1
|
Li YY, Wang XY, Li Y, Wang XM, Liao J, Wang YZ, Hong H, Yi W, Chen J. Targeting CD43 optimizes cancer immunotherapy through reinvigorating antitumor immune response in colorectal cancer. Cell Oncol (Dordr) 2023; 46:777-791. [PMID: 36920728 DOI: 10.1007/s13402-023-00794-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
PURPOSE Colorectal cancer (CRC) is one of the most common malignancies worldwide, with dramatically increasing incidence and mortality for decades. However, current therapeutic strategies for CRC, including chemotherapies and immunotherapies, have only demonstrated limited efficacy. Here, we report a novel immune molecule, CD43, that can regulate the tumor immune microenvironment (TIME) and serves as a promising target for CRC immunotherapy. METHODS The correlation of CD43 expression with CRC patient prognosis was revealed by public data analysis. CD43 knockout (KO) CRC cell lines were generated by CRISPR-Cas9 technology, and a syngenetic murine CRC model was established to investigate the in vivo function of CD43. The TIME was analyzed via immunohistochemical staining, flow cytometry and RNA-seq. Immune functions were investigated by depletion of immune subsets in vivo and T-cell functional assays in vitro, including T-cell priming, cytotoxicity, and chemotaxis experiments. RESULTS In this study, we found that high expression of CD43 was correlated with poor survival of CRC patients and the limited infiltration of CD8+ T cells in human CRC tissues. Importantly, CD43 expressed on tumor cells, rather than host cells, promoted tumor progression in a syngeneic tumor model. Loss of CD43 facilitated the infiltration of immune cells and immunological memory in the TIME of CRC tumors. Mechanistically, the protumor effect of CD43 depends on T cells, thereby attenuating T-cell-mediated cytotoxicity and cDC1-mediated antigen-specific T-cell activation. Moreover, targeting CD43 synergistically improved PD-L1 blockade immunotherapy for CRC. CONCLUSION Our findings revealed that targeting tumor-intrinsic CD43 could activate the antitumor immune response and provide particular value for optimized cancer immunotherapy by regulating the TIME in CRC patients.
Collapse
Affiliation(s)
- Yi-Yi Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.,Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xin-Yu Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.,Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.,Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiu-Mei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.,Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing Liao
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Ying-Zhao Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hai Hong
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Wei Yi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.
| | - Jun Chen
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China. .,Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Center for Precision Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
2
|
Vega-Mendoza D, Cañas-Linares A, Flores-Alcantar A, Espinosa-Neira R, Melchy-Perez E, Vera-Estrella R, Auvynet C, Rosenstein Y. CD43 (sialophorin) is involved in the induction of extracellular matrix remodeling and angiogenesis by lung cancer cells. J Cell Physiol 2021; 236:6643-6656. [PMID: 33533043 DOI: 10.1002/jcp.30308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/17/2021] [Accepted: 01/21/2021] [Indexed: 12/19/2022]
Abstract
Aberrant expression of CD43 in malignant tumors of nonhematopoietic origin such as those from lung, cervix, colon, and breast has been shown to correlate with poor prognosis, providing tumor cells with enhanced motility, anchorage-independent growth, and in vivo tumor size, while protecting the cells of NK lysis and apoptosis. To further characterize the role of CD43 in cell transformation, we tested whether interfering its expression modified the capacity of the A549 non-small cell lung cancer cells to secrete molecules contributing to malignancy. The proteomic analysis of the secretome of serum-starved A549 cells revealed that cells expressing normal levels of CD43 released significantly high levels of molecules involved in extracellular matrix organization, angiogenesis, platelet degranulation, collagen degradation, and inflammation, as compared to CD43 RNAi cells. This data reveals a novel and unexpected role for CD43 in lung cancer development, mainly in remodeling the tumor microenvironment.
