone also enhanced axon growth from wild-type sympathetic neurons. Our work reveals the first and physiologically relevant role for TWE-PRIL and suggests that it mediates reverse signalling. Summary:In vivo and in vitro studies of superior cervical ganglion neurons of April−/− mice reveal that TWE-PRIL is a physiological regulator of NGF-promoted sympathetic axon growth, acting as a reverse signalling receptor.
Collapse
Affiliation(s)
- Laura Howard
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Erin Wosnitzka
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Darian Okakpu
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Matthew A White
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Sean Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Alun M Davies
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| |
Collapse
|
16
|
Wallach D. The Tumor Necrosis Factor Family: Family Conventions and Private Idiosyncrasies. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028431. [PMID: 28847899 DOI: 10.1101/cshperspect.a028431] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The tumor necrosis factor (TNF) cytokine family and the TNF/nerve growth factor (NGF) family of their cognate receptors together control numerous immune functions, as well as tissue-homeostatic and embryonic-development processes. These diverse functions are dictated by both shared and distinct features of family members, and by interactions of some members with nonfamily ligands and coreceptors. The spectra of their activities are further expanded by the occurrence of the ligands and receptors in both membrane-anchored and soluble forms, by "re-anchoring" of soluble forms to extracellular matrix components, and by signaling initiation via intracellular domains (IDs) of both receptors and ligands. Much has been learned about shared features of the receptors as well as of the ligands; however, we still have only limited knowledge of the mechanistic basis for their functional heterogeneity and for the differences between their functions and those of similarly acting cytokines of other families.
Collapse
Affiliation(s)
- David Wallach
- Department of Biomolecular Sciences, The Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
17
|
Pintarelli G, Dassano A, Cotroneo CE, Galvan A, Noci S, Piazza R, Pirola A, Spinelli R, Incarbone M, Palleschi A, Rosso L, Santambrogio L, Dragani TA, Colombo F. Read-through transcripts in normal human lung parenchyma are down-regulated in lung adenocarcinoma. Oncotarget 2017; 7:27889-98. [PMID: 27058892 PMCID: PMC5053695 DOI: 10.18632/oncotarget.8556] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/18/2016] [Indexed: 12/26/2022] Open
Abstract
Read-through transcripts result from the continuous transcription of adjacent, similarly oriented genes, with the splicing out of the intergenic region. They have been found in several neoplastic and normal tissues, but their pathophysiological significance is unclear. We used high-throughput sequencing of cDNA fragments (RNA-Seq) to identify read-through transcripts in the non-involved lung tissue of 64 surgically treated lung adenocarcinoma patients. A total of 52 distinct read-through species was identified, with 24 patients having at least one read-through event, up to a maximum of 17 such transcripts in one patient. Sanger sequencing validated 28 of these transcripts and identified an additional 15, for a total of 43 distinct read-through events involving 35 gene pairs. Expression levels of 10 validated read-through transcripts were measured by quantitative PCR in pairs of matched non-involved lung tissue and lung adenocarcinoma tissue from 45 patients. Higher expression levels were observed in normal lung tissue than in the tumor counterpart, with median relative quantification ratios between normal and tumor varying from 1.90 to 7.78; the difference was statistically significant (P < 0.001, Wilcoxon's signed-rank test for paired samples) for eight transcripts: ELAVL1–TIMM44, FAM162B–ZUFSP, IFNAR2–IL10RB, INMT–FAM188B, KIAA1841–C2orf74, NFATC3–PLA2G15, SIRPB1–SIRPD, and SHANK3–ACR. This report documents the presence of read-through transcripts in apparently normal lung tissue, with inter-individual differences in patterns and abundance. It also shows their down-regulation in tumors, suggesting that these chimeric transcripts may function as tumor suppressors in lung tissue.
Collapse
Affiliation(s)
- Giulia Pintarelli
- Department of Predictive and Prevention Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Alice Dassano
- Department of Predictive and Prevention Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara E Cotroneo
- Department of Predictive and Prevention Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy.,Present Address: UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Antonella Galvan
- Formerly, Department of Predictive and Prevention Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Noci
- Department of Predictive and Prevention Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Rocco Piazza
- Department of Health Sciences, University of Milano-Bicocca, Monza, Italy.,Hematology and Clinical Research Unit, San Gerardo Hospital, Monza, Italy
| | - Alessandra Pirola
- Department of Health Sciences, University of Milano-Bicocca, Monza, Italy
| | - Roberta Spinelli
- Formerly, Department of Health Sciences, University of Milano-Bicocca, Monza, Italy
| | - Matteo Incarbone
- Department of Surgery, San Giuseppe Hospital, Multimedica, Milan, Italy
| | - Alessandro Palleschi
- Department of Surgery, IRCCS Fondazione Cà Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Rosso
- Department of Surgery, IRCCS Fondazione Cà Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Luigi Santambrogio
- Department of Surgery, IRCCS Fondazione Cà Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Tommaso A Dragani
- Department of Predictive and Prevention Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Colombo
- Department of Predictive and Prevention Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
18
|
Pangrazzi L, Meryk A, Naismith E, Koziel R, Lair J, Krismer M, Trieb K, Grubeck-Loebenstein B. "Inflamm-aging" influences immune cell survival factors in human bone marrow. Eur J Immunol 2017; 47:481-492. [PMID: 27995612 PMCID: PMC5434810 DOI: 10.1002/eji.201646570] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/21/2016] [Accepted: 12/14/2016] [Indexed: 01/19/2023]
Abstract
The bone marrow (BM) plays a key role in the long-term maintenance of immunological memory. However, the impact of aging on the production of survival factors for effector/memory T cells and plasma cells in the human BM has not been studied. We now show that the expression of molecules involved in the maintenance of immunological memory in the human BM changes with age. While IL-15, which protects potentially harmful CD8+ CD28- senescent T cells, increases, IL-7 decreases. IL-6, which may synergize with IL-15, is also overexpressed. In contrast, a proliferation-inducing ligand, a plasma cell survival factor, is reduced. IFN-y, TNF, and ROS accumulate in the BM in old age. IL-15 and IL-6 expression are stimulated by IFN-y and correlate with ROS levels in BM mononuclear cells. Both cytokines are reduced by incubation with the ROS scavengers N-acetylcysteine and vitamin C. IL-15 and IL-6 are also overexpressed in the BM of superoxide dismutase 1 knockout mice compared to their WT counterparts. In summary, our results demonstrate the role of inflammation and oxidative stress in age-related changes of immune cell survival factors in the BM, suggesting that antioxidants may be beneficial in counteracting immunosenescence by improving immunological memory in old age.
