1
|
Michaud SA, Pětrošová H, Sinclair NJ, Kinnear AL, Jackson AM, McGuire JC, Hardie DB, Bhowmick P, Ganguly M, Flenniken AM, Nutter LMJ, McKerlie C, Smith D, Mohammed Y, Schibli D, Sickmann A, Borchers CH. Multiple reaction monitoring assays for large-scale quantitation of proteins from 20 mouse organs and tissues. Commun Biol 2024; 7:6. [PMID: 38168632 PMCID: PMC10762018 DOI: 10.1038/s42003-023-05687-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Mouse is the mammalian model of choice to study human health and disease due to its size, ease of breeding and the natural occurrence of conditions mimicking human pathology. Here we design and validate multiple reaction monitoring mass spectrometry (MRM-MS) assays for quantitation of 2118 unique proteins in 20 murine tissues and organs. We provide open access to technical aspects of these assays to enable their implementation in other laboratories, and demonstrate their suitability for proteomic profiling in mice by measuring normal protein abundances in tissues from three mouse strains: C57BL/6NCrl, NOD/SCID, and BALB/cAnNCrl. Sex- and strain-specific differences in protein abundances are identified and described, and the measured values are freely accessible via our MouseQuaPro database: http://mousequapro.proteincentre.com . Together, this large library of quantitative MRM-MS assays established in mice and the measured baseline protein abundances represent an important resource for research involving mouse models.
Collapse
Affiliation(s)
- Sarah A Michaud
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada.
| | - Helena Pětrošová
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada
| | - Nicholas J Sinclair
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada
| | - Andrea L Kinnear
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada
| | - Angela M Jackson
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada
| | - Jamie C McGuire
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada
| | - Darryl B Hardie
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada
| | - Pallab Bhowmick
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada
| | - Milan Ganguly
- The Center for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Ann M Flenniken
- The Center for Phenogenomics, Toronto, ON, Canada
- Sinai Health Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Lauryl M J Nutter
- The Center for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Derek Smith
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada
| | - Yassene Mohammed
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, 44139, Germany
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - David Schibli
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, 44139, Germany
| | - Christoph H Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, QC, Canada.
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada.
- Department of Pathology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Laghouaouta H, Fraile L, Suárez-Mesa R, Ros-Freixedes R, Estany J, Pena RN. A genome-wide screen for resilient responses in growing pigs. Genet Sel Evol 2022; 54:50. [PMID: 35787790 PMCID: PMC9251948 DOI: 10.1186/s12711-022-00739-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Background There is a growing interest to decipher the genetic background of resilience and its possible improvement through selective breeding. The objective of the present study was to provide new insights into the genetic make-up of resilience in growing pigs by identifying genomic regions and candidate genes associated with resilience indicators. Commercial Duroc pigs were challenged with an attenuated Aujeszky vaccine at 12 weeks of age. Two resilience indicators were used: deviation from the expected body weight at 16 weeks of age given the growth curve of non-vaccinated pigs (∆BW) and the increase in acute-phase protein haptoglobin at four days post-vaccination (∆HP). Genome-wide association analyses were carried out on 445 pigs, using genotypes at 41,165 single nucleotide polymorphisms (SNPs) and single-marker and Bayesian multiple-marker regression approaches. Results Genomic regions on pig chromosomes 2, 8, 9, 11 (∆BW) and 8, 9, 13 (∆HP) were found to be associated with the resilience indicators and explained high proportions of their genetic variance. The genomic regions that were associated explained 27 and 5% of the genetic variance of ∆BW and ∆HP, respectively. These genomic regions harbour promising candidate genes that are involved in pathways related to immune response, response to stress, or signal transduction (CD6, PTGDR2, IKZF1, RNASEL and MYD88), and growth (GRB10 and LCORL). Conclusions Our study identified novel genomic regions that are associated with two resilience indicators (∆BW and ∆HP) in pigs. These associated genomic regions harbour potential candidate genes involved in immune response and growth pathways, which emphasise the strong relationship between resilience and immune response. Supplementary Information The online version contains supplementary material available at 10.1186/s12711-022-00739-1.
Collapse
Affiliation(s)
- Houda Laghouaouta
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Lorenzo Fraile
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Rafael Suárez-Mesa
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Roger Ros-Freixedes
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Joan Estany
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Ramona Natacha Pena
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain.
| |
Collapse
|
3
|
Bonaud A, Clare S, Bisio V, Sowerby JM, Yao S, Ostergaard H, Balabanian K, Smith KGC, Espéli M. Leupaxin Expression Is Dispensable for B Cell Immune Responses. Front Immunol 2020; 11:466. [PMID: 32269569 PMCID: PMC7109257 DOI: 10.3389/fimmu.2020.00466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 02/28/2020] [Indexed: 11/22/2022] Open
Abstract
The generation of a potent humoral immune response by B cells relies on the integration of signals induced by the B cell receptor, toll-like receptors and both negative and positive co-receptors. Several reports also suggest that integrin signaling plays an important role in this process. How integrin signaling is regulated in B cells is however still partially understood. Integrin activity and function are controlled by several mechanisms including regulation by molecular adaptors of the paxillin family. In B cells, Leupaxin (Lpxn) is the most expressed member of the family and in vitro studies suggest that it could dampen BCR signaling. Here, we report that Lpxn expression is increased in germinal center B cells compared to naïve B cells. Moreover, Lpxn deficiency leads to decreased B cell differentiation into plasma cells in vitro. However, Lpxn seems dispensable for the generation of a potent B cell immune response in vivo. Altogether our results suggest that Lpxn is dispensable for T-dependent and T-independent B cell immune responses.
