151
|
Tsapogas P, Mooney CJ, Brown G, Rolink A. The Cytokine Flt3-Ligand in Normal and Malignant Hematopoiesis. Int J Mol Sci 2017; 18:E1115. [PMID: 28538663 PMCID: PMC5485939 DOI: 10.3390/ijms18061115] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 12/22/2022] Open
Abstract
The cytokine Fms-like tyrosine kinase 3 ligand (FL) is an important regulator of hematopoiesis. Its receptor, Flt3, is expressed on myeloid, lymphoid and dendritic cell progenitors and is considered an important growth and differentiation factor for several hematopoietic lineages. Activating mutations of Flt3 are frequently found in acute myeloid leukemia (AML) patients and associated with a poor clinical prognosis. In the present review we provide an overview of our current knowledge on the role of FL in the generation of blood cell lineages. We examine recent studies on Flt3 expression by hematopoietic stem cells and its potential instructive action at early stages of hematopoiesis. In addition, we review current findings on the role of mutated FLT3 in leukemia and the development of FLT3 inhibitors for therapeutic use to treat AML. The importance of mouse models in elucidating the role of Flt3-ligand in normal and malignant hematopoiesis is discussed.
Collapse
Affiliation(s)
- Panagiotis Tsapogas
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Mattenstrasse 28, Basel 4058, Switzerland.
| | - Ciaran James Mooney
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edbgaston, Birmingham B15 2TT, UK.
| | - Geoffrey Brown
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edbgaston, Birmingham B15 2TT, UK.
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edbgaston, Birmingham B15 2TT, UK.
| | - Antonius Rolink
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Mattenstrasse 28, Basel 4058, Switzerland.
| |
Collapse
|
152
|
Won H, Moreira D, Gao C, Duttagupta P, Zhao X, Manuel E, Diamond D, Yuan YC, Liu Z, Jones J, D'Apuzzo M, Pal S, Kortylewski M. TLR9 expression and secretion of LIF by prostate cancer cells stimulates accumulation and activity of polymorphonuclear MDSCs. J Leukoc Biol 2017; 102:423-436. [PMID: 28533357 DOI: 10.1189/jlb.3ma1016-451rr] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/14/2017] [Accepted: 04/16/2017] [Indexed: 12/26/2022] Open
Abstract
Proinflammatory signals promote prostate tumorigenesis and progression, but their origins and downstream effects remain unclear. We recently demonstrated that the expression of an innate immune receptor, TLR9, by prostate cancer cells is critical for their tumor-propagating potential. We investigated whether cancer cell-intrinsic TLR9 signaling alters composition of the prostate tumor microenvironment. We generated Ras/Myc (RM9) and Myc-driven (Myc-CaP) prostate cancer cells expressing the tetracycline-inducible gene Tlr9 (Tlr9ON ) or the control LacZ (LacZON ). When engrafted into mice and treated with tetracycline, Tlr9ON , but not LacZON , tumors showed accelerated growth kinetics compared with tumors in PBS-treated mice. Tlr9 upregulation in cancer cells triggered the selective accumulation of CD11b+Ly6GHILy6CLO myeloid cells, phenotypically similar to PMN-MDSCs. The PMN-MDSCs from tetracycline-treated RM9-Tlr9ON tumors increased the immunosuppressive activity of the STAT3 transcription factor, thereby more potently inhibiting T cell proliferation. We identified LIF, an IL-6-type cytokine and STAT3 activator, as a potential mediator of crosstalk between TLR9-expressing prostate cancer cells and PMN-MDSCs. Antibody-mediated LIF neutralization reduced the percentage of tumor-infiltrating PMN-MDSCs and inhibited tumor growth in mice. The clinical relevance of LIF is confirmed by the correlation between TLR9 and LIF expression in prostate cancer specimens. Furthermore, blood samples from patients with prostate cancer showed elevated levels of LIF and high LIFR expression on circulating PMN-MDSCs. Our results suggest that TLR9+ prostate cancers promote immune evasion via LIF-mediated expansion and activation of PMN-MDSCs. Finally, targeting TLR9/LIF/STAT3 signaling using oligonucleotide-based inhibitors, such as CpG-STAT3dODN, can offer new opportunities for prostate cancer immunotherapy.
Collapse
Affiliation(s)
- Haejung Won
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Dayson Moreira
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Chan Gao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Priyanka Duttagupta
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Xingli Zhao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Edwin Manuel
- Department of Experimental Therapeutics, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Don Diamond
- Department of Experimental Therapeutics, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Yate-Ching Yuan
- Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Zheng Liu
- Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Jeremy Jones
- Department of Cell Biology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Massimo D'Apuzzo
- Department of Pathology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; and
| | - Sumanta Pal
- Department of Medical Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Marcin Kortylewski
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA;
| |
Collapse
|
153
|
Cao Y, Zhang E, Yang J, Yang Y, Yu J, Xiao Y, Li W, Zhou D, Li Y, Zhao B, Yan H, Lu M, Zhong M, Yan H. Frontline Science: Nasal epithelial GM-CSF contributes to TLR5-mediated modulation of airway dendritic cells and subsequent IgA response. J Leukoc Biol 2017; 102:575-587. [PMID: 28522600 DOI: 10.1189/jlb.3hi0816-368rr] [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: 09/02/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 11/24/2022] Open
Abstract
Flagellin, as a TLR5 agonist, is an established mucosal adjuvant for enhancing mucosal IgA responses by i.n. immunization. Nasal epithelial cells (NECs) are the first sentinel cells to be exposed to antigen and adjuvant in i.n. immunization, and it is suggested that they play an important role in the mucosal adjuvant activity of flagellin. However, the molecular mechanism leading to modulation and the response by flagellin-activated NECs remain unknown. We aimed to identify the soluble mediator(s) from flagellin-activated NECs that modulate the functions of airway dendritic cells (DCs) and enhance subsequent IgA response. In vitro studies showed that compared with the TLR4 agonist LPS, flagellin directly triggered slight up-regulation of CD80 on airway DCs but was insufficient to affect CD86 expression and DC-mediated IgA response. With the use of an in vitro system for culturing mouse primary NECs (mNECs), we demonstrated that flagellin-activated mNECs could functionally modulate airway DCs, which manifested as significant up-regulation of CD80/CD86 and enhancement of IgA production. The functional modulation of airway DCs was dependent on TLR5 activation of mNECs rather than direct TLR5 activation of airway DCs. With the use of cytokine array and antibody-blocking assays, we further identified that GM-CSF, a cytokine secreted from TLR5-activated mNECs, contributes to the activation of mNECs to airway DCs and subsequent IgA enhancement. In vivo blocking experiments confirmed that GM-CSF is an important factor in recombinant flagellin derived from Salmonella typhi (FliC)-induced airway DC activation and antigen-specific IgA enhancement. Our data directly demonstrate that nasal epithelial GM-CSF contributes to TLR5-mediated modulation of airway DCs and a subsequent IgA response.
Collapse
Affiliation(s)
- Yuan Cao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Ejuan Zhang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Jingyi Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Yi Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Jie Yu
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Yang Xiao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Wei Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Dihan Zhou
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Yaoming Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Bali Zhao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Hu Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Mengji Lu
- Institute of Virology, University Hospital of Essen, Essen, Germany
| | - Maohua Zhong
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| |
Collapse
|
154
|
Domingues MJ, Cao H, Heazlewood SY, Cao B, Nilsson SK. Niche Extracellular Matrix Components and Their Influence on HSC. J Cell Biochem 2017; 118:1984-1993. [PMID: 28112429 DOI: 10.1002/jcb.25905] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 12/16/2022]
Abstract
Maintenance of hematopoietic stem cells (HSC) takes place in a highly specialized microenvironment within the bone marrow. Technological improvements, especially in the field of in vivo imaging, have helped unravel the complexity of the niche microenvironment and have completely changed the classical concept from what was previously believed to be a static supportive platform, to a dynamic microenvironment tightly regulating HSC homeostasis through the complex interplay between diverse cell types, secreted factors, extracellular matrix molecules, and the expression of different transmembrane receptors. To add to the complexity, non-protein based metabolites have also been recognized as a component of the bone marrow niche. The objective of this review is to discuss the current understanding on how the different extracellular matrix components of the niche regulate HSC fate, both during embryonic development and in adulthood. Special attention will be provided to the description of non-protein metabolites, such as lipids and metal ions, which contribute to the regulation of HSC behavior. J. Cell. Biochem. 118: 1984-1993, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mélanie J Domingues
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Huimin Cao
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Shen Y Heazlewood
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Benjamin Cao
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Susan K Nilsson
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
155
|
Zhang J, Liu Q, Qiao L, Hu P, Deng G, Liang N, Xie J, Luo H, Zhang J. Novel role of granulocyte-macrophage colony-stimulating factor: antitumor effects through inhibition of epithelial-to-mesenchymal transition in esophageal cancer. Onco Targets Ther 2017; 10:2227-2237. [PMID: 28461757 PMCID: PMC5404808 DOI: 10.2147/ott.s133504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose Recent studies demonstrate the possible antitumor effects of granulocyte-macrophage colony-stimulating factor (GM-CSF); however, the exact mechanism is still unclear. The aim of our study was to analyze the effects of GM-CSF on multiple biological functions of human esophageal cancer (EC) cell lines and to explore the potential mechanism of its antitumor effects. Materials and methods Eca109/9706 human EC cells were examined. Cell proliferation, apoptosis, and migration were analyzed using cell proliferation assay, flow cytometry, and transwell assay, respectively. The expression of signaling molecules were examined by reverse transcription polymerase chain reaction and Western blot. Results Our results provide experimental evidence that GM-CSF inhibits growth and migration, as well as induction of apoptosis in EC cells. In addition, EC cells stimulated with GM-CSF were more likely to have suppressed epithelial-to-mesenchymal transition (EMT), accompanied by increased E-cadherin and decreased vimentin expression. Conclusion Our data demonstrate that GM-CSF inhibits cancer cell proliferation and migration, as well as induction of apoptosis. Moreover, our findings indicate that GM-CSF may regulate EMT through JAK2-PRMT5 signaling, and thereby exhibit its antitumor effects on EC cells.