Collapse
Affiliation(s)
- Luca Pangrazzi
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Andreas Meryk
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Erin Naismith
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Rafal Koziel
- Department of Molecular and Cell Biology, Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
| | - Julian Lair
- Department of Orthopedic Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Martin Krismer
- Department of Orthopedic Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Klemens Trieb
- Department of Orthopedic Surgery, Hospital Wels-Grieskirchen, Wels, Austria
| | - Beatrix Grubeck-Loebenstein
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
19
|
Das S, Sutoh Y, Hirano M, Han Q, Li J, Cooper MD, Herrin BR. Characterization of Lamprey BAFF-like Gene: Evolutionary Implications. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:2695-703. [PMID: 27543613 PMCID: PMC5026938 DOI: 10.4049/jimmunol.1600799] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/25/2016] [Indexed: 12/25/2022]
Abstract
BAFF (TNF superfamily [TNFSF] 13B/Blys) and APRIL (TNFSF13) are important regulatory factors for lymphocyte activation and survival in mammals. A BAFF/APRIL-like relative called BAFF- and APRIL-like molecule (BALM) has also been identified in cartilaginous and bony fishes, and we report in this study a BAFF-like gene in lampreys. Our phylogenetic analysis of these genes and a related TNFSF12 gene called TNF-like weak inducer of apoptosis (TWEAK) suggest that, whereas an ancestral homolog of BAFF and APRIL was already present in a common ancestor of jawed and jawless vertebrates, TWEAK evolved early on in the jawed vertebrate lineage. Like mammalian BAFF and APRIL, the lamprey BAFF-like gene is expressed in T-like, B-like, and innate immune cells. The predicted protein encoded by this BAFF-like gene in lampreys exhibits higher sequence similarity with mammalian BAFF than APRIL. Correspondingly, we find BAFF orthologs in all of the jawed vertebrate representatives that we examined, although APRIL and/or BALM orthologs are not identifiable in certain jawed vertebrates. For example, BALM is not identifiable in tetrapods, and APRIL is not identifiable in several bony fishes or in birds, the latter of which also lack a TWEAK-like gene. Our analysis further suggests that a hybrid molecule called TWE-PRIL, which is a product of an in-genomic fusion between APRIL and TWEAK genes evolved early in mammalian evolution.
Collapse
Affiliation(s)
- Sabyasachi Das
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Yoichi Sutoh
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Masayuki Hirano
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Qifeng Han
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Jianxu Li
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Max D Cooper
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Brantley R Herrin
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| |
Collapse
|
20
|
Abstract
The prognosis of patients with classical Hodgkin lymphoma following chemo- and radiotherapy has been excellent during the last 4 decades. However, the development of secondary malignancies is of major concern. Therefore, the reduction of radiotherapy application is a major objective of ongoing clinical trials. De-escalation of treatment may increase the risk of relapses and thus may lead to reappearance of prognostic factors. Prognostic biomarkers might help to identify patients who are at increased risk of relapse. This review summarizes the current knowledge about potential prognostic biomarkers for patients with classical Hodgkin lymphoma.
Collapse
Affiliation(s)
- Martin S Staege
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Stefanie Kewitz
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Toralf Bernig
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Caspar Kühnöl
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Christine Mauz-Körholz
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| |
Collapse
|
21
|
van Attekum M, Terpstra S, Reinen E, Kater AP, Eldering E. Macrophage-mediated chronic lymphocytic leukemia cell survival is independent of APRIL signaling. Cell Death Discov 2016; 2:16020. [PMID: 27551513 PMCID: PMC4979474 DOI: 10.1038/cddiscovery.2016.20] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/03/2016] [Indexed: 12/27/2022] Open
Abstract
Survival of chronic lymphocytic leukemia (CLL) cells is mainly driven by interactions within the lymph node (LN) microenvironment with bystander cells such as T cells or cells from the monocytic lineage. Although the survival effect by T cells is largely governed by the TNFR ligand family member CD40L, the exact mechanism of monocyte-derived cell-induced survival is not known. An important role has been attributed to the TNFR ligand, a proliferation-inducing ligand (APRIL), although the exact mechanism remained unclear. Since we detected that APRIL was expressed by CD68+ cells in CLL LN, we addressed its relevance in various aspects of CLL biology, using a novel APRIL overexpressing co-culture system, recombinant APRIL, and APRIL reporter cells. Unexpectedly, we found, that in these various systems, APRIL had no effect on survival of CLL cells, and activation of NF-κB was not enhanced on APRIL stimulation. Moreover, APRIL stity mulation did not affect CLL proliferation, neither as single stimulus nor in combination with known CLL proliferation stimuli. Furthermore, the survival effect conveyed by macrophages to CLL cells was not affected by transmembrane activator and CAML interactor-Fc, an APRIL decoy receptor. We conclude that the direct role ascribed to APRIL in CLL cell survival might be overestimated due to application of supraphysiological levels of recombinant APRIL.
Collapse
Affiliation(s)
- Mha van Attekum
- Academic Medical Center, Department of Hematology, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Academic Medical Center, Department of Experimental Immunology, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - S Terpstra
- Academic Medical Center, Department of Hematology, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Academic Medical Center, Department of Experimental Immunology, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - E Reinen
- Academic Medical Center, Department of Hematology, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Academic Medical Center, Department of Experimental Immunology, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - A P Kater
- Academic Medical Center, Department of Hematology, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - E Eldering
- Academic Medical Center, Department of Experimental Immunology, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Lu G, Wu J, Zhao G, Wang Z, Chen W, Mu S. Abundant and broad expression of transcription-induced chimeras and protein products in mammalian genomes. Biochem Biophys Res Commun 2015; 470:759-765. [PMID: 26718406 DOI: 10.1016/j.bbrc.2015.12.084] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 12/19/2015] [Indexed: 10/22/2022]
Abstract
The expression of transcription-induced chimeras (TICs) was underestimated due to strategic and logical reasons. In order to thoroughly examine TICs, systematic survey of TIC events was conducted in mammalian genomes using ESTs, followed by experimental validation using RT-PCR and real-time quantitative PCR (qPCR). The expression of ∼98% TIC events in at least one tissue or cell line from both mouse and human was verified. Besides, ∼40% TICs were broadly expressed, and ∼33% of TICs showed expression levels comparable to or higher than their upstream parental genes. We further identified putative chimeric proteins in public databases and validated two using Western blotting. GO analysis revealed that proteins resided in one multi-protein complex or functioning in metabolic or signaling pathway tended to produce fused products. Taken together, we have shown substantial evidence for the underestimated TIC events; and TICs could be a novel regulated way to further increases the proteome complexity in mammalian genomes. Possible regulation mechanisms and evolution of TICs were also discussed.