Collapse
Affiliation(s)
- Amélie Bonaud
- Inflammation Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Medicale (INSERM), Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Clamart, France
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France
| | - Simon Clare
- Wellcome Trust Genome, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Valeria Bisio
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France
| | - John M. Sowerby
- The Department of Medicine, Cambridge Biomedical, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Jeffrey Cheah Biomedical Centre Cambridge Biomedical, Cambridge Institute of Therapeutic Immunology & Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - Shugang Yao
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Hanne Ostergaard
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Karl Balabanian
- Inflammation Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Medicale (INSERM), Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Clamart, France
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France
| | - Kenneth G. C. Smith
- The Department of Medicine, Cambridge Biomedical, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Jeffrey Cheah Biomedical Centre Cambridge Biomedical, Cambridge Institute of Therapeutic Immunology & Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - Marion Espéli
- Inflammation Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Medicale (INSERM), Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Clamart, France
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France
| |
Collapse
|
4
|
Drobin K, Assadi G, Hong MG, Andersson E, Fredolini C, Forsström B, Reznichenko A, Akhter T, Ek WE, Bonfiglio F, Hansen MB, Sandberg K, Greco D, Repsilber D, Schwenk JM, D’Amato M, Halfvarson J. Targeted Analysis of Serum Proteins Encoded at Known Inflammatory Bowel Disease Risk Loci. Inflamm Bowel Dis 2019; 25:306-316. [PMID: 30358838 PMCID: PMC6327232 DOI: 10.1093/ibd/izy326] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Few studies have investigated the blood proteome of inflammatory bowel disease (IBD). We characterized the serum abundance of proteins encoded at 163 known IBD risk loci and tested these proteins for their biomarker discovery potential. METHODS Based on the Human Protein Atlas (HPA) antibody availability, 218 proteins from genes mapping at 163 IBD risk loci were selected. Targeted serum protein profiles from 49 Crohn's disease (CD) patients, 51 ulcerative colitis (UC) patients, and 50 sex- and age-matched healthy individuals were obtained using multiplexed antibody suspension bead array assays. Differences in relative serum abundance levels between disease groups and controls were examined. Replication was attempted for CD-UC comparisons (including disease subtypes) by including 64 additional patients (33 CD and 31 UC). Antibodies targeting a potentially novel risk protein were validated by paired antibodies, Western blot, immuno-capture mass spectrometry, and epitope mapping. RESULTS By univariate analysis, 13 proteins mostly related to neutrophil, T-cell, and B-cell activation and function were differentially expressed in IBD patients vs healthy controls, 3 in CD patients vs healthy controls and 2 in UC patients vs healthy controls (q < 0.01). Multivariate analyses further differentiated disease groups from healthy controls and CD subtypes from UC (P < 0.05). Extended characterization of an antibody targeting a novel, discriminative serum marker, the laccase (multicopper oxidoreductase) domain containing 1 (LACC1) protein, provided evidence for antibody on-target specificity. CONCLUSIONS Using affinity proteomics, we identified a set of IBD-associated serum proteins encoded at IBD risk loci. These candidate proteins hold the potential to be exploited as diagnostic biomarkers of IBD.
Collapse
Affiliation(s)
- Kimi Drobin
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH, Royal Institute of Technology, Stockholm, Sweden
| | - Ghazaleh Assadi
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Mun-Gwan Hong
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH, Royal Institute of Technology, Stockholm, Sweden
| | - Eni Andersson
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH, Royal Institute of Technology, Stockholm, Sweden
| | - Claudia Fredolini
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH, Royal Institute of Technology, Stockholm, Sweden
| | - Björn Forsström
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH, Royal Institute of Technology, Stockholm, Sweden
| | - Anna Reznichenko
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Tahmina Akhter
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Weronica E Ek
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ferdinando Bonfiglio
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Department of Gastrointestinal and Liver Diseases, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Mark Berner Hansen
- AstraZeneca R&D Mölndal, Innovative and Global Medicines, Mölndal, Sweden
- Digestive Disease Center, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Sandberg
- Science for Life Laboratory, Drug Discovery & Development Platform & Organic Pharmaceutical Chemistry, Department of Medicinal Chemistry, Uppsala Biomedical Center, Uppsala University, Uppsala, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Dario Greco
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Dirk Repsilber
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Jochen M Schwenk
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH, Royal Institute of Technology, Stockholm, Sweden
| | - Mauro D’Amato
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- BioDonostia Health Research Institute, San Sebastian and IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Address correspondence to: Jonas Halfvarson, PhD, Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, SE 70182, Örebro, Sweden ()
| |
Collapse
|
5
|
Zhu GH, Dai HP, Shen Q, Zhang Q. Downregulation of LPXN expression by siRNA decreases the malignant proliferation and transmembrane invasion of SHI-1 cells. Oncol Lett 2018; 17:135-140. [PMID: 30655748 PMCID: PMC6313184 DOI: 10.3892/ol.2018.9605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 08/30/2018] [Indexed: 01/03/2023] Open
Abstract
The aim of the present study was to investigate the effects of decreasing leupaxin (LPXN) expression on the proliferation and invasion of human acute monocytic leukemia SHI-1 cells. The transfection efficiency of fluorescein amidite (FAM)-small interfering RNA (siRNA) was determined using flow cytometry, and the protein expression levels of LPXN, phosphorylated (p)-c-Jun N-terminal kinase (JNK), p-p38 mitogen-activated protein kinase (p38 MAPK) and p-extracellular-signal-regulated kinase (ERK) were detected by western blot analysis. Proliferation was determined using the cell counting kit-8 reagent and cellular transmembrane invasion ability was determined using a Transwell chamber system. The gelatinase levels of matrix metalloproteinase (MMP)-2 and MMP-9 in the cell culture supernatant were also analyzed by gelatin zymography. In SHI-1 cells, the optimal transfection conditions of siRNA were a cell density of 4×105 cells/ml and a ratio of siRNA/Lipofectamine® 2000 of 200 pmol/1 µl. The highest transfection efficiency of FAM-siRNA was 74.5%. In the present study, L2-siRNA was selected to effectively decrease the expression of LPXN. Following downregulation of LPXN expression by L2-siRNA, proliferation inhibition rates increased to 27.043±2.051 and cell transmembrane invasion rates decreased to 25.270±2.145 (P<0.05). The results of the western blot analysis and the gelatin zymography indicated that downregulation of LPXN expression increased the expression of p-p38 MAPK and p-JNK, and attenuated the secretion levels of MMP-2 and MMP-9. However, downregulation of LPXN expression had no effect on p-ERK expression in SHI-1 cells. The results of the present study indicated that downregulation of LPXN expression decreased the malignant proliferation and transmembrane invasion of SHI-1 cells by activating JNK and p38 MAPK, and inhibiting MMP-2 and MMP-9 secretion.