Collapse
Affiliation(s)
- Jingxin Zhang
- Division of Oncology, Department of Graduate, Weifang Medical College, Weifang
| | - Qiqi Liu
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Shandong University
| | - Lili Qiao
- Department of Oncology, The Fifth Peoples' Hospital of Jinan, Jinan
| | - Pingping Hu
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Shandong University
| | - Guodong Deng
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Shandong University
| | - Ning Liang
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Shandong University
| | - Jian Xie
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Shandong University
| | - Hui Luo
- Department of Radiation Oncology, Henan Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jiandong Zhang
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Shandong University
| |
Collapse
|
156
|
Priam P, Krasteva V, Rousseau P, D'Angelo G, Gaboury L, Sauvageau G, Lessard JA. SMARCD2 subunit of SWI/SNF chromatin-remodeling complexes mediates granulopoiesis through a CEBPɛ dependent mechanism. Nat Genet 2017; 49:753-764. [PMID: 28369034 DOI: 10.1038/ng.3812] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 02/14/2017] [Indexed: 12/15/2022]
Abstract
Recent studies suggest that individual subunits of chromatin-remodeling complexes produce biologically specific meaning in different cell types through combinatorial assembly. Here we show that granulocyte development requires SMARCD2, a subunit of ATP-dependent SWI/SNF (BAF) chromatin-remodeling complexes. Smarcd2-deficient mice fail to generate functionally mature neutrophils and eosinophils, a phenotype reminiscent of neutrophil-specific granule deficiency (SGD) in humans, for which loss-of-function mutations in CEBPE (encoding CEBPɛ) have been reported. SMARCD2-containing SWI/SNF complexes are necessary for CEBPɛ transcription factor recruitment to the promoter of neutrophilic secondary granule genes and for granulocyte differentiation. The homologous SMARCD1 protein (63% identical at the amino acid level) cannot replace the role of SMARCD2 in granulocyte development. We find that SMARCD2 functional specificity is conferred by its divergent coiled-coil 1 and SWIB domains. Strikingly, both CEBPE and SMARCD2 loss-of-function mutations identified in patients with SGD abolish the interaction with SWI/SNF and thereby secondary granule gene expression, thus providing a molecular basis for this disease.
Collapse
Affiliation(s)
- Pierre Priam
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada.,Department of Pathology and Cellular Biology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Veneta Krasteva
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada.,Department of Pathology and Cellular Biology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Philippe Rousseau
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada
| | - Giovanni D'Angelo
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Louis Gaboury
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada.,Department of Pathology and Cellular Biology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Guy Sauvageau
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada.,Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada.,Leukemia Cell Bank of Quebec, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada.,Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Julie A Lessard
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada.,Department of Pathology and Cellular Biology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
157
|
Cucak H, Høj Thomsen L, Rosendahl A. IL-20 contributes to low grade inflammation and weight gain in the Psammomys obesus. Int Immunopharmacol 2017; 45:53-67. [DOI: 10.1016/j.intimp.2017.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 01/26/2017] [Accepted: 01/27/2017] [Indexed: 02/08/2023]
|
158
|
Amouzegar A, Mittal SK, Sahu A, Sahu SK, Chauhan SK. Mesenchymal Stem Cells Modulate Differentiation of Myeloid Progenitor Cells During Inflammation. Stem Cells 2017; 35:1532-1541. [PMID: 28295880 DOI: 10.1002/stem.2611] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 02/14/2017] [Accepted: 02/26/2017] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) possess distinct immunomodulatory properties and have tremendous potential for use in therapeutic applications in various inflammatory diseases. MSCs have been shown to regulate pathogenic functions of mature myeloid inflammatory cells, such as macrophages and neutrophils. Intriguingly, the capacity of MSCs to modulate differentiation of myeloid progenitors (MPs) to mature inflammatory cells remains unknown to date. Here, we report the novel finding that MSCs inhibit the expression of differentiation markers on MPs under inflammatory conditions. We demonstrate that the inhibitory effect of MSCs is dependent on direct cell-cell contact and that this intercellular contact is mediated through interaction of CD200 expressed by MSCs and CD200R1 expressed by MPs. Furthermore, using an injury model of sterile inflammation, we show that MSCs promote MP frequencies and suppress infiltration of inflammatory cells in the inflamed tissue. We also find that downregulation of CD200 in MSCs correlates with abrogation of their immunoregulatory function. Collectively, our study provides unequivocal evidence that MSCs inhibit differentiation of MPs in the inflammatory environment via CD200-CD200R1 interaction. Stem Cells 2017;35:1532-1541.
Collapse
Affiliation(s)
- Afsaneh Amouzegar
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Sharad K Mittal
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Anuradha Sahu
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA
| | - Srikant K Sahu
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA.,L. V. Prasad Eye Institute, Bhubaneswar, Odisha, 751024, India
| | - Sunil K Chauhan
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, 02114, USA
| |
Collapse
|
159
|
Sharapov MG, Novoselov VI, Fesenko EE, Bruskov VI, Gudkov SV. The role of peroxiredoxin 6 in neutralization of X-ray mediated oxidative stress: effects on gene expression, preservation of radiosensitive tissues and postradiation survival of animals. Free Radic Res 2017; 51:148-166. [DOI: 10.1080/10715762.2017.1289377] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- M. G. Sharapov
- Institute of Cell Biophysics, Russian Academy of Sciences, Moscow, Russia
| | - V. I. Novoselov
- Institute of Cell Biophysics, Russian Academy of Sciences, Moscow, Russia
| | - E. E. Fesenko
- Institute of Cell Biophysics, Russian Academy of Sciences, Moscow, Russia
| | - V. I. Bruskov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Moscow, Russia
| | - S. V. Gudkov
- A.M. Prokhorov General Physics Institute, Russian Academy of Sciences, Moscow, Russia
- Lobachevsky State University of Nizhni Novgorod, Nizhni Novgorod, Russia
| |
Collapse
|
160
|
Noack M, Miossec P. Selected cytokine pathways in rheumatoid arthritis. Semin Immunopathol 2017; 39:365-383. [DOI: 10.1007/s00281-017-0619-z] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/31/2017] [Indexed: 12/13/2022]
|
161
|
Immune modulation of some autoimmune diseases: the critical role of macrophages and neutrophils in the innate and adaptive immunity. J Transl Med 2017; 15:36. [PMID: 28202039 PMCID: PMC5312441 DOI: 10.1186/s12967-017-1141-8] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/03/2017] [Indexed: 12/16/2022] Open
Abstract
Macrophages and neutrophils are key components involved in the regulation of numerous chronic inflammatory diseases, infectious disorders, and especially certain autoimmune disease. However, little is known regarding the contribution of these cells to the pathogenesis of autoimmune disorders. Recent studies have aimed to clarify certain important factors affecting the immunogenicity of these cells, including the type and dose of antigen, the microenvironment of the cell-antigen encounter, and the number, subset, and phenotype of these cells, which can prevent or induce autoimmune responses. This review highlights the role of macrophage subsets and neutrophils in injured tissues, supporting their cooperation during the pathogenesis of certain autoimmune diseases.
Collapse
|
162
|
Laffont S, Seillet C, Guéry JC. Estrogen Receptor-Dependent Regulation of Dendritic Cell Development and Function. Front Immunol 2017; 8:108. [PMID: 28239379 PMCID: PMC5300975 DOI: 10.3389/fimmu.2017.00108] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/23/2017] [Indexed: 12/23/2022] Open
Abstract
Autoimmunity, infectious diseases and cancer affect women and men differently. Because they tend to develop more vigorous adaptive immune responses than men, women are less susceptible to some infectious diseases but also at higher risk of autoimmunity. The regulation of immune responses by sex-dependent factors probably involves several non-redundant mechanisms. A privileged area of study, however, concerns the role of sex steroid hormones in the biology of innate immune cells, especially dendritic cells (DCs). In recent years, our understanding of the lineage origin of DC populations has expanded, and the lineage-committing transcription factors shaping peripheral DC subsets have been identified. Both progenitor cells and mature DC subsets express estrogen receptors (ERs), which are ligand-dependent transcription factors. This suggests that estrogens may contribute to the reported sex differences in immunity by regulating DC biology. Here, we review the recent literature and highlight evidence that estrogen-dependent activation of ERα regulates the development or the functional responses of particular DC subsets. The in vitro model of GM-CSF-induced DC differentiation shows that CD11c+ CD11bint Ly6cneg cells depend on ERα activation by estrogen for their development, and for the acquisition of competence to activate naive CD4+ T lymphocytes and mount a robust pro-inflammatory cytokine response to CD40 stimulation. In this model, estrogen signaling in conjunction with GM-CSF is necessary to promote early interferon regulatory factor (Irf)-4 expression in macrophage-DC progenitors and their subsequent differentiation into IRF-4hi CD11c+ CD11bint Ly6cneg cells, closely related to the cDC2 subset. The Flt3L-induced model of DC differentiation in turn shows that ERα signaling promotes the development of conventional DC (cDC) and plasmacytoid DC (pDC) with higher capability of pro-inflammatory cytokine production in response to TLR stimulation. Likewise, cell-intrinsic ER signaling positively regulates the TLR-driven production of type I interferons (IFNs) in mouse pDCs in vivo. This effect of estrogens likely contributes to the greater proficiency of women's pDCs than men's as regards the production of type I IFNs elicited by TLR7 ligands. In summary, evidence is emerging in support of the notion that estrogen signaling regulates important aspects of cDC and pDC development and/or effector functions, in both mice and humans.
