1
|
Nunes J, Tafesse R, Mao C, Purcell M, Mo X, Zhang L, Long M, Cyr MG, Rader C, Muthusamy N. Siglec-6 as a therapeutic target for cell migration and adhesion in chronic lymphocytic leukemia. Nat Commun 2024; 15:5180. [PMID: 38890323 PMCID: PMC11189495 DOI: 10.1038/s41467-024-48678-3] [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: 01/30/2023] [Accepted: 05/08/2024] [Indexed: 06/20/2024] Open
Abstract
Siglec-6 is a lectin receptor with restricted expression in the placenta, mast cells and memory B-cells. Although Siglec-6 is expressed in patients with chronic lymphocytic leukemia (CLL), its pathophysiological role has not been elucidated. We describe here a role for Siglec-6 in migration and adhesion of CLL B cells to CLL- bone marrow stromal cells (BMSCs) in vitro and compromised migration to bone marrow and spleen in vivo. Mass spectrometry analysis revealed interaction of Siglec-6 with DOCK8, a guanine nucleotide exchange factor. Stimulation of MEC1-002 CLL cells with a Siglec-6 ligand, sTn, results in Cdc42 activation, WASP protein recruitment and F-actin polymerization, which are all associated with cell migration. Therapeutically, a Siglec-6/CD3-bispecific T-cell-recruiting antibody (T-biAb) improves overall survival in an immunocompetent mouse model and eliminates CLL cells in a patient derived xenograft model. Our findings thus reveal a migratory role for Siglec-6 in CLL, which can be therapeutically targeted using a Siglec-6 specific T-biAb.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Humans
- Animals
- Cell Movement
- Cell Adhesion
- Lectins/metabolism
- Mice
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Female
- B-Lymphocytes/metabolism
- B-Lymphocytes/immunology
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Cell Line, Tumor
- Mesenchymal Stem Cells/metabolism
- Male
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jessica Nunes
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Rakeb Tafesse
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Charlene Mao
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Matthew Purcell
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Liwen Zhang
- Campus Chemical Instrument Center, The Ohio State University, Columbus, OH, USA
| | - Meixiao Long
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Matthew G Cyr
- UF Scripps Biomedical Research, University of Florida, Jupiter, FL, USA
| | - Christoph Rader
- UF Scripps Biomedical Research, University of Florida, Jupiter, FL, USA
| | - Natarajan Muthusamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Lissek T. Aging as a Consequence of the Adaptation-Maladaptation Dilemma. Adv Biol (Weinh) 2024; 8:e2300654. [PMID: 38299389 DOI: 10.1002/adbi.202300654] [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: 11/30/2023] [Revised: 01/11/2024] [Indexed: 02/02/2024]
Abstract
In aging, the organism is unable to counteract certain harmful influences over its lifetime which leads to progressive dysfunction and eventually death, thus delineating aging as one failed process of adaptation to a set of aging stimuli. A central problem in understanding aging is hence to explain why the organism cannot adapt to these aging stimuli. The adaptation-maladaptation theory of aging proposes that in aging adaptation processes such as adaptive transcription, epigenetic remodeling, and metabolic plasticity drive dysfunction themselves over time (maladaptation) and thereby cause aging-related disorders such as cancer and metabolic dysregulation. The central dilemma of aging is thus that the set of adaptation mechanisms that the body uses to deal with internal and external stressors acts as a stressor itself and cannot be effectively counteracted. The only available option for the organism to decrease maladaptation may be a program to progressively reduce the output of adaptive cascades (e.g., via genomic methylation) which then leads to reduced physiological adaptation capacity and syndromes like frailty, immunosenescence, and cognitive decline. The adaptation-maladaptation dilemma of aging entails that certain biological mechanisms can simultaneously protect against aging as well as drive aging. The key to longevity may lie in uncoupling adaptation from maladaptation.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| |
Collapse
|
3
|
Nie K, Redmond D, Eng KW, Zhang T, Cheng S, Mathew S, Elemento O, Tam W. Mutation landscape, clonal evolution pattern, and potential pathogenic pathways in B-lymphoblastic transformation of follicular lymphoma. Leukemia 2020; 35:1203-1208. [PMID: 32788647 DOI: 10.1038/s41375-020-01014-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Kui Nie
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kenneth W Eng
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Taotao Zhang
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shuhua Cheng
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Susan Mathew
