1
|
Yee EJ, Vigil I, Sun Y, Torphy RJ, Schulick RD, Zhu Y. Group XIV C-type lectins: emerging targets in tumor angiogenesis. Angiogenesis 2024; 27:173-192. [PMID: 38468017 PMCID: PMC11021320 DOI: 10.1007/s10456-024-09907-x] [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: 10/25/2023] [Accepted: 01/23/2024] [Indexed: 03/13/2024]
Abstract
C-type lectins, distinguished by a C-type lectin binding domain (CTLD), are an evolutionarily conserved superfamily of glycoproteins that are implicated in a broad range of physiologic processes. The group XIV subfamily of CTLDs are comprised of CD93, CD248/endosialin, CLEC14a, and thrombomodulin/CD141, and have important roles in creating and maintaining blood vessels, organizing extracellular matrix, and balancing pro- and anti-coagulative processes. As such, dysregulation in the expression and downstream signaling pathways of these proteins often lead to clinically relevant pathology. Recently, group XIV CTLDs have been shown to play significant roles in cancer progression, namely tumor angiogenesis and metastatic dissemination. Interest in therapeutically targeting tumor vasculature is increasing and the search for novel angiogenic targets is ongoing. Group XIV CTLDs have emerged as key moderators of tumor angiogenesis and metastasis, thus offering substantial therapeutic promise for the clinic. Herein, we review our current knowledge of group XIV CTLDs, discuss each's role in malignancy and associated potential therapeutic avenues, briefly discuss group XIV CTLDs in the context of two other relevant lectin families, and offer future direction in further elucidating mechanisms by which these proteins function and facilitate tumor growth.
Collapse
Affiliation(s)
- Elliott J Yee
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA
| | - Isaac Vigil
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA
| | - Yi Sun
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA
| | - Robert J Torphy
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Richard D Schulick
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA.
| |
Collapse
|
2
|
Xu Y, Sun Y, Zhu Y, Song G. Structural insight into CD93 recognition by IGFBP7. Structure 2024; 32:282-291.e4. [PMID: 38218180 DOI: 10.1016/j.str.2023.12.011] [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: 07/18/2023] [Revised: 11/28/2023] [Accepted: 12/19/2023] [Indexed: 01/15/2024]
Abstract
The CD93/IGFBP7 axis proteins are key factors expressed in endothelial cells (EC) that mediate EC angiogenesis and migration. Their upregulation contributes to tumor vascular abnormality and a blockade of this interaction promotes a favorable tumor microenvironment for therapeutic interventions. However, the interactions of these proteins with each other remain unclear. In this study, we determined a partial structure of the human CD93-IGFBP7 complex comprising the EGF1 domain of CD93 and the IB domain of IGFBP7. Mutagenesis studies confirmed interactions and specificities. Cellular and mouse tumor studies demonstrated the physiological relevance of the CD93-IGFBP7 interaction in EC angiogenesis. Our study provides leads for the development of therapeutic agents to precisely disrupt unwanted CD93-IGFBP7 signaling in the tumor microenvironment. Additionally, analysis of the CD93 full-length architecture provides insights into how CD93 protrudes on the cell surface and forms a flexible platform for binding to IGFBP7 and other ligands.
Collapse
Affiliation(s)
- Yueming Xu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yi Sun
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Gaojie Song
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
3
|
Qu J, Lin L, Fu G, Zheng M, Geng J, Sun X, Xing L. The analysis of multiple omics and examination of pathological images revealed the prognostic and therapeutic significances of CD93 in lung squamous cell carcinoma. Life Sci 2024; 339:122422. [PMID: 38224815 DOI: 10.1016/j.lfs.2024.122422] [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: 08/14/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/17/2024]
Abstract
As a potent pro-angiogenic factor, the role of CD93 in the prognosis and therapeutic outcomes of lung squamous cell carcinoma (LUSC) merits exploration. In this study, we systematically collected transcriptomic, genomic, and clinical data from various public databases, as well as pathological images from hospital-operated patients. Employing statistical analysis software like R (Version 4.2.2) and GraphPad (Version 8.0), we conducted comprehensive analyses of multi-omics data. The results revealed elevated CD93 expression in LUSC tissues, closely associated with various cancer-related pathways. High CD93 expression indicated advanced clinical stage and poorer prognosis. Furthermore, CD93 contributed to resistance against chemotherapy and immunotherapy by enhancing tumor cell stemness, reducing immune cell infiltration, and inducing T cell exhaustion. Patients with low CD93 expression exhibited higher response rates to both chemotherapy and immunotherapy. Immunohistochemistry validated the significance of CD93 in LUSC. CD93 emerges as a biomarker signaling unfavorable prognosis and influencing therapeutic outcomes, suggesting a potential LUSC treatment avenue.
Collapse
Affiliation(s)
- Jialin Qu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan 250117, Shandong, China
| | - Li Lin
- Department of Respiratory Medicine, Shandong Provincial Chest Hospital, Shandong Public Health Clinical Center, Jinan 250117, Shandong, China
| | - Guangming Fu
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Mei Zheng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan 250117, Shandong, China
| | - Jiaxiao Geng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan 250117, Shandong, China
| | - Xiaorong Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China.
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan 250117, Shandong, China.
| |
Collapse
|
4
|
Xu Y, Sun Y, Zhu Y, Song G. Structural insight into CD93 recognition by IGFBP7. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.543655. [PMID: 37333140 PMCID: PMC10274810 DOI: 10.1101/2023.06.07.543655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The CD93/IGFBP7 axis are key factors expressed in endothelial cells (EC) that mediate EC angiogenesis and migration. Upregulation of them contributes to tumor vascular abnormality and blockade of this interaction promotes a favorable tumor microenvironment for therapeutic interventions. However, how these two proteins associated to each other remains unclear. In this study, we solved the human CD93-IGFBP7 complex structure to elucidate the interaction between the EGF 1 domain of CD93 and the IB domain of IGFBP7. Mutagenesis studies confirmed the binding interactions and specificities. Cellular and mouse tumor studies demonstrated the physiological relevance of the CD93-IGFBP7 interaction in EC angiogenesis. Our study provides hints for development of therapeutic agents to precisely disrupt unwanted CD93-IGFBP7 signaling in the tumor microenvironment. Additionally, analysis of the CD93 full-length architecture provides insights into how CD93 protrudes on the cell surface and forms a flexible platform for binding to IGFBP7 and other ligands.
Collapse
|
5
|
Ma K, Chen S, Chen X, Zhao X, Yang J. CD93 is Associated with Glioma-related Malignant Processes and Immunosuppressive Cell Infiltration as an Inspiring Biomarker of Survivance. J Mol Neurosci 2022; 72:2106-2124. [PMID: 36006582 DOI: 10.1007/s12031-022-02060-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022]
Abstract
Previous reports have confirmed the significance of CD93 in the progression of multiple tumors; however, there are few studies examining its immune properties for gliomas. Here, we methodically investigated the pathophysiological characteristics and clinical manifestations of gliomas. Six hundred ninety-nine glioma patients in TCGA along with 325 glioma patients in CGGA were correspondingly collected for training and validating. We analyzed and visualized total statistics using RStudio. One-way ANOVA and Student's t-test were used to assess groups' differences. All differences were considered statistically significant at the level of P < 0.05. CD93 markedly upregulated among HGG, MGMT promoter unmethylated subforms, IDH wild forms, 1p19q non-codeletion subforms, and mesenchyme type gliomas. ROC analysis illustrated the favorable applicability of CD93 in estimating mesenchyme subform. Kaplan-Meier curves together with multivariable Cox analyses upon survivance identified high-expression CD93 as a distinct prognostic variable for glioma patients. GO analysis of CD93 documented its predominant part in glioma-related immunobiological processes and inflammation responses. We examined the associations of CD93 with immune-related meta-genes, and CD93 positively correlated with HCK, LCK, MHC I, MHC II, STAT1 and IFN, while adverse with IgG. Association analyses between CD93 and gliomas-infiltrating immunocytes indicated that the infiltrating degrees of most immunocytes exhibited positive correlations with CD93, particularly these immunosuppressive subsets such as TAM, Treg, and MDSCs. CD93 is markedly associated with adverse pathology types, unfavorable survival, and immunosuppressive immunocytes infiltration among gliomas, thus identifying CD93 as a practicable marker and a promising target for glioma-based precise diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
- Kaiming Ma
- Department of Neurosurgery, Peking University Third Hospital, Haidian District, 49 North Garden Rd, Beijing, 100191, China
| | - Suhua Chen
- Department of Neurosurgery, Peking University Third Hospital, Haidian District, 49 North Garden Rd, Beijing, 100191, China
| | - Xin Chen
- Department of Neurosurgery, Peking University Third Hospital, Haidian District, 49 North Garden Rd, Beijing, 100191, China.,Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Xiaofang Zhao
- Department of Neurosurgery, Peking University Third Hospital, Haidian District, 49 North Garden Rd, Beijing, 100191, China
| | - Jun Yang
- Department of Neurosurgery, Peking University Third Hospital, Haidian District, 49 North Garden Rd, Beijing, 100191, China. .,Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China.
| |
Collapse
|
6
|
Rossi G, Giger S, Hübscher T, Lutolf MP. Gastruloids as in vitro models of embryonic blood development with spatial and temporal resolution. Sci Rep 2022; 12:13380. [PMID: 35927563 PMCID: PMC9352713 DOI: 10.1038/s41598-022-17265-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/22/2022] [Indexed: 01/01/2023] Open
Abstract
Gastruloids are three-dimensional embryonic organoids that reproduce key features of early mammalian development in vitro with unique scalability, accessibility, and spatiotemporal similarity to real embryos. Recently, we adapted the gastruloid culture conditions to promote cardiovascular development. In this work, we extended these conditions to capture features of embryonic blood development through a combination of immunophenotyping, detailed transcriptomics analysis, and identification of blood stem/progenitor cell potency. We uncovered the emergence of blood progenitor and erythroid-like cell populations in late gastruloids and showed the multipotent clonogenic capacity of these cells, both in vitro and after transplantation into irradiated mice. We also identified the spatial localization near a vessel-like plexus in the anterior portion of gastruloids with similarities to the emergence of blood stem cells in the mouse embryo. These results highlight the potential and applicability of gastruloids to the in vitro study of complex processes in embryonic blood development with spatiotemporal fidelity.