Collapse
Affiliation(s)
- Guo-Hua Zhu
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Hai-Ping Dai
- Leukemia Research Unit, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qun Shen
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China.,Department of Hematology, First Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210009, P.R. China
| | - Qi Zhang
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
6
|
Mahmoud TI, Wang J, Karnell JL, Wang Q, Wang S, Naiman B, Gross P, Brohawn PZ, Morehouse C, Aoyama J, Wasserfall C, Carter L, Atkinson MA, Serreze DV, Braley-Mullen H, Mustelin T, Kolbeck R, Herbst R, Ettinger R. Autoimmune manifestations in aged mice arise from early-life immune dysregulation. Sci Transl Med 2017; 8:361ra137. [PMID: 27798262 DOI: 10.1126/scitranslmed.aag0367] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/19/2016] [Indexed: 12/13/2022]
Abstract
Autoantibodies can be present years to decades before the onset of disease manifestations in autoimmunity. This finding suggests that the initial autoimmune trigger involves a peripheral lymphoid component, which ultimately drives disease pathology in local tissues later in life. We show that Sjögren's syndrome manifestations that develop in aged NOD.H-2h4 mice were driven by and dependent on peripheral dysregulation that arose in early life. Specifically, elimination of spontaneous germinal centers in spleens of young NOD.H-2h4 mice by transient blockade of CD40 ligand (CD40L) or splenectomy abolished Sjögren's pathology of aged mice. Strikingly, a single injection of anti-CD40L at 4 weeks of age prevented tertiary follicle neogenesis and greatly blunted the formation of key autoantibodies implicated in glandular pathology, including anti-muscarinic receptor antibodies. Microarray profiling of the salivary gland characterized the expression pattern of genes that increased with disease progression and showed that early anti-CD40L greatly repressed B cell function while having a broader effect on multiple biological pathways, including interleukin-12 and interferon signaling. A single prophylactic treatment with anti-CD40L also inhibited the development of autoimmune thyroiditis and diabetes in NOD.H-2h4 and nonobese diabetic mice, respectively, supporting a key role for CD40L in the pathophysiology of several autoimmune models. These results strongly suggest that early peripheral immune dysregulation gives rise to autoimmune manifestations later in life, and for diseases predated by autoantibodies, early prophylactic intervention with biologics may prove efficacious.
Collapse
Affiliation(s)
- Tamer I Mahmoud
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Jingya Wang
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Jodi L Karnell
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Qiming Wang
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Shu Wang
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Brian Naiman
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Phillip Gross
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Philip Z Brohawn
- Translational Sciences-Pharmacogenomics, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Chris Morehouse
- Translational Sciences-Pharmacogenomics, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Jordan Aoyama
- Translational Sciences-Pharmacogenomics, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Clive Wasserfall
- Departments of Pathology and Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Laura Carter
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Mark A Atkinson
- Departments of Pathology and Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | | | | | - Tomas Mustelin
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Roland Kolbeck
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Ronald Herbst
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Rachel Ettinger
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune LLC, Gaithersburg, MD 20878, USA.
| |
Collapse
|
7
|
Essential role for the transcription factor Bhlhe41 in regulating the development, self-renewal and BCR repertoire of B-1a cells. Nat Immunol 2017; 18:442-455. [PMID: 28250425 PMCID: PMC5363839 DOI: 10.1038/ni.3694] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 01/26/2017] [Indexed: 01/02/2023]
Abstract
Innate-like B-1a cells provide a first line of defense against pathogens, yet little is known about their transcriptional control. Here we identified an essential role of the transcription factor Bhlhe41, with a lesser contribution of Bhlhe40, in controlling late stages of B-1a cell differentiation. Bhlhe41–/–Bhlhe40–/– B-1a cells were severely reduced as compared to their wild-type counterparts. Mutant B-1a cells exhibited an abnormal cell-surface phenotype and altered B-cell receptor (BCR) repertoire exemplified by loss of the phosphatidylcholine-specific VH12/Vκ4 BCR. Expression of a pre-rearranged VH12/Vκ4 BCR failed to rescue the mutant phenotype and revealed enhanced proliferation accompanied with increased cell death. Bhlhe41 directly repressed the expression of cell cycle regulators and inhibitors of BCR signaling, while enabling pro-survival cytokine signaling. Thus, Bhlhe41 controls the development, BCR repertoire and self-renewal of B-1a cells.