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Wayne Tam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Greene JT, Mani R, Ramaswamy R, Frissora F, Yano M, Zapolnik K, Harrington B, Wasmuth R, Tran M, Mo X, McKenna M, Rangnekar VM, Byrd JC, Bondada S, Muthusamy N. Par-4 overexpression impedes leukemogenesis in the Eµ-TCL1 leukemia model through downregulation of NF-κB signaling. Blood Adv 2019; 3:1255-1266. [PMID: 30987970 PMCID: PMC6482354 DOI: 10.1182/bloodadvances.2018025973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/23/2019] [Indexed: 01/25/2023] Open
Abstract
Prostate apoptosis response 4 (Par-4) is a tumor suppressor that prevents proliferation and induces cell death in several solid tumors. However, its role in B-cell malignancies has not been elucidated. To describe the role of Par-4 in chronic lymphocytic leukemia (CLL) pathogenesis, we developed a B-cell-specific human Par-4-overexpressing mouse model of CLL using the TCL1 leukemia model. While Par-4 transgenic mice did not display any obvious defects in B-cell development or function, disease burden as evidenced by abundance of CD19+CD5+ B cells in the peripheral blood was significantly reduced in Par-4 × TCL1 mice compared with TCL1 littermates. This conferred a survival advantage on the Par-4-overexpressing mice. In addition, a B-cell-specific knockout model displayed the opposite effect, where lack of Par-4 expression resulted in accelerated disease progression and abbreviated survival in the TCL1 model. Histological and flow cytometry-based analysis of spleen and bone marrow upon euthanasia revealed comparable levels of malignant B-cell infiltration in Par-4 × TCL1 and TCL1 individuals, indicating delayed but pathologically normal disease progression in Par-4 × TCL1 mice. In vivo analysis of splenic B-cell proliferation by 5-ethynyl-2-deoxyuridine incorporation indicated >50% decreased expansion of CD19+CD5+ cells in Par-4 × TCL1 mice compared with TCL1 littermates. Moreover, reduced nuclear p65 levels were observed in Par-4 × TCL1 splenic B cells compared with TCL1, suggesting suppressed NF-κB signaling. These findings have identified an in vivo antileukemic role for Par-4 through an NF-κB-dependent mechanism in TCL1-mediated CLL-like disease progression.
Collapse
MESH Headings
- Animals
- Apoptosis Regulatory Proteins/biosynthesis
- Carcinogenesis/genetics
- Carcinogenesis/metabolism
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- Mice, Transgenic
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Signal Transduction
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- J T Greene
- The James Comprehensive Cancer Center and
| | | | | | | | - Max Yano
- The James Comprehensive Cancer Center and
| | | | | | | | - Minh Tran
- The James Comprehensive Cancer Center and
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH; and
| | - Mary McKenna
- Markey Cancer Center, University of Kentucky, Lexington, KY
| | | | | | | | | |
Collapse
|
5
|
Mani R, Mao Y, Frissora FW, Chiang CL, Wang J, Zhao Y, Wu Y, Yu B, Yan R, Mo X, Yu L, Flynn J, Jones J, Andritsos L, Baskar S, Rader C, Phelps MA, Chen CS, Lee RJ, Byrd JC, Lee LJ, Muthusamy N. Tumor antigen ROR1 targeted drug delivery mediated selective leukemic but not normal B-cell cytotoxicity in chronic lymphocytic leukemia. Leukemia 2015; 29:346-55. [PMID: 24947019 PMCID: PMC4272672 DOI: 10.1038/leu.2014.199] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 05/08/2014] [Accepted: 06/04/2014] [Indexed: 12/29/2022]
Abstract
Selective cytotoxicity to cancer cells without compromising their normal counterparts pose a huge challenge for traditional drug design. Here we developed a tumor antigen-targeted delivery of immunonanoparticle carrying a novel non-immunosuppressive FTY720 derivative OSU-2S with potent cytotoxicity against leukemic B cells. OSU-2S induces activation of protein phosphatase 2A (PP2A), phosphorylation and nuclear translocation of SHP1(S591) and deregulation of multiple cellular processes in chronic lymphocytic leukemia (CLL) resulting in potent cytotoxicity. To preclude OSU-2S-mediated effects on these ubiquitous phosphatases in unintended cells and avoid potential adverse effects, we developed an OSU-2S-targeted delivery of immunonanoparticles (2A2-OSU-2S-ILP), that mediated selective cytotoxicity of CLL but not normal B cells through targeting receptor tyrosine kinase ROR1 expressed in leukemic but not normal B cells. Developing a novel spontaneous CLL mouse model expressing human ROR1 (hROR1) in all leukemic B cells, we demonstrate the therapeutic benefit of enhanced survival with 2A2-OSU-2S-ILP in vivo. The newly developed non-immunosuppressive OSU-2S, its delivery using human CLL directed immunonanoparticles and the novel transgenic (Tg) mouse model of CLL that expresses hROR1 exclusively in leukemic B cell surface are highly innovative and can be applied to CLL and other ROR1+ malignancies including mantle cell lymphoma and acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- R Mani
- 1] Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA [2] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA [3] Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Y Mao