Collapse
Affiliation(s)
- Giuliana Rossi
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Vaud, 1015, Lausanne, Switzerland. .,Roche Institute for Translational Bioengineering (ITB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland.
| | - Sonja Giger
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Vaud, 1015, Lausanne, Switzerland
| | - Tania Hübscher
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Vaud, 1015, Lausanne, Switzerland
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Vaud, 1015, Lausanne, Switzerland. .,Institute of Chemical Sciences and Engineering, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Vaud, 1015, Lausanne, Switzerland. .,Roche Institute for Translational Bioengineering (ITB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland.
| |
Collapse
|
7
|
Riether C, Radpour R, Kallen NM, Bürgin DT, Bachmann C, Schürch CM, Lüthi U, Arambasic M, Hoppe S, Albers CE, Baerlocher GM, Ochsenbein AF. Metoclopramide treatment blocks CD93-signaling-mediated self-renewal of chronic myeloid leukemia stem cells. Cell Rep 2021; 34:108663. [PMID: 33503440 DOI: 10.1016/j.celrep.2020.108663] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 11/20/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
Abstract
Self-renewal is a key characteristic of leukemia stem cells (LSCs) responsible for the development and maintenance of leukemia. In this study, we identify CD93 as an important regulator of self-renewal and proliferation of murine and human LSCs, but not hematopoietic stem cells (HSCs). The intracellular domain of CD93 promotes gene transcription via the transcriptional regulator SCY1-like pseudokinase 1 independently of ligation of the extracellular domain. In a drug library screen, we identify the anti-emetic agent metoclopramide as an efficient blocker of CD93 signaling. Metoclopramide treatment reduces murine and human LSCs in vitro and prolongs survival of chronic myeloid leukemia (CML) mice through downregulation of pathways related to stemness and proliferation in LSCs. Overall, these results identify CD93 signaling as an LSC-specific regulator of self-renewal and proliferation and a targetable pathway to eliminate LSCs in CML.
Collapse
Affiliation(s)
- Carsten Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Ramin Radpour
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Nils M Kallen
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Damian T Bürgin
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Chantal Bachmann
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland; Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Christian M Schürch
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ursina Lüthi
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Miroslav Arambasic
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sven Hoppe
- Wirbelsäulenmedizin Bern, Hirslanden Salem-Spital, Bern, Switzerland; Department of Orthopedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Christoph E Albers
- Department of Orthopedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gabriela M Baerlocher
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland; Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Adrian F Ochsenbein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| |
Collapse
|
8
|
Chen MJ, Lummertz da Rocha E, Cahan P, Kubaczka C, Hunter P, Sousa P, Mullin NK, Fujiwara Y, Nguyen M, Tan Y, Landry S, Han A, Yang S, Lu YF, Jha DK, Vo LT, Zhou Y, North TE, Zon LI, Daley GQ, Schlaeger TM. Transcriptome Dynamics of Hematopoietic Stem Cell Formation Revealed Using a Combinatorial Runx1 and Ly6a Reporter System. Stem Cell Reports 2020; 14:956-971. [PMID: 32302558 PMCID: PMC7220988 DOI: 10.1016/j.stemcr.2020.03.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 01/01/2023] Open
Abstract
Studies of hematopoietic stem cell (HSC) development from pre-HSC-producing hemogenic endothelial cells (HECs) are hampered by the rarity of these cells and the presence of other cell types with overlapping marker expression profiles. We generated a Tg(Runx1-mKO2; Ly6a-GFP) dual reporter mouse to visualize hematopoietic commitment and study pre-HSC emergence and maturation. Runx1-mKO2 marked all intra-arterial HECs and hematopoietic cluster cells (HCCs), including pre-HSCs, myeloid- and lymphoid progenitors, and HSCs themselves. However, HSC and lymphoid potential were almost exclusively found in reporter double-positive (DP) cells. Robust HSC activity was first detected in DP cells of the placenta, reflecting the importance of this niche for (pre-)HSC maturation and expansion before the fetal liver stage. A time course analysis by single-cell RNA sequencing revealed that as pre-HSCs mature into fetal liver stage HSCs, they show signs of interferon exposure, exhibit signatures of multi-lineage differentiation gene expression, and develop a prolonged cell cycle reminiscent of quiescent adult HSCs.
Collapse
Affiliation(s)
- Michael J Chen
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| | - Edroaldo Lummertz da Rocha
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Caroline Kubaczka
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Phoebe Hunter
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Patricia Sousa
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nathaniel K Mullin
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yuko Fujiwara
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Minh Nguyen
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Yuqi Tan
- Department of Biomedical Engineering, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Samuel Landry
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Areum Han
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Song Yang
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Yi-Fen Lu
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Deepak Kumar Jha
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Linda T Vo
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Yi Zhou
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Trista E North
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Boston, MA, USA
| | - Leonard I Zon
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Boston, MA, USA; Howard Hughes Medical Institute, Harvard University, Boston, MA, USA; Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - George Q Daley
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Boston, MA, USA
| | - Thorsten M Schlaeger
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Boston, MA, USA.
| |
Collapse
|
9
|
Liang Q, Su L, Zhang D, Jiao J. CD93 negatively regulates astrogenesis in response to MMRN2 through the transcriptional repressor ZFP503 in the developing brain. Proc Natl Acad Sci U S A 2020; 117:9413-9422. [PMID: 32291340 PMCID: PMC7196765 DOI: 10.1073/pnas.1922713117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Astrogenesis is repressed in the early embryonic period and occurs in the late embryonic period. A variety of external and internal signals contribute to the sequential differentiation of neural stem cells. Here, we discovered that immune-related CD93 plays a critical negative role in the regulation of astrogenesis in the mouse cerebral cortex. We show that CD93 expression is detected in neural stem cells and neurons but not in astrocytes and declines as differentiation proceeds. Cd93 knockout increases astrogenesis at the expense of neuron production during the late embryonic period. CD93 responds to the extracellular matrix protein Multimerin 2 (MMRN2) to trigger the repression of astrogenesis. Mechanistically, CD93 delivers signals to β-Catenin through a series of phosphorylation cascades, and then β-Catenin transduces these signals to the nucleus to activate Zfp503 transcription. The transcriptional repressor ZFP503 inhibits the transcription of glial fibrillary acidic protein (Gfap) by binding to the Gfap promoter with the assistance of Grg5. Furthermore, Cd93 knockout mice exhibit autism-like behaviors. Taken together, our results reveal that CD93 is a negative regulator of the onset of astrogenesis and provide insight into therapy for psychiatric disorders.
Collapse
Affiliation(s)
- Qingli Liang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Medical School, University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Libo Su
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Medical School, University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Dongming Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Medical School, University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China;
- Medical School, University of Chinese Academy of Sciences, 100049 Beijing, China
- Co-Innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
- Innovation Academy for Stem Cell and Regeneration, 100101 Beijing, China
| |
Collapse
|
10
|
Borah S, Vasudevan D, Swain RK. C-type lectin family XIV members and angiogenesis. Oncol Lett 2019; 18:3954-3962. [PMID: 31579078 DOI: 10.3892/ol.2019.10760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022] Open
Abstract
The growth and metastasis of tumors is dependent on angiogenesis. C-type lectins are carbohydrate-binding proteins with a diverse range of functions. The C-type lectin family XIV members are transmembrane glycoproteins, and all four members of this family have been reported to regulate angiogenesis, although the detailed mechanism of action has yet to be completely elucidated. They interact with extracellular matrix proteins and mediate cell-cell adhesion by their lectin-like domain. The aim of the present study was to summarize the available information on the function and mechanism of C-type lectin family XIV in angiogenesis and discuss their potential as targets for cancer therapy.
Collapse
Affiliation(s)
- Supriya Borah
- Institute of Life Sciences, Bhubaneswar, Odisha 751023, India.,Department of Biotechnology, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | | | - Rajeeb K Swain
- Institute of Life Sciences, Bhubaneswar, Odisha 751023, India
| |
Collapse
|
11
|
Khan KA, McMurray JL, Mohammed F, Bicknell R. C-type lectin domain group 14 proteins in vascular biology, cancer and inflammation. FEBS J 2019; 286:3299-3332. [PMID: 31287944 PMCID: PMC6852297 DOI: 10.1111/febs.14985] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/21/2019] [Accepted: 07/05/2019] [Indexed: 02/06/2023]
Abstract
The C‐type lectin domain (CTLD) group 14 family of transmembrane glycoproteins consist of thrombomodulin, CD93, CLEC14A and CD248 (endosialin or tumour endothelial marker‐1). These cell surface proteins exhibit similar ectodomain architecture and yet mediate a diverse range of cellular functions, including but not restricted to angiogenesis, inflammation and cell adhesion. Thrombomodulin, CD93 and CLEC14A can be expressed by endothelial cells, whereas CD248 is expressed by vasculature associated pericytes, activated fibroblasts and tumour cells among other cell types. In this article, we review the current literature of these family members including their expression profiles, interacting partners, as well as established and speculated functions. We focus primarily on their roles in the vasculature and inflammation as well as their contributions to tumour immunology. The CTLD group 14 family shares several characteristic features including their ability to be proteolytically cleaved and engagement of some shared extracellular matrix ligands. Each family member has strong links to tumour development and in particular CD93, CLEC14A and CD248 have been proposed as attractive candidate targets for cancer therapy.
Collapse
Affiliation(s)
- Kabir A Khan
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Canada
| | - Jack L McMurray
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, UK
| | - Fiyaz Mohammed
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, UK
| | - Roy Bicknell
- Institutes of Cardiovascular Sciences and Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, UK
| |
Collapse
|
12
|
Nativel B, Ramin-Mangata S, Mevizou R, Figuester A, Andries J, Iwema T, Ikewaki N, Gasque P, Viranaïcken W. CD93 is a cell surface lectin receptor involved in the control of the inflammatory response stimulated by exogenous DNA. Immunology 2019; 158:85-93. [PMID: 31335975 DOI: 10.1111/imm.13100] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 01/08/2023] Open
Abstract
Bacterial DNA contains CpG oligonucleotide (ODN) motifs to trigger innate immune responses through the endosomal receptor Toll-like receptor 9 (TLR9). One of the cell surface receptors to capture and deliver microbial DNA to intracellular TLR9 is the C-type lectin molecule DEC-205 through its N-terminal C-type lectin-like domain (CTLD). CD93 is a cell surface protein and member of the lectin group XIV with a CTLD. We hypothesized that CD93 could interact with CpG motifs, and possibly serve as a novel receptor to deliver bacterial DNA to endosomal TLR9. Using ELISA and tryptophan fluorescence binding studies we observed that the soluble histidine-tagged CD93-CTLD was specifically binding to CpG ODN and bacterial DNA. Moreover, we found that CpG ODN could bind to CD93-expressing IMR32 neuroblastoma cells and induced more robust interleukin-6 secretion when compared with mock-transfected IMR32 control cells. Our data argue for a possible contribution of CD93 to control cell responsiveness to bacterial DNA in a manner reminiscent of DEC-205. We postulate that CD93 may act as a receptor at plasma membrane for DNA or CpG ODN and to grant delivery to endosomal TLR9.