Collapse
Affiliation(s)
- Daniela Vega-Mendoza
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico.,Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Alicia Cañas-Linares
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico.,Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Angel Flores-Alcantar
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Roberto Espinosa-Neira
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico.,División de Investigación Básica, Laboratorio de Epigenética del Cáncer, Instituto Nacional de Cancerología, Ciudad de México, Mexico
| | - Erika Melchy-Perez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Rosario Vera-Estrella
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Constance Auvynet
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Yvonne Rosenstein
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| |
Collapse
|
3
|
de Jong G, Bartels L, Kedde M, Verdegaal EME, Gillissen MA, Levie SE, Cercel MG, van Hal-van Veen SE, Fatmawati C, van de Berg D, Yasuda E, Claassen YB, Bakker AQ, van der Burg SH, Schotte R, Villaudy J, Spits H, Hazenberg MD, van Helden PM, Wagner K. Melanoma cells can be eliminated by sialylated CD43 × CD3 bispecific T cell engager formats in vitro and in vivo. Cancer Immunol Immunother 2020; 70:1569-1581. [PMID: 33225419 DOI: 10.1007/s00262-020-02780-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/22/2020] [Indexed: 01/13/2023]
Abstract
Targeted cancer therapy with monoclonal antibodies has proven successful for different cancer types but is limited by the availability of suitable antibody targets. CD43s, a unique sialylated form of CD43 expressed by hematologic malignancies, is a recently identified target and antibodies interacting with CD43s may have therapeutic potential against acute myeloid leukemia (AML) and myelodysplastic syndrome. CD43s is recognized by the human antibody AT1413, that was derived from a high-risk AML patient who successfully cleared leukemia after allogeneic stem cell transplantation. Here we observed that AT1413 binds also to certain non-hematopoietic tumor cells, particularly melanoma and breast cancer. AT1413 immune precipitated CD43s from melanoma cells confirming that it recognizes the same target on melanoma as on AML. AT1413 induced antibody-dependent cellular cytotoxicity against short-term cultured patient-derived melanoma samples. However, AT1413 was unable to affect the growth of melanoma cells in vivo. To increase the efficacy of AT1413 as a therapeutic antibody, we generated two different formats of bispecific T-cell engaging antibodies (TCEs): one binding bivalently (bTCE) and the other monovalently (knob-in-hole; KiH) to both CD43s and CD3ε. In vitro, these TCEs redirected T-cell cytotoxicity against melanoma cells with differences in potencies. To investigate their effects in vivo, we grafted mice that harbor a human immune system with the melanoma cell line A375. Treatment with both AT1413 bTCE and AT1413 KiH significantly reduced tumor outgrowth in these mice. These data indicate a broad therapeutic potential of AT1413 that includes AML and CD43s-expressing solid tumors that originate from CD43-negative tissues.
Collapse
Affiliation(s)
- G de Jong
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands
| | - L Bartels
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands
| | - M Kedde
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - E M E Verdegaal
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - M A Gillissen
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands
| | - S E Levie
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - M G Cercel
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | | | - C Fatmawati
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - D van de Berg
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - E Yasuda
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - Y B Claassen
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - A Q Bakker
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - S H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - R Schotte
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - J Villaudy
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - H Spits
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - M D Hazenberg
- Department of Hematology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands.,Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
| | - P M van Helden
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.