Collapse
Affiliation(s)
- Sophie Laffont
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS , Toulouse , France
| | - Cyril Seillet
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Jean-Charles Guéry
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS , Toulouse , France
| |
Collapse
|
163
|
Måløy M, Måløy F, Jakobsen P, Olav Brandsdal B. Dynamic self-organisation of haematopoiesis and (a)symmetric cell division. J Theor Biol 2017; 414:147-164. [DOI: 10.1016/j.jtbi.2016.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/06/2016] [Accepted: 11/30/2016] [Indexed: 12/24/2022]
|
164
|
Borriello F, Iannone R, Di Somma S, Loffredo S, Scamardella E, Galdiero MR, Varricchi G, Granata F, Portella G, Marone G. GM-CSF and IL-3 Modulate Human Monocyte TNF-α Production and Renewal in In Vitro Models of Trained Immunity. Front Immunol 2017; 7:680. [PMID: 28138327 PMCID: PMC5237654 DOI: 10.3389/fimmu.2016.00680] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 12/21/2016] [Indexed: 12/25/2022] Open
Abstract
GM-CSF and IL-3 are hematopoietic cytokines that also modulate the effector functions of several immune cell subsets. In particular, GM-CSF and IL-3 exert a significant control on monocyte and macrophage effector functions, as assessed in experimental models of inflammatory and autoimmune diseases and also in human studies. Here, we sought to investigate the mechanisms and the extent to which GM-CSF and IL-3 modulate the pro-inflammatory, LPS-mediated, activation of human CD14+ monocytes taking into account the new concept of trained immunity (i.e., the priming stimulus modulates the response to subsequent stimuli mainly by inducing chromatin remodeling and increased transcription at relevant genetic loci). We demonstrate that GM-CSF and IL-3 priming enhances TNF-α production upon subsequent LPS stimulation (short-term model of trained immunity) in a p38- and SIRT2-dependent manner without increasing TNF primary transcript levels (a more direct measure of transcription), thus supporting a posttranscriptional regulation of TNF-α in primed monocytes. GM-CSF and IL-3 priming followed by 6 days of resting also results in increased TNF-α production upon LPS stimulation (long-term model of trained immunity). In this case, however, GM-CSF and IL-3 priming induces a c-Myc-dependent monocyte renewal and increase in cell number that is in turn responsible for heightened TNF-α production. Overall, our results provide insights to understand the biology of monocytes in health and disease conditions in which the hematopoietic cytokines GM-CSF and IL-3 play a role and also extend our knowledge of the cellular and molecular mechanisms of trained immunity.
Collapse
Affiliation(s)
- Francesco Borriello
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Raffaella Iannone
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Sarah Di Somma
- Department of Translational Medical Sciences, University of Naples Federico II , Naples , Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Eloise Scamardella
- Department of Translational Medical Sciences, University of Naples Federico II , Naples , Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Francescopaolo Granata
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Giuseppe Portella
- Department of Translational Medical Sciences, University of Naples Federico II , Naples , Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| |
Collapse
|
165
|
López C, Zamorano P, Teuber S, Salas M, Otth C, Hidalgo MA, Concha I, Zambrano A. Interleukin-3 Prevents Cellular Death Induced by Oxidative Stress in HEK293 Cells. J Cell Biochem 2017; 118:1330-1340. [PMID: 27862234 DOI: 10.1002/jcb.25790] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/08/2016] [Indexed: 11/07/2022]
Abstract
Interleukin-3 (IL-3) is a well-characterized growth factor in hematopoietic cells, but it is also expressed in other cell types with poorly described functions. Many studies have provided evidence that IL-3 plays an important role in cell survival. We have previously shown that IL-3 is able to increase glucose uptake in HEK293 cells, suggesting that this factor requires sustained glucose metabolism to promote cell survival. In this study, we demonstrate that IL-3 contributes to cell survival under oxidative stress, a prominent feature in the pathophysiology of cancer, diabetes, and neurodegenerative diseases, as well as in the aging process. Our results suggest a molecular mechanism that involves signaling pathways mediated by PI-3k/Akt and Erk. Altogether, these findings show an important role for IL-3 in supporting the viability of non-hematopoietic systems. J. Cell. Biochem. 118: 1330-1340, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Camila López
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Patricia Zamorano
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Stefanie Teuber
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Mónica Salas
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Carola Otth
- Facultad de Medicina, Instituto de Microbiología Clínica, Valdivia, Los Ríos, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - María A Hidalgo
- Facultad de Ciencias Veterinarias, Instituto de Farmacología, Universidad Austral de Chile, Valdivia, Los Ríos, Chile
| | - Ilona Concha
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Angara Zambrano
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
166
|
Elias HK, Bryder D, Park CY. Molecular mechanisms underlying lineage bias in aging hematopoiesis. Semin Hematol 2017; 54:4-11. [DOI: 10.1053/j.seminhematol.2016.11.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
167
|
Anti-colony-stimulating factor therapies for inflammatory and autoimmune diseases. Nat Rev Drug Discov 2016; 16:53-70. [DOI: 10.1038/nrd.2016.231] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
168
|
Permissive roles of cytokines interleukin-7 and Flt3 ligand in mouse B-cell lineage commitment. Proc Natl Acad Sci U S A 2016; 113:E8122-E8130. [PMID: 27911806 DOI: 10.1073/pnas.1613316113] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Hematopoietic cells are continuously generated throughout life from hematopoietic stem cells, thus making hematopoiesis a favorable system to study developmental cell lineage commitment. The main factors incorporating environmental signals to developing hematopoietic cells are cytokines, which regulate commitment of hematopoietic progenitors to the different blood lineages by acting either in an instructive or a permissive manner. Fms-like tyrosine kinase-3 (Flt3) ligand (FL) and Interleukin-7 (IL-7) are cytokines pivotal for B-cell development, as manifested by the severely compromised B-cell development in their absence. However, their precise role in regulating B-cell commitment has been the subject of debate. In the present study we assessed the rescue of B-cell commitment in mice lacking IL-7 but simultaneously overexpressing FL. Results obtained demonstrate that FL overexpression in IL-7-deficient mice rescues B-cell commitment, resulting in significant Ebf1 and Pax5 expression in Ly6D+CD135+CD127+CD19- precursors and subsequent generation of normal numbers of CD19+ B-cell progenitors, therefore indicating that IL-7 can be dispensable for commitment to the B-cell lineage. Further analysis of Ly6D+CD135+CD127+CD19- progenitors in IL-7- or FL-deficient mice overexpressing Bcl2, as well as in IL-7 transgenic mice suggests that both FL and IL-7 regulate B-cell commitment in a permissive manner: FL by inducing proliferation of Ly6D+CD135+CD127+CD19- progenitors and IL-7 by providing survival signals to these progenitors.
Collapse
|
169
|
G-CSF-induced sympathetic tone provokes fever and primes antimobilizing functions of neutrophils via PGE2. Blood 2016; 129:587-597. [PMID: 27827823 DOI: 10.1182/blood-2016-07-725754] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/01/2016] [Indexed: 12/22/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) is widely used for peripheral blood stem/progenitor mobilization. G-CSF causes low-grade fever that is ameliorated by nonsteroidal anti-inflammatory drugs (NSAIDs), suggesting the activation of arachidonic acid (AA) cascade. How G-CSF regulated this reaction was assessed. G-CSF treatment in mice resulted in fever, which was canceled in prostaglandin E synthase (mPGES-1)-deficient mice. Mobilization efficiency was twice as high in chimeric mice lacking mPGES-1, specifically in hematopoietic cells, suggesting that prostaglandin E2 (PGE2) from hematopoietic cells modulated the bone marrow (BM) microenvironment. Neutrophils from steady-state BM constitutively expressed mPGES-1 and significantly enhanced PGE2 production in vitro by β-adrenergic stimulation, but not by G-CSF, which was inhibited by an NSAID. Although neutrophils expressed all β-adrenergic receptors, only β3-agonist induced this phenomenon. Liquid chromatography-tandem mass spectrometry traced β-agonist-induced PGE2 synthesis from exogenous deuterium-labeled AA. Spontaneous PGE2 production was highly efficient in Gr-1high neutrophils among BM cells from G-CSF-treated mice. In addition to these in vitro data, the in vivo depletion of Gr-1high neutrophils disrupted G-CSF-induced fever. Furthermore, sympathetic denervation eliminated both neutrophil priming for PGE2 production and fever during G-CSF treatment. Thus, sympathetic tone-primed BM neutrophils were identified as one of the major PGE2 producers. PGE2 upregulated osteopontin, specifically in preosteoblasts, to retain progenitors in the BM via EP4 receptor. Thus, the sympathetic nervous system regulated neutrophils as an indispensable PGE2 source to modulate BM microenvironment and body temperature. This study provided a novel mechanistic insight into the communication of the nervous system, BM niche components, and hematopoietic cells.