Collapse
Affiliation(s)
- Brice Nativel
- GRI, Groupe de recherche en immunopathologie, EA4517, Université de la Réunion, Saint-Denis, France.,Université de La Réunion, INSERM 1188, Diabète athérothombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Stéphane Ramin-Mangata
- GRI, Groupe de recherche en immunopathologie, EA4517, Université de la Réunion, Saint-Denis, France.,Université de La Réunion, INSERM 1188, Diabète athérothombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Rudy Mevizou
- GRI, Groupe de recherche en immunopathologie, EA4517, Université de la Réunion, Saint-Denis, France
| | - Audrey Figuester
- GRI, Groupe de recherche en immunopathologie, EA4517, Université de la Réunion, Saint-Denis, France
| | - Jessica Andries
- GRI, Groupe de recherche en immunopathologie, EA4517, Université de la Réunion, Saint-Denis, France
| | - Thomas Iwema
- GRI, Groupe de recherche en immunopathologie, EA4517, Université de la Réunion, Saint-Denis, France
| | - Nobunao Ikewaki
- Laboratories of Clinical Immunology, Department of Animal Pharmaceutical Science, Welfare School of Pharmaceutical Sciences, Kyushu University of Health, Miyazaki, Japan
| | - Philippe Gasque
- GRI, Groupe de recherche en immunopathologie, EA4517, Université de la Réunion, Saint-Denis, France.,Université de La Réunion, INSERM 1187, CNRS, 9192, IRD 249, UM 134 Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Saint-Denis de La Réunion, France.,Laboratoire d'Immunologie Clinique et Expérimentale, ZOI (LICE-OI). CHU site Bellepierre, Saint-Denis de La Réunion, France
| | - Wildriss Viranaïcken
- GRI, Groupe de recherche en immunopathologie, EA4517, Université de la Réunion, Saint-Denis, France.,Université de La Réunion, INSERM 1187, CNRS, 9192, IRD 249, UM 134 Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Saint-Denis de La Réunion, France
| |
Collapse
|
13
|
Griffiths MR, Botto M, Morgan BP, Neal JW, Gasque P. CD93 regulates central nervous system inflammation in two mouse models of autoimmune encephalomyelitis. Immunology 2018; 155:346-355. [PMID: 29923617 DOI: 10.1111/imm.12974] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/23/2018] [Accepted: 06/13/2018] [Indexed: 01/10/2023] Open
Abstract
Microglia and non-professional immune cells (endothelial cells, neurons) participate in the recognition and removal of pathogens and tissue debris in the injured central nervous system through major pro-inflammatory processes. However, the mechanisms involved in regulating these responses remain ill-characterized. We herein show that CD93, also known as complement C1qRp/AA4 stem cell marker, has an important role in the regulation of inflammatory processes. The role of CD93 was evaluated in two models of neuroinflammation. We used the MOG-experimental autoimmune encephalomyelitis (EAE) model and the antibody-dependent EAE (ADEAE), which were induced in wild-type and CD93 knockout mice. We found that CD93 was highly expressed by neurons, endothelial cells and microglia (ramified >> amoeboid). Astrocytes and oligodendrocytes did not to express CD93. We further observed that CD93-deficient (CD93-/- ) mice presented a more robust brain and spinal cord inflammation in EAE and ADEAE. Encephalitis in CD93-/- was characterized by increased numbers of infiltrating M1 macrophages (CD11c+ CD206- ) and amoeboid microglia exhibiting a more activated phenotype (Tomato Lectinhigh Cox2high ). Damage to and leakage through the blood-brain barrier was increased in CD93-/- animals and was associated with a more robust neuronal injury when compared with wild-type EAE mice. We propose that CD93 is an important neuro-immune regulator to control central nervous system inflammation.
Collapse
Affiliation(s)
- Mark R Griffiths
- BIIG, Brain Inflammation and Immunity Group, Cardiff University School of Medicine, Cardiff, UK
| | - Marina Botto
- Centre for Complement and Inflammation Research, Department of Medicine, Imperial College, London, UK
| | - Bryan Paul Morgan
- Complement Biology Group, Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - James W Neal
- Neuropathology Department, Cardiff University School of Medicine, Cardiff, UK
| | - Philippe Gasque
- BIIG, Brain Inflammation and Immunity Group, Cardiff University School of Medicine, Cardiff, UK.,GRI EA4517, Immunopathology and infectious disease grouping, CHU, CYROI, Université de La Réunion, Sainte-Clotilde, La Réunion, France.,CNRS 9192, INSERM U1187, IRD 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Plateforme Technologique CYROI, Université de La Réunion, Sainte-Clotilde, La Réunion, France.,Laboratoire de Biologie, secteur : Laboratoire d'immunologie clinique et expérimentale ZOI (LICE OI), CHU La Réunion site Félix Guyon, St Denis, La Réunion, France
| |
Collapse
|
14
|
Induction of hemangiosarcoma in mice after chronic treatment with S1P-modulator siponimod and its lack of relevance to rat and human. Arch Toxicol 2018; 92:1877-1891. [PMID: 29556671 PMCID: PMC5962627 DOI: 10.1007/s00204-018-2189-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/13/2018] [Indexed: 11/30/2022]
Abstract
A high incidence of hemangiosarcoma (HSA) was observed in mice treated for 2 years with siponimod, a sphingosine-1-phosphate receptor 1 (S1P1) functional antagonist, while no such tumors were observed in rats under the same treatment conditions. In 3-month rat (90 mg/kg/day) and 9-month mouse (25 and 75 mg/kg/day) in vivo mechanistic studies, vascular endothelial cell (VEC) activation was observed in both species, but VEC proliferation and persistent increases in circulating placental growth factor 2 (PLGF2) were only seen in the mouse. In mice, these effects were sustained over the 9-month study duration, while in rats increased mitotic gene expression was present at day 3 only and PLGF2 was induced only during the first week of treatment. In the mouse, the persistent VEC activation, mitosis induction, and PLGF2 stimulation likely led to sustained neo-angiogenesis which over life-long treatment may result in HSA formation. In rats, despite sustained VEC activation, the transient mitotic and PLGF2 stimuli did not result in the formation of HSA. In vitro, the mouse and rat primary endothelial cell cultures mirrored their respective in vivo findings for cell proliferation and PLGF2 release. Human VECs, like rat cells, were unresponsive to siponimod treatment with no proliferative response and no release of PLGF2 at all tested concentrations. Hence, it is suggested that the human cells also reproduce a lack of in vivo response to siponimod. In conclusion, the molecular mechanisms leading to siponimod-induced HSA in mice are considered species specific and likely irrelevant to humans.
Collapse
|
15
|
Multimerin-2 is a ligand for group 14 family C-type lectins CLEC14A, CD93 and CD248 spanning the endothelial pericyte interface. Oncogene 2017; 36:6097-6108. [PMID: 28671670 PMCID: PMC5671938 DOI: 10.1038/onc.2017.214] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 04/06/2017] [Accepted: 04/14/2017] [Indexed: 12/27/2022]
Abstract
The C-type lectin domain containing group 14 family members CLEC14A and CD93 are proteins expressed by endothelium and are implicated in tumour angiogenesis. CD248 (alternatively known as endosialin or tumour endothelial marker-1) is also a member of this family and is expressed by tumour-associated fibroblasts and pericytes. Multimerin-2 (MMRN2) is a unique endothelial specific extracellular matrix protein that has been implicated in angiogenesis and tumour progression. We show that the group 14 C-type lectins CLEC14A, CD93 and CD248 directly bind to MMRN2 and only thrombomodulin of the family does not. Binding to MMRN2 is dependent on a predicted long-loop region in the C-type lectin domain and is abrogated by mutation within the domain. CLEC14A and CD93 bind to the same non-glycosylated coiled-coil region of MMRN2, but the binding of CD248 occurs on a distinct non-competing region. CLEC14A and CD248 can bind MMRN2 simultaneously and this occurs at the interface between endothelium and pericytes in human pancreatic cancer. A recombinant peptide of MMRN2 spanning the CLEC14A and CD93 binding region blocks CLEC14A extracellular domain binding to the endothelial cell surface as well as increasing adherence of human umbilical vein endothelial cells to the active peptide. This MMRN2 peptide is anti-angiogenic in vitro and reduces tumour growth in mouse models. These findings identify novel protein interactions involving CLEC14A, CD93 and CD248 with MMRN2 as targetable components of vessel formation.
Collapse
|
16
|
Talos F, Mitrofanova A, Bergren SK, Califano A, Shen MM. A computational systems approach identifies synergistic specification genes that facilitate lineage conversion to prostate tissue. Nat Commun 2017; 8:14662. [PMID: 28429718 PMCID: PMC5413950 DOI: 10.1038/ncomms14662] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 01/21/2017] [Indexed: 12/19/2022] Open
Abstract
To date, reprogramming strategies for generating cell types of interest have been facilitated by detailed understanding of relevant developmental regulatory factors. However, identification of such regulatory drivers often represents a major challenge, as specific gene combinations may be required for reprogramming. Here we show that a computational systems approach can identify cell type specification genes (master regulators) that act synergistically, and demonstrate its application for reprogramming of fibroblasts to prostate tissue. We use three such master regulators (FOXA1, NKX3.1 and androgen receptor, AR) in a primed conversion strategy starting from mouse fibroblasts, resulting in prostate tissue grafts with appropriate histological and molecular properties that respond to androgen-deprivation. Moreover, generation of reprogrammed prostate does not require traversal of a pluripotent state. Thus, we describe a general strategy by which cell types and tissues can be generated even with limited knowledge of the developmental pathways required for their specification in vivo.
