1
|
Park SD, Saunders AS, Reidy MA, Bender DE, Clifton S, Morris KT. A review of granulocyte colony-stimulating factor receptor signaling and regulation with implications for cancer. Front Oncol 2022; 12:932608. [PMID: 36033452 PMCID: PMC9402976 DOI: 10.3389/fonc.2022.932608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/15/2022] [Indexed: 12/29/2022] Open
Abstract
Granulocyte colony-stimulating factor receptor (GCSFR) is a critical regulator of granulopoiesis. Studies have shown significant upregulation of GCSFR in a variety of cancers and cell types and have recognized GCSFR as a cytokine receptor capable of influencing both myeloid and non-myeloid immune cells, supporting pro-tumoral actions. This systematic review aims to summarize the available literature examining the mechanisms that control GCSFR signaling, regulation, and surface expression with emphasis on how these mechanisms may be dysregulated in cancer. Experiments with different cancer cell lines from breast cancer, bladder cancer, glioma, and neuroblastoma are used to review the biological function and underlying mechanisms of increased GCSFR expression with emphasis on actions related to tumor proliferation, migration, and metastasis, primarily acting through the JAK/STAT pathway. Evidence is also presented that demonstrates a differential physiological response to aberrant GCSFR signal transduction in different organs. The lifecycle of the receptor is also reviewed to support future work defining how this signaling axis becomes dysregulated in malignancies.
Collapse
Affiliation(s)
- Sungjin David Park
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Apryl S. Saunders
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Megan A. Reidy
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Dawn E. Bender
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Shari Clifton
- Department of Information Management, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Katherine T. Morris
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
- *Correspondence: Katherine T. Morris,
| |
Collapse
|
2
|
Sommerfeld S, Mundim AV, Silva RR, Queiroz JS, Rios MP, Notário FO, Medeiros Ronchi AA, Beletti ME, Franco RR, Espindola FS, Goulart LR, Fonseca BB. Physiological Changes in Chicken Embryos Inoculated with Drugs and Viruses Highlight the Need for More Standardization of this Animal Model. Animals (Basel) 2022; 12:1156. [PMID: 35565581 PMCID: PMC9099557 DOI: 10.3390/ani12091156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/08/2022] [Accepted: 04/22/2022] [Indexed: 01/01/2023] Open
Abstract
Several studies have been developed using the Gallus gallus embryo as an experimental model to study the toxicity of drugs and infections. Studies that seek to standardize the evaluated parameters are needed to better understand and identify the viability of CEs as an experimental model. Therefore, we sought to verify whether macroscopic, histopathological, blood count, metabolites and/or enzymes changes and oxidative stress in CE of different ages are specific to the model. To achieve this goal, in ovo assays were performed by injecting a virus (Gammacoronavirus) and two drugs (filgrastim and dexamethasone) that cause known changes in adult animals. Although congestion and inflammatory infiltrate were visible in the case of viral infections, the white blood cell count and inflammation biomarkers did not change. Filgrastim (FG) testing did not increase granulocytes as we expected. On the other hand, CE weight and red blood cell count were lower with dexamethasone (DX), whereas white blood cell count and biomarkers varied depended on the stage of CE development. Our work reinforces the importance of standardization and correct use of the model so that the results of infection, toxicity and pharmacokinetics are reproducible.
Collapse
Affiliation(s)
- Simone Sommerfeld
- School of Veterinary Medicine, Federal University of Uberlândia, Uberlândia 38402-018, Brazil; (A.V.M.); (R.R.S.); (J.S.Q.); (M.P.R.); (F.O.N.); (A.A.M.R.); (B.B.F.)
| | - Antonio Vicente Mundim
- School of Veterinary Medicine, Federal University of Uberlândia, Uberlândia 38402-018, Brazil; (A.V.M.); (R.R.S.); (J.S.Q.); (M.P.R.); (F.O.N.); (A.A.M.R.); (B.B.F.)
| | - Rogério Reis Silva
- School of Veterinary Medicine, Federal University of Uberlândia, Uberlândia 38402-018, Brazil; (A.V.M.); (R.R.S.); (J.S.Q.); (M.P.R.); (F.O.N.); (A.A.M.R.); (B.B.F.)
| | - Jéssica Santos Queiroz
- School of Veterinary Medicine, Federal University of Uberlândia, Uberlândia 38402-018, Brazil; (A.V.M.); (R.R.S.); (J.S.Q.); (M.P.R.); (F.O.N.); (A.A.M.R.); (B.B.F.)
| | - Maisa Paschoal Rios
- School of Veterinary Medicine, Federal University of Uberlândia, Uberlândia 38402-018, Brazil; (A.V.M.); (R.R.S.); (J.S.Q.); (M.P.R.); (F.O.N.); (A.A.M.R.); (B.B.F.)
| | - Fabiana Oliveira Notário
- School of Veterinary Medicine, Federal University of Uberlândia, Uberlândia 38402-018, Brazil; (A.V.M.); (R.R.S.); (J.S.Q.); (M.P.R.); (F.O.N.); (A.A.M.R.); (B.B.F.)
| | - Alessandra Aparecida Medeiros Ronchi
- School of Veterinary Medicine, Federal University of Uberlândia, Uberlândia 38402-018, Brazil; (A.V.M.); (R.R.S.); (J.S.Q.); (M.P.R.); (F.O.N.); (A.A.M.R.); (B.B.F.)
| | - Marcelo Emílio Beletti
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38405-319, Brazil;
| | - Rodrigo Rodrigues Franco
- Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38405-319, Brazil; (R.R.F.); (F.S.E.); (L.R.G.)
| | - Foued Salmen Espindola
- Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38405-319, Brazil; (R.R.F.); (F.S.E.); (L.R.G.)
| | - Luiz Ricardo Goulart
- Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38405-319, Brazil; (R.R.F.); (F.S.E.); (L.R.G.)
| | - Belchiolina Beatriz Fonseca
- School of Veterinary Medicine, Federal University of Uberlândia, Uberlândia 38402-018, Brazil; (A.V.M.); (R.R.S.); (J.S.Q.); (M.P.R.); (F.O.N.); (A.A.M.R.); (B.B.F.)
- Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38405-319, Brazil; (R.R.F.); (F.S.E.); (L.R.G.)
| |
Collapse
|
3
|
Mouchemore KA, Anderson RL. Immunomodulatory effects of G-CSF in cancer: Therapeutic implications. Semin Immunol 2021; 54:101512. [PMID: 34763974 DOI: 10.1016/j.smim.2021.101512] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/23/2021] [Indexed: 01/04/2023]
Abstract
Numerous preclinical studies have reported a pro-tumour role for granulocyte colony-stimulating factor (G-CSF) that is predominantly mediated by neutrophils and MDSCs, the major G-CSF receptor expressing populations. In the presence of G-CSF (either tumour-derived or exogenous) these myeloid populations commonly exhibit a T cell suppressive phenotype. However, the direct effects of this cytokine on other immune lineages, such as T and NK cells, are not as well established. Herein we discuss the most recent data relating to the effect of G-CSF on the major immune populations, exclusively in the context of cancer. Recent publications have drawn attention to the other tumour-promoting effects of G-CSF on myeloid cells, including NETosis, promotion of cancer stemness and skewed differentiation of bone marrow progenitors towards myelopoiesis. Although G-CSF is safely and commonly used as a supportive therapy to prevent or treat chemotherapy-associated neutropenia in cancer patients, we also discuss the potential impacts of G-CSF on other anti-cancer treatments. Importantly, considerations for immune checkpoint blockade are highlighted, as many publications report a T cell suppressive effect of G-CSF that may diminish the effectiveness of this immunotherapy.
Collapse
Affiliation(s)
- Kellie A Mouchemore
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Robin L Anderson
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
4
|
Ribatti D, Tamma R. Hematopoietic growth factors and tumor angiogenesis. Cancer Lett 2018; 440-441:47-53. [PMID: 30312730 DOI: 10.1016/j.canlet.2018.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/26/2018] [Accepted: 10/08/2018] [Indexed: 01/13/2023]
Abstract
Angiogenesis is regulated by numerous "classic" factors such as vascular endothelial growth factor (VEGF) and many other endogenous "non-classic"peptides, including erythropoietin (Epo), and granulocyte-/granulocyte macrophage colony stimulating factor (G-/GM-CSF). The latter play an important regulatory role in angiogenesis, especially under pathological conditions and constitute a crosslink between angiogenesis and hematopoiesis. This article reviews studies on the ability of hematopoietic cytokines to affect several endothelial cell functions in tumor angiogenesis. These findings in all these studies support the hypothesis formulated at the beginning of this century that a common ancestral cell, the hemangioblast, gives rise to cells of both the endothelial and the hematopoietic lineages.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
5
|
Peng W. Neuroprotective effects of G-CSF administration in microglia-mediated reactive T cell activation in vitro. Immunol Res 2018. [PMID: 28646409 DOI: 10.1007/s12026-017-8928-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
G-CSF is a growth factor that has known neuroprotective effects in a variety of experimental brain injury models. As both antigen-presenting microglia and reactive T cells are key components in the development and progression of EAE, the aim of this study is to investigate the neuroprotective effects of recombinant human G-CSF, as administered in microglia-mediated reactive T cell assay in vitro. Our results indicate that G-CSF treatment has no apparent effect for the resting un-activated microglia. G-CSF pre-protection of microglia increased protective cytokine IL-4 production and effectively inhibited the productions of NO and other inflammatory mediators (IFN-γ, TNF-α, IL-1β, IL-17, and chemokine MCP-1) after LPS stimulation. G-CSF suppressed the proliferative response of microglia-mediated MOG35-55 reactive T cells. G-CSF-microglia-T cells increased IL-4 and IL-10 secretions and decreased IFN-γ, TNF-α, and IL-17 productions. G-CSF significantly elevated CD4+CD25+ regulatory T cell subset in microglia-mediated reactive T cells. Moreover, G-CSF inhibited MHC-II expression of microglia after LPS activation or in the interactions of microglia and reactive T cells. G-CSF administration induced the apoptosis and enhanced the G0/G1 to S phase transition and elevated the gene expression of apoptosis markers in microglia-mediated reactive T cells after stimulated by specific antigen MOG35-55. These findings reveal that G-CSF administration potently neuroprotects the central nervous system (CNS) from immune-mediated damage in microglia-mediated reactive T cell activation. Apoptosis of reactive T cells in CNS is important in attenuating the development of autoimmune CNS diseases. G-CSF administration has neuroprotective effects in CNS and the potential to be a therapeutic agent in multiple sclerosis.