Collapse
Affiliation(s)
- Guanting Lu
- Department of Blood Transfusion, Tangdu Hospital, The Fourth Military Medical University, 710032, Xi'an, China
| | - Jin Wu
- Department of Clinical Oncology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | | | - Zhiqiang Wang
- Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Weihua Chen
- Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
| | - Shijie Mu
- Department of Blood Transfusion, Tangdu Hospital, The Fourth Military Medical University, 710032, Xi'an, China.
| |
Collapse
|
23
|
Functional characterization of BC039389-GATM and KLK4-KRSP1 chimeric read-through transcripts which are up-regulated in renal cell cancer. BMC Genomics 2015; 16:247. [PMID: 25888189 PMCID: PMC4422297 DOI: 10.1186/s12864-015-1446-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2015] [Indexed: 11/10/2022] Open
Abstract
Background Chimeric read-through RNAs are transcripts originating from two directly adjacent genes (<10 kb) on the same DNA strand. Although they are found in next-generation whole transcriptome sequencing (RNA-Seq) data on a regular basis, investigating them further has usually been refrained from. Therefore, their expression patterns or functions in general, and in oncogenesis in particular, are poorly understood. Results We used paired-end RNA-Seq and a specifically designed computational data analysis pipeline (FusionSeq) to nominate read-through events in a small discovery set of renal cell carcinomas (RCC) and confirmed them in a larger validation cohort. 324 read-through events were called overall; 22/27 (81%) selected nominees passed validation with conventional PCR and were sequenced at the junction region. We frequently identified various isoforms of a given read-through event. 2/22 read-throughs were up-regulated: BC039389-GATM was higher expressed in RCC compared to benign adjacent kidney; KLK4-KRSP1 was expressed in 46/169 (27%) RCCs, but rarely in normal tissue. KLK4-KRSP1 expression was associated with worse clinical outcome in the patient cohort. In cell lines, both read-throughs influenced molecular mechanisms (i.e. target gene expression or migration/invasion) in a way that counteracted the effect of the respective parent transcript GATM or KLK4. Conclusions Our data suggests that the up-regulation of read-through RNA chimeras in tumors is not random but causes regulatory effects on cellular mechanisms and may impact patient survival. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1446-z) contains supplementary material, which is available to authorized users.
Collapse
|
24
|
Effect of TACI signaling on humoral immunity and autoimmune diseases. J Immunol Res 2015; 2015:247426. [PMID: 25866827 PMCID: PMC4381970 DOI: 10.1155/2015/247426] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/04/2015] [Indexed: 02/02/2023] Open
Abstract
Transmembrane activator and calcium-modulating cyclophilin ligand interactor (TACI) is one of the receptors of B cell activating factor of the tumor necrosis factor family (BAFF) and a proliferation-inducing ligand (APRIL). TACI is a regulator in the immune responses. TACI inhibits B cell expansion and promotes the differentiation and survival of plasma cells. The mechanisms underlying these effects probably involve changed expressions of some crucial molecules, such as B lymphocyte induced maturation protein-1 (Blimp-1) and inducible T-cell costimulator ligand (ICOSL) in B cells and/or plasma cells. However, abnormal TACI signaling may relate to autoimmune disorders. Common variable immune deficiency (CVID) patients with heterozygous mutations in TACI alleles increase susceptibility to autoimmune diseases. Taci−/− mice and BAFF transgenic mice both develop signs of human SLE. These findings that indicate inappropriate levels of TACI signaling may disrupt immune system balance, thereby promoting the development of autoimmune diseases. In this review, we summarize the basic characteristics of the TACI ligands BAFF and APRIL, and detail the research findings on the role of TACI in humoral immunity. We also discuss the possible mechanisms underlying the susceptibility of CVID patients with TACI mutations to autoimmune diseases and the role of TACI in the pathogenesis of SLE.
Collapse
|
25
|
Weldon AJ, Moldovan I, Cabling MG, Hernandez EA, Hsu S, Gonzalez J, Parra A, Benitez A, Daoud N, Colburn K, Payne KJ. Surface APRIL Is Elevated on Myeloid Cells and Is Associated with Disease Activity in Patients with Rheumatoid Arthritis. J Rheumatol 2015; 42:749-59. [PMID: 25729037 DOI: 10.3899/jrheum.140630] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To assess surface APRIL (a proliferation-inducing ligand; CD256) expression by circulating myeloid cells in rheumatoid arthritis (RA) and to determine its relationship to disease activity. METHODS Peripheral blood mononuclear cells (PBMC) and plasma were obtained from patients with RA and healthy donors. PBMC were stained for flow cytometry to detect surface APRIL and blood cell markers to identify circulating myeloid cell subsets. Based on CD14 and CD16 phenotypes, monocyte subsets described as classical (CD14+CD16-), intermediate (CD14+CD16+), and nonclassical (CD14loCD16+) were identified. Levels of surface APRIL expression were measured by flow cytometry and median fluorescence intensity was used for comparisons. Levels of soluble APRIL in the plasma were determined by ELISA. Disease activity was measured by the Disease Activity Score in 28 joints. RESULTS In patients with RA, total myeloid cells showed expression of surface APRIL that correlated with disease activity and with plasma APRIL levels observed in these patients. In healthy donors, classical monocytes were composed of > 80% of circulating monocytes. However, in patients with RA, the intermediate and nonclassical subsets were elevated and made up the majority of circulating monocytes. In contrast to healthy donors, where high levels of surface APRIL were only observed in nonclassical monocytes, patients with RA showed high levels of surface APRIL expression by all circulating monocyte subsets. CONCLUSION Surface APRIL is elevated in circulating myeloid cells in patients with RA where it is highly correlated with disease activity. Patients with RA also showed skewing of monocytes toward subsets associated with secretion of tumor necrosis factor-α and/or interleukin 1β.