Collapse
Affiliation(s)
- Flaminia Talos
- Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Urology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| | - Antonina Mitrofanova
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| | - Sarah K Bergren
- Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Urology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| | - Andrea Califano
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| | - Michael M Shen
- Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Urology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| |
Collapse
|
17
|
Galvagni F, Nardi F, Maida M, Bernardini G, Vannuccini S, Petraglia F, Santucci A, Orlandini M. CD93 and dystroglycan cooperation in human endothelial cell adhesion and migration adhesion and migration. Oncotarget 2017; 7:10090-103. [PMID: 26848865 PMCID: PMC4891106 DOI: 10.18632/oncotarget.7136] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 01/22/2016] [Indexed: 02/07/2023] Open
Abstract
CD93 is a transmembrane glycoprotein predominantly expressed in endothelial cells. Although CD93 displays proangiogenic activity, its molecular function in angiogenesis still needs to be clarified. To get molecular insight into the biological role of CD93 in the endothelium, we performed proteomic analyses to examine changes in the protein profile of endothelial cells after CD93 silencing. Among differentially expressed proteins, we identified dystroglycan, a laminin-binding protein involved in angiogenesis, whose expression is increased in vascular endothelial cells within malignant tumors. Using immunofluorescence, FRET, and proximity ligation analyses, we observed a close interaction between CD93 and β-dystroglycan. Moreover, silencing experiments showed that CD93 and dystroglycan promoted endothelial cell migration and organization into capillary-like structures. CD93 proved to be phosphorylated on tyrosine 628 and 644 following cell adhesion on laminin through dystroglycan. This phosphorylation was shown to be necessary for a proper endothelial migratory phenotype. Moreover, we showed that during cell spreading phosphorylated CD93 recruited the signaling protein Cbl, which in turn was phosphorylated on tyrosine 774. Altogether, our results identify a new signaling pathway which is activated by the cooperation between CD93 and dystroglycan and involved in the control of endothelial cell function.
Collapse
Affiliation(s)
- Federico Galvagni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Federica Nardi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Marco Maida
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Giulia Bernardini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Silvia Vannuccini
- Department of Molecular and Developmental Medicine, Obstetrics and Gynecology, University of Siena, 53100 Siena, Italy
| | - Felice Petraglia
- Department of Molecular and Developmental Medicine, Obstetrics and Gynecology, University of Siena, 53100 Siena, Italy
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Maurizio Orlandini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| |
Collapse
|
18
|
Tosi GM, Caldi E, Parolini B, Toti P, Neri G, Nardi F, Traversi C, Cevenini G, Marigliani D, Nuti E, Bacci T, Galvagni F, Orlandini M. CD93 as a Potential Target in Neovascular Age-Related Macular Degeneration. J Cell Physiol 2016; 232:1767-1773. [DOI: 10.1002/jcp.25689] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/11/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Gian Marco Tosi
- Ophthalmology Unit of the Department of Medicine, Surgery and Neuroscience; University of Siena; Siena Italy
| | - Elena Caldi
- Department of Biotechnology, Chemistry and Pharmacy; University of Siena; Siena Italy
| | | | - Paolo Toti
- Department of Medical Biotechnologies; University of Siena; Siena Italy
| | - Giovanni Neri
- Ophthalmology Unit of the Department of Medicine, Surgery and Neuroscience; University of Siena; Siena Italy
| | - Federica Nardi
- Department of Biotechnology, Chemistry and Pharmacy; University of Siena; Siena Italy
| | - Claudio Traversi
- Ophthalmology Unit of the Department of Medicine, Surgery and Neuroscience; University of Siena; Siena Italy
| | - Gabriele Cevenini
- Department of Medical Biotechnologies; University of Siena; Siena Italy
| | - Davide Marigliani
- Ophthalmology Unit of the Department of Medicine, Surgery and Neuroscience; University of Siena; Siena Italy
| | - Elisabetta Nuti
- Ophthalmology Unit of the Department of Medicine, Surgery and Neuroscience; University of Siena; Siena Italy
| | - Tommaso Bacci
- Ophthalmology Unit of the Department of Medicine, Surgery and Neuroscience; University of Siena; Siena Italy
| | - Federico Galvagni
- Department of Biotechnology, Chemistry and Pharmacy; University of Siena; Siena Italy
| | - Maurizio Orlandini
- Department of Biotechnology, Chemistry and Pharmacy; University of Siena; Siena Italy
| |
Collapse
|
19
|
Elevated expression of CD93 promotes angiogenesis and tumor growth in nasopharyngeal carcinoma. Biochem Biophys Res Commun 2016; 476:467-474. [PMID: 27255994 DOI: 10.1016/j.bbrc.2016.05.146] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 05/27/2016] [Indexed: 01/08/2023]
Abstract
CD93, also known as the complement component C1q receptor (C1qRp), has been reported to promote the progression of some cancer types. However, the expression and physiological significance of CD93 in nasopharyngeal carcinoma (NPC) remain largely elusive. In this study, we first examined the expression of CD93 in NPC and experimentally manipulated its expression. We observed that vascular CD93 expression is elevated in NPC and is correlated with T classification, N classification, distant metastasis, clinical stage and poor prognosis (all P < 0.05). In addition, overexpression of CD93 promoted angiogenesis in vitro. What's more, we found that CD93 was highly expressed in NPC tissues and cells, and the regulation of CD93 on cell proliferation was determined by cell counting kit (CCK)-8 assay and cell cycle analyses. Our findings provide unique insight into the pathogenesis of NPC and underscore the need to explore novel therapeutic targets such as CD93 to improve NPC treatment.
Collapse
|
20
|
Yu YH, Narayanan G, Sankaran S, Ramasamy S, Chan SY, Lin S, Chen J, Yang H, Srivats H, Ahmed S. Purification, Visualization, and Molecular Signature of Neural Stem Cells. Stem Cells Dev 2015; 25:189-201. [PMID: 26464067 PMCID: PMC4770853 DOI: 10.1089/scd.2015.0190] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Neural stem cells (NSCs) are isolated from primary brain tissue and propagated as a heterogeneous mix of cells, including neural progenitors. To date, NSCs have not been purified in vitro to allow study of their biology and utility in regenerative medicine. In this study, we identify C1qR1 as a novel marker for NSCs and show that it can be used along with Lewis-X (LeX) to yield a highly purified population of NSCs. Using time-lapse microscopy, we are able to follow NSCs forming neurospheres, allowing their visualization. Finally, using single-cell polymerase chain reaction (PCR), we determine the molecular signature of NSCs. The single-cell PCR data suggest that along with the Notch and Shh pathways, the Hippo pathway plays an important role in NSC activity.
Collapse
Affiliation(s)
- Yuan Hong Yu
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Gunaseelan Narayanan
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Shvetha Sankaran
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Srinivas Ramasamy
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Shi Yu Chan
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Shuping Lin
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Jinmiao Chen
- 2 Bioinformatics Laboratory , Singapore Immunology Network, Singapore, Singapore
| | - Henry Yang
- 2 Bioinformatics Laboratory , Singapore Immunology Network, Singapore, Singapore
| | - Hariharan Srivats
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Sohail Ahmed
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| |
Collapse
|
21
|
Iwasaki M, Liedtke M, Gentles AJ, Cleary ML. CD93 Marks a Non-Quiescent Human Leukemia Stem Cell Population and Is Required for Development of MLL-Rearranged Acute Myeloid Leukemia. Cell Stem Cell 2015; 17:412-21. [PMID: 26387756 DOI: 10.1016/j.stem.2015.08.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/28/2015] [Accepted: 08/10/2015] [Indexed: 12/16/2022]
Abstract
Leukemia stem cells (LSCs) are thought to share several properties with hematopoietic stem cells (HSCs), including cell-cycle quiescence and a capacity for self-renewal. These features are hypothesized to underlie leukemic initiation, progression, and relapse, and they also complicate efforts to eradicate leukemia through therapeutic targeting of LSCs without adverse effects on HSCs. Here, we show that acute myeloid leukemias (AMLs) with genomic rearrangements of the MLL gene contain a non-quiescent LSC population. Although human CD34(+)CD38(-) LSCs are generally highly quiescent, the C-type lectin CD93 is expressed on a subset of actively cycling, non-quiescent AML cells enriched for LSC activity. CD93 expression is functionally required for engraftment of primary human AML LSCs and leukemogenesis, and it regulates LSC self-renewal predominantly by silencing CDKN2B, a major tumor suppressor in AML. Thus, CD93 expression identifies a predominantly cycling, non-quiescent leukemia-initiating cell population in MLL-rearranged AML, providing opportunities for selective targeting and eradication of LSCs.
Collapse
Affiliation(s)
- Masayuki Iwasaki
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michaela Liedtke
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew J Gentles
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael L Cleary
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Langenkamp E, Zhang L, Lugano R, Huang H, Elhassan TEA, Georganaki M, Bazzar W, Lööf J, Trendelenburg G, Essand M, Pontén F, Smits A, Dimberg A. Elevated Expression of the C-Type Lectin CD93 in the Glioblastoma Vasculature Regulates Cytoskeletal Rearrangements That Enhance Vessel Function and Reduce Host Survival. Cancer Res 2015; 75:4504-16. [DOI: 10.1158/0008-5472.can-14-3636] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 07/26/2015] [Indexed: 11/16/2022]
|
23
|
McGrath KE, Frame JM, Fegan KH, Bowen JR, Conway SJ, Catherman SC, Kingsley PD, Koniski AD, Palis J. Distinct Sources of Hematopoietic Progenitors Emerge before HSCs and Provide Functional Blood Cells in the Mammalian Embryo. Cell Rep 2015; 11:1892-904. [PMID: 26095363 DOI: 10.1016/j.celrep.2015.05.036] [Citation(s) in RCA: 285] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/29/2015] [Accepted: 05/19/2015] [Indexed: 12/11/2022] Open
Abstract
Hematopoietic potential arises in mammalian embryos before adult-repopulating hematopoietic stem cells (HSCs). At embryonic day 9.5 (E9.5), we show the first murine definitive erythro-myeloid progenitors (EMPs) have an immunophenotype distinct from primitive hematopoietic progenitors, maturing megakaryocytes and macrophages, and rare B cell potential. EMPs emerge in the yolk sac with erythroid and broad myeloid, but not lymphoid, potential. EMPs migrate to the fetal liver and rapidly differentiate, including production of circulating neutrophils by E11.5. Although the surface markers, transcription factors, and lineage potential associated with EMPs overlap with those found in adult definitive hematopoiesis, they are present in unique combinations or proportions that result in a specialized definitive embryonic progenitor. Furthermore, we find that embryonic stem cell (ESC)-derived hematopoiesis recapitulates early yolk sac hematopoiesis, including primitive, EMP, and rare B cell potential. EMPs do not have long-term potential when transplanted in immunocompromised adults, but they can provide transient adult-like RBC reconstitution.