Collapse
Affiliation(s)
- Wei Peng
- Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, People's Republic of China. .,Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Sderot Churchill, Jerusalem, 91240, Israel.
| |
Collapse
|
6
|
Kohlstedt K, Trouvain C, Frömel T, Mudersbach T, Henschler R, Fleming I. Role of the angiotensin-converting enzyme in the G-CSF-induced mobilization of progenitor cells. Basic Res Cardiol 2018; 113:18. [PMID: 29549541 DOI: 10.1007/s00395-018-0677-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 03/15/2018] [Indexed: 12/22/2022]
Abstract
In addition to being a peptidase, the angiotensin-converting enzyme (ACE) can be phosphorylated and involved in signal transduction. We evaluated the role of ACE in granulocyte-colony-stimulating factor (G-CSF)-induced hematopoietic progenitor cell (HPC) mobilization and detected a significant increase in mice-lacking ACE. Transplantation experiments revealed that the loss of ACE in the HPC microenvironment rather than in the HPCs increased mobilization. Indeed, although ACE was expressed by a small population of bone-marrow cells, it was more strongly expressed by endosteal bone. Interestingly, there was a physical association of ACE with the G-CSF receptor (CD114), and G-CSF elicited ACE phosphorylation on Ser1270 in vivo and in vitro. A transgenic mouse expressing a non-phosphorylatable ACE (ACES/A) mutant demonstrated increased G-CSF-induced HPC mobilization and decreased G-CSF-induced phosphorylation of STAT3 and STAT5. These results indicate that ACE expression/phosphorylation in the bone-marrow niche interface negatively regulates G-CSF-induced signaling and HPC mobilization.
Collapse
Affiliation(s)
- Karin Kohlstedt
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Caroline Trouvain
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Timo Frömel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Thomas Mudersbach
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Reinhard Henschler
- Blood Donor Services Zürich and Chur, Swiss Red Cross, Zurich, Switzerland
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany. .,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt am Main, Germany.
| |
Collapse
|
7
|
McNamee EN, Biette KA, Hammer J, Harris R, Miyazawa H, Lee JJ, Furuta GT, Masterson JC. Targeting granulocyte-macrophage colony-stimulating factor in epithelial and vascular remodeling in experimental eosinophilic esophagitis. Allergy 2017; 72:1232-1242. [PMID: 27926989 DOI: 10.1111/all.13105] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a chronic antigen-mediated clinicopathologic disease of the esophagus characterized by an eosinophil-predominant inflammatory infiltrate. A clinical hallmark is extensive tissue remodeling including basal zone hyperplasia, fibrosis, and angiogenesis. However, the cellular mechanisms responsible for these processes are not fully defined. We hypothesized that targeting granulocyte-macrophage colony-stimulating factor (GM-CSF; an agonist cytokine linked with eosinophil survival and activation) would be protective in a preclinical model of EoE. METHODS Eosinophilic esophagitis-like esophageal inflammation was induced in the L2-IL5OXA EoE mouse model, and GM-CSF production was assessed by mRNA and protein analyses. Granulocyte-macrophage colony-stimulating factor-receptor-alpha expression patterns were examined by flow cytometric and immunofluorescence analysis. L2-IL5OXA EoE mice were treated with anti-GM-CSF neutralizing antibody or isotype control and assessed for histopathological indices of eosinophilia, epithelial hyperplasia, and angiogenesis by immunohistochemistry and RT-PCR. RESULTS Significantly increased levels of esophageal GM-CSF expression was detected in the L2-IL5OXA mouse EoE model during active inflammation. Granulocyte-macrophage colony-stimulating factor-receptor-alpha was predominantly expressed on esophageal eosinophils during EoE, in addition to select cells within the lamina propria. Anti-GM-CSF neutralization in L2-IL5OXA EoE mice resulted in a significant diminution of epithelial eosinophilia in addition to basal cell hyperplasia and vascular remodeling. This treatment response was independent of effects on esophageal eosinophil maturation or activation. CONCLUSION Granulocyte-macrophage colony-stimulating factor is a potential therapeutic target to reduce esophageal eosinophilia and remodeling.
Collapse
Affiliation(s)
- E. N. McNamee
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
- Department of Anesthesiology; University of Colorado School of Medicine; Aurora CO USA
| | - K. A. Biette
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| | - J. Hammer
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| | - R. Harris
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| | - H. Miyazawa
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
| | - J. J. Lee
- Department of Biochemistry and Molecular Biology; Mayo Clinic; Scottsdale AZ USA
| | - G. T. Furuta
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| | - J. C. Masterson
- Department of Pediatrics; Gastrointestinal Eosinophilic Diseases Program; Section of Pediatric Gastroenterology, Hepatology and Nutrition; University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Children's Hospital Colorado; Aurora CO USA
- Department of Medicine; Mucosal Inflammation Program; University of Colorado School of Medicine; Aurora CO USA
| |
Collapse
|
8
|
Campbell IK, Leong D, Edwards KM, Rayzman V, Ng M, Goldberg GL, Wilson NJ, Scalzo-Inguanti K, Mackenzie-Kludas C, Lawlor KE, Wicks IP, Brown LE, Baz Morelli A, Panousis C, Wilson MJ, Nash AD, McKenzie BS, Andrews AE. Therapeutic Targeting of the G-CSF Receptor Reduces Neutrophil Trafficking and Joint Inflammation in Antibody-Mediated Inflammatory Arthritis. THE JOURNAL OF IMMUNOLOGY 2016; 197:4392-4402. [DOI: 10.4049/jimmunol.1600121] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/08/2016] [Indexed: 01/01/2023]
|
9
|
Christensen AD, Haase C, Cook AD, Hamilton JA. Granulocyte colony-stimulating factor (G-CSF) plays an important role in immune complex-mediated arthritis. Eur J Immunol 2016; 46:1235-45. [DOI: 10.1002/eji.201546185] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/22/2015] [Accepted: 01/29/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Anne D. Christensen
- Department of Medicine; University of Melbourne; Parkville Victoria Australia
- Novo Nordisk A/S; Måløv Denmark
| | | | - Andrew D. Cook
- Department of Medicine; University of Melbourne; Parkville Victoria Australia
| | - John A. Hamilton
- Department of Medicine; University of Melbourne; Parkville Victoria Australia
| |
Collapse
|
10
|
Ribatti D. The discovery of angiogenic growth factors: the contribution of Italian scientists. Vasc Cell 2014; 6:8. [PMID: 24690161 PMCID: PMC3974417 DOI: 10.1186/2045-824x-6-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/26/2014] [Indexed: 12/31/2022] Open
Abstract
Angiogenesis is regulated, under both physiological and pathological conditions, by numerous “non-classic” pro-angiogenic factors, including fibroblast growth factor-2 (FGF-2), vascular endothelial growth factor (VEGF), and placental growth factor (PlGF), and “non-classic” pro-angiogenic factors, including granulocyte colony stimulating factor (G-CSF), granulocyte macrophage colony stimulating factor (GM-CSF), and erythropoietin (EPO). In the context of the most important discoveries in this field, this review article summarizes the important role played by the Italian scientists in the course of the last twenty years.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Policlinico - Piazza G, Cesare, 11, 70124 Bari, Italy.
| |
Collapse
|
11
|
Zgheib A, Pelletier-Bonnier É, Levros LC, Annabi B. Selective JAK/STAT3 signalling regulates transcription of colony stimulating factor-2 and -3 in Concanavalin-A-activated mesenchymal stromal cells. Cytokine 2013; 63:187-93. [PMID: 23688618 DOI: 10.1016/j.cyto.2013.04.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 03/28/2013] [Accepted: 04/23/2013] [Indexed: 12/13/2022]
|
12
|
Lee AS, Kim D, Wagle SR, Lee JE, Jung YJ, Kang KP, Lee S, Park SK, Kim W. Granulocyte colony-stimulating factor induces in vitro lymphangiogenesis. Biochem Biophys Res Commun 2013; 436:565-70. [DOI: 10.1016/j.bbrc.2013.05.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/13/2013] [Indexed: 01/05/2023]
|
13
|
Zgheib A, Lamy S, Annabi B. Epigallocatechin gallate targeting of membrane type 1 matrix metalloproteinase-mediated Src and Janus kinase/signal transducers and activators of transcription 3 signaling inhibits transcription of colony-stimulating factors 2 and 3 in mesenchymal stromal cells. J Biol Chem 2013; 288:13378-86. [PMID: 23548906 PMCID: PMC3650376 DOI: 10.1074/jbc.m113.456533] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/20/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND CSF-2 and CSF-3 confer proangiogenic and immunomodulatory properties to mesenchymal stromal cells (MSCs). RESULTS Transcriptional regulation of CSF-2 and CSF-3 in concanavalin A-activated MSCs requires MT1-MMP signaling and is inhibited by EGCG. CONCLUSION The chemopreventive properties of diet-derived EGCG alter MT1-MMP-mediated intracellular signaling. SIGNIFICANCE Pharmacological targeting of MSCs proangiogenic functions may prevent their contribution to tumor development. Epigallocatechin gallate (EGCG), a major form of tea catechins, possesses immunomodulatory and antiangiogenic effects, both of which contribute to its chemopreventive properties. In this study, we evaluated the impact of EGCG treatment on the expression of colony-stimulating factors (CSF) secreted from human bone marrow-derived mesenchymal stromal cells (MSCs), all of which also contribute to the immunomodulatory and angiogenic properties of these cells. MSCs were activated with concanavalin A (ConA), a Toll-like receptor (TLR)-2 and TLR-6 agonist as well as a membrane type 1 matrix metalloproteinase (MT1-MMP) inducer, which increased granulocyte macrophage-CSF (GM-CSF, CSF-2), granulocyte CSF (G-CSF, CSF-3), and MT1-MMP gene expression. EGCG antagonized the ConA-induced CSF-2 and CSF-3 gene expression, and this process required an MT1-MMP-mediated sequential activation of the Src and JAK/STAT pathways. Gene silencing of MT1-MMP expression further demonstrated its requirement in the phosphorylation of Src and STAT3, whereas overexpression of a nonphosphorylatable MT1-MMP mutant (Y573F) abrogated CSF-2 and CSF-3 transcriptional increases. Given that MSCs are recruited within vascularizing tumors and are believed to contribute to tumor angiogenesis, possibly through secretion of CSF-2 and CSF-3, our study suggests that diet-derived polyphenols such as EGCG may exert chemopreventive action through pharmacological targeting of the MT1-MMP intracellular signaling.