- 1] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA [2] Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA [3] Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, OH, USA
| | - F W Frissora
- 1] Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA [2] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - C-L Chiang
- 1] Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA [2] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - J Wang
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Y Zhao
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Y Wu
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, OH, USA
| | - B Yu
- 1] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA [2] Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, OH, USA
| | - R Yan
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - X Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - L Yu
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - J Flynn
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - J Jones
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - L Andritsos
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - S Baskar
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - C Rader
- Department of Cancer Biology and Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL, USA
| | - M A Phelps
- 1] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA [2] Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - C-S Chen
- 1] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA [2] Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA [3] Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - R J Lee
- 1] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA [2] Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA [3] Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, OH, USA
| | - J C Byrd
- 1] Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA [2] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA [3] Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA [4] Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - L J Lee
- 1] Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, OH, USA [2] Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - N Muthusamy
- 1] Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA [2] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA [3] Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
6
|
MAP Kinase Cascades in Antigen Receptor Signaling and Physiology. Curr Top Microbiol Immunol 2015; 393:211-231. [PMID: 26275875 DOI: 10.1007/82_2015_481] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) play roles in a cell type and context-dependent manner to convert extracellular stimuli to a variety of cellular responses, thereby directing cells to proliferation, differentiation, survival, apoptosis, and migration. Studies of genetically engineered mice or chemical inhibitors specific to each MAPK signaling pathway revealed that MAPKs have various, but non-redundant physiologically important roles among different families. MAPK cascades are obviously integrated in the B cell receptor signaling pathways as critical components to drive B cell-mediated immunity.
Collapse
|
7
|
The CD37-targeted antibody-drug conjugate IMGN529 is highly active against human CLL and in a novel CD37 transgenic murine leukemia model. Leukemia 2014; 28:1501-10. [PMID: 24445867 PMCID: PMC4090271 DOI: 10.1038/leu.2014.32] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 12/27/2013] [Accepted: 01/08/2014] [Indexed: 12/18/2022]
Abstract
Therapeutic regimens for chronic lymphocytic leukemia (CLL) have increasingly utilized monoclonal antibodies since the chimeric anti-CD20 antibody rituximab was introduced. Despite improved clinical outcomes, current CLL therapies are not curative. Therefore, antibodies with greater efficacy and novel targets are desirable. One promising target is CD37, a tetraspanin protein highly expressed on malignant B-cells in CLL and non-Hodgkin lymphoma. Although several novel CD37-directed therapeutics are emerging, detailed preclinical evaluation of these agents is limited by lack of appropriate animal models with spontaneous leukemia expressing the human CD37 (hCD37) target. To address this, we generated a murine CLL model that develops transplantable hCD37+ leukemia. Subsequently, we engrafted healthy mice with this leukemia to evaluate IMGN529, a novel hCD37-targeting antibody-drug conjugate. IMGN529 rapidly eliminated peripheral blood leukemia and improved overall survival. In contrast, the antibody component of IMGN529 could not alter disease course despite exhibiting substantial in vitro cytotoxicity. Furthermore, IMGN529 is directly cytotoxic to human CLL in vitro, depletes B-cells in patient whole blood and promotes killing by macrophages and natural killer cells. Our results demonstrate the utility of a novel mouse model for evaluating anti-human CD37 therapeutics and highlight the potential of IMGN529 for treatment of CLL and other CD37-positive B-cell malignancies.