Collapse
Affiliation(s)
- Kathleen E McGrath
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jenna M Frame
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Katherine H Fegan
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - James R Bowen
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Simon J Conway
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Seana C Catherman
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Paul D Kingsley
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Anne D Koniski
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - James Palis
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
24
|
Orlandini M, Galvagni F, Bardelli M, Rocchigiani M, Lentucci C, Anselmi F, Zippo A, Bini L, Oliviero S. The characterization of a novel monoclonal antibody against CD93 unveils a new antiangiogenic target. Oncotarget 2015; 5:2750-60. [PMID: 24809468 PMCID: PMC4058042 DOI: 10.18632/oncotarget.1887] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The inhibition of tumor angiogenesis is one of the main challenges in cancer therapy. With the aim of developing monoclonal antibodies able to inhibit angiogenesis, we immunized mice with proliferating human umbilical vein endothelial cells. We generated a library of monoclonal antibodies able to recognize antigens expressed on endothelial cells and screened the antibodies for their ability to inhibit endothelial cell proliferation, migration, and sprouting in vitro. Here, we show that the antibody, designated as 4E1, is able to neutralize the formation of new vessels both in vitro and in vivo without affecting endothelial cell survival. By mass spectrometry we identified CD93 as the antigen bound by 4E1 and mapped the recognized epitope. CD93 is a transmembrane protein heavily glycosylated preferentially expressed in the vascular endothelium. CD93 silencing by lentiviral-mediated small hairpin RNA expression impairs human endothelial cell proliferation, migration, and sprouting. Altogether these findings reveal 4E1 as a novel antiangiogenic antibody and identify CD93 as a new target suitable for antiangiogenic therapy.
Collapse
Affiliation(s)
- Maurizio Orlandini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2 - 53100 Siena, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Liu C, Cui Z, Wang S, Zhang D. CD93 and GIPC expression and localization during central nervous system inflammation. Neural Regen Res 2015; 9:1995-2001. [PMID: 25598782 PMCID: PMC4283283 DOI: 10.4103/1673-5374.145383] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2014] [Indexed: 12/15/2022] Open
Abstract
CD93 and GAIP-interacting protein, C termius (GIPC) have been shown to interactively alter phagocytic processes of immune cells. CD93 and GIPC expression and localization during central nervous system inflammation have not yet been reported. In this study, we established a rat model of brain inflammation by lipopolysaccharide injection to the lateral ventricle. In the brain of rats with inflammation, western blots showed increased CD93 expression that decreased over time. GIPC expression was unaltered. Immunohistochemistry demonstrated extensive distribution of CD93 expression mainly in cell membranes in the cerebral cortex. After lipopolysaccharide stimulation, CD93 expression increased and then reduced, with distinct staining in the cytoplasm and nucleus. Double immunofluorescence staining in cerebral cortex of normal rats showed that CD93 and GIPC widely expressed in resting microglia and neurons. CD93 was mainly expressed in microglial and neuronal cell membranes, while GIPC was expressed in both cell membrane and cytoplasm. In the cerebral cortex at 9 hours after model establishment, CD93-immunoreactive signal diminished in microglial membrane, with cytoplasmic translocation and aggregation detected. GIPC localization was unaltered in neurons and microglia. These results are the first to demonstrate CD93 participation in pathophysiological processes of central nervous system inflammation.
Collapse
Affiliation(s)
- Chun Liu
- Experimental Animal Center, Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, Nantong, Jiangsu Province, China
| | - Zhichao Cui
- Department of Pathogen Biology, Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Shengjie Wang
- Experimental Animal Center, Key Laboratory of Inflammation and Molecular Drug Targets of Jiangsu Province, Nantong University, Nantong, Jiangsu Province, China
| | - Dongmei Zhang
- Department of Pathogen Biology, Medical School of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
26
|
Olsen RS, Lindh M, Vorkapic E, Andersson RE, Zar N, Löfgren S, Dimberg J, Matussek A, Wågsäter D. CD93 gene polymorphism is associated with disseminated colorectal cancer. Int J Colorectal Dis 2015; 30:883-90. [PMID: 26008729 PMCID: PMC4471320 DOI: 10.1007/s00384-015-2247-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2015] [Indexed: 02/04/2023]
Abstract
PURPOSE Cluster of differentiation 93 (CD93) is involved in apoptosis and inflammation and has a suggested role in angiogenesis, and all of which are involved in the development and dissemination of cancer. We evaluated the expression of CD93 and the association with two single nucleotide polymorphisms (SNPs), rs2749812 and rs2749817, as possible biomarkers in colorectal cancer (CRC). METHODS Tissue levels and plasma levels of CD93 were measured using an enzyme-linked immunosorbent assay (ELISA). Expression of CD93 was determined by immunohistochemistry, western blot and gene expression analysis. Genotype frequencies were established for the SNPs by real-time polymerase chain reaction (PCR), and the association with tumour stage and survival was analysed. RESULTS Total CD93 levels were 82% higher (P < 0.001) in tumours compared to matched normal tissues. Mean levels of soluble CD93 in plasma were 30% lower (P < 0.001) in the patients compared to the controls. The T/T genotype of SNP rs2749817 was more common in stage IV patients, with consequently higher risk of CRC death (T/T vs. C/C and C/T; hazard ratio (HR) = 1.73, 95% confidence interval (CI) = 1.11-2.67, P = 0.014), and was associated with a higher risk of CRC recurrence after radical operation (T/T vs. C/C and C/T; HR = 2.07, CI = 1.22-3.51, P = 0.007). CONCLUSIONS We showed that the T/T genotype of SNP rs2749817 is associated with disseminated cancer at diagnosis and an increased recurrence rate after radical operation. Patients with this genotype may benefit from early identification.
Collapse
Affiliation(s)
- Renate S Olsen
- Division of Drug Research, Department of Medical and Health Sciences, Faculty of Health Sciences, University of Linköping, 58185, Linköping, Sweden,
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Modica L, Iotti G, D’Avola A, Blasi F. Prep1 (pKnox1) regulates mouse embryonic HSC cycling and self-renewal affecting the Stat1-Sca1 IFN-dependent pathway. PLoS One 2014; 9:e107916. [PMID: 25233378 PMCID: PMC4169458 DOI: 10.1371/journal.pone.0107916] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/25/2014] [Indexed: 01/27/2023] Open
Abstract
A hypomorphic Prep1 mutation results in embryonic lethality at late gestation with a pleiotropic embryonic phenotype that includes defects in all hematopoietic lineages. Reduced functionality of the hematopoietic stem cells (HSCs) compartment might be responsible for the hematopoietic phenotype observed at mid-gestation. In this paper we demonstrate that Prep1 regulates the number of HSCs in fetal livers (FLs), their clonogenic potential and their ability to de novo generate the hematopoietic system in ablated hosts. Furthermore, we show that Prep1 controls the self-renewal ability of the FL HSC compartment as demonstrated by serial transplantation experiments. The premature exhaustion of Prep1 mutant HSCs correlates with the reduced quiescent stem cell pool thus suggesting that Prep1 regulates the self-renewal ability by controlling the quiescence/proliferation balance. Finally, we show that in FL HSCs Prep1 absence induces the interferon signaling pathway leading to premature cycling and exhaustion of fetal HSCs.
Collapse
Affiliation(s)
- Livia Modica
- Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| | - Giorgio Iotti
- Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| | | | - Francesco Blasi
- Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
- * E-mail:
| |
Collapse
|
28
|
Sonoda Y. Human CD34-negative Hematopoietic Stem Cells. STEM CELL BIOLOGY AND REGENERATIVE MEDICINE 2014. [DOI: 10.1007/978-1-4939-1001-4_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
29
|
Hanel W, Marchenko N, Xu S, Xiaofeng Yu S, Weng W, Moll U. Two hot spot mutant p53 mouse models display differential gain of function in tumorigenesis. Cell Death Differ 2013; 20:898-909. [PMID: 23538418 PMCID: PMC3679454 DOI: 10.1038/cdd.2013.17] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 01/04/2013] [Accepted: 01/07/2013] [Indexed: 12/21/2022] Open
Abstract
Mutant p53 proteins not only lose their tumor-suppressor function but some acquire oncogenic gain of function (GOF). The published mutp53 knock-in (KI) alleles (R172H, R270H, R248W) manifest GOF by broader tumor spectrum and more metastasis compared with the p53-null allele, but do not shorten survival. However, whether GOF also occurs with other mutations and whether they are all biologically equal is unknown. To answer this, we created novel humanized mutp53 KI mice harboring the hot spot alleles R248Q and G245S. Intriguingly, their impact was very different. Compared with p53-null mice, R248Q/- mice had accelerated onset of all tumor types and shorter survival, thus unprecedented strong GOF. In contrast, G245S/- mice were similar to null mice in tumor latency and survival. This was associated with a twofold higher T-lymphoma proliferation in R248Q/- mice compared with G245S/- and null mice. Moreover, R248Q/- hematopoietic and mesenchymal stem cells were expanded relative to G245S/- and null mice, the first indication that GOF also acts by perturbing pretumorous progenitor pools. Importantly, these models closely mirror Li-Fraumeni patients who show higher tumor numbers, accelerated onset and shorter tumor-free survival by 10.5 years when harboring codon R248Q mutations as compared with Li-Fraumeni patients with codon G245S mutations or p53 deletions/loss. Conversely, both KI alleles caused a modest broadening of tumor spectrum with enhanced Akt signaling compared with null mice. These models are the first in vivo proof for differential oncogenic strength among p53 GOF alleles, with genotype-phenotype correlations borne out in humans.