Collapse
Affiliation(s)
- Alain Zgheib
- From the Laboratoire d'Oncologie Moléculaire, Centre de Recherche BIOMED, Département de Chimie, Université du Québec à Montreal, Montreal, Quebec, Canada H3C 3P8
| | - Sylvie Lamy
- From the Laboratoire d'Oncologie Moléculaire, Centre de Recherche BIOMED, Département de Chimie, Université du Québec à Montreal, Montreal, Quebec, Canada H3C 3P8
| | - Borhane Annabi
- From the Laboratoire d'Oncologie Moléculaire, Centre de Recherche BIOMED, Département de Chimie, Université du Québec à Montreal, Montreal, Quebec, Canada H3C 3P8
| |
Collapse
|
14
|
Sessa R, Seano G, di Blasio L, Gagliardi PA, Isella C, Medico E, Cotelli F, Bussolino F, Primo L. The miR-126 regulates Angiopoietin-1 signaling and vessel maturation by targeting p85β. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1925-35. [DOI: 10.1016/j.bbamcr.2012.07.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 06/18/2012] [Accepted: 07/23/2012] [Indexed: 10/28/2022]
|
15
|
Jana S, Patel H. Expression of human granulocyte colony stimulating factor (hG-CSF) in colon adenocarcinoma cell line (Caco-2). Biotechnol Lett 2012; 34:1791-6. [PMID: 22714276 PMCID: PMC3483470 DOI: 10.1007/s10529-012-0977-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 05/25/2012] [Indexed: 11/25/2022]
Abstract
Growth and progression of many cancer cells are mediated by alterations in the microenvironment often caused by an aberrant expression of growth factors and receptors. There is no report on expression of growth factor granulocyte colony-stimulating factor (G-CSF) in the experimental model, colon adenocarcinoma cell line (Caco2), that is commonly used in drug permeability assays. We hypothesize that in vitro, the Caco2 model is associated with a constitutive neo-expression of the hematopoietic G-CSF thereby causing an autocrine stimulation of Caco2 growth and proliferation in vitro. To test our hypothesis, we analyzed mRNA and protein expression of G-CSF in Caco2 cells using reverse transcriptase-PCR and SDS-PAGE. G-CSF mRNA and protein were detected in Caco2 cells. Expression of G-CSF protein was similar at different passages of this cell line. The expression of G-CSF has a significant role in the autocrine regulation of Caco2 cell growth and proliferation.
Collapse
Affiliation(s)
- Snehasis Jana
- Drug Metabolism and Pharmacokinetics-Toxicology Division, Sai Advantium Pharma Ltd, Building 1, Plot No 2, Chrysalis Enclave, International Biotech Park, Phase-II, Hinjewadi, Pune 411057, India.
| | | |
Collapse
|
16
|
Cavalloni G, Peraldo-Neia C, Sarotto I, Gammaitoni L, Migliardi G, Soster M, Marchiò S, Aglietta M, Leone F. Antitumor activity of Src inhibitor saracatinib (AZD-0530) in preclinical models of biliary tract carcinomas. Mol Cancer Ther 2012; 11:1528-38. [PMID: 22452946 DOI: 10.1158/1535-7163.mct-11-1020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Biliary tract carcinoma (BTC) has a poor prognosis due to limited treatment options. There is, therefore, an urgent need to identify new targets and to design innovative therapeutic approaches. Among potential candidate molecules, we evaluated the nonreceptor tyrosine kinase Src, observing promising antitumor effects of its small-molecule inhibitor saracatinib in BTC preclinical models. The presence of an active Src protein was investigated by immunohistochemistry in 19 surgical samples from patients with BTC. Upon saracatinib treatment, the phosphorylation of Src and of its downstream transducers was evaluated in the BTC cell lines TFK-1, EGI-1, HuH28, and TGBC1-TKB. The effect of saracatinib on proliferation and migration was analyzed in these same cell lines, and its antitumor activity was essayed in EGI-1 mouse xenografts. Saracatinib-modulated transcriptome was profiled in EGI-1 cells and in tumor samples of the xenograft model. Src was activated in about 80% of the human BTC samples. In cultured BTC cell lines, low-dose saracatinib counteracted the activation of Src and of its downstream effectors, increased the fraction of cells in G(0)-G(1) phase, and inhibited cell migration. At high concentrations (median dose from 2.26-6.99 μmol/L), saracatinib was also capable of inhibiting BTC cell proliferation. In vivo, saracatinib treatment resulted in delayed tumor growth, associated with an impaired vascular network. Here, we provide a demonstration that the targeted inhibition of Src kinase by saracatinib is of therapeutic benefit in preclinical models of BTC. We propose our results as a basis for the design of saracatinib-based clinical applications.
Collapse
Affiliation(s)
- Giuliana Cavalloni
- Department of Medical Oncology, University of Turin Medical School, Turin, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Angiogenic activity of classical hematopoietic cytokines. Leuk Res 2012; 36:537-43. [PMID: 22386730 DOI: 10.1016/j.leukres.2012.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 01/24/2012] [Accepted: 02/07/2012] [Indexed: 12/24/2022]
Abstract
Hematopoiesis is regulated by several cytokines with pleiotropic activity. Several evidences have clearly demonstrated that these molecules, formerly regarded as specific for the hematopoietic system, also affect certain endothelial cell functions and that hematopoietic factors clearly influence angiogenesis. This review article summarizes the most important literature data concerning this inconvertible relationship.
Collapse
|
18
|
A key role for G-CSF-induced neutrophil production and trafficking during inflammatory arthritis. Blood 2008; 112:5193-201. [PMID: 18824600 DOI: 10.1182/blood-2008-02-139535] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
We have previously shown that G-CSF-deficient (G-CSF(-/-)) mice are markedly protected from collagen-induced arthritis (CIA), which is the major murine model of rheumatoid arthritis, and now investigate the mechanisms by which G-CSF can promote inflammatory disease. Serum G-CSF levels were significantly elevated during CIA. Reciprocal bone marrow chimeras using G-CSF(-/-), G-CSFR(-/-), and wild-type (WT) mice identified nonhematopoietic cells as the major producers of G-CSF and hematopoietic cells as the major responders to G-CSF during CIA. Protection against CIA was associated with relative neutropenia. Depletion of neutrophils or blockade of the neutrophil adhesion molecule, Mac-1, dramatically attenuated the progression of established CIA in WT mice. Intravital microscopy of the microcirculation showed that both local and systemic administration of G-CSF significantly increased leukocyte trafficking into tissues in vivo. G-CSF-induced trafficking was Mac-1 dependent, and G-CSF up-regulated CD11b expression on neutrophils. Multiphoton microscopy of synovial vessels in the knee joint during CIA revealed significantly fewer adherent Gr-1(+) neutrophils in G-CSF(-/-) mice compared with WT mice. These data confirm a central proinflammatory role for G-CSF in the pathogenesis of inflammatory arthritis, which may be due to the promotion of neutrophil trafficking into inflamed joints, in addition to G-CSF-induced neutrophil production.
Collapse
|
19
|
Pamphilon D, Nacheva E, Navarrete C, Madrigal A, Goldman J. The use of granulocytecolony-stimulating factor in volunteer unrelated hemopoietic stem cell donors. Transfusion 2008; 48:1495-501. [DOI: 10.1111/j.1537-2995.2008.01694.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Scott G, Bowman S, Smith T, Flower R, Bolton C. Glutamate-stimulated peroxynitrite production in a brain-derived endothelial cell line is dependent on N-methyl-D-aspartate (NMDA) receptor activation. Biochem Pharmacol 2007; 73:228-36. [PMID: 17118345 PMCID: PMC1855445 DOI: 10.1016/j.bcp.2006.09.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Revised: 09/19/2006] [Accepted: 09/20/2006] [Indexed: 11/04/2022]
Abstract
There is accumulating and convincing evidence indicating a role for glutamate in the pathogenesis of the human demyelinating disease multiple sclerosis (MS). Studies in experimental autoimmune encephalomyelitis (EAE), the animal model of MS, demonstrate that pharmacological inhibition of specific glutamate receptors suppresses neurological symptoms and prevents blood-brain barrier (BBB) breakdown. The mechanisms through which glutamate influences BBB function during EAE remain unclear. Glutamate triggers the production of nitric oxide and superoxide, which can lead to the formation of peroxynitrite (ONOO(-)). Recent studies have implicated ONOO(-) in the loss of neurovascular integrity during EAE. We propose that glutamate contributes to BBB breakdown via the actions of ONOO(-). The present investigation examined glutamate-induced ONOO(-) formation in the b.End3 brain-derived endothelial cell line. b.End3 cells were incubated with a concentration range of glutamate and ONOO(-) production was assessed over time. Results showed a concentration- and time-dependent increase in ONOO(-) levels in glutamate-treated cells that were suppressed by selective and non-selective inhibitors of ONOO(-)-mediated reactions. Specific activation of b.End3-associated NMDA receptors also resulted in a concentration-dependent increase in ONOO(-) production. The ability of b.End3 cells to respond to the presence of glutamate was confirmed through the detection of NMDA receptor immnuoreactivity in cell extracts. In addition, the use of the NMDA receptor antagonists MK-801 and memantine reduced glutamate-mediated ONOO(-) generation from b.End3 cells. The data reinforce the important relationship between glutamate and the NMDA receptor, positioned at neurovascular sites, which may be of particular relevance to the pathogenesis of demyelinating disease.