| | - K Wagner
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Sánchez NC, Medrano-Jiménez E, Aguilar-León D, Pérez-Martínez L, Pedraza-Alva G. Tumor Necrosis Factor-Induced miR-146a Upregulation Promotes Human Lung Adenocarcinoma Metastasis by Targeting Merlin. DNA Cell Biol 2020; 39:484-497. [PMID: 31999471 DOI: 10.1089/dna.2019.4620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Inflammation plays a key role in carcinogenesis and metastasis. This process involves the inactivation of tumor suppressor molecules, yet the molecular mechanisms by which inflammation impairs tumor suppressors are not completely understood. In this study, we show that proinflammatory signals such as tumor necrosis factor (TNF) support lung cancer metastasis by reducing the levels of the tumor suppressor Merlin through regulation of miR-146a. Immunodeficient mice inoculated with A549 cells expressing high miR-146a levels and low Merlin protein levels exhibited reduced survival, which correlated with the number of metastatic nodes formed. Accordingly, restoring Merlin protein levels inhibited metastasis and increased survival of the mice. Consistent with these results, we found that elevated miR-146a expression levels correlated with low Merlin protein levels in human lung adenocarcinoma. Furthermore, human invasive and metastatic tumors showed higher TNF and miR-146a levels, but lower Merlin protein levels than noninvasive tumors. These findings indicate that upregulation of miR-146a by TNF in lung adenocarcinoma promotes Merlin protein inhibition and metastasis. Thus, we suggest that the ratio between miR-146a and Merlin protein levels could be a relevant molecular biomarker that can predict lung cancer progression and that the TNF/miR-146a/Merlin pathway is a promising new therapeutic target to inhibit lung adenocarcinoma progression.
Collapse
Affiliation(s)
- Nilda C Sánchez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México.,Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos
| | - Elisa Medrano-Jiménez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Diana Aguilar-León
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, México
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| |
Collapse
|
5
|
Ma XB, Zhong YP, Zheng Y, Jiang J, Wang YP. Coexpression of CD5 and CD43 predicts worse prognosis in diffuse large B-cell lymphoma. Cancer Med 2018; 7:4284-4295. [PMID: 30019388 PMCID: PMC6144247 DOI: 10.1002/cam4.1674] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/20/2018] [Indexed: 12/15/2022] Open
Abstract
Both CD5 and CD43 are expressed on the surface of B lymphocytes of definite phase and associated with the adverse outcome in diffuse large B-cell lymphoma (DLBCL). However, the relationship between CD5 and CD43 expression and the prognostic value of CD5/CD43 coexpression in DLBCL are unknown. We herein determined the correlation between CD5 and CD43 expression, as separate factors or in combination, with the clinicopathological features and survival of 200 patients with DLBCL receiving standard chemotherapy with or without rituximab. Among these DLBCL patients, CD5 expression, CD43 expression, and CD5/CD43 coexpression were detected in 18 (9%), 57 (27%), and 10 (5%) patients, respectively, and all were positively correlated with advanced age and nongerminal cell type. CD5-positive and CD43-positive DLBCL patients had poorer event-free survival (EFS, P < 0.001) and overall survival (OS, P < 0.001) than CD5-negative and CD43-negative patients, respectively. CD5/CD43 coexpression was correlated with a significantly worse prognosis than CD5 or CD43 expression alone. Univariate analysis showed that CD5 expression, CD43 expression, and CD5/CD43 coexpression were all adverse prognostic factors for DLBCL patient survival, and CD5/CD43 coexpression was associated with a greater relative risk for recurrence and death than either CD5 or CD43 expression alone. Multivariate analysis demonstrated that CD5/CD43 coexpression was an independent prognostic factor for EFS (P < 0.001) and OS (P < 0.001) in DLBCL. In conclusion, our data indicate that DLBCL patients with CD5/CD43 coexpression represent a specific subgroup with a significantly worse prognosis than those expressing either marker alone.