Collapse
|
8
|
Elucidation of tonic and activated B-cell receptor signaling in Burkitt's lymphoma provides insights into regulation of cell survival. Proc Natl Acad Sci U S A 2016; 113:5688-93. [PMID: 27155012 DOI: 10.1073/pnas.1601053113] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Burkitt's lymphoma (BL) is a highly proliferative B-cell neoplasm and is treated with intensive chemotherapy that, because of its toxicity, is often not suitable for the elderly or for patients with endemic BL in developing countries. BL cell survival relies on signals transduced by B-cell antigen receptors (BCRs). However, tonic as well as activated BCR signaling networks and their relevance for targeted therapies in BL remain elusive. We have systematically characterized and compared tonic and activated BCR signaling in BL by quantitative phosphoproteomics to identify novel BCR effectors and potential drug targets. We identified and quantified ∼16,000 phospho-sites in BL cells. Among these sites, 909 were related to tonic BCR signaling, whereas 984 phospho-sites were regulated upon BCR engagement. The majority of the identified BCR signaling effectors have not been described in the context of B cells or lymphomas yet. Most of these newly identified BCR effectors are predicted to be involved in the regulation of kinases, transcription, and cytoskeleton dynamics. Although tonic and activated BCR signaling shared a considerable number of effector proteins, we identified distinct phosphorylation events in tonic BCR signaling. We investigated the functional relevance of some newly identified BCR effectors and show that ACTN4 and ARFGEF2, which have been described as regulators of membrane-trafficking and cytoskeleton-related processes, respectively, are crucial for BL cell survival. Thus, this study provides a comprehensive dataset for tonic and activated BCR signaling and identifies effector proteins that may be relevant for BL cell survival and thus may help to develop new BL treatments.
Collapse
|
9
|
Zhou G, Stevenson MM, Geary TG, Xia J. Comprehensive Transcriptome Meta-analysis to Characterize Host Immune Responses in Helminth Infections. PLoS Negl Trop Dis 2016; 10:e0004624. [PMID: 27058578 PMCID: PMC4826001 DOI: 10.1371/journal.pntd.0004624] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/21/2016] [Indexed: 12/19/2022] Open
Abstract
Helminth infections affect more than a third of the world’s population. Despite very broad phylogenetic differences among helminth parasite species, a systemic Th2 host immune response is typically associated with long-term helminth infections, also known as the “helminth effect”. Many investigations have been carried out to study host gene expression profiles during helminth infections. The objective of this study is to determine if there is a common transcriptomic signature characteristic of the helminth effect across multiple helminth species and tissue types. To this end, we performed a comprehensive meta-analysis of publicly available gene expression datasets. After data processing and adjusting for study-specific effects, we identified ~700 differentially expressed genes that are changed consistently during helminth infections. Functional enrichment analyses indicate that upregulated genes are predominantly involved in various immune functions, including immunomodulation, immune signaling, inflammation, pathogen recognition and antigen presentation. Down-regulated genes are mainly involved in metabolic process, with only a few of them are involved in immune regulation. This common immune gene signature confirms previous observations and indicates that the helminth effect is robust across different parasite species as well as host tissue types. To the best of our knowledge, this study is the first comprehensive meta-analysis of host transcriptome profiles during helminth infections. Many studies have been conducted to understand the immune modulatory effects in helminth infections. To determine whether there is a common transcriptomic signature characteristic of the helminth effect, we performed a comprehensive meta-analysis of publicly available gene expression datasets. The results revealed a distinct pattern of gene expression that is consistent across multiple helminth species and host tissue types, with upregulated genes dominated by those involved in immune regulation, Th2 immunity and inflammatory responses.
Collapse
Affiliation(s)
- Guangyan Zhou
- Institute of Parasitology, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Centre for Host-Parasite Interactions, McGill University, Sainte Anne de Bellevue, Quebec, Canada
| | - Mary M. Stevenson
- Centre for Host-Parasite Interactions, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Departments of Medicine and Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Timothy G. Geary
- Institute of Parasitology, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Centre for Host-Parasite Interactions, McGill University, Sainte Anne de Bellevue, Quebec, Canada
| | - Jianguo Xia
- Institute of Parasitology, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Centre for Host-Parasite Interactions, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Department of Animal Science, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- * E-mail:
| |
Collapse
|
10
|
Abe A, Yamamoto Y, Iba S, Kanie T, Okamoto A, Tokuda M, Inaguma Y, Yanada M, Morishima S, Mizuta S, Akatsuka Y, Okamoto M, Kameyama T, Mayeda A, Emi N. ETV6-LPXN fusion transcript generated by t(11;12)(q12.1;p13) in a patient with relapsing acute myeloid leukemia with NUP98-HOXA9. Genes Chromosomes Cancer 2016; 55:242-50. [PMID: 26542893 DOI: 10.1002/gcc.22327] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/14/2015] [Accepted: 10/15/2015] [Indexed: 11/09/2022] Open
Abstract
ETV6, which encodes an ETS family transcription factor, is frequently rearranged in human leukemias. We show here that a patient with acute myeloid leukemia with t(7;11)(p15;p15) gained, at the time of relapse, t(11;12)(q12.1;p13) with a split ETV6 FISH signal. Using 3'-RACE PCR analysis, we found that ETV6 was fused to LPXN at 11q12.1, which encodes leupaxin. ETV6-LPXN, an in-frame fusion between exon 4 of ETV6 and exon 2 of LPXN, did not transform the interleukin-3-dependent 32D myeloid cell line to cytokine independence; however, an enhanced proliferative response was observed when these cells were treated with G-CSF without inhibition of granulocytic differentiation. The 32D and human leukemia cell lines each transduced with ETV6-LPXN showed enhanced migration towards the chemokine CXCL12. We show here for the first time that LPXN is a fusion partner of ETV6 and present evidence indicating that ETV6-LPXN plays a crucial role in leukemia progression through enhancing the response to G-CSF and CXCL12.