Collapse
Affiliation(s)
- G.S. Scott
- Centre of Biochemical Pharmacology & Experimental Pathology, The William Harvey Research Institute, St. Bartholomew's Hospital Medical College and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - S.R. Bowman
- Centre of Biochemical Pharmacology & Experimental Pathology, The William Harvey Research Institute, St. Bartholomew's Hospital Medical College and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - T. Smith
- NeuMatRx, Truro, Cornwall TR3 6NT, United Kingdom
| | - R.J. Flower
- Centre of Biochemical Pharmacology & Experimental Pathology, The William Harvey Research Institute, St. Bartholomew's Hospital Medical College and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - C. Bolton
- Centre of Biochemical Pharmacology & Experimental Pathology, The William Harvey Research Institute, St. Bartholomew's Hospital Medical College and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
21
|
Nishida M, Hamaoka K. How Does G-CSF Act on the Kidney during Acute Tubular Injury? ACTA ACUST UNITED AC 2006; 104:e123-8. [PMID: 16902315 DOI: 10.1159/000094962] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent findings in stem cell research have demonstrated multi-lineage plasticity of bone marrow cells, and also the contribution of hematopoietic bone marrow stem cells to the regeneration of injured organs including the kidney. These findings suggested the possibility of the use of granulocyte colony-stimulating factor (G-CSF) as a therapeutic option to regenerate injured organs. Recently, several studies regarding the effect of G-CSF on renal function have been reported in mouse models of acute renal failure. This series of experiments provided potentially important information regarding the treatment of patients with renal injury. This review summarizes the possible actions of G-CSF on the kidney, especially during acute tubular injury caused by toxic or ischemic insults.
Collapse
Affiliation(s)
- Masashi Nishida
- Department of Pediatric Cardiology and Nephrology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan.
| | | |
Collapse
|
22
|
Capoccia BJ, Shepherd RM, Link DC. G-CSF and AMD3100 mobilize monocytes into the blood that stimulate angiogenesis in vivo through a paracrine mechanism. Blood 2006; 108:2438-45. [PMID: 16735597 PMCID: PMC1895560 DOI: 10.1182/blood-2006-04-013755] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
There is compelling evidence that circulating angiogenic cells exist that are able to home to sites of vascular injury and stimulate angiogenesis. However, the number of angiogenic cells in the blood is low, limiting their delivery to sites of ischemia. Treatment with certain cytokines may mobilize angiogenic cells into the blood, potentially circumventing this limitation. Herein, we show that treatment with granulocyte colony-stimulating factor (G-CSF) or AMD3100, a novel CXCR4 antagonist, significantly stimulated angiogenesis in a murine model of acute hindlimb ischemia. The kinetics of angiogenic-cell mobilization by these agents appears to be distinct, with more rapid revascularization observed in AMD3100-treated mice. Combination treatment with G-CSF and AMD3100 resulted in the earliest and most complete recovery in blood flow to the ischemic hindlimb. Adoptive transfer of mobilized blood mononuclear cells, while potently stimulating angiogenesis, did not result in the significant incorporation of donor cells into the neoendothelium. Cell-fractionation studies showed that it is the monocyte population in the blood that mediates angiogenesis in this model. Collectively, these data suggest that monocytes mobilized into the blood by G-CSF or AMD3100 stimulate angiogenesis at sites of ischemia through a paracrine mechanism.
Collapse
Affiliation(s)
- Benjamin J Capoccia
- Division of Oncology, Washington University School of Medicine, Box 8007, 660 South Euclid Ave, St Louis, MO 63110, USA
| | | | | |
Collapse
|
23
|
Ohki Y, Heissig B, Sato Y, Akiyama H, Zhu Z, Hicklin DJ, Shimada K, Ogawa H, Daida H, Hattori K, Ohsaka A. Granulocyte colony-stimulating factor promotes neovascularization by releasing vascular endothelial growth factor from neutrophils. FASEB J 2005; 19:2005-7. [PMID: 16223785 DOI: 10.1096/fj.04-3496fje] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The granulocyte colony-stimulating factor (G-CSF) promotes angiogenesis. However, the exact mechanism is not known. We demonstrate that vascular endothelial growth factor (VEGF) was released by Gr-1+CD11b- neutrophils but not Gr-1-CD11b+ monocytes prestimulated with G-CSF in vitro and in vivo. Similarly, in vivo, concomitant with an increase in neutrophil numbers in circulation, G-CSF augmented plasma VEGF level in vivo. Local G-CSF administration into ischemic tissue increased capillary density and provided a functional vasculature and contributed to neovascularization of ischemic tissue. Blockade of the VEGF pathway abrogated G-CSF-induced angiogenesis. On the other hand, as we had shown previously, VEGF can induce endothelial progenitor cell (EPC) mobilization. Here, we show that G-CSF also augmented the number of circulating VEGF receptor-2 (VEGFR2) EPCs as compared with untreated controls. Blocking the VEGF/VEGFR1, but to a much lesser extent, the VEGF/VEGFR2 pathway in G-CSF-treated animals delayed tissue revascularization in a hindlimb model. These data clearly show that G-CSF modulates angiogenesis by increasing myelomonocytic cells (VEGFR1+ neutrophils) and their release of VEGF. Our results indicated that administration of G-CSF into ischemic tissue provides a novel and safe therapeutic strategy to improve neovascularization.
Collapse
Affiliation(s)
- Yuichi Ohki
- Department of Cardiology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Yang X, Liu F, Xu Z, Chen C, Wu X, Li G, Li J. Expression of granulocyte colony stimulating factor receptor in human colorectal cancer. Postgrad Med J 2005; 81:333-7. [PMID: 15879049 PMCID: PMC1743273 DOI: 10.1136/pgmj.2004.024646] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIMS To discover if human colorectal cancer expresses granulocyte colony stimulating factor receptor (G-CSFR) and if granulocyte colony stimulating factor (G-CSF) plays an important part in the development and progression of human colorectal cancer. METHODS Forty two specimens of colorectal cancer and normal colorectal mucosa were investigated, taken from the colon or rectum in group of colorectal cancer patients. Immunohistochemistry and reverse transcription polymerase chain reaction (RT-PCR) technique were used to show G-CSFR expression. The relation between expression of G-CSFR and clinical or pathological factors was analysed. RESULTS Immucohistochemical analyses showed that G-CSFR was expressed in the human colorectal cancer (25 of 42, 59.52%) and seemed to be up-regulated compared with the normal mucosa (14 of 42, 33.33%, p<0.001). In pronounced contrast with mostly strongly positive tumours, corresponding normal colorectal mucosa was negative or weakly positive. A significant correlation was found between G-CSFR expression and tumour stage (p = 0.001), tumour differentiation (p<0.001), but there was no significant relation between the expression of G-CSFR and the age, sex, and tumour size (p = 0.346, p = 0.686, p = 0.459). In RT-PCR, 21 of all 42 tumours had G-CSFR mRNA expression, while only 11 of 42 normal colorectal mucosa had such expression. CONCLUSIONS These data show that G-CSFR is commonly expressed in human colorectal cancers, thus supporting a possible role for G-CSF in colorectal cancer physiology.
Collapse
Affiliation(s)
- X Yang
- Medical College, Nanjing University, Jingsu, China.
| | | | | | | | | | | | | |
Collapse
|
25
|
Iwanaga K, Takano H, Ohtsuka M, Hasegawa H, Zou Y, Qin Y, Odaka K, Hiroshima K, Tadokoro H, Komuro I. Effects of G-CSF on cardiac remodeling after acute myocardial infarction in swine. Biochem Biophys Res Commun 2005; 325:1353-9. [PMID: 15555576 DOI: 10.1016/j.bbrc.2004.10.149] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2004] [Indexed: 10/26/2022]
Abstract
We examined whether granulocyte colony-stimulating factor (G-CSF) prevents cardiac dysfunction and remodeling after myocardial infarction (MI) in large animals. MI was produced by ligation of left anterior descending coronary artery in swine. G-CSF (10 microg/kg/day, once a day) was injected subcutaneously from 24h after ligation for 7 days. Echocardiographic examination revealed that the G-CSF treatment induced improvement of cardiac function and attenuation of cardiac remodeling at 4 weeks after MI. In the ischemic region, the number of apoptotic endothelial cells was smaller and the number of vessels was larger in the G-CSF treatment group than in control group. Moreover, vascular endothelial growth factor was more abundantly expressed and Akt was more strongly activated in the ischemic region of the G-CSF treatment group than of control group. These findings suggest that G-CSF prevents cardiac dysfunction and remodeling after MI in large animals.