Collapse
|
170
|
Abu-Khader A, Pasha R, Ward GCD, Boisjoli G, Pineault N. Characterization of the growth modulatory activities of osteoblast conditioned media on cord blood progenitor cells. Cytotechnology 2016; 68:2257-2269. [PMID: 27757713 DOI: 10.1007/s10616-016-0019-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/19/2016] [Indexed: 12/25/2022] Open
Abstract
Engraftment outcomes are strongly correlated with the numbers of hematopoietic stem and progenitor cells (HSPC) infused. Expansion of umbilical cord blood (CB) HSPC has gained much interest lately since infusion of expanded HSPC can accelerate engraftment and improve clinical outcomes. Many novel protocols based on different expansion strategies of HSPC and their downstream derivatives are under development. Herein, we describe the production and properties of serum-free medium (SFM) conditioned with mesenchymal stromal cells derived-osteoblasts (OCM) for the expansion of umbilical CB cells and progenitors. After optimization of the conditioning length, we show that OCM increased the production of human CB total nucleated cells and CD34+ cells by 1.8-fold and 1.5-fold over standard SFM, respectively. Production of immature CD34+ subpopulations enriched in hematopoietic stem cells was also improved with a shorter conditioning period. Moreover, we show that the growth modulatory activities of OCM on progenitor expansion are regulated by both soluble factors and non-soluble cellular elements. Finally, the growth and differentiation modulatory activities of OCM were fully retained after high dose-ionizing irradiation and highly stable when OCM is stored frozen. In summary, our results suggest that OCM efficiently mimics some of the natural regulatory activities of osteoblasts on HSPC and highlight the marked expansion potentials of SFM conditioned with osteoblasts.
Collapse
Affiliation(s)
- Ahmad Abu-Khader
- Centre for Innovation, Canadian Blood Services, 1800 Alta Vista, Ottawa, ON, K1G 4J5, Canada
| | - Roya Pasha
- Centre for Innovation, Canadian Blood Services, 1800 Alta Vista, Ottawa, ON, K1G 4J5, Canada
| | - Gwendoline C D Ward
- Centre for Innovation, Canadian Blood Services, 1800 Alta Vista, Ottawa, ON, K1G 4J5, Canada
| | - Gavin Boisjoli
- Centre for Innovation, Canadian Blood Services, 1800 Alta Vista, Ottawa, ON, K1G 4J5, Canada
| | - Nicolas Pineault
- Centre for Innovation, Canadian Blood Services, 1800 Alta Vista, Ottawa, ON, K1G 4J5, Canada. .,Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
171
|
Abstract
The cause of Crohn’s disease (CD) has posed a conundrum for at least a century. A large body of work coupled with recent technological advances in genome research have at last started to provide some of the answers. Initially this review seeks to explain and to differentiate between bowel inflammation in the primary immunodeficiencies that generally lead to very early onset diffuse bowel inflammation in humans and in animal models, and the real syndrome of CD. In the latter, a trigger, almost certainly enteric infection by one of a multitude of organisms, allows the faeces access to the tissues, at which stage the response of individuals predisposed to CD is abnormal. Direct investigation of patients’ inflammatory response together with genome-wide association studies (GWAS) and DNA sequencing indicate that in CD the failure of acute inflammation and the clearance of bacteria from the tissues, and from within cells, is defective. The retained faecal products result in the characteristic chronic granulomatous inflammation and adaptive immune response. In this review I will examine the contemporary evidence that has led to this understanding, and look for explanations for the recent dramatic increase in the incidence of this disease.
Collapse
|
172
|
Stiehl T, Lutz C, Marciniak-Czochra A. Emergence of heterogeneity in acute leukemias. Biol Direct 2016; 11:51. [PMID: 27733173 PMCID: PMC5062896 DOI: 10.1186/s13062-016-0154-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Leukemias are malignant proliferative disorders of the blood forming system. Sequencing studies demonstrate that the leukemic cell population consists of multiple clones. The genetic relationship between the different clones, referred to as the clonal hierarchy, shows high interindividual variability. So far, the source of this heterogeneity and its clinical relevance remain unknown. We propose a mathematical model to study the emergence and evolution of clonal heterogeneity in acute leukemias. The model allows linking properties of leukemic clones in terms of self-renewal and proliferation rates to the structure of the clonal hierarchy. RESULTS Computer simulations imply that the self-renewal potential of the first emerging leukemic clone has a major impact on the total number of leukemic clones and on the structure of their hierarchy. With increasing depth of the clonal hierarchy the self-renewal of leukemic clones increases, whereas the proliferation rates do not change significantly. The emergence of deep clonal hierarchies is a complex process that is facilitated by a cooperativity of different mutations. CONCLUSION Comparison of patient data and simulation results suggests that the self-renewal of leukemic clones increases with the emergence of clonal heterogeneity. The structure of the clonal hierarchy may serve as a marker for patient prognosis. REVIEWERS This article was reviewed by Marek Kimmel, Tommaso Lorenzi and Tomasz Lipniacki.
Collapse
Affiliation(s)
- Thomas Stiehl
- Institute of Applied Mathematics, Heidelberg University, Im Neuenheimer Feld 205, Heidelberg, 69120, Germany. .,Interdisciplinary Center for Scientific Computing, Heidelberg University, Im Neuenheimer Feld 205, Heidelberg, 69120, Germany. .,Bioquant Center, Heidelberg University, Im Neuenheimer Feld 297, Heidelberg, 69120, Germany.
| | - Christoph Lutz
- Department of Medicine V, Heidelberg University, Im Neuenheimer Feld 410, Heidelberg, 69120, Germany
| | - Anna Marciniak-Czochra
- Institute of Applied Mathematics, Heidelberg University, Im Neuenheimer Feld 205, Heidelberg, 69120, Germany.,Interdisciplinary Center for Scientific Computing, Heidelberg University, Im Neuenheimer Feld 205, Heidelberg, 69120, Germany.,Bioquant Center, Heidelberg University, Im Neuenheimer Feld 297, Heidelberg, 69120, Germany
| |
Collapse
|
173
|
Abstract
The cause of Crohn's disease (CD) has posed a conundrum for at least a century. A large body of work coupled with recent technological advances in genome research have at last started to provide some of the answers. Initially this review seeks to explain and to differentiate between bowel inflammation in the primary immunodeficiencies that generally lead to very early onset diffuse bowel inflammation in humans and in animal models, and the real syndrome of CD. In the latter, a trigger, almost certainly enteric infection by one of a multitude of organisms, allows the faeces access to the tissues, at which stage the response of individuals predisposed to CD is abnormal. Direct investigation of patients' inflammatory response together with genome-wide association studies (GWAS) and DNA sequencing indicate that in CD the failure of acute inflammation and the clearance of bacteria from the tissues, and from within cells, is defective. The retained faecal products result in the characteristic chronic granulomatous inflammation and adaptive immune response. In this review I will examine the contemporary evidence that has led to this understanding, and look for explanations for the recent dramatic increase in the incidence of this disease.
Collapse
|
174
|
Kandalla PK, Sarrazin S, Molawi K, Berruyer C, Redelberger D, Favel A, Bordi C, de Bentzmann S, Sieweke MH. M-CSF improves protection against bacterial and fungal infections after hematopoietic stem/progenitor cell transplantation. J Exp Med 2016; 213:2269-2279. [PMID: 27811055 PMCID: PMC5068229 DOI: 10.1084/jem.20151975] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 09/01/2016] [Indexed: 02/01/2023] Open
Abstract
Myeloablative treatment preceding hematopoietic stem cell (HSC) and progenitor cell (HS/PC) transplantation results in severe myeloid cytopenia and susceptibility to infections in the lag period before hematopoietic recovery. We have previously shown that macrophage colony-stimulating factor (CSF-1; M-CSF) directly instructed myeloid commitment in HSCs. In this study, we tested whether this effect had therapeutic benefit in improving protection against pathogens after HS/PC transplantation. M-CSF treatment resulted in an increased production of mature myeloid donor cells and an increased survival of recipient mice infected with lethal doses of clinically relevant opportunistic pathogens, namely the bacteria Pseudomonas aeruginosa and the fungus Aspergillus fumigatus M-CSF treatment during engraftment or after infection efficiently protected from these pathogens as early as 3 days after transplantation and was effective as a single dose. It was more efficient than granulocyte CSF (G-CSF), a common treatment of severe neutropenia, which showed no protective effect under the tested conditions. M-CSF treatment showed no adverse effect on long-term lineage contribution or stem cell activity and, unlike G-CSF, did not impede recovery of HS/PCs, thrombocyte numbers, or glucose metabolism. These results encourage potential clinical applications of M-CSF to prevent severe infections after HS/PC transplantation.