Collapse
Affiliation(s)
- Xiao-Bo Ma
- Department of Pathology, First Hospital of Jilin University, Changchun, China
| | - Yan-Ping Zhong
- Department of Pathology, First Hospital of Jilin University, Changchun, China
| | - Yan Zheng
- Department of Pathology, First Hospital of Jilin University, Changchun, China
| | - Jing Jiang
- Division of Clinical Epidemiology, First Hospital of Jilin University, Changchun, China
| | - Yin-Ping Wang
- Department of Pathology, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Stepanova DS, Semenova G, Kuo YM, Andrews AJ, Ammoun S, Hanemann CO, Chernoff J. An Essential Role for the Tumor-Suppressor Merlin in Regulating Fatty Acid Synthesis. Cancer Res 2017; 77:5026-5038. [PMID: 28729415 DOI: 10.1158/0008-5472.can-16-2834] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 05/17/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022]
Abstract
Neurofibromatosis type 2 (NF2) is an autosomal dominant disorder characterized by the development of multiple tumors in the central nervous system, most notably schwannomas, and meningiomas. Mutational inactivation of the NF2 gene encoding the protein Merlin is found in most sporadic and inherited schwannomas, but the molecular mechanisms underlying neoplastic changes in schwannoma cells remain unclear. We report here that Nf2-deficient cells display elevated expression levels of key enzymes involved in lipogenesis and that this upregulation is caused by increased activity of Torc1. Inhibition or knockdown of fatty acid synthase (FASN), the enzyme that catalyzes the formation of palmitic acid from malonyl-CoA, drove NF2-deficient cells into apoptosis. Treatment of NF2-mutant cells with agents that inhibit the production of malonyl-CoA reduced their sensitivity to FASN inhibitors. Collectively, these results suggest that the altered lipid metabolism found in NF2-mutant cells renders them sensitive to elevated levels of malonyl-CoA, as occurs following blockade of FASN, suggesting new targeted strategies in the treatment of NF2-deficient tumors. Cancer Res; 77(18); 5026-38. ©2017 AACR.
Collapse
Affiliation(s)
- Dina S Stepanova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Galina Semenova
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yin-Ming Kuo
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Andrew J Andrews
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Sylwia Ammoun
- Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, United Kingdom
| | - C Oliver Hanemann
- Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, United Kingdom
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
7
|
Interaction of the CD43 Sialomucin with the Mycobacterium tuberculosis Cpn60.2 Chaperonin Leads to Tumor Necrosis Factor Alpha Production. Infect Immun 2017; 85:IAI.00915-16. [PMID: 28069816 DOI: 10.1128/iai.00915-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/18/2016] [Indexed: 12/23/2022] Open
Abstract
Mycobacterium tuberculosis is the causal agent of tuberculosis. Tumor necrosis factor alpha (TNF-α), transforming growth factor β (TGF-β), and gamma interferon (IFN-γ) secreted by activated macrophages and lymphocytes are considered essential to contain Mycobacterium tuberculosis infection. The CD43 sialomucin has been reported to act as a receptor for bacilli through its interaction with the chaperonin Cpn60.2, facilitating mycobacterium-macrophage contact. We report here that Cpn60.2 induces both human THP-1 cells and mouse-derived bone marrow-derived macrophages (BMMs) to produce TNF-α and that this production is CD43 dependent. In addition, we present evidence that the signaling pathway leading to TNF-α production upon interaction with Cpn60.2 requires active Src family kinases, phospholipase C-γ (PLC-γ), phosphatidylinositol 3-kinase (PI3K), p38, and Jun N-terminal protein kinase (JNK), both in BMMs and in THP-1 cells. Our data highlight the role of CD43 and Cpn60.2 in TNF-α production and underscore an important role for CD43 in the host-mycobacterium interaction.