Collapse
Affiliation(s)
- Koji Iwanaga
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Hamamoto M, Tomita S, Nakatani T, Yutani C, Yamashiro S, Sueda T, Yagihara T, Kitamura S. Granulocyte-colony stimulating factor directly enhances proliferation of human troponin I-positive cells derived from idiopathic dilated cardiomyopathy through specific receptors. J Heart Lung Transplant 2004; 23:1430-7. [PMID: 15607674 DOI: 10.1016/j.healun.2003.09.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2003] [Accepted: 09/18/2003] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Our previous study showed that granulocyte-colony stimulating factor (G-CSF) enhanced bone-marrow-cell migration into the injured heart and that bone-marrow cells differentiated into cardiomyocytes. However, the number of bone-marrow-derived cardiomyocytes seems too small to have a direct, positive impact on pump function. Therefore, we hypothesized that G-CSF directly could affect the host myocardium through G-CSF receptors (G-CSFRs). METHODS In experiment 1, we cultured normal mouse heart cells with G-CSF at concentrations of 0, 1, 10, 50, and 100 ng/ml. In experiment 2, we cultured heart cells derived from a recipient with idiopathic cardiomyopathy (IDCM) after heart transplantation. We compared the total number of heart cells and Ki67- and troponin I (TnI)-positive cells with/without G-CSF at 50 ng/ml. We also performed immunochemical staining of the heart specimen from a recipient with IDCM using a rabbit polyclonal anti-G-CSFR antibody. RESULTS In experiment 1, mouse heart cells with G-CSF (50 ng/ml) proliferated maximally. In experiment 2, the total numbers of heart cells, Ki67-positive cells. TnI-positive cells, Ki67- and TnI-double-positive cells in the G-CSF group were greater than those in the non-G-CSF group at Days 14 and 28 (p <0.05). In the IDCM heart, G-CSFRs on cardiomyocytes were expressed heterogeneously and widely. CONCLUSIONS Granulocyte-colony stimulating factor directly enhanced the proliferation of TnI-positive cells derived from a recipient with IDCM through the G-CSFR.
Collapse
Affiliation(s)
- Masaki Hamamoto
- Department of Regenerative Medicine and Tissue Engineering, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Dose-dense adjuvant breast cancer chemotherapy is a new treatment strategy that aims to improve tumour control by using more frequent cytotoxic dosing together with continuous granulocyte colony-stimulating factor (G-CSF) to minimize neutropaenia. In addition to stimulating neutrophil proliferation, G-CSF mobilizes neutrophils from the bone marrow through proteolytic disruption of the chemokine receptor CXCR4 and its chemotactic ligand CXCL12. As breast cancers also express CXCR4 and oestrogen induces CXCL12, the success of dose-dense treatment could partly reflect inhibition of CXCR4-dependent micrometastatic homing and/or paracrine survival, and suggests a benefit of adjuvant oestrogen suppression for patients with oestrogen-receptor-negative, CXCR4-positive disease.
Collapse
Affiliation(s)
- Richard J Epstein
- Department of Medicine, The University of Hong Kong, Room 404, Professorial Block, Queen Mary Hospital, Pokfulam Road, Hong Kong.
| |
Collapse
|
28
|
Zhang Y, Woodward VK, Shelton JM, Richardson JA, Zhou XJ, Link D, Kielar ML, Jeyarajah DR, Lu CY. Ischemia-reperfusion induces G-CSF gene expression by renal medullary thick ascending limb cells in vivo and in vitro. Am J Physiol Renal Physiol 2004; 286:F1193-201. [PMID: 14734360 DOI: 10.1152/ajprenal.00379.2002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ischemic acute renal failure involves not only the kidney but also extrarenal organs such as the bone marrow that produces inflammatory cells. By ELISA and RNase protection assays, we now show that renal ischemia-reperfusion increases serum concentrations of granulocyte macrophage colony-stimulating factor (G-CSF) protein and increases both G-CSF mRNA and protein in the ischemic kidney. In situ hybridization localized the increased G-CSF mRNA to tubule cells, including medullary thick ascending limb cells (mTAL), in the outer medulla. We also show that mTAL produce G-CSF protein and increase G-CSF mRNA after stimulation by reactive oxygen species in vitro. The production of G-CSF by the kidney after ischemia-reperfusion provides a means of communication from the injured kidney to the bone marrow. This supports the known inflammatory response to ischemia.
Collapse
Affiliation(s)
- Ying Zhang
- Div. of Nephrology (Internal Medicine Univ. of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8856, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Romiti E, Meacci E, Donati C, Formigli L, Zecchi-Orlandini S, Farnararo M, Ito M, Bruni P. Neutral ceramidase secreted by endothelial cells is released in part associated with caveolin-1. Arch Biochem Biophys 2003; 417:27-33. [PMID: 12921776 DOI: 10.1016/s0003-9861(03)00212-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neutral ceramidase (CDase) is a key enzyme of sphingomyelin (SM) metabolism implicated in cell signaling triggered by a variety of extracellular ligands. Previously it was shown that in murine endothelial cells a portion of neutral CDase is localized in detergent-resistant light membranes. In this study subcellular distribution of neutral CDase was further investigated. In accordance with the previous finding, the enzyme was identified in caveolae. Moreover, the same protein was detected in medium-speed supernatant of cell-conditioned medium, accounting for CDase activity measurable in the medium at neutral pH. Notably, these cells released also the caveolae-scaffolding protein caveolin-1 (cav-1). Interestingly, secreted neutral CDase and cav-1 coimmunoprecipitated. In addition, acid sphingomyelinase (SMase) activity was detectable in cav-1 immunocomplexes. These findings are consistent with the view that neutral CDase is released, in part, in association with cav-1 together with acid SMase. It remains to be established whether the here-identified secreted cav-1-enriched complex acts as platform to facilitate extracellular metabolism of SM.
Collapse
Affiliation(s)
- Elena Romiti
- Dipartimento di Scienze Biochimiche, Università degli Studi di Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Heeschen C, Aicher A, Lehmann R, Fichtlscherer S, Vasa M, Urbich C, Mildner-Rihm C, Martin H, Zeiher AM, Dimmeler S. Erythropoietin is a potent physiologic stimulus for endothelial progenitor cell mobilization. Blood 2003; 102:1340-6. [PMID: 12702503 DOI: 10.1182/blood-2003-01-0223] [Citation(s) in RCA: 667] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence suggests that postnatal neovascularization involves the recruitment of circulating endothelial progenitor cells (EPCs). Hematopoietic and endothelial cell lineages share common progenitors. Cytokines formerly thought to be specific for the hematopoietic system have only recently been shown to affect several functions in endothelial cells. Accordingly, we investigated the stimulatory potential of erythropoietin (Epo) on EPC mobilization and neovascularization. The bone marrow of Epo-treated mice showed a significant increase in number and proliferation of stem and progenitor cells as well as in colony-forming units. The number of isolated EPCs and CD34+/flk-1+ precursor cells was significantly increased in spleen and peripheral blood of Epo-treated mice compared with phosphate-buffered saline-treated mice. In in vivo models of postnatal neovascularization, Epo significantly increased inflammation- and ischemia-induced neovascularization. The physiologic relevance of these findings was investigated in patients with coronary heart disease. In a multivariate regression model, serum levels of Epo and vascular endothelial growth factor were significantly associated with the number of stem and progenitor cells in the bone marrow as well as with the number and function of circulating EPCs. In conclusion, the present study suggests that Epo stimulates postnatal neovascularization at least in part by enhancing EPC mobilization from the bone marrow. Epo appears to physiologically regulate EPC mobilization in patients with ischemic heart disease. Thus, Epo serum levels may help in identifying patients with impaired EPC recruitment capacity.
Collapse
Affiliation(s)
- Christopher Heeschen
- Department of Internal Medicine IV and Hematology, University of Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hall AV, Jevnikar AM. Significance of endothelial cell survival programs for renal transplantation. Am J Kidney Dis 2003; 41:1140-54. [PMID: 12776265 DOI: 10.1016/s0272-6386(03)00345-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Initial and longer term kidney transplant function is determined in part by the renal allograft microcirculation because it provides a thromboresistant surface, regulates cellular infiltration, and elaborates paracrine and autocrine growth and survival factors. Loss of endothelial-derived signaling mediators accelerates vascular injury and endothelial cell (EC) death. EC apoptosis is implicated in accelerated allograft vasculopathy and premature loss of organ function. Renal allograft EC injury and replacement by recipient-derived repair mechanisms has long been proposed to influence allograft acceptance and function. Repair of cellular injury in allografts is linked with cell-survival mechanisms, but few precise indicators exist to predict recovery and repair in organ transplants. The significance of the growth phenotype of the microvascular endothelium for acute and longer term renal allograft survival is presented.
Collapse
Affiliation(s)
- Anne V Hall
- Division of Nephrology and Multi-Organ Transplant Program, University of Western Ontario, London Health Sciences Centre, London, Ontario, Canada
| | | |
Collapse
|
32
|
Semerad CL, Liu F, Gregory AD, Stumpf K, Link DC. G-CSF is an essential regulator of neutrophil trafficking from the bone marrow to the blood. Immunity 2002; 17:413-23. [PMID: 12387736 DOI: 10.1016/s1074-7613(02)00424-7] [Citation(s) in RCA: 324] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neutrophils are released from the bone marrow in a regulated fashion to maintain homeostatic levels in the blood and to respond to physiological stresses, including infection. We show that under basal conditions granulocyte colony-stimulating factor (G-CSF) is an essential regulator of neutrophil release from the bone marrow. Nonredundant signals generated by the membrane-proximal 87 amino acids of the G-CSF receptor (G-CSFR) are sufficient to mediate this response. Surprisingly, G-CSFR expression on neutrophils is neither necessary nor sufficient for their mobilization from the bone marrow, suggesting that G-CSF induces neutrophil mobilization indirectly through the generation of trans-acting signals. Evidence is provided suggesting that downregulation of stromal cell-derived factor 1 expression in the bone marrow may represent such a signal.