Collapse
Affiliation(s)
- Prashanth K Kandalla
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, 13288 Marseille, France
| | - Sandrine Sarrazin
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, 13288 Marseille, France
| | - Kaaweh Molawi
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, 13288 Marseille, France.,Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft, 13125 Berlin, Germany
| | - Carole Berruyer
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, 13288 Marseille, France
| | - David Redelberger
- Centre National de la Recherche Scientifique, Laboratoire d' Ingenierie des Systemes Macromoleculaires, Institut de Microbiologie de la Mediterranee, Aix Marseille Université, 13402 Marseille, France
| | - Anne Favel
- Institute National de la Recherche Agronomique, Unite Mixte de Recherche 1163 BBF, Aix Marseille Université, 13288 Marseille, France
| | - Christophe Bordi
- Centre National de la Recherche Scientifique, Laboratoire d' Ingenierie des Systemes Macromoleculaires, Institut de Microbiologie de la Mediterranee, Aix Marseille Université, 13402 Marseille, France
| | - Sophie de Bentzmann
- Centre National de la Recherche Scientifique, Laboratoire d' Ingenierie des Systemes Macromoleculaires, Institut de Microbiologie de la Mediterranee, Aix Marseille Université, 13402 Marseille, France
| | - Michael H Sieweke
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, 13288 Marseille, France .,Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft, 13125 Berlin, Germany
| |
Collapse
|
175
|
Yaparla A, Wendel ES, Grayfer L. The unique myelopoiesis strategy of the amphibian Xenopus laevis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 63:136-143. [PMID: 27234705 DOI: 10.1016/j.dci.2016.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/18/2016] [Accepted: 05/18/2016] [Indexed: 06/05/2023]
Abstract
Myeloid progenitors reside within specific hematopoietic organs and commit to progenitor lineages bearing megakaryocyte/erythrocyte (MEP) or granulocyte/macrophage potentials (GMP) within these sites. Unlike other vertebrates, the amphibian Xenopus laevis committed macrophage precursors are absent from the hematopoietic subcapsular liver and instead reside within their bone marrow. Presently, we demonstrate that while these frogs' liver-derived cells are unresponsive to recombinant forms of principal X. laevis macrophage (colony-stimulating factor-1; CSF-1) and granulocyte (CSF-3) growth factors, bone marrow cells cultured with CSF-1 and CSF-3 exhibit respectively archetypal macrophage and granulocyte morphology, gene expression and functionalities. Moreover, we demonstrate that liver, but not bone marrow cells possess erythropoietic capacities when stimulated with a X. laevis erythropoietin. Together, our findings indicate that X. laevis retain their MEP within the hematopoietic liver while sequestering their GMP to the bone marrow, thus marking a very novel myelopoietic strategy as compared to those seen in other jawed vertebrate species.
Collapse
Affiliation(s)
- Amulya Yaparla
- Department of Biological Sciences, George Washington University, Washington, DC, USA
| | - Emily S Wendel
- Department of Biological Sciences, George Washington University, Washington, DC, USA
| | - Leon Grayfer
- Department of Biological Sciences, George Washington University, Washington, DC, USA.
| |
Collapse
|
176
|
Gorjifard S, Goldszmid RS. Microbiota-myeloid cell crosstalk beyond the gut. J Leukoc Biol 2016; 100:865-879. [PMID: 27605211 DOI: 10.1189/jlb.3ri0516-222r] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/11/2016] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota is a complex and dynamic microbial ecosystem that plays a fundamental role in host physiology. Locally, the gut commensal microbes/host symbiotic relationship is vital for barrier fortification, nutrient absorption, resistance against intestinal pathogens, and the development and maintenance of the mucosal immune system. It is now clear that the effects of the indigenous intestinal flora extend beyond the gut, ranging from shaping systemic immune responses to metabolic and behavioral functions. However, the underlying mechanisms of the gut microbiota/systemic immune system interactions remain largely unknown. Myeloid cells respond to microbial signals, including those derived from commensals, and initiate innate and adaptive immune responses. In this review, we focus on the impact of the gut microbiota on myeloid cells at extraintestinal sites. In particular, we discuss how commensal-derived signals affect steady-state myelopoiesis and cellular function and how that influences the response to infection and cancer therapy.
Collapse
Affiliation(s)
- Sayeh Gorjifard
- Inflammatory Cell Dynamics Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Romina S Goldszmid
- Inflammatory Cell Dynamics Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
177
|
Identification of factors promoting ex vivo maintenance of mouse hematopoietic stem cells by long-term single-cell quantification. Blood 2016; 128:1181-92. [DOI: 10.1182/blood-2016-03-705590] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/14/2016] [Indexed: 12/11/2022] Open
Abstract
Key Points
AFT024-induced HSC maintenance correlates with early survival/proliferation whereas early death is a major reason for HSC loss in culture. Dermatopontin is required for ex vivo HSC maintenance, and also improves HSC clonogenicity in stroma-based and stroma-free cultures.
Collapse
|
178
|
MPL expression on AML blasts predicts peripheral blood neutropenia and thrombocytopenia. Blood 2016; 128:2253-2257. [PMID: 27574191 DOI: 10.1182/blood-2016-04-711986] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/19/2016] [Indexed: 11/20/2022] Open
Abstract
Although the molecular pathways that cause acute myeloid leukemia (AML) are increasingly well understood, the pathogenesis of peripheral blood cytopenia, a major cause of AML mortality, remains obscure. A prevailing assumption states that AML spatially displaces nonleukemic hematopoiesis from the bone marrow. However, examining an initial cohort of 223 AML patients, we found no correlation between bone marrow blast content and cytopenia, questioning the displacement theory. Measuring serum concentration of thrombopoietin (TPO), a key regulator of hematopoietic stem cells and megakaryocytes, revealed loss of physiologic negative correlation with platelet count in AML cases with blasts expressing MPL, the thrombopoietin (scavenging) receptor. Mechanistic studies demonstrated that MPLhi blasts could indeed clear TPO, likely therefore leading to insufficient cytokine levels for nonleukemic hematopoiesis. Microarray analysis in an independent multicenter study cohort of 437 AML cases validated MPL expression as a central predictor of thrombocytopenia and neutropenia in AML. Moreover, t(8;21) AML cases demonstrated the highest average MPL expression and lowest average platelet and absolute neutrophil counts among subgroups. Our work thus explains the pathophysiology of peripheral blood cytopenia in a relevant number of AML cases.
Collapse
|
179
|
Papp S, Moderzynski K, Rauch J, Heine L, Kuehl S, Richardt U, Mueller H, Fleischer B, Osterloh A. Liver Necrosis and Lethal Systemic Inflammation in a Murine Model of Rickettsia typhi Infection: Role of Neutrophils, Macrophages and NK Cells. PLoS Negl Trop Dis 2016; 10:e0004935. [PMID: 27548618 PMCID: PMC4993389 DOI: 10.1371/journal.pntd.0004935] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/31/2016] [Indexed: 12/31/2022] Open
Abstract
Rickettsia (R.) typhi is the causative agent of endemic typhus, an emerging febrile disease that is associated with complications such as pneumonia, encephalitis and liver dysfunction. To elucidate how innate immune mechanisms contribute to defense and pathology we here analyzed R. typhi infection of CB17 SCID mice that are congenic to BALB/c mice but lack adaptive immunity. CB17 SCID mice succumbed to R. typhi infection within 21 days and showed high bacterial load in spleen, brain, lung, and liver. Most evident pathological changes in R. typhi-infected CB17 SCID mice were massive liver necrosis and splenomegaly due to the disproportionate accumulation of neutrophils and macrophages (MΦ). Both neutrophils and MΦ infiltrated the liver and harbored R. typhi. Both cell populations expressed iNOS and produced reactive oxygen species (ROS) and, thus, exhibited an inflammatory and bactericidal phenotype. Surprisingly, depletion of neutrophils completely prevented liver necrosis but neither altered bacterial load nor protected CB17 SCID mice from death. Furthermore, the absence of neutrophils had no impact on the overwhelming systemic inflammatory response in these mice. This response was predominantly driven by activated MΦ and NK cells both of which expressed IFNγ and is considered as the reason of death. Finally, we observed that iNOS expression by MΦ and neutrophils did not correlate with R. typhi uptake in vivo. Moreover, we demonstrate that MΦ hardly respond to R. typhi in vitro. These findings indicate that R. typhi enters MΦ and also neutrophils unrecognized and that activation of these cells is mediated by other mechanisms in the context of tissue damage in vivo.
Collapse
Affiliation(s)
- Stefanie Papp
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Kristin Moderzynski
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jessica Rauch
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Liza Heine
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Svenja Kuehl
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Ulricke Richardt
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Heidelinde Mueller
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Bernhard Fleischer
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Osterloh
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
180
|
Wojtowicz EE, Lechman ER, Hermans KG, Schoof EM, Wienholds E, Isserlin R, van Veelen PA, Broekhuis MJC, Janssen GMC, Trotman-Grant A, Dobson SM, Krivdova G, Elzinga J, Kennedy J, Gan OI, Sinha A, Ignatchenko V, Kislinger T, Dethmers-Ausema B, Weersing E, Alemdehy MF, de Looper HWJ, Bader GD, Ritsema M, Erkeland SJ, Bystrykh LV, Dick JE, de Haan G. Ectopic miR-125a Expression Induces Long-Term Repopulating Stem Cell Capacity in Mouse and Human Hematopoietic Progenitors. Cell Stem Cell 2016; 19:383-96. [PMID: 27424784 DOI: 10.1016/j.stem.2016.06.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 04/01/2016] [Accepted: 06/15/2016] [Indexed: 12/25/2022]
Abstract
Umbilical cord blood (CB) is a convenient and broadly used source of hematopoietic stem cells (HSCs) for allogeneic stem cell transplantation. However, limiting numbers of HSCs remain a major constraint for its clinical application. Although one feasible option would be to expand HSCs to improve therapeutic outcome, available protocols and the molecular mechanisms governing the self-renewal of HSCs are unclear. Here, we show that ectopic expression of a single microRNA (miRNA), miR-125a, in purified murine and human multipotent progenitors (MPPs) resulted in increased self-renewal and robust long-term multi-lineage repopulation in transplanted recipient mice. Using quantitative proteomics and western blot analysis, we identified a restricted set of miR-125a targets involved in conferring long-term repopulating capacity to MPPs in humans and mice. Our findings offer the innovative potential to use MPPs with enhanced self-renewal activity to augment limited sources of HSCs to improve clinical protocols.