Collapse
Affiliation(s)
- W Hanel
- Department of Pathology, Stony Brook University, Stony Brook ,NY, USA
- Medical Scientist Program, Stony Brook University, Stony Brook, NY, USA
| | - N Marchenko
- Department of Pathology, Stony Brook University, Stony Brook ,NY, USA
| | - S Xu
- Department of Pathology, Stony Brook University, Stony Brook ,NY, USA
| | | | - W Weng
- Ingenious Targeting Inc., Stony Brook, NY, USA
| | - U Moll
- Department of Pathology, Stony Brook University, Stony Brook ,NY, USA
| |
Collapse
|
30
|
Li H, Usas A, Poddar M, Chen CW, Thompson S, Ahani B, Cummins J, Lavasani M, Huard J. Platelet-rich plasma promotes the proliferation of human muscle derived progenitor cells and maintains their stemness. PLoS One 2013; 8:e64923. [PMID: 23762264 PMCID: PMC3676442 DOI: 10.1371/journal.pone.0064923] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/20/2013] [Indexed: 01/01/2023] Open
Abstract
Human muscle-derived progenitor cells (hMDPCs) offer great promise for muscle cell-based regenerative medicine; however, prolonged ex-vivo expansion using animal sera is necessary to acquire sufficient cells for transplantation. Due to the risks associated with the use of animal sera, the development of a strategy for the ex vivo expansion of hMDPCs is required. The purpose of this study was to investigate the efficacy of using platelet-rich plasma (PRP) for the ex-vivo expansion of hMDPCs. Pre-plated MDPCs, myoendothelial cells, and pericytes are three populations of hMDPCs that we isolated by the modified pre-plate technique and Fluorescence Activated Cell Sorting (FACS), respectively. Pooled allogeneic human PRP was obtained from a local blood bank, and the effect that thrombin-activated PRP-releasate supplemented media had on the ex-vivo expansion of the hMDPCs was tested against FBS supplemented media, both in vitro and in vivo. PRP significantly enhanced short and long-term cell proliferation, with or without FBS supplementation. Antibody-neutralization of PDGF significantly blocked the mitogenic/proliferative effects that PRP had on the hMDPCs. A more stable and sustained expression of markers associated with stemness, and a decreased expression of lineage specific markers was observed in the PRP-expanded cells when compared with the FBS-expanded cells. The in vitro osteogenic, chondrogenic, and myogenic differentiation capacities of the hMDPCs were not altered when expanded in media supplemented with PRP. All populations of hMDPCs that were expanded in PRP supplemented media retained their ability to regenerate myofibers in vivo. Our data demonstrated that PRP promoted the proliferation and maintained the multi-differentiation capacities of the hMDPCs during ex-vivo expansion by maintaining the cells in an undifferentiated state. Moreover, PDGF appears to be a key contributing factor to the beneficial effect that PRP has on the proliferation of hMDPCs.
Collapse
Affiliation(s)
- Hongshuai Li
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Arvydas Usas
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Minakshi Poddar
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Chien-Wen Chen
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Seth Thompson
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Bahar Ahani
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - James Cummins
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mitra Lavasani
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Johnny Huard
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
31
|
Alexandrova EM, Moll UM. Generation of p53-deficient induced pluripotent stem cells from mouse embryo fibroblasts. Methods Mol Biol 2013; 962:157-64. [PMID: 23150445 DOI: 10.1007/978-1-62703-236-0_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Here we describe a method for generating induced pluripotent stem (iPS) cells from mouse embryonic fibroblasts (MEFs). Recombinant retroviruses carrying human transcription factors for Klf4, Oct3/4, Sox2, with or without c-Myc, are used to transduce early passage MEFs several times. Based on morphologic criteria, the resulting iPS colonies are picked manually at first, and then propagated and expanded by standard methods. iPS cells can then be differentiated into virtually any cell type or lineage, thus allowing for discoveries of new functions of p53 and mutant p53.
Collapse
|
32
|
Kao YC, Jiang SJ, Pan WA, Wang KC, Chen PK, Wei HJ, Chen WS, Chang BI, Shi GY, Wu HL. The epidermal growth factor-like domain of CD93 is a potent angiogenic factor. PLoS One 2012; 7:e51647. [PMID: 23272129 PMCID: PMC3525571 DOI: 10.1371/journal.pone.0051647] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 11/02/2012] [Indexed: 12/15/2022] Open
Abstract
Human CD93, an epidermal growth factor (EGF)-like domain containing transmembrane protein, is predominantly expressed in the vascular endothelium. Studies have shown that AA4, the homolog of CD93 in mice, may mediate cell migration and angiogenesis in endothelial cells. Soluble CD93 has been detected in the plasma of healthy individuals. However, the role of soluble CD93 in the endothelium remains unclear. Recombinant soluble CD93 proteins with EGF-like domains (rCD93D123, with domains 1, 2, and 3; and rCD93D23, with domains 2 and 3) were generated to determine their functions in angiogenesis. We found that rCD93D23 was more potent than rCD93D123 in stimulating the proliferation and migration of human umbilical vein endothelial cells (HUVECs). Production of matrix-metalloproteinase 2 increased after the HUVECs were treated with rCD93D23. Further, in a tube formation assay, rCD93D23 induced cell differentiation of HUVECs through phosphoinositide 3-kinase/Akt/endothelial nitric oxide synthase and extracellular signal-regulated kinases-1/2 signaling. Moreover, rCD93D23 promoted blood vessel formation in a Matrigel-plug assay and an oxygen-induced retinopathy model in vivo. Our findings suggest that the soluble EGF-like domain containing CD93 protein is a novel angiogenic factor acting on the endothelium.
Collapse
Affiliation(s)
- Yuan-Chung Kao
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Shinn-Jong Jiang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Wen-An Pan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Kuan-Chieh Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Po-Ku Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Hsi-Ju Wei
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Sheng Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Bi-Ing Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Guey-Yueh Shi
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
- Center for Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- * E-mail: (GYS); (HLW)
| | - Hua-Lin Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cardiovascular Research Center, National Cheng Kung University, Tainan, Taiwan
- Center for Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- * E-mail: (GYS); (HLW)
| |
Collapse
|
33
|
Yanaba K, Asano Y, Noda S, Akamata K, Aozasa N, Taniguchi T, Takahashi T, Ichimura Y, Toyama T, Sumida H, Kuwano Y, Tada Y, Sugaya M, Kadono T, Sato S. Augmented production of soluble CD93 in patients with systemic sclerosis and clinical association with severity of skin sclerosis. Br J Dermatol 2012; 167:542-7. [PMID: 22540233 DOI: 10.1111/j.1365-2133.2012.11020.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND The cell surface protein CD93, expressed on endothelial and myeloid cells, mediates phagocytosis, inflammation and cell adhesion. A soluble form of CD93 (sCD93) is released during inflammation. OBJECTIVES To determine the serum sCD93 level and its association with clinical parameters in patients with systemic sclerosis (SSc). METHODS Serum sCD93 levels were examined by enzyme-linked immunosorbent assay in 59 patients with SSc, 24 patients with systemic lupus erythematosus and 47 healthy individuals. The expression of CD93 in skin tissues was examined immunohistochemically. In a retrospective longitudinal study, sera from 11 patients with SSc were analysed. RESULTS Serum sCD93 levels were increased in patients with SSc compared with healthy individuals (P<0·001). Patients with diffuse cutaneous SSc showed greater levels of sCD93 than those with limited cutaneous SSc (P<0·01) or systemic lupus erythematosus (P<0·01). Serum sCD93 levels correlated positively with the severity of skin sclerosis. Strong CD93 immunostaining was observed on endothelial cells in lesional skin tissues. In the longitudinal study, sCD93 levels decreased in parallel with improvement in skin sclerosis. CONCLUSIONS Serum sCD93 levels are increased in patients with SSc and correlate with the severity and activity of skin sclerosis. CD93 may contribute to the development of skin fibrosis in SSc.
Collapse
Affiliation(s)
- K Yanaba
- Department of Dermatology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Two-factor reprogramming of somatic cells to pluripotent stem cells reveals partial functional redundancy of Sox2 and Klf4. Cell Death Differ 2012; 19:1268-76. [PMID: 22539002 DOI: 10.1038/cdd.2012.45] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Ectopic expression of defined sets of transcription factors in somatic cells enables them to adopt the qualities of pluripotency. Mouse embryonic fibroblasts (MEFs) are the classic target cell used to elucidate the core principles of nuclear reprogramming. However, their phenotypic and functional heterogeneity represents a major hurdle for mechanistic studies aimed at defining the molecular nature of cellular plasticity. We show that reducing the complexity of MEFs by flow cytometry allows the isolation of discrete cell subpopulations that can be efficiently reprogrammed to pluripotency with fewer genes. Using these FACS-sorted cells, we performed a systematic side-by-side analysis of the reprogramming efficiency with different two- and three-factor combinations of Oct4, Sox2 and Klf4. We show that introduction of exogenous Oct4 with either Sox2 or Klf4 does not directly convert MEFs to a pluripotent state. Instead, each combination of factors disrupts the normal cellular homeostasis and establishes transient states characterized by the concurrent expression of mixed lineage markers. These cells convert into induced pluripotent stem cells in a stochastic fashion. Our data suggest that there is a partial functional redundancy between Sox2 and Klf4 in the disruption of cellular homeostasis and activation of regulatory networks that define pluripotency.
Collapse
|
35
|
Gomez G, Lee JH, Veldman MB, Lu J, Xiao X, Lin S. Identification of vascular and hematopoietic genes downstream of etsrp by deep sequencing in zebrafish. PLoS One 2012; 7:e31658. [PMID: 22438865 PMCID: PMC3306315 DOI: 10.1371/journal.pone.0031658] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 01/17/2012] [Indexed: 11/18/2022] Open
Abstract
The transcription factor etsrp/Er71/Etv2 is a master control gene for vasculogenesis in all species studied to date. It is also required for hematopoiesis in zebrafish and mice. Several novel genes expressed in vasculature have been identified through transcriptional profiling of zebrafish embryos overexpressing etsrp by microarrays. Here we re-examined this transcriptional profile by Illumina RNA-sequencing technology, revealing a substantially increased number of candidate genes regulated by etsrp. Expression studies of 50 selected candidate genes from this dataset resulted in the identification of 39 new genes that are expressed in vascular cells. Regulation of these genes by etsrp was confirmed by their ectopic induction in etsrp overexpressing and decreased expression in etsrp deficient embryos. Our studies demonstrate the effectiveness of the RNA-sequencing technology to identify biologically relevant genes in zebrfish and produced a comprehensive profile of genes previously unexplored in vascular endothelial cell biology.