Collapse
|
8
|
Schjerven H, Ayongaba EF, Aghajanirefah A, McLaughlin J, Cheng D, Geng H, Boyd JR, Eggesbø LM, Lindeman I, Heath JL, Park E, Witte ON, Smale ST, Frietze S, Müschen M. Genetic analysis of Ikaros target genes and tumor suppressor function in BCR-ABL1 + pre-B ALL. J Exp Med 2017; 214:793-814. [PMID: 28190001 PMCID: PMC5339667 DOI: 10.1084/jem.20160049] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 10/03/2016] [Accepted: 01/12/2017] [Indexed: 01/19/2023] Open
Abstract
Schjerven et al. compare mouse and human models of pre–B ALL to define conserved target genes and pathways of the tumor suppressor Ikaros, revealing CTNND1 and the early hematopoietic cell-surface receptors SPN (CD43) and CD34 as novel Ikaros targets that each confer oncogenic growth advantage. Inactivation of the tumor suppressor gene encoding the transcriptional regulator Ikaros (IKZF1) is a hallmark of BCR-ABL1+ precursor B cell acute lymphoblastic leukemia (pre–B ALL). However, the mechanisms by which Ikaros functions as a tumor suppressor in pre–B ALL remain poorly understood. Here, we analyzed a mouse model of BCR-ABL1+ pre–B ALL together with a new model of inducible expression of wild-type Ikaros in IKZF1 mutant human BCR-ABL1+ pre–B ALL. We performed integrated genome-wide chromatin and expression analyses and identified Ikaros target genes in mouse and human BCR-ABL1+ pre–B ALL, revealing novel conserved gene pathways associated with Ikaros tumor suppressor function. Notably, genetic depletion of different Ikaros targets, including CTNND1 and the early hematopoietic cell surface marker CD34, resulted in reduced leukemic growth. Our results suggest that Ikaros mediates tumor suppressor function by enforcing proper developmental stage–specific expression of multiple genes through chromatin compaction at its target genes.
Collapse
Affiliation(s)
- Hilde Schjerven
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Etapong F Ayongaba
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143.,Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Ali Aghajanirefah
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Jami McLaughlin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Donghui Cheng
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095
| | - Huimin Geng
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Joseph R Boyd
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont, Burlington, VT 05405
| | - Linn M Eggesbø
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143.,Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Ida Lindeman
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143.,Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Jessica L Heath
- Department of Pediatrics, University of Vermont, Burlington, VT 05405.,Department of Biochemistry, University of Vermont, Burlington, VT 05405
| | - Eugene Park
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Owen N Witte
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095.,Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Stephen T Smale
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095.,Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Seth Frietze
- Department of Medical Laboratory and Radiation Science, University of Vermont, Burlington, VT 05405
| | - Markus Müschen
- Department of Systems Biology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Pasadena, CA 91016
| |
Collapse
|
9
|
Bravo-Adame ME, Vera-Estrella R, Barkla BJ, Martínez-Campos C, Flores-Alcantar A, Ocelotl-Oviedo JP, Pedraza-Alva G, Rosenstein Y. An alternative mode of CD43 signal transduction activates pro-survival pathways of T lymphocytes. Immunology 2016; 150:87-99. [PMID: 27606486 DOI: 10.1111/imm.12670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 08/20/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023] Open
Abstract
CD43 is one of the most abundant co-stimulatory molecules on a T-cell surface; it transduces activation signals through its cytoplasmic domain, contributing to modulation of the outcome of T-cell responses. The aim of this study was to uncover new signalling pathways regulated by this sialomucin. Analysis of changes in protein abundance allowed us to identify pyruvate kinase isozyme M2 (PKM2), an enzyme of the glycolytic pathway, as an element potentially participating in the signalling cascade resulting from the engagement of CD43 and the T-cell receptor (TCR). We found that the glycolytic activity of this enzyme was not significantly increased in response to TCR+CD43 co-stimulation, but that PKM2 was tyrosine phosphorylated, suggesting that it was performing moonlight functions. We report that phosphorylation of both Y105 of PKM2 and of Y705 of signal transducer and activator of transcription 3 was induced in response to TCR+CD43 co-stimulation, resulting in activation of the mitogen-activated protein kinase kinase 5/extracellular signal-regulated kinase 5 (MEK5/ERK5) pathway. ERK5 and the cAMP response element binding protein (CREB) were activated, and c-Myc and nuclear factor-κB (p65) nuclear localization, as well as Bad phosphorylation, were augmented. Consistent with this, expression of human CD43 in a murine T-cell hybridoma favoured cell survival. Altogether, our data highlight novel signalling pathways for the CD43 molecule in T lymphocytes, and underscore a role for CD43 in promoting cell survival through non-glycolytic functions of metabolic enzymes.