Collapse
Affiliation(s)
- Akihiro Abe
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Yukiya Yamamoto
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Sachiko Iba
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Tadaharu Kanie
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Akinao Okamoto
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Masutaka Tokuda
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoko Inaguma
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Masamitsu Yanada
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Satoko Morishima
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Shuichi Mizuta
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoshiki Akatsuka
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Masataka Okamoto
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| | - Toshiki Kameyama
- Division of Gene Expression Mechanism, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Akila Mayeda
- Division of Gene Expression Mechanism, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Nobuhiko Emi
- Department of Hematology, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
11
|
Xu H, Chaudhri VK, Wu Z, Biliouris K, Dienger-Stambaugh K, Rochman Y, Singh H. Regulation of bifurcating B cell trajectories by mutual antagonism between transcription factors IRF4 and IRF8. Nat Immunol 2015; 16:1274-81. [DOI: 10.1038/ni.3287] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/31/2015] [Indexed: 12/17/2022]
|
12
|
Zhang M, March ME, Lane WS, Long EO. A signaling network stimulated by β2 integrin promotes the polarization of lytic granules in cytotoxic cells. Sci Signal 2014; 7:ra96. [PMID: 25292215 DOI: 10.1126/scisignal.2005629] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cytotoxic lymphocytes kill target cells through the polarized release of the contents of intracellular perforin-containing granules. In natural killer (NK) cells, the binding of β2 integrin to members of the intercellular adhesion molecule family is sufficient to promote not only the adhesion of NK cells to target cells but also the polarization of intracellular lytic granules toward the target. We used NK cells in an experimental system designed to enable us to study the polarization of lytic granules in the absence of their release through degranulation, as well as β2 integrin signaling independently of inside-out signals from other receptors. Through a proteomics approach, we identified a signaling network centered on an integrin-linked kinase (ILK)-Pyk2-paxillin core that was required for granule and microtubule-organizing center (MTOC) polarization. The conserved Cdc42-Par6 signaling pathway, which controls cell polarity, was also activated by ILK and was required for granule polarization toward the target cell. A subset of the signaling components required for polarization contributed also to the convergence of granules on the MTOC. These results delineate two connected signaling networks that are stimulated upon β2 integrin engagement and control the polarization of the MTOC and associated lytic granules toward the site of contact with target cells to mediate cellular cytotoxicity.
Collapse
Affiliation(s)
- Minggang Zhang
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Michael E March
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - William S Lane
- Mass Spectrometry and Proteomics Resource Laboratory, FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Eric O Long
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
13
|
Sakata A, Ohmori T, Nishimura S, Suzuki H, Madoiwa S, Mimuro J, Kario K, Sakata Y. Paxillin is an intrinsic negative regulator of platelet activation in mice. Thromb J 2014; 12:1. [PMID: 24383745 PMCID: PMC3904695 DOI: 10.1186/1477-9560-12-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 12/09/2013] [Indexed: 01/04/2023] Open
Abstract
Background Paxillin is a LIM domain protein localized at integrin-mediated focal adhesions. Although paxillin is thought to modulate the functions of integrins, little is known about the contribution of paxillin to signaling pathways in platelets. Here, we studied the role of paxillin in platelet activation in vitro and in vivo. Methods and results We generated paxillin knockdown (Pxn-KD) platelets in mice by transplanting bone marrow cells transduced with a lentiviral vector carrying a short hairpin RNA sequence, and confirmed that paxillin expression was significantly reduced in platelets derived from the transduced cells. Pxn-KD platelets showed a slight increased in size and augmented integrin αIIbβ3 activation following stimulation of multiple receptors including glycoprotein VI and G protein-coupled receptors. Thromboxane A2 biosynthesis and the release of α-granules and dense granules in response to agonist stimulation were also enhanced in Pxn-KD platelets. However, Pxn-KD did not increase tyrosine phosphorylation or intracellular calcium mobilization. Intravital imaging confirmed that Pxn-KD enhanced thrombus formation in vivo. Conclusions Our findings suggest that paxillin negatively regulates several common platelet signaling pathways, resulting in the activation of integrin αIIbβ3 and release reactions.
Collapse
Affiliation(s)
| | - Tsukasa Ohmori
- Research Division of Cell and Molecular Medicine, Center for Molecular Medicine, Jichi Medical University School of Medicine, 3111-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan.
| | | | | | | | | | | | | |
Collapse
|
14
|
Ananthakrishnan AN, Oxford EC, Nguyen DD, Sauk J, Yajnik V, Xavier RJ. Genetic risk factors for Clostridium difficile infection in ulcerative colitis. Aliment Pharmacol Ther 2013; 38:522-30. [PMID: 23848254 PMCID: PMC3755009 DOI: 10.1111/apt.12425] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 06/10/2013] [Accepted: 07/02/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with inflammatory bowel disease (IBD) are at higher risk for Clostridium difficile infection (CDI). Disruption of gut microbiome and interaction with the intestinal immune system are essential mechanisms for pathogenesis of both CDI and IBD. Whether genetic polymorphisms associated with susceptibility to IBD are also associated with risk of CDI is unknown. AIMS To use a well-characterised and genotyped cohort of patients with UC to (i) identify clinical risk factors for CDI; (ii) examine if any of the IBD genetic risk loci were associated with CDI; and (iii) to compare the performance of predictive models using clinical and genetic risk factors in determining risk of CDI. METHODS We used a prospective registry of patients from a tertiary referral hospital. Medical record review was performed to identify all ulcerative colitis (UC) patients within the registry with a history of CDI. All patients were genotyped on the Immunochip. We examined the association between the 163 risk loci for IBD and risk of CDI using a dominant genetic model. Model performance was examined using receiver operating characteristics curves. RESULTS The study included 319 patients of whom 29 developed CDI (9%). Female gender and pancolitis were associated with increased risk, while use of anti-TNF was protective against CDI. Six genetic polymorphisms including those at TNFRSF14 [Odds ratio (OR) 6.0, P-value 0.01] were associated with increased risk while 2 loci were inversely associated. On multivariate analysis, none of the clinical parameters retained significance after adjusting for genetics. Presence of at least one high-risk locus was associated with an increase in risk for CDI (20% vs. 1%) (P = 6 × 10⁻⁹). Compared to 11% for a clinical model, the genetic loci explained 28% of the variance in CDI risk and had a greater AUROC. CONCLUSION Host genetics may influence susceptibility to Clostridium difficile infection in patients with ulcerative colitis.