Collapse
Affiliation(s)
- Gustavo Gomez
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | | | | | | | | | | |
Collapse
|
36
|
Dravid GG, Crooks GM. The challenges and promises of blood engineered from human pluripotent stem cells. Adv Drug Deliv Rev 2011; 63:331-41. [PMID: 21232565 DOI: 10.1016/j.addr.2010.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 12/02/2010] [Accepted: 12/09/2010] [Indexed: 12/21/2022]
Abstract
The concept that stem cells can be used to replace and regenerate tissue was founded over half a century ago using hematopoietic stem cells in the clinical field of bone marrow transplantation. The development of human embryonic stem cell lines and patient-specific induced pluripotent stem cells has the potential to overcome the problem presented by shortages of immunologically compatible hematopoietic stem cell donors. This review summarizes the current advances made and limitations to be overcome in order to realize the full potential of engineering blood from pluripotent stem cells for clinical use.
Collapse
|
37
|
Coskun S, Hirschi KK. Establishment and regulation of the HSC niche: Roles of osteoblastic and vascular compartments. ACTA ACUST UNITED AC 2011; 90:229-42. [PMID: 21181885 DOI: 10.1002/bdrc.20194] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hematopoietic stem cells (HSC) are multi-potent cells that function to generate a lifelong supply of all blood cell types. During mammalian embryogenesis, sites of hematopoiesis change over the course of gestation: from extraembryonic yolk sac and placenta, to embryonic aorta-gonad-mesonephros region, fetal liver, and finally fetal bond marrow where HSC reside postnatally. These tissues provide microenviroments for de novo HSC formation, as well as HSC maturation and expansion. Within adult bone marrow, HSC self-renewal and differentiation are thought to be regulated by two major cellular components within their so-called niche: osteoblasts and vascular endothelial cells. This review focuses on HSC generation within, and migration to, different tissues during development, and also provides a summary of major regulatory factors provided by osteoblasts and vascular endothelial cells within the adult bone marrow niche.
Collapse
Affiliation(s)
- Suleyman Coskun
- Center for Cell and Gene Therapy, Baylor College of Medicine; Houston, Texas, 77030, USA
| | | |
Collapse
|
38
|
|
39
|
Abstract
Despite significant research on the role of inflammation and immunosurveillance in the immunologic microenvironment of tumors, little attention has been given to the oncogenic capabilities of the complement cascade. The recent finding that complement may contribute to tumor growth suggests an insidious relationship between complement and cancer, especially in light of evidence that complement facilitates cellular proliferation and regeneration. We address the hypothesis that complement proteins promote carcinogenesis and suggest mechanisms by which complement can drive the fundamental features of cancer. Evidence shows that this diverse family of innate immune proteins facilitates dysregulation of mitogenic signaling pathways, sustained cellular proliferation, angiogenesis, insensitivity to apoptosis, invasion and migration, and escape from immunosurveillance. Given that the traditionally held functions for the complement system include innate immunity and cancer defense, our review suggests a new way of thinking about the role of complement proteins in neoplasia.
Collapse
Affiliation(s)
- Martin J Rutkowski
- Department of Neurological Surgery, University of California at San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
40
|
Jeon JW, Jung JG, Shin EC, Choi HI, Kim HY, Cho ML, Kim SW, Jang YS, Sohn MH, Moon JH, Cho YH, Hoe KL, Seo YS, Park YW. Soluble CD93 Induces Differentiation of Monocytes and Enhances TLR Responses. THE JOURNAL OF IMMUNOLOGY 2010; 185:4921-7. [DOI: 10.4049/jimmunol.0904011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
41
|
Irion S, Clarke RL, Luche H, Kim I, Morrison SJ, Fehling HJ, Keller GM. Temporal specification of blood progenitors from mouse embryonic stem cells and induced pluripotent stem cells. Development 2010; 137:2829-39. [PMID: 20659975 DOI: 10.1242/dev.042119] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The efficient and reproducible generation of differentiated progenitors from pluripotent stem cells requires the recapitulation of appropriate developmental stages and pathways. Here, we have used the combination of activin A, BMP4 and VEGF under serum-free conditions to induce hematopoietic differentiation from both embryonic and induced pluripotent stem cells, with the aim of modeling the primary sites of embryonic hematopoiesis. We identified two distinct Flk1-positive hematopoietic populations that can be isolated based on temporal patterns of emergence. The earliest arising population displays characteristics of yolk sac hematopoiesis, whereas a late developing Flk1-positive population appears to reflect the para-aortic splanchnopleura hematopoietic program, as it has reduced primitive erythroid capacity and substantially enhanced myeloid and lymphoid potential compared with the earlier wave. These differences between the two populations are accompanied by differences in the expression of Sox17 and Hoxb4, as well as in the cell surface markers AA4.1 and CD41. Together, these findings support the interpretation that the two populations are representative of the early sites of mammalian hematopoiesis.
Collapse
Affiliation(s)
- Stefan Irion
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
42
|
Rutkowski MJ, Sughrue ME, Kane AJ, Ahn BJ, Fang S, Parsa AT. The complement cascade as a mediator of tissue growth and regeneration. Inflamm Res 2010; 59:897-905. [PMID: 20517706 PMCID: PMC2945462 DOI: 10.1007/s00011-010-0220-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 04/02/2010] [Accepted: 05/11/2010] [Indexed: 12/16/2022] Open
Abstract
Recent evidence has demonstrated that the complement cascade is involved in a variety of physiologic and pathophysiologic processes in addition to its role as an immune effector. Research in a variety of organ systems has shown that complement proteins are direct participants in maintenance of cellular turnover, healing, proliferation and regeneration. As a physiologic housekeeper, complement proteins maintain tissue integrity in the absence of inflammation by disposing of cellular debris and waste, a process critical to the prevention of autoimmune disease. Developmentally, complement proteins influence pathways including hematopoietic stem cell engraftment, bone growth, and angiogenesis. They also provide a potent stimulus for cellular proliferation including regeneration of the limb and eye in animal models, and liver proliferation following injury. Here, we describe the complement cascade as a mediator of tissue growth and regeneration.
Collapse
Affiliation(s)
- Martin J Rutkowski
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | |
Collapse
|
43
|
Harhausen D, Prinz V, Ziegler G, Gertz K, Endres M, Lehrach H, Gasque P, Botto M, Stahel PF, Dirnagl U, Nietfeld W, Trendelenburg G. CD93/AA4.1: a novel regulator of inflammation in murine focal cerebral ischemia. THE JOURNAL OF IMMUNOLOGY 2010; 184:6407-17. [PMID: 20439917 DOI: 10.4049/jimmunol.0902342] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The stem-cell marker CD93 (AA4.1/C1qRp) has been described as a potential complement C1q-receptor. Its exact molecular function, however, remains unknown. By using global expression profiling we showed that CD93-mRNA is highly induced after transient focal cerebral ischemia. CD93 protein is upregulated in endothelial cells, but also in selected macrophages and microglia. To elucidate the potential functional role of CD93 in postischemic brain damage, we used mice with a targeted deletion of the CD93 gene. After 30 min of occlusion of the middle cerebral artery and 3 d of reperfusion these mice displayed increased leukocyte infiltration into the brain, increased edema, and significantly larger infarct volumes (60.8 +/- 52.2 versus 23.9 +/- 16.6 mm(3)) when compared with wild-type (WT) mice. When the MCA was occluded for 60 min, after 2 d of reperfusion the CD93 knockout mice still showed more leukocytes in the brain, but the infarct volumes were not different from those seen in WT animals. To further explore CD93-dependent signaling pathways, we determined global transcription profiles and compared CD93-deficient and WT mice at various time points after induction of focal cerebral ischemia. We found a highly significant upregulation of the chemokine CCL21/Exodus-2 in untreated and treated CD93-deficient mice at all time points. Induction of CCL21 mRNA and protein was confirmed by PCR and immunohistochemistry. CCL21, which was formerly shown to be released by damaged neurons and to activate microglia, contributes to neurodegeneration. Thus, we speculate that CD93-neuroprotection is mediated via suppression of the neuroinflammatory response through downregulation of CCL21.
Collapse
Affiliation(s)
- Denise Harhausen
- Experimentelle Neurologie, Charité-Universitätsmedizin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Complement and the central nervous system: emerging roles in development, protection and regeneration. Immunol Cell Biol 2010; 88:781-6. [PMID: 20404838 DOI: 10.1038/icb.2010.48] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
As expanding research reveals the novel ability of complement proteins to promote proliferation and regeneration of tissues throughout the body, the concept of the complement cascade as an innate immune effector has changed rapidly. In particular, its interactions with the central nervous system have provided a wealth of information regarding the ability of complement proteins to mediate neurogenesis, synaptogenesis, cell migration, neuroprotection, proliferation and regeneration. At numerous phases of the neuronal and glial cell cycle, complement proteins exert direct or indirect influence over their behavior and fate. Neuronal stem cells differentiate and migrate in response to complement, and it prevents injury and death in adult cells in response to toxic agents. Furthermore, complement proteins promote survival via anti-apoptotic actions, and can facilitate clearance and regeneration of injured tissues in various models of CNS disease. In summary, we highlight the protean abilities of complement proteins in the central nervous system, underscoring an exciting avenue of research that has yielded greater understanding of complement's role in central nervous system health and disease.
Collapse
|
45
|
Zekavat G, Mozaffari R, Arias VJ, Rostami SY, Badkerhanian A, Tenner AJ, Nichols KE, Naji A, Noorchashm H. A novel CD93 polymorphism in non-obese diabetic (NOD) and NZB/W F1 mice is linked to a CD4+ iNKT cell deficient state. Immunogenetics 2010; 62:397-407. [PMID: 20387063 PMCID: PMC2875467 DOI: 10.1007/s00251-010-0442-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 03/16/2010] [Indexed: 01/25/2023]
Abstract
In the present study, we characterize a polymorphism in the CD93 molecule, originally identified as the receptor for the C1q complement component (i.e., C1qRp, or AA4.1) in non-obese diabetic (NOD) mice. This allele carries a coding polymorphism in the first epidermal growth factor-like domain of CD93, which results in an amino acid substitution from Asn→His at position 264. This polymorphism does not appear to influence protein translation or ecto-domain cleavage, as CD93 is detectable in bone-marrow-derived macrophage and B-cell precursor lysates and in soluble form in the serum. The NOD CD93 isoform causes a phenotypic aberrancy in the early B-cell developmental stages (i.e., pro-, pre-, immature, and transitional), likely related to a conformational variation. Interestingly, the NZB/W F1 strain, which serves as a murine model of Lupus, also expresses an identical CD93 sequence polymorphism. Cd93 is located within the NOD Idd13 locus and is also tightly linked to the NZB/W F1 Wbw1 and Nkt2 disease susceptibility loci, which are thought to regulate natural killer T (NKT) cell homeostasis. Consistent with this genetic linkage, we found B6 CD93−/− and B6.NODIdd13 mice to be susceptible to a profound CD4+ NKT cell deficient state. These data suggest that Cd93 may be an autoimmune susceptibility gene residing within the Idd13 locus, which plays a role in regulating absolute numbers of CD4+ NKT cells.