Collapse
Affiliation(s)
- Maria Elena Bravo-Adame
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México.,Posgrado en Ciencias Bioquímicas, UNAM, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Rosario Vera-Estrella
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Bronwyn J Barkla
- Southern Cross Plant Science, Southern Cross University, Lismore, NSW, Australia
| | - Cecilia Martínez-Campos
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México.,Posgrado en Ciencias Bioquímicas, UNAM, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Angel Flores-Alcantar
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Jose Pablo Ocelotl-Oviedo
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Gustavo Pedraza-Alva
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Yvonne Rosenstein
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| |
Collapse
|
10
|
Candeias MM, Hagiwara M, Matsuda M. Cancer-specific mutations in p53 induce the translation of Δ160p53 promoting tumorigenesis. EMBO Rep 2016; 17:1542-1551. [PMID: 27702985 DOI: 10.15252/embr.201541956] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 09/06/2016] [Indexed: 12/29/2022] Open
Abstract
Wild-type p53 functions as a tumour suppressor while mutant p53 possesses oncogenic potential. Until now it remains unclear how a single mutation can transform p53 into a functionally distinct gene harbouring a new set of original cellular roles. Here we show that the most common p53 cancer mutants express a larger number and higher levels of shorter p53 protein isoforms that are translated from the mutated full-length p53 mRNA. Cells expressing mutant p53 exhibit "gain-of-function" cancer phenotypes, such as enhanced cell survival, proliferation, invasion and adhesion, altered mammary tissue architecture and invasive cell structures. Interestingly, Δ160p53-overexpressing cells behave in a similar manner. In contrast, an exogenous or endogenous mutant p53 that fails to express Δ160p53 due to specific mutations or antisense knock-down loses pro-oncogenic potential. Our data support a model in which "gain-of-function" phenotypes induced by p53 mutations depend on the shorter p53 isoforms. As a conserved wild-type isoform, Δ160p53 has evolved during millions of years. We thus provide a rational explanation for the origin of the tumour-promoting functions of p53 mutations.
Collapse
Affiliation(s)
- Marco M Candeias
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies Kyoto University, Kyoto, Japan .,Department of Anatomy and Developmental Biology, Graduate School of Medicine Kyoto University, Kyoto, Japan.,Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisboa, Portugal
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine Kyoto University, Kyoto, Japan.,Medical Research Support Center, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Michiyuki Matsuda
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies Kyoto University, Kyoto, Japan.,Department of Pathology and Biology of Diseases, Graduate School of Medicine Kyoto University, Kyoto, Japan
| |
Collapse
|
11
|
Pérez-García EI, Meza-Sosa KF, López-Sevilla Y, Camacho-Concha N, Sánchez NC, Pérez-Martínez L, Pedraza-Alva G. Merlin negative regulation by miR-146a promotes cell transformation. Biochem Biophys Res Commun 2015; 468:594-600. [PMID: 26549232 DOI: 10.1016/j.bbrc.2015.10.156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 10/29/2015] [Indexed: 01/01/2023]
Abstract
Inactivation of the tumor suppressor Merlin, by deleterious mutations or by protein degradation via sustained growth factor receptor signaling-mediated mechanisms, results in cell transformation and tumor development. In addition to these mechanisms, here we show that, miRNA-dependent negative regulation of Merlin protein levels also promotes cell transformation. We provide experimental evidences showing that miR-146a negatively regulates Merlin protein levels through its interaction with an evolutionary conserved sequence in the 3´ untranslated region of the NF2 mRNA. Merlin downregulation by miR-146a in A549 lung epithelial cells resulted in enhanced cell proliferation, migration and tissue invasion. Accordingly, stable miR-146a-transfectant cells formed tumors with metastatic capacity in vivo. Together our results uncover miRNAs as yet another negative mechanism controlling Merlin tumor suppressor functions.