Collapse
Affiliation(s)
- Abby Jones Weldon
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University.
| | - Ioana Moldovan
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Marven G Cabling
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Elvin A Hernandez
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Sheri Hsu
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Jennifer Gonzalez
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Andrea Parra
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Abigail Benitez
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Nasim Daoud
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Keith Colburn
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Kimberly J Payne
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| |
Collapse
|
26
|
Staines K, Hunt LG, Young JR, Butter C. Evolution of an expanded mannose receptor gene family. PLoS One 2014; 9:e110330. [PMID: 25390371 PMCID: PMC4229073 DOI: 10.1371/journal.pone.0110330] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 09/21/2014] [Indexed: 01/22/2023] Open
Abstract
Sequences of peptides from a protein specifically immunoprecipitated by an antibody, KUL01, that recognises chicken macrophages, identified a homologue of the mammalian mannose receptor, MRC1, which we called MRC1L-B. Inspection of the genomic environment of the chicken gene revealed an array of five paralogous genes, MRC1L-A to MRC1L-E, located between conserved flanking genes found either side of the single MRC1 gene in mammals. Transcripts of all five genes were detected in RNA from a macrophage cell line and other RNAs, whose sequences allowed the precise definition of spliced exons, confirming or correcting existing bioinformatic annotation. The confirmed gene structures were used to locate orthologues of all five genes in the genomes of two other avian species and of the painted turtle, all with intact coding sequences. The lizard genome had only three genes, one orthologue of MRC1L-A and two orthologues of the MRC1L-B antigen gene resulting from a recent duplication. The Xenopus genome, like that of most mammals, had only a single MRC1-like gene at the corresponding locus. MRC1L-A and MRC1L-B genes had similar cytoplasmic regions that may be indicative of similar subcellular migration and functions. Cytoplasmic regions of the other three genes were very divergent, possibly indicating the evolution of a new functional repertoire for this family of molecules, which might include novel interactions with pathogens.
Collapse
Affiliation(s)
| | | | | | - Colin Butter
- The Pirbright Institute, Compton, United Kingdom
| |
Collapse
|
27
|
Figgett WA, Vincent FB, Saulep-Easton D, Mackay F. Roles of ligands from the TNF superfamily in B cell development, function, and regulation. Semin Immunol 2014; 26:191-202. [PMID: 24996229 DOI: 10.1016/j.smim.2014.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 06/09/2014] [Indexed: 01/01/2023]
Abstract
Most ligands from the tumour necrosis factor (TNF) superfamily play very important roles in the immune system, and particularly so in B lymphocyte biology. TNF ligands are essential to many aspects of normal B cell biology from development in the bone marrow to maturation in the periphery as well as for activation and differentiation into germinal centre, memory or plasma cells. TNF ligands also influence other aspects of B cell biology such as their ability to present antigens or regulate immune responses. Importantly, inadequate regulation of many TNF ligands is associated with B cell disorders including autoimmunity and cancers. As a result, inhibitors of a number of TNF ligands have been tested in the clinic, with some becoming very successful approved treatments alleviating B cell-mediated pathologies.
Collapse
Affiliation(s)
- William A Figgett
- Department of Immunology, Monash University, Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), Commercial Road, Melbourne, Victoria 3004, Australia
| | - Fabien B Vincent
- Department of Immunology, Monash University, Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), Commercial Road, Melbourne, Victoria 3004, Australia
| | - Damien Saulep-Easton
- Department of Immunology, Monash University, Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), Commercial Road, Melbourne, Victoria 3004, Australia
| | - Fabienne Mackay
- Department of Immunology, Monash University, Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), Commercial Road, Melbourne, Victoria 3004, Australia.
| |
Collapse
|
28
|
Liu XG, Hou M. Immune thrombocytopenia and B-cell-activating factor/a proliferation-inducing ligand. Semin Hematol 2014; 50 Suppl 1:S89-99. [PMID: 23664525 DOI: 10.1053/j.seminhematol.2013.03.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Primary immune thrombocytopenia (ITP) is an organ-specific autoimmune disorder characterized by autoantibody-mediated enhanced platelet destruction and dysmegakaryocytopoiesis. B cells have been demonstrated to play critical roles in the pathophysiology of ITP. B-cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL) are crucial cytokines supporting survival and differentiation of B cells, and dysregulation of BAFF/APRIL is involved in the pathogenesis of B-cell related autoimmune diseases including ITP. Currently ongoing clinical trials using BAFF and/or APRIL-blocking agents have yielded positive results in human systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA), further confirming the pathological role of BAFF/APRIL in autoimmunity. This review will describe the function of BAFF/APRIL and address the feasibility of BAFF/APRIL inhibition in the management of ITP.
Collapse
Affiliation(s)
- Xin-guang Liu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, PR China
| | | |
Collapse
|
29
|
Abstract
Systemic lupus erythematosus (SLE) is characterized by multisystem immune-mediated injury in the setting of autoimmunity to nuclear antigens. The clinical heterogeneity of SLE, the absence of universally agreed clinical trial end points, and the paucity of validated therapeutic targets have, historically, contributed to a lack of novel treatments for SLE. However, in 2011, a therapeutic monoclonal antibody that neutralizes the cytokine TNF ligand superfamily member 13B (also known as B-cell-activating factor of the TNF family [BAFF]), belimumab, became the first targeted therapy for SLE to have efficacy in a randomized clinical trial. Because of its specificity, the efficacy of belimumab provides an opportunity to increase understanding of SLE pathophysiology. Although belimumab depletes B cells, this effect is not as powerful as that of other B-cell-directed therapies that have not been proven efficacious in randomized clinical trials. In this article, therefore, we review results suggesting that neutralizing BAFF can have effects on the immune system other than depletion of B cells. We also identify aspects of the BAFF system for which data in relation to SLE are still missing, and we suggest studies to investigate the pathogenesis of SLE and ways to refine anti-BAFF therapies. The role of a related cytokine, TNF ligand superfamily member 13 (also known as a proliferation-inducing ligand [APRIL]) in SLE is much less well understood, and hence this review focuses on BAFF.
Collapse
|
30
|
Stohl W. Therapeutic targeting of the BAFF/APRIL axis in systemic lupus erythematosus. Expert Opin Ther Targets 2014; 18:473-89. [DOI: 10.1517/14728222.2014.888415] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
31
|
Sindhava VJ, Scholz JL, Stohl W, Cancro MP. APRIL mediates peritoneal B-1 cell homeostasis. Immunol Lett 2014; 160:120-7. [PMID: 24512739 DOI: 10.1016/j.imlet.2014.01.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 01/31/2014] [Indexed: 01/13/2023]
Abstract
BLyS (B lymphocyte stimulator) family cytokines and receptors play key roles in B-2 cell maturation and survival, but their importance for B-1 cells remains less clear. Here we use knockout mice to show that APRIL (A proliferation-inducing ligand), but not BLyS, plays a role in peritoneal B-1 cell maintenance. APRIL likely exerts its effects on peritoneal B-1 cells through binding to HSPG (heparan sulfate proteoglycans) rather than to the TACI (transmembrane activator and cyclophilin ligand interactor) receptor. Finally, we show that peritoneal macrophages express high levels of APRIL message, and are a likely local source of the cytokine in this anatomic locale.