Collapse
Affiliation(s)
- Craig L Semerad
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
33
|
Pu JJ, Li C, Rodriguez M, Banerjee D. Cloning and structural characterization of ECTACC, a new member of the transforming acidic coiled coil (TACC) gene family: cDNA sequence and expression analysis in human microvascular endothelial cells. Cytokine 2001; 13:129-37. [PMID: 11161455 DOI: 10.1006/cyto.2000.0812] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Erythropoietin (Epo) transduces mitogenic and chemoattractant signals to human endothelial cells. Identifications of Epo-responsive genes are important for understanding the molecular nature of Epo signaling in endothelial cells. The effects of Epo on differential expression of various genes were examined in human microvascular endothelial cells (HMVEC) by differential display reverse transcriptase polymerase chain reaction (RT-PCR). In the current study we obtained from Epo-treated HMVEC a cDNA fragment with characteristics of the 3' end of mRNA. Using the cDNA fragment, we then selectively isolated a full-length clone by screening an unamplified endothelial cell cDNA library followed by 5' rapid amplification of cDNA ends by polymerase chain reaction (RACE-PCR). The nucleotide sequence of the longest cDNA revealed an open reading frame of 3311 nucleotides that encodes a protein consisting of approximately 906 amino acids with a predicted MW of approximately 100 kDa. The nucleotide sequence of the cDNA is nearly identical to that of transforming acidic coiled coil-containing (TACC2) and anti-zuai-1 (AZU-1) cDNA clones except at the 5'- and 3'-ends. Northern blot analysis showed an increase in endothelial-TACC-related mRNA levels in Epo-treated cells in comparison to that of the control cells. Endothelial-TACC-related mRNA was highly expressed in heart and skeletal muscle tissue. Placenta and brain tissue exhibited low levels of expression of endothelial-TACC-related gene. Southern blot analysis of genomic DNA from somatic cell hybrids showed that endothelial-TACC-related cDNA maps to chromosome 10. Immunofluorescence microscopy and the occurrence of several putative phosphorylation and SH3 binding sites on the deduced protein suggest that endothelial-TACC-related protein may be involved in Epo signaling cascades in endothelial cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Carrier Proteins
- Cells, Cultured
- Chromosome Mapping
- Chromosomes, Human, Pair 10/genetics
- Cloning, Molecular
- Cricetinae
- DNA, Complementary/biosynthesis
- DNA, Complementary/isolation & purification
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Fetal Proteins
- Fluorescent Antibody Technique, Indirect
- Gene Amplification
- Gene Expression Profiling
- Gene Library
- Humans
- Mice
- Microcirculation/chemistry
- Microcirculation/cytology
- Microcirculation/metabolism
- Microtubule-Associated Proteins/chemistry
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/isolation & purification
- Microtubule-Associated Proteins/metabolism
- Molecular Sequence Data
- Multigene Family
- Nuclear Proteins
- RNA, Messenger/biosynthesis
- Sequence Analysis, DNA
- Tumor Suppressor Proteins
Collapse
Affiliation(s)
- J J Pu
- Department of Membrane Biochemistry II, The Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10021, USA
| | | | | | | |
Collapse
|
34
|
Barbucci R, Lamponi S, Magnani A, Peluso G, Petillo O. Metal complexes with linear and crosslinked polysaccharides as mediators of angiogenesis. POLYM ADVAN TECHNOL 2001. [DOI: 10.1002/pat.141] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Marelli-Berg FM, Peek E, Lidington EA, Stauss HJ, Lechler RI. Isolation of endothelial cells from murine tissue. J Immunol Methods 2000; 244:205-15. [PMID: 11033033 DOI: 10.1016/s0022-1759(00)00258-1] [Citation(s) in RCA: 156] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The isolation and long-term culture of murine endothelial cells (ECs) has often proven a difficult task. In this paper we describe a quick, efficient protocol for the isolation of microvascular endothelial cells from murine tissues. Murine lung or heart are mechanically minced and enzymatically digested with collagenase and trypsin. The single cell suspension obtained is then incubated with an anti-CD31 antibody, anti-CD105 antibody and with biotinylated isolectin B-4. Pure EC populations are finally obtained by magnetic bead separation using rat anti-mouse Ig- and streptavidin-conjugated microbeads. EC cultures are subsequently expanded and characterised. The surface molecule expression by the primary cultures of murine EC obtained from lung and heart tissue is analysed and compared to that of a murine endothelioma and of primary cultures of murine renal tubular epithelial cells. The phenotype and morphology of these cultures remain stable over 10-15 passages in culture, and no overgrowth of contaminating cells of non-endothelial origin is observed at any stage.
Collapse
Affiliation(s)
- F M Marelli-Berg
- Department of Immunology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, W12 ONN, London, UK.
| | | | | | | | | |
Collapse
|
36
|
Barbucci R, Magnani A, Lamponi S, Mitola S, Ziche M, Morbidelli L, Bussolino F. Cu(II) and Zn(II) complexes with hyaluronic acid and its sulphated derivative. Effect on the motility of vascular endothelial cells. J Inorg Biochem 2000; 81:229-37. [PMID: 11065187 DOI: 10.1016/s0162-0134(00)00127-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
With the aim of improving the compatibility of biomaterials to be used for the construction of cardiovascular prosthesis, we have designed bioactive macromolecules resulting from chemical modifications of hyaluronic acid (Hyal). The stability constants of Cu(II) and Zn(II) complexes with the sulphated derivative of hyaluronic acid (HyalS3.5) were evaluated. Two different complexes have been found for each metal ion, CuL, Cu(OH)2L and ZnL, Zn(OH)2L (L means the disaccharide unit of the ligands) in aqueous solution at 37 degrees C. The dihydroxo Cu(II) complex was present in high percentage at pH=7.4. On the contrary, the Zn(II) ion was present with a relatively low percentage of both complexes. The ability to stimulate endothelial cell adhesion and migration was evaluated for Hyal, HyalS3.5 and their complexes with Cu(II) and Zn(II) ions. The results revealed that Hyal and [Cu(OH)2HyalS3.5](4.5)- induced cell adhesion, while [ZnHyalS3.5](2.5)- and [Zn(OH)2HyalS3.5](4.5)- inhibited the process. The chemotactic activity of increasing concentrations of the above complexes was also evaluated, demonstrating that [Cu(OH)2HyalS3.5](4.5)- complex at 1 microM concentration was the most active in inducing cell migration. These results have been also strengthened by analysing adherent cell migration in agarose. In conclusion, sulphated hyaluronic acid coordinated to Cu(II) seems to be a promising matrix molecule for the construction of cardiovascular prosthesis.
Collapse
Affiliation(s)
- R Barbucci
- Department of Chemical and Biosystem Sciences and Technologies, University of Siena, Italy.
| | | | | | | | | | | | | |
Collapse
|
37
|
Bhattacharya V, Shi Q, Ishida A, Sauvage LR, Hammond WP, Wu MH. Administration of granulocyte colony-stimulating factor enhances endothelialization and microvessel formation in small-caliber synthetic vascular grafts. J Vasc Surg 2000; 32:116-23. [PMID: 10876212 DOI: 10.1067/mva.2000.107308] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The purpose of this study was to determine whether systemic administration of granulocyte colony-stimulating factor (G-CSF) would promote endothelialization for small-caliber Dacron vascular grafts. METHODS We implanted 4-mm preclotted Dacron grafts in both carotids of 12 dogs. For a fair comparison, all dogs had a comparable platelet aggregation profile with platelet aggregation scores less than 30. Five dogs served as controls, and the others were given 7-day subcutaneous injections of G-CSF (10 microg/kg per day), starting on the seventh postoperative day. The effect of G-CSF was evaluated by white blood cell count, which showed a 3.7-fold (+/- 2.7-fold) increase at the end of treatment. Grafts were harvested at 4 weeks. All G-CSF grafts were patent, and one control occluded. Endothelial-like cell coverage averaged 80.8% on G-CSF grafts, but only 35.6% for control grafts (P <.0004). With the exclusion of the anastomotic pannus healing factor, the difference in endothelial-like cell coverage was even greater (68.5% vs 9.8%; P <.0001). Immunocytochemical staining and electron microscopy studies demonstrated endothelial cells. Light microscopy also showed that there were more microvessels on and in the G-CSF grafts than in the control grafts. This study suggests that G-CSF can enhance early endothelialization of small-caliber vascular grafts. Further studies to determine the proper dosage and timing are needed before clinical application can be recommended.
Collapse
Affiliation(s)
- V Bhattacharya
- Hope Heart Institute, Providence Seattle Medical Center, Seattle, WA, USA
| | | | | | | | | | | |
Collapse
|
38
|
Expression of the G-CSF receptor on hematopoietic progenitor cells is not required for their mobilization by G-CSF. Blood 2000. [DOI: 10.1182/blood.v95.10.3025] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The mechanisms that regulate hematopoietic progenitor cell (HPC) mobilization from the bone marrow to blood have not yet been defined. HPC mobilization by granulocyte colony-stimulating factor (G-CSF), cyclophosphamide (CY), or interleukin-8 but not flt-3 ligand is markedly impaired in G-CSF receptor–deficient (G-CSFR–deficient) mice. G-CSFR is expressed on mature hematopoietic cells, HPCs, and stromal cells, which suggests that G-CSFR signals in one or more of these cell types was required for mobilization by these agents. To define the cell type(s) responsible for G-CSF–dependent mobilization, a series of chimeric mice were generated using bone marrow transplantation. Mobilization studies in these chimeras demonstrated that expression of the G-CSFR on transplantable hematopoietic cells but not stromal cells is required for CY- or G-CSF–induced mobilization. Moreover, in irradiated mice reconstituted with both wild type and G-CSFR–deficient bone marrow cells, treatment with CY or G-CSF resulted in the equal mobilization of both types of HPCs. This result held true for a broad spectrum of HPCs including colony-forming cells, CD34+lineage− and Sca+ lineage−cells, and long-term culture initiating cells. Collectively, these data provide the first definitive evidence that expression of the G-CSFR on HPCs is not required for their mobilization by G-CSF and suggest a model in which G-CSFR–dependent signals act in trans to mobilize HPCs from the bone marrow.