Collapse
Affiliation(s)
- Erick I Pérez-García
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. 62210, Mexico
| | - Karla F Meza-Sosa
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. 62210, Mexico
| | - Yaxem López-Sevilla
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. 62210, Mexico
| | - Nohemi Camacho-Concha
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. 62210, Mexico
| | - Nilda C Sánchez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. 62210, Mexico
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. 62210, Mexico
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. 62210, Mexico.
| |
Collapse
|
12
|
CD43 expression in diffuse large B-cell lymphoma, not otherwise specified: CD43 is a marker of adverse prognosis. Hum Pathol 2015; 46:593-9. [PMID: 25682152 DOI: 10.1016/j.humpath.2015.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/24/2014] [Accepted: 01/02/2015] [Indexed: 12/28/2022]
Abstract
CD43 (leukosialin) is a transmembrane glycoprotein expressed in a variety of hematopoietic cells, including B lymphocytes, and a variety of malignancies including lymphoma, leukemia, and solid tumors. CD43 plays an important role in the development of many diseases, and coexpression of CD43 and CD20 on peripheral B cells is a predictive factor of hematopoietic malignancy. Although CD43 is expressed in approximately 25% of diffuse large B-cell lymphomas (DLBCLs), its prognostic significance remains unclear. To analyze CD43 expression in DLBCL, not otherwise specified (DLBCL, NOS), and assess its prognostic value, we analyzed clinical data from 160 patients with DLBCL, NOS. We observed that CD43 expression was detected in 47 (29.4%) of 160 cases. CD43 expression was positively correlated with old age (>60 years), high serum lactate dehydrogenase level, B symptoms, non-germinal center type, and DLBCL, NOS, mortality. Patients with CD43-positive DLBCL, NOS, had poorer overall survival (P < .001, log-rank test) and event-free survival (P < .001, log-rank test) than CD43-negative patients. Univariate analysis showed that CD43 expression, age, sex, Ann Arbor stage, International Prognostic Index category, and germinal center phenotype were prognostic factors for DLBCL, NOS, patient survival. Multivariate analysis showed that CD43 expression was an independent significant prognostic factor for event-free survival (P < .001) and overall survival (P < .001). Based on these data, we conclude that CD43 expression is a novel adverse prognostic factor for patients with DLBCL, NOS.
Collapse
|
13
|
MiR-7 promotes epithelial cell transformation by targeting the tumor suppressor KLF4. PLoS One 2014; 9:e103987. [PMID: 25181544 PMCID: PMC4151986 DOI: 10.1371/journal.pone.0103987] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 07/06/2014] [Indexed: 12/30/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous small non-coding RNAs that have a pivotal role in the post-transcriptional regulation of gene expression and their misregulation is common in different types of cancer. Although it has been shown that miR-7 plays an oncogenic role in different cellular contexts, the molecular mechanisms by which miR-7 promotes cell transformation are not well understood. Here we show that the transcription factor KLF4 is a direct target of miR-7 and present experimental evidence indicating that the regulation of KLF4 by miR-7 has functional implications in epithelial cell transformation. Stable overexpression of miR-7 into lung and skin epithelial cells enhanced cell proliferation, cell migration and tumor formation. Alteration of these cellular functions by miR-7 resulted from misregulation of KLF4 target genes involved in cell cycle control. miR-7-induced tumors showed decreased p21 and increased Cyclin D levels. Taken together, these findings indicate that miR-7 acts as an oncomiR in epithelial cells in part by directly regulating KLF4 expression. Thus, we conclude that miR-7 acts as an oncomiR in the epithelial cellular context, where through the negative regulation of KLF4-dependent signaling pathways, miR-7 promotes cellular transformation and tumor growth.
Collapse
|