Collapse
Affiliation(s)
- Vishal J Sindhava
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, United States
| | - Jean L Scholz
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, United States
| | - William Stohl
- Division of Rheumatology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, United States
| | - Michael P Cancro
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, United States.
| |
Collapse
|
32
|
Gardam S, Brink R. Non-Canonical NF-κB Signaling Initiated by BAFF Influences B Cell Biology at Multiple Junctures. Front Immunol 2014; 4:509. [PMID: 24432023 PMCID: PMC3880999 DOI: 10.3389/fimmu.2013.00509] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 12/24/2013] [Indexed: 01/13/2023] Open
Abstract
It has been more than a decade since it was recognized that the nuclear factor of kappa light polypeptide gene enhancer in B cells (NF-κB) transcription factor family was activated by two distinct pathways: the canonical pathway involving NF-κB1 and the non-canonical pathway involving NF-κB2. During this time a great deal of evidence has been amassed on the ligands and receptors that activate these pathways, the cytoplasmic adapter molecules involved in transducing the signals from receptors to nucleus, and the resulting physiological outcomes within body tissues. In contrast to NF-κB1 signaling, which can be activated by a wide variety of receptors, the NF-κB2 pathway is typically only activated by a subset of receptor and ligand pairs belonging to the tumor necrosis factor (TNF) family. Amongst these is B cell activating factor of the TNF family (BAFF) and its receptor BAFFR. Whilst BAFF is produced by many cell types throughout the body, BAFFR expression appears to be restricted to the hematopoietic lineage and B cells in particular. For this reason, the main physiological outcomes of BAFF mediated NF-κB2 activation are confined to B cells. Indeed BAFF mediated NF-κB2 signaling contributes to peripheral B cell survival and maturation as well as playing a role in antibody responses and long term maintenance plasma cells. Thus the importance BAFF and NF-κB2 permeates the entire B cell lifespan and impacts on this important component of the immune system in a variety of ways.
Collapse
Affiliation(s)
- Sandra Gardam
- Immunology Division, Garvan Institute of Medical Research , Darlinghurst, NSW , Australia
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research , Darlinghurst, NSW , Australia ; St. Vincent's Clinical School, University of New South Wales , Darlinghurst, NSW , Australia
| |
Collapse
|
33
|
Dore-Duffy P. Pericytes and adaptive angioplasticity: the role of tumor necrosis factor-like weak inducer of apoptosis (TWEAK). Methods Mol Biol 2014; 1135:35-52. [PMID: 24510853 DOI: 10.1007/978-1-4939-0320-7_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The TNF superfamily member TWEAK has emerged as a pleiotropic cytokine that regulates many cellular functions that include immune/inflammatory activity, angiogenesis, cell proliferation, and fate. TWEAK through its inducible receptor, FGF-inducible molecule 14 (Fn14), can induce both beneficial and deleterious activity that has a profound effect on cell survival. Thus it is highly likely that TWEAK and Fn14 expressed by cells of the neurovascular unit help regulate and maintain vascular and tissue homeostasis. In this chapter we discuss the expression of TWEAK and Fn14 signaling in the cerebral microvascular pericyte. Pericytes are a highly enigmatic population of microvascular cells that are important in regulatory pathways that modulate physiological angiogenesis in response to chronic mild hypoxic stress. A brief introduction will identify the microvascular pericyte. A more detailed discussion of pericyte TWEAK signaling during adaptive angioplasticity will follow.
Collapse
Affiliation(s)
- Paula Dore-Duffy
- Division of Neuroimmunology, Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
34
|
Lian M, Fang J, Han D, Ma H, Wang R, Yang F. The up-regulation expression of APRIL is a marker of glottic malignant disease. Eur Arch Otorhinolaryngol 2013; 271:2781-7. [PMID: 24276470 DOI: 10.1007/s00405-013-2826-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 11/13/2013] [Indexed: 12/11/2022]
Abstract
A proliferation-inducing ligand (APRIL) is a member of the tumor necrosis factor (TNF) family. Recent studies have implied that APRIL is closely related to solid tumors and hematological tumors, indicating that APRIL could be a potential marker to diagnose glottic malignant disease. The purpose of this study was to investigate the difference of the APRIL mRNA and protein expression in glottic malignant disease, corresponding adjacent non-neoplastic tissues and glottic benign lesion, and detect the influence of different clinical parameter in glottic carcinoma. The APRIL mRNA expression in the glottic carcinoma, corresponding adjacent non-neoplastic tissues and glottic polypus tissue samples from patients was detected by qRT-PCR. Moreover, we studied the APRIL protein expression in pathological sections of other patients with glottic carcinoma or glottic polypus using immunohistochemistry. All the patients with different clinical parameter underwent surgery. Using qRT-PCR, we revealed an up-regulation of APRIL mRNA expression in glottic carcinoma as compared to glottic polypus and corresponding adjacent non-neoplastic tissues, but no significant difference with T stages, histopathological differentiation grade or lymph node metastasis in glottic carcinoma. The result of the immunohistochemistry was the same, with no influence of different clinical parameter in glottic carcinoma. These results strongly suggest that APRIL could be a potential diagnosed marker to distinguish glottic malignant disease from glottic benign lesion, and it may play an important role in the development of glottic malignant disease.
Collapse
Affiliation(s)
- Meng Lian
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China,
| | | | | | | | | | | |
Collapse
|
35
|
Knockdown of specific host factors protects against influenza virus-induced cell death. Cell Death Dis 2013; 4:e769. [PMID: 23949218 PMCID: PMC3763457 DOI: 10.1038/cddis.2013.296] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 07/12/2013] [Accepted: 07/15/2013] [Indexed: 12/15/2022]
Abstract
Cell death is a characteristic consequence of cellular infection by influenza virus. Mounting evidence indicates the critical involvement of host-mediated cellular death pathways in promoting efficient influenza virus replication. Furthermore, it appears that many signaling pathways, such as NF-κB, formerly suspected to solely promote cell survival, can also be manipulated to induce cell death. Current understanding of the cell death pathways involved in influenza virus-mediated cytopathology and in virus replication is limited. This study was designed to identify host genes that are required for influenza-induced cell death. The approach was to perform genome-wide lentiviral-mediated human gene silencing in A549 cells and determine which genes could be silenced to provide resistance to influenza-induced cell death. The assay proved to be highly reproducible with 138 genes being identified in independent screens. The results were independently validated using siRNA to each of these candidates. Graded protection was observed in this screen with the silencing of any of 19 genes, each providing >85% protection. Three gene products, TNFSF13 (APRIL), TNFSF12-TNFSF13 (TWE-PRIL) and USP47, were selected because of the high levels of protection conferred by their silencing. Protein and mRNA silencing and protection from influenza-induced cell death was confirmed using multiple shRNA clones and siRNA, indicating the specificity of the effects. USP47 knockdown prevented proper viral entry into the host cell, whereas TNFSF12-13/TNFSF13 knockdown blocked a late stage in viral replication. This screening approach offers the means to identify a large number of potential candidates for the analysis of viral-induced cell death. These results may also have much broader applicability in defining regulatory mechanisms involved in cell survival.