Collapse
|
8
|
Heydarian M, Luperchio TR, Cutler J, Mitchell CJ, Kim MS, Pandey A, Sollner-Webb B, Reddy K. Prediction of Gene Activity in Early B Cell Development Based on an Integrative Multi-Omics Analysis. ACTA ACUST UNITED AC 2014; 7. [PMID: 25544807 PMCID: PMC4276347 DOI: 10.4172/jpb.1000302] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
An increasingly common method for predicting gene activity is genome-wide chromatin immuno-precipitation of ‘active’ chromatin modifications followed by massively parallel sequencing (ChIP-seq). In order to understand better the relationship between developmentally regulated chromatin landscapes and regulation of early B cell development, we determined how differentially active promoter regions were able to predict relative RNA and protein levels at the pre-pro-B and pro-B stages. Herein, we describe a novel ChIP-seq quantification method (cRPKM) to identify active promoters and a multi-omics approach that compares promoter chromatin status with ongoing active transcription (GRO-seq), steady state mRNA (RNA-seq), inferred mRNA stability, and relative proteome abundance measurements (iTRAQ). We demonstrate that active chromatin modifications at promoters are good indicators of transcription and steady state mRNA levels. Moreover, we found that promoters with active chromatin modifications exclusively in one of these cell states frequently predicted the differential abundance of proteins. However, we found that many genes whose promoters have non-differential but active chromatin modifications also displayed changes in abundance of their cognate proteins. As expected, this large class of developmentally and differentially regulated proteins that was uncoupled from chromatin status used mostly post-transcriptional mechanisms. Strikingly, the most differentially abundant protein in our B-cell development system, 2410004B18Rik, was regulated by a post-transcriptional mechanism, which further analyses indicated was mediated by a micro-RNA. These data highlight how this integrated multi-omics data set can be a useful resource in uncovering regulatory mechanisms. This data can be accessed at: https://usegalaxy.org/u/thereddylab/p/prediction-of-gene-activity-based-on-an-integrative-multi-omics-analysis
Collapse
Affiliation(s)
- Mohammad Heydarian
- Johns Hopkins University, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, USA ; Johns Hopkins University, Center for Epigenetics, 855 North Wolfe Street, Baltimore, USA
| | - Teresa Romeo Luperchio
- Johns Hopkins University, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, USA ; Johns Hopkins University, Center for Epigenetics, 855 North Wolfe Street, Baltimore, USA
| | - Jevon Cutler
- Johns Hopkins University, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, USA ; Johns Hopkins University, Center for Epigenetics, 855 North Wolfe Street, Baltimore, USA ; Johns Hopkins University, McKusick-Nathans Institute of Genetic Medicine, 733 North, Broadway Avenue, Baltimore, USA
| | - Christopher J Mitchell
- Johns Hopkins University, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, USA ; Johns Hopkins University, McKusick-Nathans Institute of Genetic Medicine, 733 North, Broadway Avenue, Baltimore, USA
| | - Min-Sik Kim
- Johns Hopkins University, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, USA ; Johns Hopkins University, McKusick-Nathans Institute of Genetic Medicine, 733 North, Broadway Avenue, Baltimore, USA
| | - Akhilesh Pandey
- Johns Hopkins University, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, USA ; Johns Hopkins University, McKusick-Nathans Institute of Genetic Medicine, 733 North, Broadway Avenue, Baltimore, USA
| | - Barbara Sollner-Webb