Collapse
Affiliation(s)
- Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | | | - Deanna D Nguyen
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Jenny Sauk
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Vijay Yajnik
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Ramnik J Xavier
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA,Center for Computational and Integrative Biology, MGH, Boston, MA,Broad Institute, Cambridge, MA
| |
Collapse
|
15
|
Vijai J, Kirchhoff T, Schrader KA, Brown J, Dutra-Clarke AV, Manschreck C, Hansen N, Rau-Murthy R, Sarrel K, Przybylo J, Shah S, Cheguri S, Stadler Z, Zhang L, Paltiel O, Ben-Yehuda D, Viale A, Portlock C, Straus D, Lipkin SM, Lacher M, Robson M, Klein RJ, Zelenetz A, Offit K. Susceptibility loci associated with specific and shared subtypes of lymphoid malignancies. PLoS Genet 2013; 9:e1003220. [PMID: 23349640 PMCID: PMC3547842 DOI: 10.1371/journal.pgen.1003220] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 10/18/2012] [Indexed: 12/31/2022] Open
Abstract
The genetics of lymphoma susceptibility reflect the marked heterogeneity of diseases that comprise this broad phenotype. However, multiple subtypes of lymphoma are observed in some families, suggesting shared pathways of genetic predisposition to these pathologically distinct entities. Using a two-stage GWAS, we tested 530,583 SNPs in 944 cases of lymphoma, including 282 familial cases, and 4,044 public shared controls, followed by genotyping of 50 SNPs in 1,245 cases and 2,596 controls. A novel region on 11q12.1 showed association with combined lymphoma (LYM) subtypes. SNPs in this region included rs12289961 near LPXN, (P(LYM) = 3.89×10(-8), OR = 1.29) and rs948562 (P(LYM) = 5.85×10(-7), OR = 1.29). A SNP in a novel non-HLA region on 6p23 (rs707824, P(NHL) = 5.72×10(-7)) was suggestive of an association conferring susceptibility to lymphoma. Four SNPs, all in a previously reported HLA region, 6p21.32, showed genome-wide significant associations with follicular lymphoma. The most significant association with follicular lymphoma was for rs4530903 (P(FL) = 2.69×10(-12), OR = 1.93). Three novel SNPs near the HLA locus, rs9268853, rs2647046, and rs2621416, demonstrated additional variation contributing toward genetic susceptibility to FL associated with this region. Genes implicated by GWAS were also found to be cis-eQTLs in lymphoblastoid cell lines; candidate genes in these regions have been implicated in hematopoiesis and immune function. These results, showing novel susceptibility regions and allelic heterogeneity, point to the existence of pathways of susceptibility to both shared as well as specific subtypes of lymphoid malignancy.
Collapse
Affiliation(s)
- Joseph Vijai
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Cancer Biology and Genetics Program, Sloan-Kettering Institute, New York, New York, United States of America
| | - Tomas Kirchhoff
- New York University Cancer Institute, New York University School of Medicine, New York, New York, United States of America
| | - Kasmintan A. Schrader
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Cancer Biology and Genetics Program, Sloan-Kettering Institute, New York, New York, United States of America
| | - Jennifer Brown
- Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Ana Virginia Dutra-Clarke
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Christopher Manschreck
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Nichole Hansen
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Rohini Rau-Murthy
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Kara Sarrel
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Jennifer Przybylo
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Sohela Shah
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Cancer Biology and Genetics Program, Sloan-Kettering Institute, New York, New York, United States of America
| | - Srujana Cheguri
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Zsofia Stadler
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Liying Zhang
- Diagnostic Molecular Genetics Laboratory, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ora Paltiel
- Department of Hematology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Dina Ben-Yehuda
- Department of Hematology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Agnes Viale
- Genomics Core, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Carol Portlock
- Lymphoma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - David Straus
- Lymphoma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Steven M. Lipkin
- Weill Cornell Medical Center, New York, New York, United States of America
| | - Mortimer Lacher
- Lymphoma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Mark Robson
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Robert J. Klein
- Cancer Biology and Genetics Program, Sloan-Kettering Institute, New York, New York, United States of America
| | - Andrew Zelenetz
- Lymphoma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Kenneth Offit
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Cancer Biology and Genetics Program, Sloan-Kettering Institute, New York, New York, United States of America
- Lymphoma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
16
|
De Braekeleer E, Douet-Guilbert N, Morel F, Le Bris MJ, Férec C, De Braekeleer M. RUNX1 translocations and fusion genes in malignant hemopathies. Future Oncol 2011; 7:77-91. [PMID: 21174539 DOI: 10.2217/fon.10.158] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The RUNX1 gene, located in chromosome 21q22, is crucial for the establishment of definitive hematopoiesis and the generation of hematopoietic stem cells in the embryo. It contains a 'Runt homology domain' as well as transcription activation and inhibition domains. RUNX1 can act as activator or repressor of target gene expression depending upon the large number of transcription factors, coactivators and corepressors that interact with it. Translocations involving chromosomal band 21q22 are regularly identified in leukemia patients. Most of them are associated with a rearrangement of RUNX1. Indeed, at present, 55 partner chromosomal bands have been described but the partner gene has solely been identified in 21 translocations at the molecular level. All the translocations that retain Runt homology domains but remove the transcription activation domain have a leukemogenic effect by acting as dominant negative inhibitors of wild-type RUNX1 in transcription activation.