Collapse
|
39
|
Expression of the G-CSF receptor on hematopoietic progenitor cells is not required for their mobilization by G-CSF. Blood 2000. [DOI: 10.1182/blood.v95.10.3025.010k32_3025_3031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mechanisms that regulate hematopoietic progenitor cell (HPC) mobilization from the bone marrow to blood have not yet been defined. HPC mobilization by granulocyte colony-stimulating factor (G-CSF), cyclophosphamide (CY), or interleukin-8 but not flt-3 ligand is markedly impaired in G-CSF receptor–deficient (G-CSFR–deficient) mice. G-CSFR is expressed on mature hematopoietic cells, HPCs, and stromal cells, which suggests that G-CSFR signals in one or more of these cell types was required for mobilization by these agents. To define the cell type(s) responsible for G-CSF–dependent mobilization, a series of chimeric mice were generated using bone marrow transplantation. Mobilization studies in these chimeras demonstrated that expression of the G-CSFR on transplantable hematopoietic cells but not stromal cells is required for CY- or G-CSF–induced mobilization. Moreover, in irradiated mice reconstituted with both wild type and G-CSFR–deficient bone marrow cells, treatment with CY or G-CSF resulted in the equal mobilization of both types of HPCs. This result held true for a broad spectrum of HPCs including colony-forming cells, CD34+lineage− and Sca+ lineage−cells, and long-term culture initiating cells. Collectively, these data provide the first definitive evidence that expression of the G-CSFR on HPCs is not required for their mobilization by G-CSF and suggest a model in which G-CSFR–dependent signals act in trans to mobilize HPCs from the bone marrow.
Collapse
|
40
|
van Eeden SF, Terashima T. Interleukin 8 (IL-8) and the release of leukocytes from the bone marrow. Leuk Lymphoma 2000; 37:259-71. [PMID: 10752978 DOI: 10.3109/10428190009089427] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Interleukin 8 (IL-8) is produced by various cells upon stimulation and influences a variety of functions of leukocytes in particular neutrophils. Systemic administration of IL-8 induces a rapid neutropenia associated by sequestration of neutrophils in the lung that is followed by a neutrophilia characterized by the rapid release of neutrophils from the bone marrow. These cells are released predominantly from the bone marrow venous sinusoids. In addition, several studies have shown the potential role of IL-8 in hematopoiesis and trafficking of hematopoietic stem cells. Systemic administration of IL-8 induces a rapid mobilization of progenitors from the bone marrow with long-term myelo-lymphoid repopulation capacity. It has been employed clinically to mobilize hematopoietic progenitor cells into the peripheral blood and used for autologous or allogeneic bone marrow transplantation. The mechanism for these effects of IL-8 is largely speculative. This report summarizes current ideas on the possible mechanisms how IL-8 influences cell trafficking in and from the bone marrow.
Collapse
Affiliation(s)
- S F van Eeden
- Pulmonary Research Laboratory, University of British Columbia, St. Pauls Hospital, Vancouver, Canada.
| | | |
Collapse
|
41
|
Ribatti D, Vacca A, Roncali L, Dammacco F. Hematopoiesis and angiogenesis: a link between two apparently independent processes. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2000; 9:13-9. [PMID: 10738967 DOI: 10.1089/152581600319577] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In early ontogeny, hematopoiesis is closely associated with angiogenesis. This article reviews recent studies on the role of angiogenic factors that regulate the proliferation and differentiation of endothelial cells in promoting hematopoietic cell growth and studies on the ability of hematopoietic cytokines to affect several endothelial cell functions. The findings in all these studies support the hypothesis formulated at the beginning of this century that a common ancestral cell, the hemangioblast, gives rise to cells of both the endothelial and the hematopoietic lineages.
Collapse
Affiliation(s)
- D Ribatti
- Institute of Human Anatomy, Histology, and Embryology, University of Bari Medical School, Italy
| | | | | | | |
Collapse
|
42
|
Romiti E, Vasta V, Meacci E, Farnararo M, Linke T, Ferlinz K, Sandhoff K, Bruni P. Characterization of sphingomyelinase activity released by thrombin-stimulated platelets. Mol Cell Biochem 2000; 205:75-81. [PMID: 10821424 DOI: 10.1023/a:1007041329052] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In this study we report that human platelets display neutral (nSMase) and acid sphingomyelinase (aSMase) as well as acid ceramidase (aCerase) activity. Cell activation by thrombin resulted in a marked decrease of intracellular aSMase activity, accompanied by the release of enzyme into the medium. In contrast, thrombin treatment did not affect aCerase activity. Two major protein bands of 73 and 70 kDa were recognized by aSMase antibodies in resting platelet lysates and in the medium of stimulated cells. Phorbol esters together with the calcium ionophore A23187 fully reproduced thrombin action on aSMase release. The secreted enzymatic activity was insensitive to digestion with endoglycosidase H but it was stimulated by Zn2+, although to a limited extent compared to aSMase constitutively released by murine endothelial cells. Taken together, these data suggest that secreted aSMase does not originate from the lysosomal compartment but rather from other platelet vesicles.
Collapse
Affiliation(s)
- E Romiti
- Dipartimento di Scienze Biochimiche, Università di Firenze, Florence, Italy
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Dentelli P, Sorbo LD, Rosso A, Molinar A, Garbarino G, Camussi G, Pegoraro L, Brizzi MF. Human IL-3 Stimulates Endothelial Cell Motility and Promotes In Vivo New Vessel Formation. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.4.2151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract
Angiogenesis is a critical process for growth of new capillary blood vessels from preexisting capillaries and postcapillary venules, both in physiological and pathological conditions. Endothelial cell proliferation is a major component of angiogenesis and it is regulated by several growth factors. It has been previously shown that the human hemopoietic growth factor IL-3 (hIL-3), predominantly produced by activated T lymphocytes, stimulates both endothelial cell proliferation and functional activation. In the present study, we report that hIL-3 is able to induce directional migration and tube formation of HUVEC. The in vivo neoangiogenetic effect of hIL-3 was also demonstrated in a murine model in which Matrigel was used for the delivery of the cytokine, suggesting a role of hIL-3 in sustaining neoangiogenesis. Challenge of HUVEC with hIL-3 lead to the synthesis of platelet-activating factor (PAF), which was found to act as secondary mediator for hIL-3-mediated endothelial cell motility but not for endothelial cell proliferation. Consistent with the role of STAT5 proteins in regulating IL-3-mediated mitogenic signals, we herein report that, in hIL-3-stimulated HUVEC, the recruitment of STAT5A and STAT5B, by the β common (βc) subunit of the IL-3R, was not affected by PAF receptor blockade.
Collapse
Affiliation(s)
| | - Lorenzo Del Sorbo
- †Fisiopatologia Clinica,Università di Torino, Torino, Italy
- Dipartimento di
| | | | | | | | | | | | | |
Collapse
|
44
|
Kobayashi S, Imamura M, Hashino S, Noto S, Mori A, Tanaka J, Naohara T, Kasai M, Asaka M. Possible role of granulocyte colony-stimulating factor in increased serum soluble interleukin-2 receptor-alpha levels after allogeneic bone marrow transplantation. Leuk Lymphoma 1999; 33:559-66. [PMID: 10342583 DOI: 10.3109/10428199909058460] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Serum soluble interleukin-2 receptor - alpha (sIL-2R) levels markedly increased at the engraftment period in patients who underwent allogeneic bone marrow transplantation (BMT). Since serum G-CSF levels increased during G-CSF administration and decreased after the cessation, increased sIL-2R levels appeared to be induced by G-CSF administration. There was no increase in sIL-2R levels in a patient given macrophage colony-stimulating factor (M-CSF). The sIL-2R levels at the engraftment period and the onset of acute graft-versus-host disease (GVHD) were higher in patients who developed acute GVHD during G-CSF administration than in those who developed acute GVHD after G-CSF cessation. This finding suggests that G-CSF administration may possibly augment acute GVHD. However, it appears to be unlikely, because in the entire population, 18 of 35 patients had acute GVHD while only 6 of 17 patients had acute GVHD during G-CSF administration. Further analysis is still needed in order to draw definite conclusions. Preconditioning regimens did not appear to affect the sIL-2R levels, when the variable frequencies of methotrexate (MTX) administration were compared.
Collapse
Affiliation(s)
- S Kobayashi
- Third Department of Internal Medicine, Hokkaido University School of Medicine, Sapporo Hokuyu Hospital, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Human Erythropoietin Induces a Pro-Angiogenic Phenotype in Cultured Endothelial Cells and Stimulates Neovascularization In Vivo. Blood 1999. [DOI: 10.1182/blood.v93.8.2627.408k21_2627_2636] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic and endothelial cell lineages share common progenitors. Accordingly, cytokines formerly thought to be specific for the hematopoietic system have been shown to affect several functions in endothelial cells, including angiogenesis. In this study, we investigated the angiogenic potential of erythropoietin (Epo), the main hormone regulating proliferation, differentiation, and survival of erythroid cells. Epo receptors (EpoRs) have been identified in the human EA.hy926 endothelial cell line by Western blot analysis. Also, recombinant human Epo (rHuEpo) stimulates Janus Kinase-2 (JAK-2) phosphorylation, cell proliferation, and matrix metalloproteinase-2 (MMP-2) production in EA.hy926 cells and significantly enhances their differentiation into vascular structures when seeded on Matrigel. In vivo, rHuEpo induces a potent angiogenic response in the chick embryo chorioallantoic membrane (CAM). Accordingly, endothelial cells of the CAM vasculature express EpoRs, as shown by immunostaining with an anti-EpoR antibody. The angiogenic response of CAM blood vessels to rHuEpo was comparable to that elicited by the prototypic angiogenic cytokine basic fibroblast growth factor (FGF2), it occurred in the absence of a significant mononuclear cell infiltrate, and it was not mimicked by endothelin-1 (ET-1) treatment. Taken together, these data demonstrate the ability of Epo to interact directly with endothelial cells and to elicit an angiogenic response in vitro and in vivo and thus act as a bona fide direct angiogenic factor.