- Johns Hopkins University, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, USA
| | - Karen Reddy
- Johns Hopkins University, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, USA ; Johns Hopkins University, Center for Epigenetics, 855 North Wolfe Street, Baltimore, USA
| |
Collapse
|
9
|
|
10
|
Muthusamy N, Chen HC, Rajgolikar G, Butz KG, Frissora FW, Gronostajski RM. Recombination activation gene-2-deficient blastocyst complementation analysis reveals an essential role for nuclear factor I-A transcription factor in T-cell activation. Int Immunol 2011; 23:385-90. [PMID: 21602176 DOI: 10.1093/intimm/dxr025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Nuclear factor I (NFI)-A is a member of the NFI family of transcription factors implicated in regulation of granulocyte differentiation. However, its role in the lymphoid lineage is not known. NFI-A deficiency results in perinatal lethality, thus precluding analysis of the role of NFI-A in lymphocyte development and function. Using recombination activation gene-2-deficient (RAG-2(-/-)) blastocysts and embryonic stem cells with homozygous NFI-A gene deletion, we show an essential role for NFI-A in T-cell activation. NFI-A(-/-)→RAG-2(-/-) chimeric mice had normal distributions of CD4(-)CD8(-) double negative, CD4(+)CD8(+) double positive, CD4(+)CD8(-) and CD4(-)CD8(+)-single positive cells in the thymus and CD4(+)CD8(-) and CD4(-)CD8(+) cells in spleen and lymph nodes. However, NFI-A(-/-)→RAG-2(-)(/)(-) mice had severely reduced thymus size and hypocellularity. The decrease in thymocytes and peripheral T cells in NFI-A(-/-)→RAG-2(-/-) chimeric mice is attributed to proliferative defects associated with decreased blast transformation, CD69 expression and DNA synthesis in response to T antigen receptor stimulation. Interestingly, NFI-A-null T cells showed increased levels of c-myc transcription that is inhibited in response to antigen receptor-mediated activation. These studies demonstrate for the first time a requirement for the NFI-A transcription factor in antigen receptor-induced T-cell activation events.
Collapse
Affiliation(s)
- Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine,The OSU Comprehensive Cancer Center, The Ohio State University,Columbus, OH 43210, USA.
| | | | | | | | | | | |
Collapse
|
11
|
Pei DS, Yang XJ, Liu W, Guikema JEJ, Schrader CE, Strauss PR. A novel regulatory circuit in base excision repair involving AP endonuclease 1, Creb1 and DNA polymerase beta. Nucleic Acids Res 2010; 39:3156-65. [PMID: 21172930 PMCID: PMC3082881 DOI: 10.1093/nar/gkq1142] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
DNA repair is required to maintain genome stability in stem cells and early embryos. At critical junctures, oxidative damage to DNA requires the base excision repair (BER) pathway. Since early zebrafish embryos lack the major polymerase in BER, DNA polymerase ß, repair proceeds via replicative polymerases, even though there is ample polb mRNA. Here, we report that Polb protein fails to appear at the appropriate time in development when AP endonuclease 1 (Apex), the upstream protein in BER, is knocked down. Because polb contains a Creb1 binding site, we examined whether knockdown of Apex affects creb1. Apex knockdown results in loss of Creb1 and Creb complex members but not Creb1 phosphorylation. This effect is independent of p53. Although both apex and creb1 mRNA rescue Creb1 and Polb after Apex knockdown, Apex is not a co-activator of creb1 transcription. This observation has broad significance, as similar results occur when Apex is inhibited in B cells from apex+/− mice. These results describe a novel regulatory circuit involving Apex, Creb1 and Polb and provide a mechanism for lethality of Apex loss in higher eukaryotes.