Collapse
Affiliation(s)
- Edyta E Wojtowicz
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Karin G Hermans
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Erwin M Schoof
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Erno Wienholds
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Ruth Isserlin
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Peter A van Veelen
- Departments of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Mathilde J C Broekhuis
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - George M C Janssen
- Departments of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Aaron Trotman-Grant
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Stephanie M Dobson
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gabriela Krivdova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jantje Elzinga
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - James Kennedy
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Olga I Gan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Ankit Sinha
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Vladimir Ignatchenko
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Bertien Dethmers-Ausema
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - Ellen Weersing
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - Mir Farshid Alemdehy
- Department of Hematology, Erasmus University Medical Center Cancer Institute, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Hans W J de Looper
- Department of Hematology, Erasmus University Medical Center Cancer Institute, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Martha Ritsema
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - Stefan J Erkeland
- Department of Immunology, Erasmus University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands
| | - Leonid V Bystrykh
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Gerald de Haan
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands.
| |
Collapse
|
181
|
Type 2 Interleukin-4 Receptor Signaling in Neutrophils Antagonizes Their Expansion and Migration during Infection and Inflammation. Immunity 2016; 45:172-84. [DOI: 10.1016/j.immuni.2016.06.025] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 03/29/2016] [Accepted: 06/23/2016] [Indexed: 12/23/2022]
|
182
|
Choi W, Ji YW, Ham HY, Yeo A, Noh H, Jin SE, Song JS, Kim HC, Kim EK, Lee HK. Gr-1intCD11b+ myeloid-derived suppressor cells accumulate in corneal allograft and improve corneal allograft survival. J Leukoc Biol 2016; 100:1453-1463. [PMID: 27370015 DOI: 10.1189/jlb.5a1115-508rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 05/18/2016] [Accepted: 06/12/2016] [Indexed: 11/24/2022] Open
Abstract
We identified the characteristics of myeloid-derived suppressor cells (MDSCs) and investigated their mechanism of induction and their functional role in allograft rejection using a murine corneal allograft model. In mice, MDSCs coexpress CD11b and myeloid differentiation antigen Gr-1. Gr-1+CD11b+ cells infiltrated allografted corneas between 4 d and 4 wk after surgery; however, the frequencies of Gr-1+CD11b+ cells were not different between accepted and rejected allografts or in peripheral blood or BM. Of interest, Gr-1intCD11b+ cells, but not Gr-1hiCD11b+ cells, infiltrated the accepted graft early after surgery and expressed high levels of immunosuppressive cytokines, including IL-10, TGF-β, and TNF-related apoptosis-inducing ligand. This population remained until 4 wk after surgery. In vitro, only high dose (>100 ng/ml) of IFN-γ plus GM-CSF could induce immunosuppressive cytokine expression in Gr-1intCD11b+ cells. Furthermore, adoptive transfer of Gr-1intCD11b+ cells reduced T cell infiltration, which improved graft survival. In conclusion, high-dose IFN-γ in allograft areas is essential for development of Gr-1intCD11b+ MDSCs in corneal allografts, and subtle environmental changes in the early period of the allograft can result in a large difference in graft survival.
Collapse
Affiliation(s)
- Wungrak Choi
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Woo Ji
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea.,Corneal Dystrophy Research Institute, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Hwa-Yong Ham
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Areum Yeo
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyemi Noh
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Su-Eon Jin
- College of Pharmacy, Yonsei University, Incheon, Korea
| | - Jong Suk Song
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Hyeon Chang Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eung Kwon Kim
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea.,Corneal Dystrophy Research Institute, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyung Keun Lee
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea; .,Corneal Dystrophy Research Institute, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
183
|
Abstract
Evidence presented over the last few years indicates that the hematopoietic stem cell (HSC) compartment comprises not just one but a number of different cell populations. Based on HSCs’ proliferation and engraftment potential, it has been suggested that there are two classes of HSC, with long- and short-term engraftment potential. HSC heterogeneity seems to involve differentiation capacities as well, since it has been shown that some HSC clones are able to give rise to both myeloid and lymphoid progeny, whereas others are lymphoid deficient. It has been recognized that HSC function depends on intrinsic cell regulators, which are modulated by external signals. Among the former, we can include transcription factors and non-coding RNAs as well as epigenetic modifiers. Among the latter, cytokines and extracellular matrix molecules have been implicated. Understanding the elements and mechanisms that regulate HSC populations is of significant relevance both in biological and in clinical terms, and research in this area still has to face several complex and exciting challenges.
Collapse
Affiliation(s)
- Hector Mayani
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, IMSS National Medical Center, Mexico City, Mexico
| |
Collapse
|
184
|
Kim AR, Sankaran VG. Development of autologous blood cell therapies. Exp Hematol 2016; 44:887-94. [PMID: 27345108 DOI: 10.1016/j.exphem.2016.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/21/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation and blood cell transfusions are performed commonly in patients with a variety of blood disorders. Unfortunately, these donor-derived cell therapies are constrained due to limited supplies, infectious risk factors, a lack of appropriately matched donors, and the risk of immunologic complications from such products. The use of autologous cell therapies has been proposed to overcome these shortcomings. One can derive such therapies directly from hematopoietic stem and progenitor cells of individuals, which can then be manipulated ex vivo to produce the desired modifications or differentiated to produce a particular target population. Alternatively, pluripotent stem cells, which have a theoretically unlimited self-renewal capacity and an ability to differentiate into any desired cell type, can be used as an autologous starting source for such manipulation and differentiation approaches. Such cell products can also be used as a delivery vehicle for therapeutics. In this review, we highlight recent advances and discuss ongoing challenges for the in vitro generation of autologous hematopoietic cells that can be used for cell therapy.
Collapse
Affiliation(s)
- Ah Ram Kim
- Division of Hematology/Oncology, Boston Children's Hospital, and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital, and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
185
|
Sallman DA, Cluzeau T, Basiorka AA, List A. Unraveling the Pathogenesis of MDS: The NLRP3 Inflammasome and Pyroptosis Drive the MDS Phenotype. Front Oncol 2016; 6:151. [PMID: 27379212 PMCID: PMC4909736 DOI: 10.3389/fonc.2016.00151] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/03/2016] [Indexed: 12/31/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are characterized by bone marrow cytological dysplasia and ineffective hematopoiesis in the setting of recurrent somatic gene mutations and chromosomal abnormalities. The underlying pathogenic mechanisms that drive a common clinical phenotype from a diverse array of genetic abnormalities have only recently begun to emerge. Accumulating evidence has highlighted the integral role of the innate immune system in upregulating inflammatory cytokines via NF-κB activation in the pathogenesis of MDS. Recent investigations implicate activation of the NLRP3 inflammasome in hematopoietic stem/progenitor cells as a critical convergence signal in MDS with consequent clonal expansion and pyroptotic cell death though caspase-1 maturation. Specifically, the alarmin S100A9 and/or founder gene mutations trigger pyroptosis through the generation of reactive oxygen species leading to assembly and activation of the redox-sensitive NLRP3 inflammasome and β–catenin, assuring propagation of the MDS clone. More importantly, targeted inhibition of varied steps in this pathway restore effective hematopoiesis. Together, delineation of the role of pyroptosis in the clinical phenotype of MDS patients has identified novel therapeutic strategies that offer significant promise in the treatment of MDS.
Collapse
Affiliation(s)
- David A Sallman
- Malignant Hematology Department, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL , USA
| | - Thomas Cluzeau
- Hematology Department, Centre Hospitalier Universitaire of Nice, Nice, France; Faculty of Medicine, University Nice Sophia Antipolis, Nice, France; Mediterranean Center of Molecular Medicine, INSERM U1065, Nice, France; French Group of Myelodysplasia, Paris, France
| | - Ashley A Basiorka
- Cancer Biology Ph.D. Program, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida , Tampa, FL , USA
| | - Alan List
- Malignant Hematology Department, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL , USA
| |
Collapse
|
186
|
Kraakman MJ, Dragoljevic D, Kammoun HL, Murphy AJ. Is the risk of cardiovascular disease altered with anti-inflammatory therapies? Insights from rheumatoid arthritis. Clin Transl Immunology 2016; 5:e84. [PMID: 27350883 PMCID: PMC4910124 DOI: 10.1038/cti.2016.31] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/12/2016] [Accepted: 04/12/2016] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of mortality worldwide. Atherosclerosis is the most common form of CVD, which is complex and multifactorial with an elevated risk observed in people with either metabolic or inflammatory diseases. Accumulating evidence now links obesity with a state of chronic low-grade inflammation and has renewed our understanding of this condition and its associated comorbidities. An emerging theme linking disease states with atherosclerosis is the increased production of myeloid cells, which can initiate and exacerbate atherogenesis. Although anti-inflammatory drug treatments exist and have been successfully used to treat inflammatory conditions such as rheumatoid arthritis (RA), a commonly observed side effect is dyslipidemia, inadvertently, a major risk factor for the development of atherosclerosis. The mechanisms leading to dyslipidemia associated with anti-inflammatory drug use and whether CVD risk is actually increased by this dyslipidemia are of great therapeutic importance and currently remain poorly understood. Here we review recent data providing links between inflammation, hematopoiesis, dyslipidemia and CVD risk in the context of anti-inflammatory drug use.