Collapse
|
36
|
Vincent FB, Saulep-Easton D, Figgett WA, Fairfax KA, Mackay F. The BAFF/APRIL system: emerging functions beyond B cell biology and autoimmunity. Cytokine Growth Factor Rev 2013; 24:203-15. [PMID: 23684423 PMCID: PMC7108297 DOI: 10.1016/j.cytogfr.2013.04.003] [Citation(s) in RCA: 325] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The BAFF system plays a key role in the development of autoimmunity, especially in systemic lupus erythematosus (SLE). This often leads to the assumption that BAFF is mostly a B cell factor with a specific role in autoimmunity. Focus on BAFF and autoimmunity, driven by pharmaceutical successes with the recent approval of a novel targeted therapy Belimumab, has relegated other potential roles of BAFF to the background. Far from being SLE-specific, the BAFF system has a much broader relevance in infection, cancer and allergy. In this review, we provide the latest views on additional roles of the BAFF system in health and diseases, as well as an update on BAFF and autoimmunity, with particular focus on current clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Fabienne Mackay
- Corresponding author at: Department of Immunology, Monash University, Central Clinical School, Alfred Hospital, 89 Commercial Road, Melbourne, Victoria 3004, Australia. Tel.: +61 3 99030713; fax: +61 3 99030038.
| |
Collapse
|
37
|
Molecular structure, expression pattern and functional characterisation of APRIL in an aquatic mammal. Int Immunopharmacol 2013; 16:171-7. [PMID: 23597427 DOI: 10.1016/j.intimp.2013.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 03/25/2013] [Accepted: 03/25/2013] [Indexed: 11/22/2022]
Abstract
The Yangtze finless porpoise (Neophocaena phocaenoides asiaorientalis) is listed on the First Order of Protected Animals in China and was identified as an endangered species by the International Union for Conservation of Nature and Natural Resources (IUCN) in 2011. A proliferation inducing ligand (APRIL), belonging to the tumour necrosis factor (TNF) family, is critical for immune regulation. In this study, we identified a finless porpoise APRIL cDNA (fAPRIL) by RACE (rapid amplification of cDNA ends) strategies, from the Yangtze finless porpoise (fAPRIL). This gene encodes 247 amino acids containing a predicted transmembrane domain and a TNF domain, and phylogenetic analysis of the APRIL sequence indicated that finless porpoises are closely related to Artiodactyla. In vitro, soluble fAPRIL (fsAPRIL) not only promoted the survival/proliferation of the mouse spleen lymphocytes, but also bound specifically to the surface of the B cells. The results of this study contribute to our understanding of the immune mechanisms in the finless porpoise and other aquatic mammals.
Collapse
|
38
|
Antibody deficiency associated with an inherited autosomal dominant mutation in TWEAK. Proc Natl Acad Sci U S A 2013; 110:5127-32. [PMID: 23493554 DOI: 10.1073/pnas.1221211110] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mutations in the TNF family of proteins have been associated with inherited forms of immune deficiency. Using an array-based sequencing assay, we identified an autosomal-dominant deficiency in TNF-like weak inducer of apoptosis (TWEAK; TNFSF12) in a kindred with recurrent infection and impaired antibody responses to protein and polysaccharide vaccines. This mutation occurs in the sixth exon of TWEAK and results in the amino acid substitution R145C within the conserved TNF-homology domain of the full-length protein. TWEAK mutant protein formed high molecular weight aggregates under nonreducing conditions, suggesting an increased propensity for intermolecular interactions. As a result, mutant TWEAK associated with B-cell-activating factor (BAFF) protein and down-regulated the BAFF-mediated activation of the noncanonical NF-κB pathway through inhibition of p100 processing to p52, resulting in inhibition of BAFF-dependent B-cell survival and proliferation. As BAFF mediates T-cell-independent isotype switching and B-cell survival, our data implicate TWEAK as a disease-susceptibility gene for a humoral immunodeficiency.
Collapse
|
39
|
Sindhava VJ, Scholz JL, Cancro MP. Roles for BLyS family members in meeting the distinct homeostatic demands of innate and adaptive B cells. Front Immunol 2013; 4:37. [PMID: 23443938 PMCID: PMC3580333 DOI: 10.3389/fimmu.2013.00037] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/31/2013] [Indexed: 11/13/2022] Open
Abstract
B-1 and B-2 B cell populations have different progenitors, receptor diversity, anatomic location, and functions – suggesting vastly differing requisites for homeostatic regulation. There is evidence that the B lymphocyte stimulator (BLyS) family of cytokines and receptors, key factors in the homeostatic regulation of B-2 B cell subsets, is also a major player in the B-1 compartment. Here we review the development and differentiation of these two primary B cell lineages and their immune functions. We discuss evidence that BLyS or a proliferation-inducing ligand (APRIL) availability in different anatomic sites, coupled with signature BLyS receptor expression patterns on different B cell subsets, may be important for homeostatic regulation of B-1 as well as B-2 populations. Finally, we extend our working model of B cell homeostasis to integrate B-1s.