Collapse
|
17
|
Chen PW, Kroog GS. Leupaxin is similar to paxillin in focal adhesion targeting and tyrosine phosphorylation but has distinct roles in cell adhesion and spreading. Cell Adh Migr 2011; 4:527-40. [PMID: 20543562 DOI: 10.4161/cam.4.4.12399] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Focal adhesion (FA) formation is induced by extracellular matrix-stimulated integrin clustering and activation of receptors for diffusible factors. Leupaxin (LPXN) is a member of the paxillin family of FA proteins expressed in many cancer cell lines. We found activation of gastrin-releasing peptide receptor (GRPr) by bombesin (BN) stimulated LPXN translocation from cytoplasm to FAs. Using mutagenesis, we identified LIM3 as the primary FA targeting domain for LPXN and showed BN-induced LPXN tyrosine phosphorylation on residues 22, 62 and 72. A LIM3 point mutant of LPXN failed to target to FAs and had no BN-stimulated tyrosine phosphorylation. Conversely, a non-phosphorylatable mutant (Y22/62/72F) translocated to FAs after BN addition. Stimulation of FA formation using vinblastine also induced LPXN translocation and tyrosine phosphorylation. Therefore, dynamic LPXN tyrosine phosphorylation requires translocation to FAs. LPXN and paxillin had opposite roles in adhesion to collagen I (CNI) in MDA-MB-231 breast cancer cells. LPXN siRNA stimulated whereas paxillin siRNA inhibited cell adhesion. Knockdown of both LPXN and paxillin behaved similarly to paxillin knockdown alone, suggesting LPXN’s function in adhesion might depend on paxillin. Additionally, LPXN regulated cell spreading on CNI but not on fibronectin whereas paxillin knockdown suppressed spreading on both substrates. These results demonstrate that although LPXN and paxillin’s FA targeting and tyrosine phosphorylation are similar, each protein has distinct functions.
Collapse
Affiliation(s)
- Pei-Wen Chen
- Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | |
Collapse
|
18
|
Patients with drug-free long-term graft function display increased numbers of peripheral B cells with a memory and inhibitory phenotype. Kidney Int 2010; 78:503-13. [PMID: 20531452 DOI: 10.1038/ki.2010.162] [Citation(s) in RCA: 216] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Several transplant patients maintain stable kidney graft function in the absence of immunosuppression. Here we compared the characteristics of their peripheral B cells to that of others who had stable graft function but were under pharmacologic immunosuppression, to patients with chronic rejection and to healthy volunteers. In drug-free long-term graft function (DF) there was a significant increase in both absolute cell number and frequency of total B cells; particularly activated, memory and early memory B cells. These increased B-cell numbers were associated with a significantly enriched transcriptional B-cell profile. Costimulatory/migratory molecules (B7-2/CD80, CD40, and CD62L) were upregulated in B cells; particularly in memory CD19(+)IgD(-)CD38(+/-)CD27(+) B cells in these patients. Their purified B cells, however, responded normally to a polyclonal stimulation and did not have cytokine polarization. This phenotype was associated with the following specific characteristics which include an inhibitory signal (decreased FcgammaRIIA/FcgammaRIIB ratio); a preventive signal of hyperactive B-cell response (an increase in BANK1, which negatively modulates CD40-mediated AKT activation); an increased number of B cells expressing CD1d and CD5; an increased BAFF-R/BAFF ratio that could explain why these patients have more peripheral B cells; and a specific autoantibody profile. Thus, our findings show that patients with DF have a particular blood B-cell phenotype that may contribute to the maintenance of long-term graft function.
Collapse
|
19
|
Tanaka T, Moriwaki K, Murata S, Miyasaka M. LIM domain-containing adaptor, leupaxin, localizes in focal adhesion and suppresses the integrin-induced tyrosine phosphorylation of paxillin. Cancer Sci 2010; 101:363-8. [PMID: 19917054 PMCID: PMC11158308 DOI: 10.1111/j.1349-7006.2009.01398.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Focal adhesion (FA) consists of multiple cellular proteins including paxillin and serves as a center for adhesion-mediated signaling. The assembly and disassembly of FAs is regulated by locally produced intracellular signals, and tyrosine phosphorylation of paxillin has been implicated in this process. A Lin-11 Isl-1 Mec-3 (LIM) domain-containing adaptor protein, leupaxin, a member of the paxillin family, is expressed in leukocytes as well as in certain cancer cells, and shares overall structural characteristics with paxillin. However, it remains unknown whether leupaxin and paxillin cooperate with or antagonize each other in integrin signaling. Here we show that leupaxin potently represses the tyrosine phosphorylation of paxillin. When expressed in mouse thymoma BW5147 cells bound to ICAM-1, leupaxin accumulated in FA-like patches in the cell periphery. When expressed in NIH3T3 and HEK293T cells, leupaxin localized to FAs upon cell adhesion to fibronectin and strongly suppressed the integrin-induced tyrosine phosphorylation of paxillin. In integrin-stimulated HEK293T cells, leupaxin's LIM3 domain appeared essential for selective FA localization and the suppression of paxillin tyrosine phosphorylation. Leupaxin's LD3 motif, which is critical for stable association with FAK, was dispensable for leupaxin's suppressive ability. In addition, leupaxin reduced the spreading of NIH3T3 cells on fibronectin, which required both the LD3 motif and LIM3 domain. When expressed in human leukocytic K562 cells, leupaxin significantly suppressed integrin alpha5beta1-mediated cell adhesion to fibronectin and the tyrosine phosphorylation of paxillin. These findings indicate that leupaxin functions as a paxillin counterpart that potently suppresses the tyrosine phosphorylation of paxillin during integrin signaling.