Collapse
Affiliation(s)
- Michael J Kraakman
- Department of Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Dragana Dragoljevic
- Department of Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| | - Helene L Kammoun
- Department of Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| | - Andrew J Murphy
- Department of Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
187
|
Barminko J, Reinholt B, Baron MH. Development and differentiation of the erythroid lineage in mammals. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 58:18-29. [PMID: 26709231 PMCID: PMC4775370 DOI: 10.1016/j.dci.2015.12.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/15/2015] [Accepted: 12/15/2015] [Indexed: 05/02/2023]
Abstract
The red blood cell (RBC) is responsible for performing the highly specialized function of oxygen transport, making it essential for survival during gestation and postnatal life. Establishment of sufficient RBC numbers, therefore, has evolved to be a major priority of the postimplantation embryo. The "primitive" erythroid lineage is the first to be specified in the developing embryo proper. Significant resources are dedicated to producing RBCs throughout gestation. Two transient and morphologically distinct waves of hematopoietic progenitor-derived erythropoiesis are observed in development before hematopoietic stem cells (HSCs) take over to produce "definitive" RBCs in the fetal liver. Toward the end of gestation, HSCs migrate to the bone marrow, which becomes the primary site of RBC production in the adult. Erythropoiesis is regulated at various stages of erythroid cell maturation to ensure sufficient production of RBCs in response to physiological demands. Here, we highlight key aspects of mammalian erythroid development and maturation as well as differences among the primitive and definitive erythroid cell lineages.
Collapse
Affiliation(s)
- Jeffrey Barminko
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brad Reinholt
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Margaret H Baron
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
188
|
Early production of human neutrophils and platelets posttransplant is severely compromised by growth factor exposure. Exp Hematol 2016; 44:635-40. [PMID: 27090409 DOI: 10.1016/j.exphem.2016.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 01/24/2023]
Abstract
The critical human cells that produce neutrophils and platelets within 3 weeks in recipients of hematopoietic transplants are thought to produce these mature blood cells with the same kinetics in sublethally irradiated immunodeficient mice. Quantification of their numbers indicates their relative underrepresentation in cord blood (CB), likely explaining the clinical inadequacy of single CB units in rescuing hematopoiesis in myelosuppressed adult patients. We here describe that exposure of CD34(+) CB cells ex vivo to growth factors that markedly expand their numbers and colony-forming cell content also rapidly (within 24 hours) produce a significant and sustained net loss of their original short-term repopulating activity. This loss of short-term in vivo repopulating activity affects early platelet production faster than early neutrophil output, consistent with their origin from distinct input populations. Moreover, this growth factor-mediated loss is not abrogated by published strategies to increase progenitor homing despite evidence that the effect on rapid neutrophil production is paralleled in time and amount by a loss of the homing of their committed clonogenic precursors to the bone marrow. These results highlight the inability of in vitro or phenotype assessments to reliably predict clinical engraftment kinetics of cultured CB cells.
Collapse
|
189
|
Kasahara S, Jhingran A, Dhingra S, Salem A, Cramer RA, Hohl TM. Role of Granulocyte-Macrophage Colony-Stimulating Factor Signaling in Regulating Neutrophil Antifungal Activity and the Oxidative Burst During Respiratory Fungal Challenge. J Infect Dis 2016; 213:1289-98. [PMID: 26908736 PMCID: PMC4799674 DOI: 10.1093/infdis/jiw054] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/28/2016] [Indexed: 12/13/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a pleiotropic cytokine that plays a critical role in regulating myeloid cell host defense. In this study, we demonstrated that GM-CSF signaling plays an essential role in antifungal defense against Aspergillus fumigatus. Mice that lack the GM-CSF receptor β chain (GM-CSFRβ) developed invasive hyphal growth and exhibited impaired survival after pulmonary challenge with A. fumigatus conidia. GM-CSFRβ signaling regulated the recruitment of inflammatory monocytes to infected lungs, but not the recruitment of effector neutrophils. Cell-intrinsic GM-CSFRβ signaling mediated neutrophil and inflammatory monocyte antifungal activity, because lung GM-CSFRβ(-/-) leukocytes exhibited impaired conidial killing compared with GM-CSFRβ(+/+) counterparts in mixed bone marrow chimeric mice. GM-CSFRβ(-/-) neutrophils exhibited reduced (hydrogenated) nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity in vivo. Conversely, administration of recombinant GM-CSF enhanced neutrophil NADPH oxidase function, conidiacidal activity, and lung fungal clearance in A. fumigatus-challenged mice. Thus, our study illustrates the functional role of GM-CSFRβ signaling on lung myeloid cell responses against inhaled A. fumigatus conidia and demonstrates a benefit for systemic GM-CSF administration.
Collapse
Affiliation(s)
| | | | - Sourabh Dhingra
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Anand Salem
- Infectious Disease Service, Department of Medicine
| | - Robert A Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth University, Hanover, New Hampshire
| | - Tobias M Hohl
- Infectious Disease Service, Department of Medicine Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
190
|
Park HJ, Li J, Hannah R, Biddie S, Leal-Cervantes AI, Kirschner K, Flores Santa Cruz D, Sexl V, Göttgens B, Green AR. Cytokine-induced megakaryocytic differentiation is regulated by genome-wide loss of a uSTAT transcriptional program. EMBO J 2016; 35:580-94. [PMID: 26702099 PMCID: PMC4801948 DOI: 10.15252/embj.201592383] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 11/29/2022] Open
Abstract
Metazoan development is regulated by transcriptional networks, which must respond to extracellular cues including cytokines. The JAK/STAT pathway is a highly conserved regulatory module, activated by many cytokines, in which tyrosine-phosphorylated STATs (pSTATs) function as transcription factors. However, the mechanisms by which STAT activation modulates lineage-affiliated transcriptional programs are unclear. We demonstrate that in the absence of thrombopoietin (TPO), tyrosine-unphosphorylated STAT5 (uSTAT5) is present in the nucleus where it colocalizes with CTCF and represses a megakaryocytic transcriptional program. TPO-mediated phosphorylation of STAT5 triggers its genome-wide relocation to STAT consensus sites with two distinct transcriptional consequences, loss of a uSTAT5 program that restrains megakaryocytic differentiation and activation of a canonical pSTAT5-driven program which includes regulators of apoptosis and proliferation. Transcriptional repression by uSTAT5 reflects restricted access of the megakaryocytic transcription factor ERG to target genes. These results identify a previously unrecognized mechanism of cytokine-mediated differentiation.
Collapse
Affiliation(s)
- Hyun Jung Park
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Juan Li
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Rebecca Hannah
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Simon Biddie
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Ana I Leal-Cervantes
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Kristina Kirschner
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - David Flores Santa Cruz
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, Veterinary University Vienna, Vienna, Austria
| | - Berthold Göttgens
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Anthony R Green
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
191
|
Ryu SH, Na HY, Sohn M, Han SM, Choi W, In H, Hong S, Jeon H, Seo JY, Ahn J, Park CG. Reduced expression of granule proteins during extended survival of eosinophils in splenocyte culture with GM-CSF. Immunol Lett 2016; 173:7-20. [PMID: 26969350 DOI: 10.1016/j.imlet.2016.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 03/03/2016] [Accepted: 03/07/2016] [Indexed: 01/21/2023]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a multifaceted hematopoietic cytokine and the culture of mouse bone marrow with GM-CSF produces a variety of myeloid cells including granulocytes, macrophages, and dendritic cells. In the present study, we cultured mouse splenocytes with GM-CSF and examined the changes in hematopoietic cell populations over a week. Most of the splenic hematopoietic cells disappeared significantly from culture within 6days with or without the presence of GM-CSF. Among the splenic granulocyte populations, only eosinophils fully survived throughout the culture with GM-CSF for more than a week. During 10days of culture with GM-CSF, splenic eosinophils maintained their morphology as well as most of their surface molecules at high levels, including CCR3 and Siglec F. Meanwhile, the expression of mRNAs encoding major basic protein-1 (MBP-1) and eosinophil peroxidase (EPO), two major eosinophil-derived granule proteins, was diminished significantly from the cultured eosinophils. EPO assays also revealed that eosinophils in culture for more than 5days retained 30% or less EPO activity compared to those in uncultured splenocytes. In contrast, culture of splenocytes with GM-CSF did not change the capacity of eosinophils to migrate in response to eotaxin-1. Our results indicate that mouse splenic eosinophils are effectively cultured for lengthy periods while their expression of eosinophil-derived granule proteins is specifically suppressed. The relevance of these findings to eosinophilic inflammatory response is discussed.
Collapse
Affiliation(s)
- Seul Hye Ryu
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hye Young Na
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Moah Sohn
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sun Murray Han
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Wanho Choi
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyunju In
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sookyung Hong
- Laboratory of Molecular Virology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyejin Jeon
- Laboratory of Molecular Virology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jun-Young Seo
- Laboratory of Molecular Virology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jongcheol Ahn
- WOOJUNG Life Science Research Center, WOOJUNGBSC, Suwon, Gyeonggi-do 16229, Republic of Korea
| | - Chae Gyu Park
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
192
|
Abstract
Our understanding of cancer biology has been radically transformed over recent years with a more realistic grasp of its multilayered cellular and genetic complexity. These advances are being translated into more selective and effective treatment of cancers and, although there are still considerable challenges, particularly with drug resistance and metastatic disease, many patients with otherwise lethal malignancies now enjoy protracted remissions or cure. One largely unheralded theme of this story is the extent to which new biological insights and novel clinical applications have their origins with leukaemia and related blood cell cancers, including lymphoma. In this Timeline article, I review the remarkable and ground-breaking role that studies in leukaemia have had at the forefront of this progress.