Collapse
Affiliation(s)
- Vishal J Sindhava
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| | | | | |
Collapse
|
40
|
Zhi-Chun L, Qiao-Ling Z, Zhi-Qin L, Xiao-Zhao L, Xiao-xia Z, Rong T. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) mediates p38 mitogen-activated protein kinase activation and signal transduction in peripheral blood mononuclear cells from patients with lupus nephritis. Inflammation 2012; 35:935-43. [PMID: 22009442 DOI: 10.1007/s10753-011-9396-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Forty-two patients with systemic lupus erythematosus (SLE), including 26 patients with renal damage and 16 without, and 20 healthy controls were included in the study. The isolated peripheral blood mononuclear cells (PBMCs) were treated with a p38 inhibitor (SB203580) or anti-tumor necrosis factor-like weak inducer of apoptosis (TWEAK) mAb, with or without phytohemagglutinin/phorbol myristate acetate (PHA/PMA) stimulation. Western blot experiments were used to evaluate the protein expression of TWEAK and p38 MAPK in PBMCs .Next, the contents of interleukin-10 (IL-10) and monocyte chemoattractant protein-1 (MCP-1) in the supernatant were measured by ELISA. The results showed that expression of TWEAK protein in PBMCs from lupus nephritis patients was significantly higher than that from SLE patients without renal damage and healthy controls. PHA/PMA simulation could upregulate the productions of TWEAK and p-p38MAPK in PBMCs from patients with SLE. Anti-TWEAK mAb treatment downregulated both TWEAK and p-p38 MAPK expression in PBMCs, as well as IL-10 and MCP-1 in the supernatant; SB203580 had the same effect on cytokine production in PBMC, but had no effect on the expression of TWEAK. Our results suggested that TWEAK-p38 MAPK-IL-10, MCP-1 signaling pathway in PBMC played an important pathogenic role in lupus nephritis.
Collapse
Affiliation(s)
- Liu Zhi-Chun
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Suzhou University, Suzhou, China
| | | | | | | | | | | |
Collapse
|
41
|
The complexity of the BAFF TNF-family members: Implications for autoimmunity. J Autoimmun 2012; 39:189-98. [DOI: 10.1016/j.jaut.2012.05.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 05/20/2012] [Indexed: 11/30/2022]
|
42
|
Liu ZC, Zhou QL. Tumor necrosis factor-like weak inducer of apoptosis and its potential roles in lupus nephritis. Inflamm Res 2012; 61:277-84. [PMID: 22297307 DOI: 10.1007/s00011-011-0420-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 10/26/2011] [Accepted: 12/14/2011] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a recently identified proinflammatory cytokine of the TNF superfamily that functions through binding to Fn14 receptor in target cells. TWEAK has multiple biological activities. Studies show that TWEAK plays an important role in immune inflammatory diseases. Recent work has revealed that TWEAK may play an important role in the pathogenesis of kidney damage, including in systemic lupus erythematosus (SLE), where its concentration in urine was correlated with the level of activity of lupus nephritis (LN). OBJECTIVE The major focus of this review is to discuss the recent studies on TWEAK and its possible role in the pathogenesis of LN, and the therapeutic potential of modulating this pathway in LN. RESULTS AND CONCLUSION TWEAK plays a key role in the pathogenesis of LN through activation of multiple down-signaling pathway, inducing proinflammatory cytokines and chemokines, affecting cell proliferation/apoptosis and inducing renal IgG deposition. TWEAK blockade may be a novel therapeutic approach to reducing renal damage in SLE.
Collapse
Affiliation(s)
- Zhi-Chun Liu
- Department of Rheumatology, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, People’s Republic of China
| | | |
Collapse
|
43
|
An improved understanding of TNFL/TNFR interactions using structure-based classifications. Trends Biochem Sci 2012; 37:353-63. [PMID: 22789664 DOI: 10.1016/j.tibs.2012.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 05/22/2012] [Accepted: 06/05/2012] [Indexed: 12/20/2022]
Abstract
Tumor Necrosis Factor Ligand (TNFL)-Tumor Necrosis Factor Receptor (TNFR) interactions control key cellular processes; however, the molecular basis of the specificity of these interactions remains poorly understood. Using the T-RMSD (tree based on root mean square deviation), a newly developed structure-based sequence clustering method, we have re-analyzed the available structural data to re-interpret the interactions between TNFLs and TNFRs. This improves the classification of both TNFLs and TNFRs, such that the new groups defined here are in much stronger agreement with structural and functional features than existing schemes. Our clustering approach also identifies traces of a convergent evolutionary process for TNFLs and TNFRs, leading us to propose the co-evolution of TNFLs and the third cysteine rich domain (CRD) of large TNFRs.
Collapse
|
44
|
Frenkel-Morgenstern M, Lacroix V, Ezkurdia I, Levin Y, Gabashvili A, Prilusky J, Del Pozo A, Tress M, Johnson R, Guigo R, Valencia A. Chimeras taking shape: potential functions of proteins encoded by chimeric RNA transcripts. Genome Res 2012; 22:1231-42. [PMID: 22588898 PMCID: PMC3396365 DOI: 10.1101/gr.130062.111] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chimeric RNAs comprise exons from two or more different genes and have the potential to encode novel proteins that alter cellular phenotypes. To date, numerous putative chimeric transcripts have been identified among the ESTs isolated from several organisms and using high throughput RNA sequencing. The few corresponding protein products that have been characterized mostly result from chromosomal translocations and are associated with cancer. Here, we systematically establish that some of the putative chimeric transcripts are genuinely expressed in human cells. Using high throughput RNA sequencing, mass spectrometry experimental data, and functional annotation, we studied 7424 putative human chimeric RNAs. We confirmed the expression of 175 chimeric RNAs in 16 human tissues, with an abundance varying from 0.06 to 17 RPKM (Reads Per Kilobase per Million mapped reads). We show that these chimeric RNAs are significantly more tissue-specific than non-chimeric transcripts. Moreover, we present evidence that chimeras tend to incorporate highly expressed genes. Despite the low expression level of most chimeric RNAs, we show that 12 novel chimeras are translated into proteins detectable in multiple shotgun mass spectrometry experiments. Furthermore, we confirm the expression of three novel chimeric proteins using targeted mass spectrometry. Finally, based on our functional annotation of exon organization and preserved domains, we discuss the potential features of chimeric proteins with illustrative examples and suggest that chimeras significantly exploit signal peptides and transmembrane domains, which can alter the cellular localization of cognate proteins. Taken together, these findings establish that some chimeric RNAs are translated into potentially functional proteins in humans.
Collapse
|
45
|
Targeting of colorectal cancer growth, metastasis, and anti-apoptosis in BALB/c nude mice via APRIL siRNA. Mol Cell Biochem 2011; 363:1-10. [PMID: 22170570 DOI: 10.1007/s11010-011-1151-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 11/03/2011] [Indexed: 12/30/2022]
Abstract
A proliferation-inducing ligand (APRIL) is overexpressed in most tumor cells and tissues, especially in tumors of the alimentary system, such as colorectal cancer (CRC), gastric cancer, and liver cancer. RNA interference (RNAi) has been proved to be a powerful tool for gene knockdown and holds great promise for the treatment of cancer. In this study, the efficacy of RNAi targeting APRIL was analyzed via relevant experiments on human CRC xenografted in BALB/c nude mice. Both the mRNA and protein levels of APRIL were examined after intratumoral injection of APRIL small interfering RNA (siRNA). Meanwhile, pathological tools were utilized to observe the alterations on the aspects of proliferation, metastasis, apoptosis and cellular necrosis by means of detecting proliferating cell nuclear antigen, Ki-67, MMP-2, MMP-9, TIMP-3, TIMP-4, Bcl-2, Bax and Bcl-xL of CRC. In addition, terminal deoxyribonucleotidyl transferase-mediated dUTP-digoxigenin nick end-labeling (TUNEL) and hematoxylin and eosin staining were also conducted to examine cell apoptosis and necrosis. It was found that grafted human colorectal tumor growth and metastasis were obviously inhibited while tumor cell apoptosis and necrosis were induced after in vivo APRIL siRNA injection into nude mice. The data indicated that silencing of the APRIL gene using RNAi may serve as a novel therapeutic strategy for treatment of CRC.