Collapse
Affiliation(s)
- Toshiyuki Tanaka
- Laboratory of Immunodynamics, Department of Microbiology and Immunology, Osaka University, Graduate School of Medicine, Osaka, Japan.
| | | | | | | |
Collapse
|
20
|
Dai HP, Xue YQ, Zhou JW, Li AP, Wu YF, Pan JL, Wang Y, Zhang J. LPXN, a member of the paxillin superfamily, is fused to RUNX1 in an acute myeloid leukemia patient with a t(11;21)(q12;q22) translocation. Genes Chromosomes Cancer 2009; 48:1027-36. [PMID: 19760607 DOI: 10.1002/gcc.20704] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
RUNX1 (previously AML1) is involved in multiple recurrent chromosomal rearrangements in hematological malignances. Recently, we identified a novel fusion between RUNX1 and LPXN from an acute myeloid leukemia (AML) patient with t(11;21)(q12;q22). This translocation generated four RUNX1/LPXN and one LPXN/RUNX1 chimeric transcripts. Two representative RUNX1/LPXN fusion proteins, RL and RLs, were both found to localize in the nucleus and could bring the CBFB protein into the nucleus like the wild-type RUNX1. Both fusion proteins inhibit the ability of RUNX1 to transactivate the CSF1R promoter, probably through competition for its target sequences. Unlike RL and RLs, the LPXN/RUNX1 fusion protein LR was found to localize in the cytoplasm. Thus, we believe it has little impact on the transcriptional activity of RUNX1. We also found that fusion proteins RL, RLs, LR, and wild-type LPXN could confer NIH3T3 cells with malignant transformation characteristics such as more rapid growth, the ability to form colonies in soft agar, and the ability to form solid tumors in the subcutaneous tissue of the BALB/c nude mice. Taken together, our data indicated that the RUNX1/LPXN and LPXN/RUNX1 fusion proteins may play important roles in leukemogenesis and that deregulation of cell adhesion pathways may be pathogenetically important in AML. Our study also suggests that LPXN may play an important role in carcinogenesis.
Collapse
Affiliation(s)
- Hai-Ping Dai
- Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Focal adhesion proteins connect IgE receptors to the cytoskeleton as revealed by micropatterned ligand arrays. Proc Natl Acad Sci U S A 2008; 105:17238-44. [PMID: 19004813 DOI: 10.1073/pnas.0802138105] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Patterned surfaces that present specific ligands in spatially defined arrays are used to examine structural linkages between clustered IgE receptors (IgE-Fc epsilonRI) and the cytoskeleton in rat basophilic leukemia (RBL) mast cells. We showed with fluorescence microscopy that cytoskeletal F-actin concentrates in the same regions as cell surface IgE-Fc epsilonRI that bind to the micrometer-size patterned ligands. However, the proteins mediating these cytoskeletal connections and their functional relevance were not known. We now show that whereas the adaptor proteins ezrin and moesin do not detectably concentrate with the array of clustered IgE-Fc epsilonRI, focal adhesion proteins vinculin, paxillin, and talin, which are known to link F-actin with integrins, accumulate in these regions on the same time scale as F-actin. Moreover, colocalization of these focal adhesion proteins with clustered IgE-Fc epsilonRI is enhanced after addition of fibronectin-RGD peptides. Significantly, the most prominent rat basophilic leukemia cell integrin (alpha5) avoids the patterned regions occupied by the ligands and associates preferentially with exposed regions of the silicon substrate. Thus, spatial separation provided by the patterned surface reveals that particular focal adhesion proteins, which connect to the actin cytoskeleton, associate with ligand-cross-linked IgE-Fc epsilonRI, independently of integrins. We investigated the functional role of one of these proteins, paxillin, in IgE-Fc epsilonRI-mediated signaling by using small interfering RNA. From these results, we determine that paxillin reduces stimulated phosphorylation of the Fc epsilonRI beta subunit but enhances stimulated Ca(2+) release from intracellular stores. The results suggest that paxillin associated with clustered IgE-Fc epsilonRI has a net positive effect on Fc epsilonRI signaling.
Collapse
|
22
|
Sundberg-Smith LJ, DiMichele LA, Sayers RL, Mack CP, Taylor JM. The LIM protein leupaxin is enriched in smooth muscle and functions as an serum response factor cofactor to induce smooth muscle cell gene transcription. Circ Res 2008; 102:1502-11. [PMID: 18497331 DOI: 10.1161/circresaha.107.170357] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Leupaxin is a LIM domain-containing adapter protein belonging to the paxillin family that has been previously reported to be preferentially expressed in hematopoietic cells. Herein, we identified leupaxin in a screen for focal adhesion kinase binding partners in aortic smooth muscle, and we show that leupaxin is enriched in human and mouse vascular smooth muscle and that leupaxin expression is dynamically regulated during development. In addition, our studies reveal that leupaxin can undergo cytoplasmic/nuclear shuttling and functions as an serum response factor cofactor in the nucleus. We found that leupaxin forms a complex with serum response factor and associates with CArG-containing regions of smooth muscle promoters and that ectopic expression of leupaxin induces smooth muscle marker gene expression in both 10T1/2 cells and rat aortic smooth muscle cells. Subsequent studies indicated that enhanced focal adhesion kinase activity (induced by fibronectin or expression of constitutively active focal adhesion kinase) attenuates the nuclear accumulation of leupaxin and limits the ability of leupaxin to enhance serum response factor-dependent gene transcription. Thus, these studies indicate that modulation of the subcellular localization of serum response factor cofactors is 1 mechanism by which extracellular matrix-dependent signals may regulate phenotypic switching of smooth muscle cells.
Collapse
|