Collapse
Affiliation(s)
- Ghazal Zekavat
- Harrison Department of Surgical Research, University of Pennsylvania School of Medicine, Philadelphia, PA USA
| | - Raha Mozaffari
- Harrison Department of Surgical Research, University of Pennsylvania School of Medicine, Philadelphia, PA USA
| | - Vanessa J. Arias
- School of Medicine, University of California Irvine, Irvine, CA USA
| | - Susan Y. Rostami
- Harrison Department of Surgical Research, University of Pennsylvania School of Medicine, Philadelphia, PA USA
| | - Armen Badkerhanian
- Harrison Department of Surgical Research, University of Pennsylvania School of Medicine, Philadelphia, PA USA
| | - Andrea J. Tenner
- School of Medicine, University of California Irvine, Irvine, CA USA
| | - Kim E. Nichols
- Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Ali Naji
- Harrison Department of Surgical Research, University of Pennsylvania School of Medicine, Philadelphia, PA USA
| | - Hooman Noorchashm
- Harrison Department of Surgical Research, University of Pennsylvania School of Medicine, Philadelphia, PA USA
| |
Collapse
|
46
|
Kuraoka M, Liao D, Yang K, Allgood SD, Levesque MC, Kelsoe G, Ueda Y. Activation-induced cytidine deaminase expression and activity in the absence of germinal centers: insights into hyper-IgM syndrome. THE JOURNAL OF IMMUNOLOGY 2009; 183:3237-48. [PMID: 19667096 DOI: 10.4049/jimmunol.0901548] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Somatic hypermutation normally occurs as a consequence of the expression of activation-induced cytidine deaminase (AID) by Ag-activated, mature B cells during T cell-dependent germinal center responses. Nonetheless, despite their inability to express CD154 and initiate GC responses, patients with type 1 hyper-IgM syndrome (HIGM1) support populations of IgM(+)IgD(+)CD27(+) B cells that express mutated Ig genes. The origin of these mutated B cells is unknown; the IgM(+)IgD(+)CD27(+) cells do not express AID and appear to acquire mutations independent of stringent selection by Ag. Here, we demonstrate that immature/transitional 1 B cells from the bone marrow of CD154-deficient mice express AID and acquire Ig mutations that lack the hallmarks of antigenic selection via BCR signaling. Comparable levels of AID expression was found in developmentally immature B cells recovered from murine fetal liver and from human immature/transitional 1 B cells recovered from umbilical cord blood. AID expression in human fetal liver was also robust, approaching that of human tonsil tissue and the human germinal center B cell line, Ramos. These observations led us to conclude that AID expression in developing human B cells is the origin of the mutated IgM(+)IgD(+)CD27(+) B cells present in HIGM1 patients, and we propose that both mice and humans share a latent, AID-dependent pathway for the preimmune diversification of B lymphocytes that is more prominent in chicken, sheep, and rabbits.
Collapse
Affiliation(s)
- Masayuki Kuraoka
- Department of Immunology, Duke University, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Expression of AA4.1 marks lymphohematopoietic progenitors in early mouse development. Proc Natl Acad Sci U S A 2009; 106:8953-8. [PMID: 19458045 DOI: 10.1073/pnas.0904090106] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The hematopoietic system of mice is established during the early to midgestational stage of development. However, the earliest lymphohematopoietic progenitors that appear during mouse development have been less well characterized compared with the hematopoietic stem cell compartment of fetal liver and bone marrow. We isolated the earliest lymphohematopoietic progenitors by using embryonic stem (ES) cell culture in vitro. Cells with the c-Kit(+)Lin(-) cell surface phenotype were present abundantly in ES cells cocultured with stromal cell lines. We further separated the cells into two distinct cell subsets based on AA4.1 expression. Although AA4.1(+) and AA4.1(-) cells had equivalent potency to generate myeloid cell lineages, the lymphoid potential in ES-cell-derived cells was largely restricted to the cells expressing AA4.1. The same cell type was present abundantly in the early yolk sac and in fewer numbers (approximately 5% of that in the yolk sac) in the caudal half of the developing embryos. These data suggest that AA4.1 is a cell surface marker that can identify the earliest lymphohematopoietic progenitors in mouse development.
Collapse
|
48
|
Feres KJ, Ischenko I, Hayman MJ. The RON receptor tyrosine kinase promotes MSP-independent cell spreading and survival in breast epithelial cells. Oncogene 2008; 28:279-88. [PMID: 18836480 PMCID: PMC2628450 DOI: 10.1038/onc.2008.383] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The recepteur d’origne nantais (RON) is a receptor tyrosine kinase (RTK) in the scatter factor family, which includes the c-Met receptor. RON exhibits increased expression in a significant number of human breast cancer tissues as well as in many established breast cancer cell lines. Recent studies have indicated that in addition to ligand-dependent signaling events, RON also promotes signals in the absence of its only known ligand, the macrophage stimulating protein, when expressed in epithelial cells. In the current study, we found that when expressed in MCF-10A breast epithelial cells, RON exhibits both MSP-dependent and MSP-independent signaling, which lead to distinct biological outcomes. In the absence of MSP, RON signaling promotes cell survival, increased cell spreading and enhanced migration in response to other growth factors. However, both RON-mediated proliferation and migration require the addition of MSP in MCF-10A cells. Both MSP-dependent and MSP-independent signaling by RON is mediated in part by Src-family kinases. These data suggest that RON has two alternative modes of signaling that can contribute to oncogenic behavior in normal breast epithelial cells.
Collapse
Affiliation(s)
- K J Feres
- Department of Molecular Genetics & Microbiology, Stony Brook University, Stony Brook, NY, USA
| | | | | |
Collapse
|
49
|
Zayas J, Spassov DS, Nachtman RG, Jurecic R. Murine hematopoietic stem cells and multipotent progenitors express truncated intracellular form of c-kit receptor. Stem Cells Dev 2008; 17:343-53. [PMID: 18447649 DOI: 10.1089/scd.2007.0101] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The c-kit receptor plays a vital role in self-renewal and differentiation of hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs). We have discovered that besides c-kit, the murine multipotent HSC/MPP-like cell line EML expresses the transcript and protein for a truncated intracellular form of c-kit receptor, called tr-kit. Notably, the tr-kit transcript and protein levels were down-regulated during cytokine-induced differentiation of the HSC/MPP-like cell line EML into myeloerythroid lineages. These findings prompted us to analyze tr-kit expression in purified murine fetal liver and bone marrow cell populations containing long-term repopulating (LTR) HSCs, short-term repopulating (STR) HSCs, MPPs, lineage-committed progenitors, and immature blood cells. Remarkably, these studies have revealed that in contrast to more widespread expression of c-kit, tr-kit is transcribed solely in cell populations enriched for LTR-HSCs, STR-HSCs, and MPPs. On the other hand, cell populations in which HSCs and MPPs are either present at a much lower frequency or are absent altogether, cells representing more advanced stages of differentiation into lymphoid and myeloid lineages do not express tr-kit. The observation that tr-kit is co-expressed with c-kit only in more primitive HSC- and MPP-enriched cell populations raises an exciting possibility that tr-kit functions either as a new component of the stem cell factor (SCF)/c-kit pathway or is involved in a novel signaling pathway, present exclusively in HSC and MPPs. Taken together, these findings necessitate functional characterization of tr-kit and analysis of its potential role in the self-renewal, proliferation, and/or differentiation of HSC and multipotent progenitors.
Collapse
Affiliation(s)
- Jennifer Zayas
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | | | |
Collapse
|
50
|
Nguyen HX, Galvan MD, Anderson AJ. Characterization of early and terminal complement proteins associated with polymorphonuclear leukocytes in vitro and in vivo after spinal cord injury. J Neuroinflammation 2008; 5:26. [PMID: 18578885 PMCID: PMC2443364 DOI: 10.1186/1742-2094-5-26] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 06/25/2008] [Indexed: 02/01/2023] Open
Abstract
Background The complement system has been suggested to affect injury or disease of the central nervous system (CNS) by regulating numerous physiological events and pathways. The activation of complement following traumatic CNS injury can also result in the formation and deposition of C5b-9 membrane attack complex (C5b-9/MAC), causing cell lysis or sublytic effects on vital CNS cells. Although complement proteins derived from serum/blood-brain barrier breakdown can contribute to injury or disease, infiltrating immune cells may represent an important local source of complement after injury. As the first immune cells to infiltrate the CNS within hours post-injury, polymorphonuclear leukocytes (PMNs) may affect injury through mechanisms associated with complement-mediated events. However, the expression/association of both early and terminal complement proteins by PMNs has not been fully characterized in vitro, and has not observed previously in vivo after traumatic spinal cord injury (SCI). Method We investigated the expression of complement mRNAs using rt-PCR and the presence of complement proteins associated with PMNs using immunofluroescence and quantitative flow cytometry. Results Stimulated or unstimulated PMNs expressed mRNAs encoding for C1q, C3, and C4, but not C5, C6, C7 or C9 in culture. Complement protein C1q or C3 was also detected in less than 30% of cultured PMNs. In contrast, over 70% of PMNs that infiltrated the injured spinal cord were associated with C1q, C3, C7 and C5b-9/MAC 3 days post-SCI. The localization/association of C7 or C5b-9/MAC with infiltrating PMNs in the injured spinal cord suggests the incorporation or internalization of C7 or C5b-9/MAC bound cellular debris by infiltrating PMNs because C7 and C5b-9/MAC were mostly localized to granular vesicles within PMNs at the spinal cord epicenter region. Furthermore, PMN presence in the injured spinal cord was observed for many weeks post-SCI, suggesting that this infiltrating cell population could chronically affect complement-mediated events and SCI pathogenesis after trauma. Conclusion Data presented here provide the first characterization of early and terminal complement proteins associated with PMNs in vitro and in vivo after SCI. Data also suggest a role for PMNs in the local internalization or deliverance of complement and complement activation in the post-SCI environment.
Collapse
Affiliation(s)
- Hal X Nguyen
- Physical Medicine & Rehabilitation, 1105 Gillespie Neuroscience Research Facility, University of California, Irvine, CA 92697-4292, USA.
| | | | | |
Collapse
|