Collapse
|
46
|
Kekeeva TV, Zavalishina LE, Frank GA, Zaletaev DV. Fusion genes and transcripts in neoplasia. Mol Biol 2011. [DOI: 10.1134/s0026893311050086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
47
|
BAFF and selection of autoreactive B cells. Trends Immunol 2011; 32:388-94. [PMID: 21752714 DOI: 10.1016/j.it.2011.06.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 05/24/2011] [Accepted: 06/06/2011] [Indexed: 11/21/2022]
Abstract
B cell activating factor (BAFF) is a crucial survival factor for transitional and mature B cells, and is a promising therapeutic target for systemic lupus erythematosus (SLE). A BAFF inhibitor, belimumab, is the first new drug in 50 years to be approved for the treatment of SLE. However, the mechanism of action of this drug is not entirely clear. In this review we will focus on the role of the BAFF-APRIL signaling pathway in the selection of autoreactive B cells, and discuss whether altered selection is the mechanism for the therapeutic efficacy of BAFF inhibition in SLE.
Collapse
|
48
|
Major chimpanzee-specific structural changes in sperm development-associated genes. Funct Integr Genomics 2011; 11:507-17. [PMID: 21484476 DOI: 10.1007/s10142-011-0220-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 03/14/2011] [Accepted: 03/15/2011] [Indexed: 10/18/2022]
Abstract
A comprehensive analysis of transcriptional structures of chimpanzee sperm development-associated genes is of significant interest for deeply understanding sperm development and male reproductive process. In this study, we sequenced 7,680 clones from a chimpanzee testis full-length cDNA library and obtained 1,933 nonredundant high-quality full-length cDNA sequences. Comparative analysis between human and chimpanzee showed that 78 sperm development-associated genes, most of which were yet uncharacterized, had undergone severe structural changes (mutations at the start/stop codons, INDELs, alternative splicing variations and fusion forms) on genomic and transcript levels throughout chimpanzee evolution. Specifically, among the 78 sperm development-associated genes, 39 including ODF2, UBC, and CD59 showed markedly chimpanzee-specific structural changes. Through dN/dS analysis, we found that 56 transcripts (including seven sperm development-associated genes) had values of greater than one when comparing human and chimpanzee DNA sequences, whereas the values were less than one when comparing humans and orangutans. Gene ontology annotation and expression profiling showed that the chimpanzee testis transcriptome was enriched with genes that are associated with chimpanzee male germ cell development. Taken together, our study provides the first comprehensive molecular evidence that many chimpanzee sperm development-associated genes had experienced severe structural changes over the course of evolution on genomic and transcript levels.
Collapse
|
49
|
Nacu S, Yuan W, Kan Z, Bhatt D, Rivers CS, Stinson J, Peters BA, Modrusan Z, Jung K, Seshagiri S, Wu TD. Deep RNA sequencing analysis of readthrough gene fusions in human prostate adenocarcinoma and reference samples. BMC Med Genomics 2011; 4:11. [PMID: 21261984 PMCID: PMC3041646 DOI: 10.1186/1755-8794-4-11] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 01/24/2011] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Readthrough fusions across adjacent genes in the genome, or transcription-induced chimeras (TICs), have been estimated using expressed sequence tag (EST) libraries to involve 4-6% of all genes. Deep transcriptional sequencing (RNA-Seq) now makes it possible to study the occurrence and expression levels of TICs in individual samples across the genome. METHODS We performed single-end RNA-Seq on three human prostate adenocarcinoma samples and their corresponding normal tissues, as well as brain and universal reference samples. We developed two bioinformatics methods to specifically identify TIC events: a targeted alignment method using artificial exon-exon junctions within 200,000 bp from adjacent genes, and genomic alignment allowing splicing within individual reads. We performed further experimental verification and characterization of selected TIC and fusion events using quantitative RT-PCR and comparative genomic hybridization microarrays. RESULTS Targeted alignment against artificial exon-exon junctions yielded 339 distinct TIC events, including 32 gene pairs with multiple isoforms. The false discovery rate was estimated to be 1.5%. Spliced alignment to the genome was less sensitive, finding only 18% of those found by targeted alignment in 33-nt reads and 59% of those in 50-nt reads. However, spliced alignment revealed 30 cases of TICs with intervening exons, in addition to distant inversions, scrambled genes, and translocations. Our findings increase the catalog of observed TIC gene pairs by 66%.We verified 6 of 6 predicted TICs in all prostate samples, and 2 of 5 predicted novel distant gene fusions, both private events among 54 prostate tumor samples tested. Expression of TICs correlates with that of the upstream gene, which can explain the prostate-specific pattern of some TIC events and the restriction of the SLC45A3-ELK4 e4-e2 TIC to ERG-negative prostate samples, as confirmed in 20 matched prostate tumor and normal samples and 9 lung cancer cell lines. CONCLUSIONS Deep transcriptional sequencing and analysis with targeted and spliced alignment methods can effectively identify TIC events across the genome in individual tissues. Prostate and reference samples exhibit a wide range of TIC events, involving more genes than estimated previously using ESTs. Tissue specificity of TIC events is correlated with expression patterns of the upstream gene. Some TIC events, such as MSMB-NCOA4, may play functional roles in cancer.
Collapse
Affiliation(s)
- Serban Nacu
- Department of Bioinformatics and Molecular Biology, Genentech, Inc, South San Francisco, California 94080, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kimberley F, Guadagnoli M, van Eenennaam H, Medema JP. A Proliferation-Inducing Ligand (APRIL): The Development of Antagonistic Agents as Potential Therapeutics and Deciphering the Role of Heparan Sulphate Proteoglycans (HSPGs) in APRIL Signalling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 691:501-6. [DOI: 10.1007/978-1-4419-6612-4_52] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|