Collapse
Affiliation(s)
- De-Sheng Pei
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
12
|
Abstract
Antigen receptors on the surface of B lymphocytes trigger adaptive immune responses after encountering their cognate antigens but also control a series of antigen-independent checkpoints during B cell development. These physiological processes are regulated by the expression and function of cell surface receptors, intracellular signaling molecules, and transcription factors. The function of these proteins can be altered by a dynamic array of post-translational modifications, using two interconnected mechanisms. These modifications can directly induce an altered conformational state in the protein target of the modification itself. In addition, they can create new binding sites for other protein partners, thereby contributing to where and when such multiple protein assemblies are activated within cells. As a new type of post-transcriptional regulator, microRNAs have emerged to influence the development and function of B cells by affecting the expression of target mRNAs.
Collapse
|
13
|
|
14
|
Gururajan M, Simmons A, Dasu T, Spear BT, Calulot C, Robertson DA, Wiest DL, Monroe JG, Bondada S. Early growth response genes regulate B cell development, proliferation, and immune response. THE JOURNAL OF IMMUNOLOGY 2008; 181:4590-602. [PMID: 18802061 DOI: 10.4049/jimmunol.181.7.4590] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Egr-1 (early growth response gene-1) is an immediate early gene encoding a zinc finger motif-containing transcription factor. Upon cross-linking of BCR, mature B cells undergo proliferation with an increase in Egr-1 message. Immature B lymphoma cells that express Egr-1 message and protein constitutively are growth inhibited when Egr-1 is down-regulated by negative signals from BCR or by antisense oligonucleotides. To test the hypothesis that Egr-1 is important for B cell development, we examined B cells from primary and secondary lymphoid organs in Egr-1(-/-) mice. Marginal zone B cell development was arrested in these mice, whereas the B cells in all other compartments were increased. To test the hypothesis that Egr-1 function may be partially compensated by other Egr family members, we developed transgenic mice expressing a dominant negative form of Egr-1, which lacks the trans activation domain but retains the DNA-binding domain, in a B cell-specific manner. There was a decrease in B lymphopoiesis in the bone marrow accompanied by a reduction in splenic immature and mature B cells as well as marginal zone B cells in the transgenic mice. Moreover, transgenic mice respond poorly to BCR cross-linking in vitro and T-independent and T-dependent Ags in vivo.
Collapse
Affiliation(s)
- Murali Gururajan
- Departments of Microbiology, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
The B cell receptor (BCR) activates the ERK kinases, but what does ERK do? In this issue of Immunity, Yasuda et al. (2008) show that ERK controls a transcription factor network by which the pre-BCR drives early B cell development.
Collapse
Affiliation(s)
- Michael R Gold
- Department of Microbiology and Immunology and I(3) and CELL Research Groups, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
16
|
Erk kinases link pre-B cell receptor signaling to transcriptional events required for early B cell expansion. Immunity 2008; 28:499-508. [PMID: 18356083 DOI: 10.1016/j.immuni.2008.02.015] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Revised: 02/06/2008] [Accepted: 02/06/2008] [Indexed: 11/23/2022]
Abstract
The pre-B cell receptor (pre-BCR) plays a crucial role in the development of immature B cells. Although certain aspects of proximal pre-BCR signaling have been studied, the intermediate signal transducers and the distal transcription modulators are poorly characterized. Here, we demonstrate that deletion of both Erk1 and Erk2 kinases was associated with defective pre-BCR-mediated cell expansion as well as a block in the transition of pro-B to pre-B cells. Phosphorylation of transcription factors Elk1 and CREB was mediated by Erk, and a dominant-negative mutation in the Erk-mediated phosphorylation sites of Elk1 or CREB suppressed pre-BCR-mediated cell expansion as well as expression of genes including Myc, which is involved in the cell-cycle progression. Together, our results identify a crucial role for Erk kinases in regulating B cell development by initiating transcriptional regulatory network and thereby pre-BCR-mediated cell expansion.
Collapse
|