Collapse
|
46
|
Human Erythropoietin Induces a Pro-Angiogenic Phenotype in Cultured Endothelial Cells and Stimulates Neovascularization In Vivo. Blood 1999. [DOI: 10.1182/blood.v93.8.2627] [Citation(s) in RCA: 395] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Hematopoietic and endothelial cell lineages share common progenitors. Accordingly, cytokines formerly thought to be specific for the hematopoietic system have been shown to affect several functions in endothelial cells, including angiogenesis. In this study, we investigated the angiogenic potential of erythropoietin (Epo), the main hormone regulating proliferation, differentiation, and survival of erythroid cells. Epo receptors (EpoRs) have been identified in the human EA.hy926 endothelial cell line by Western blot analysis. Also, recombinant human Epo (rHuEpo) stimulates Janus Kinase-2 (JAK-2) phosphorylation, cell proliferation, and matrix metalloproteinase-2 (MMP-2) production in EA.hy926 cells and significantly enhances their differentiation into vascular structures when seeded on Matrigel. In vivo, rHuEpo induces a potent angiogenic response in the chick embryo chorioallantoic membrane (CAM). Accordingly, endothelial cells of the CAM vasculature express EpoRs, as shown by immunostaining with an anti-EpoR antibody. The angiogenic response of CAM blood vessels to rHuEpo was comparable to that elicited by the prototypic angiogenic cytokine basic fibroblast growth factor (FGF2), it occurred in the absence of a significant mononuclear cell infiltrate, and it was not mimicked by endothelin-1 (ET-1) treatment. Taken together, these data demonstrate the ability of Epo to interact directly with endothelial cells and to elicit an angiogenic response in vitro and in vivo and thus act as a bona fide direct angiogenic factor.
Collapse
|
47
|
The Granulocyte Colony-Stimulating Factor Receptor Is Required for the Mobilization of Murine Hematopoietic Progenitors Into Peripheral Blood by Cyclophosphamide or Interleukin-8 But Not Flt-3 Ligand. Blood 1997. [DOI: 10.1182/blood.v90.7.2522] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Hematopoietic progenitor cells (HPC) can be mobilized from the bone marrow into the peripheral circulation in response to a number of stimuli including hematopoietic growth factors, cytotoxic agents, and certain chemokines. Despite significant differences in their biological activities, these stimuli result in the mobilization of HPC with a similar phenotype, suggesting that a common mechanism for mobilization may exist. In this study, the role of granulocyte colony-stimulating factor (G-CSF) in progenitor mobilization was examined using G-CSF receptor (G-CSFR)–deficient mice. In contrast to wild-type mice, no increase in circulating colony-forming cells (CFU-C), CD34+ lineage− progenitors, or day 12 colony-forming unit-spleen progenitors (CFU-S) was detected in G-CSFR–deficient mice after cyclophosphamide administration. This defect was not due to a failure to regenerate HPC following cyclophosphamide administration as the number of CFU-C in the bone marrow of G-CSFR–deficient mice was increased relative to wild-type mice. Likewise, no increase in circulating CFU-C was detected in G-CSFR–deficient mice following interleukin-8 (IL-8) administration. In contrast, mobilization of HPC in response to flt-3 ligand was nearly normal. These results show that the G-CSFR is required for mobilization in response to cyclophosphamide or IL-8 but not flt-3 ligand and suggest that the G-CSFR may play an important and previously unexpected role in HPC migration.
Collapse
|
48
|
The Granulocyte Colony-Stimulating Factor Receptor Is Required for the Mobilization of Murine Hematopoietic Progenitors Into Peripheral Blood by Cyclophosphamide or Interleukin-8 But Not Flt-3 Ligand. Blood 1997. [DOI: 10.1182/blood.v90.7.2522.2522_2522_2528] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic progenitor cells (HPC) can be mobilized from the bone marrow into the peripheral circulation in response to a number of stimuli including hematopoietic growth factors, cytotoxic agents, and certain chemokines. Despite significant differences in their biological activities, these stimuli result in the mobilization of HPC with a similar phenotype, suggesting that a common mechanism for mobilization may exist. In this study, the role of granulocyte colony-stimulating factor (G-CSF) in progenitor mobilization was examined using G-CSF receptor (G-CSFR)–deficient mice. In contrast to wild-type mice, no increase in circulating colony-forming cells (CFU-C), CD34+ lineage− progenitors, or day 12 colony-forming unit-spleen progenitors (CFU-S) was detected in G-CSFR–deficient mice after cyclophosphamide administration. This defect was not due to a failure to regenerate HPC following cyclophosphamide administration as the number of CFU-C in the bone marrow of G-CSFR–deficient mice was increased relative to wild-type mice. Likewise, no increase in circulating CFU-C was detected in G-CSFR–deficient mice following interleukin-8 (IL-8) administration. In contrast, mobilization of HPC in response to flt-3 ligand was nearly normal. These results show that the G-CSFR is required for mobilization in response to cyclophosphamide or IL-8 but not flt-3 ligand and suggest that the G-CSFR may play an important and previously unexpected role in HPC migration.
Collapse
|
49
|
Dong QG, Bernasconi S, Lostaglio S, De Calmanovici RW, Martin-Padura I, Breviario F, Garlanda C, Ramponi S, Mantovani A, Vecchi A. A general strategy for isolation of endothelial cells from murine tissues. Characterization of two endothelial cell lines from the murine lung and subcutaneous sponge implants. Arterioscler Thromb Vasc Biol 1997; 17:1599-604. [PMID: 9301641 DOI: 10.1161/01.atv.17.8.1599] [Citation(s) in RCA: 183] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A rapid, reproducible method for the isolation of murine endothelial cells (ECs) has been developed. Murine ECs were highly enriched by collagenase digestion of mechanically minced lung and subcutaneous sponge implants followed by specific selection with rat anti-mouse CD31 (i.e., PECAM-1) monoclonal antibody-coated magnetic beads (Dynabeads). Pure EC populations were isolated from primary cultures by a second cycle of immunomagnetic selection. The cells from the lung were then cloned by a limiting-dilution method to exclude the possibility of nonendothelial cell contamination. Of the 300 cells plated, 29 clones (approximately 10%) were obtained. The clones were positive for CD31 as measured by flow cytometry, and one clone from the lungs (1G11) and the cells from sponge implants (designated as SIECs) were then subjected to subsequent culture in vitro for 40 and 30 passages (up to 5 months), respectively. Characterization was performed on cells between passage 3 and 10. Both cell types formed contact-inhibited monolayers on gelatin and capillary-like "tubes" on Matrigel. However, 1G11 cells exhibited a "cobblestone" morphology, whereas SIECs had a fibroblast-like appearance at confluence. By flow cytometry and enzyme-linked immunosorbent assay, these cells constitutively expressed CD31, VE-cadherin (cadherin-5), CD34, ICAM-1, VCAM-1, and P-selectin. After stimulation with 30 ng/mL of tumor necrosis factor-alpha, the cells became positive for E-selectin (at 4 hours poststimulation) and the expression of ICAM-1, VCAM-1, and P-selectin was upregulated (after 24 hours of stimulation). The presence of VE-cadherin in 1G11 cells and SIECs was confirmed by fluorescence microscopy and Northern blot analysis. The phenotype and morphology of both cell types were stable during 5 months of culture, and there was no evidence of overgrowth by contaminating cells. Taken together, the approach outlined herein may provide a general strategy for the isolation and culture of ECs from a variety of murine tissues. The general strategy outlined here is simple, effective, and flexible, allowing the inclusion of further positive or negative selection steps.
Collapse
Affiliation(s)
- Q G Dong
- Istituto Ricerche Farmacologiche Mario Negri, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Dong QG, Graziani A, Garlanda C, De Calmanovici RW, Arese M, Soldi R, Vecchi A, Mantovani A, Bussolino F. Anti-tumor activity of cytokines against opportunistic vascular tumors in mice. Int J Cancer 1996; 65:700-8. [PMID: 8598325 DOI: 10.1002/(sici)1097-0215(19960301)65:5<700::aid-ijc23>3.0.co;2-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Polyoma middle T (PmT)-transformed endothelial cells may represent a unique murine model for human opportunistic vascular tumors. The present study was designed to evaluate the anti-tumor potential of a panel of 13 cytokines against murine PmT-transformed endothelial cells. Interferon gamma (IFNgamma) and transforming growth factor beta 1 (TGFbeta1) substantially decreased in a dose-dependent manner the proliferation of a panel of 6 PmT-transformed cell lines. IFNalpha and tumor necrosis factor alpha(TNFalpha) had marginal anti-proliferative activity, whereas other molecules (interleukins-1, -2, -4, -6 and -13, IFNbeta, leukemia inhibitory factor, oncostatin M, granulocyte-macrophage colony-stimulating factor) caused no growth inhibition. IFNgamma and TGFbeta1 were therefore selected for further analysis of their mechanism of action and in vivo relevance. IFNgamma and TGFbeta1 reduced the activity of phosphatidylinositol-3-kinase and the production of phosphatidylinositol 3,4-biphosphate, without modifying the tyrosine kinase(s) activity associated with PmT. IFNgamma and TGFbeta1 were also tested for their ability to modify the in vivo growth of the PmT-transformed endothelial cells H5V in syngeneic C57B1/6 mice. Treatment with IFNnu and TGFbeta1 significantly delayed tumor growth and increased survival time. In contrast, treatment with IFNalpha and TNFalpha failed to prolong survival. In nude mice, IFNgamma and TGFbeta1 had a transient effect on tumor growth but no effect on survival, suggesting a contribution of T cells to the in vivo anti-tumor activity of these cytokines.
Collapse
Affiliation(s)
- Q G Dong
- Istituto Ricerche Farmacologiche Mario Negri, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|