Collapse
Affiliation(s)
- Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, Brookes Lawley Building, 15 Cotswold Road, Sutton SM2 5NG, UK
| |
Collapse
|
193
|
Lundie RJ, Webb LM, Marley AK, Phythian-Adams AT, Cook PC, Jackson-Jones LH, Brown S, Maizels RM, Boon L, O'Keeffe M, MacDonald AS. A central role for hepatic conventional dendritic cells in supporting Th2 responses during helminth infection. Immunol Cell Biol 2015; 94:400-10. [PMID: 26657145 PMCID: PMC4817239 DOI: 10.1038/icb.2015.114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/24/2015] [Accepted: 11/25/2015] [Indexed: 01/23/2023]
Abstract
Dendritic cells (DCs) are the key initiators of T-helper (Th) 2 immune responses against the parasitic helminth Schistosoma mansoni. Although the liver is one of the main sites of antigen deposition during infection with this parasite, it is not yet clear how distinct DC subtypes in this tissue respond to S. mansoni antigens in vivo, or how the liver microenvironment might influence DC function during establishment of the Th2 response. In this study, we show that hepatic DC subsets undergo distinct activation processes in vivo following murine infection with S. mansoni. Conventional DCs (cDCs) from schistosome-infected mice upregulated expression of the costimulatory molecule CD40 and were capable of priming naive CD4(+) T cells, whereas plasmacytoid DCs (pDCs) upregulated expression of MHC class II, CD86 and CD40 but were unable to support the expansion of either naive or effector/memory CD4(+) T cells. Importantly, in vivo depletion of pDCs revealed that this subset was dispensable for either maintenance or regulation of the hepatic Th2 effector response during acute S. mansoni infection. Our data provides strong evidence that S. mansoni infection favors the establishment of an immunogenic, rather than tolerogenic, liver microenvironment that conditions cDCs to initiate and maintain Th2 immunity in the context of ongoing antigen exposure.
Collapse
Affiliation(s)
- Rachel J Lundie
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, UK.,Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia
| | - Lauren M Webb
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Angela K Marley
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, UK
| | | | - Peter C Cook
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Lucy H Jackson-Jones
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, UK
| | - Sheila Brown
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Rick M Maizels
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, UK
| | - Louis Boon
- EPIRUS Biopharmaceuticals, Utrecht, The Netherlands
| | - Meredith O'Keeffe
- Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Andrew S MacDonald
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| |
Collapse
|
194
|
Abstract
Granulocyte-macrophage colony stimulating factor (GM-CSF) is a growth factor first identified as an inducer of differentiation and proliferation of granulocytes and macrophages derived from haematopoietic progenitor cells. Later studies have shown that GM-CSF is involved in a wide range of biological processes in both innate and adaptive immunity, with its production being tightly linked to the response to danger signals. Given that the functions of GM-CSF span multiple tissues and biological processes, this cytokine has shown potential as a new and important therapeutic target in several autoimmune and inflammatory disorders - particularly in rheumatoid arthritis. Indeed, GM-CSF was one of the first cytokines detected in human synovial fluid from inflamed joints. Therapies that target GM-CSF or its receptor have been tested in preclinical studies with promising results, further supporting the potential of targeting the GM-CSF pathway. In this Review, we discuss our expanding view of the biology of GM-CSF, outline what has been learnt about GM-CSF from studies of animal models and human diseases, and summarize the results of early phase clinical trials evaluating GM-CSF antagonism in inflammatory disorders.
Collapse
|
195
|
Globular adiponectin induces leukocytosis and mobilizes hematopoietic progenitor cells in mice. Tissue Eng Regen Med 2015. [DOI: 10.1007/s13770-015-0040-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
196
|
Kobayashi H, Suda T, Takubo K. How hematopoietic stem/progenitors and their niche sense and respond to infectious stress. Exp Hematol 2015; 44:92-100. [PMID: 26646990 DOI: 10.1016/j.exphem.2015.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/18/2015] [Accepted: 11/21/2015] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) play important roles in fighting systemic infection as they supply immune cells in a demand-adapted manner. Various mechanisms govern HSPC responses to infection, including cytokine signaling, niche function, and direct sensing of pathogen-derived molecules by HSPCs themselves. Here we review recent advances in our understanding of HSPC responses to infection and also consider newly identified STING-mediated machinery recognizing bacteria-derived cyclic dinucleotides.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Toshio Suda
- Cancer Science Institute, National University of Singapore, Singapore
| | - Keiyo Takubo
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan.
| |
Collapse
|
197
|
G'Sell RT, Gaffney PM, Powell DW. A20-Binding Inhibitor of NF-κB Activation 1 is a Physiologic Inhibitor of NF-κB: A Molecular Switch for Inflammation and Autoimmunity. Arthritis Rheumatol 2015; 67:2292-302. [PMID: 26097105 DOI: 10.1002/art.39245] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/09/2015] [Indexed: 01/24/2023]
Affiliation(s)
- Rachel T G'Sell
- University of Louisville School of Medicine, Louisville, Kentucky
| | | | - David W Powell
- University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
198
|
Havixbeck JJ, Barreda DR. Neutrophil Development, Migration, and Function in Teleost Fish. BIOLOGY 2015; 4:715-34. [PMID: 26561837 PMCID: PMC4690015 DOI: 10.3390/biology4040715] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 12/23/2022]
Abstract
It is now widely recognized that neutrophils are sophisticated cells that are critical to host defense and the maintenance of homeostasis. In addition, concepts such as neutrophil plasticity are helping to define the range of phenotypic profiles available to cells in this group and the physiological conditions that contribute to their differentiation. Herein, we discuss key features of the life of a teleost neutrophil including their development, migration to an inflammatory site, and contributions to pathogen killing and the control of acute inflammation. The potent anti-microbial mechanisms elicited by these cells in bony fish are a testament to their long-standing evolutionary contributions in host defense. In addition, recent insights into their active roles in the control of inflammation prior to induction of apoptosis highlight their importance to the maintenance of host integrity in these early vertebrates. Overall, our goal is to summarize recent progress in our understanding of this cell type in teleost fish, and to provide evolutionary context for the contributions of this hematopoietic lineage in host defense and an efficient return to homeostasis following injury or infection.
Collapse
Affiliation(s)
- Jeffrey J Havixbeck
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G2P5, Canada.
| | - Daniel R Barreda
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G2P5, Canada.
| |
Collapse
|
199
|
Bartels M, Murphy K, Rieter E, Bruin M. Understanding chronic neutropenia: life is short. Br J Haematol 2015; 172:157-69. [PMID: 26456767 DOI: 10.1111/bjh.13798] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The pathophysiological mechanisms underlying chronic neutropenia are extensive, varying from haematopoietic stem cell disorders resulting in defective neutrophil production, to accelerated apoptosis of neutrophil progenitors or circulating mature neutrophils. While the knowledge concerning genetic defects associated with congenital neutropenia or bone marrow failure is increasing rapidly, the functional role and consequences of these genetic alterations is often not well understood. In addition, there is a large group of diseases, including primary immunodeficiencies and metabolic diseases, in which chronic neutropenia is one of the symptoms, while there is no clear bone marrow pathology or haematopoietic stem cell dysfunction. Altogether, these disease entities illustrate the complexity of normal neutrophil development, the functional role of the (bone marrow) microenvironment and the increased propensity to undergo apoptosis, which is typical for neutrophils. The large variety of disorders associated with chronic neutropenia makes classification almost impossible and possibly not desirable, based on the clinical phenotypes. However, a better understanding of the regulation of normal myeloid differentiation and neutrophil development is of great importance in the diagnostic evaluation of unexplained chronic neutropenia. In this review we propose insights in the pathophysiology of chronic neutropenia in the context of the functional role of key players during normal neutrophil development, neutrophil release and neutrophil survival.
Collapse
Affiliation(s)
- Marije Bartels
- Department of Paediatric Haematology and Stem Cell Transplantation, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Kate Murphy
- Department of Paediatric Haematology and Stem Cell Transplantation, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Ester Rieter
- Department of Paediatric Haematology and Stem Cell Transplantation, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Marrie Bruin
- Department of Paediatric Haematology and Stem Cell Transplantation, University Medical Centre Utrecht, Utrecht, the Netherlands
| |
Collapse
|
200
|
Gao J, Swaminathan S, Pai N, Johnson Z, Chen YH, Peterson L, Goolsby C. Flow cytometric detection of altered signaling in myelodysplastic syndrome and cytopenia. Leuk Res 2015; 39:1396-404. [PMID: 26410459 DOI: 10.1016/j.leukres.2015.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 08/17/2015] [Accepted: 09/06/2015] [Indexed: 10/23/2022]
Abstract
Multiparameter flow cytometric analysis allows for precise evaluation of growth factor stimulated intracellular signaling in distinct immunophenotype defined hematopoetic populations. Our analysis of intracellular phosphoprotein in response to major hematopoietic growth factors or cytokines showed several interesting findings. Although there was no characteristic signaling abnormality that was diagnostic for MDS, MDS cases were often associated with more signaling aberrancies involving more cellular populations. Higher than average response in the CD34(+)CD117(+) progenitor cells to Flt3 ligand and stem cell factor stimulation was frequently associated with high risk features or disease progression in MDS. Although preliminary results hint an adverse prognostic role of dysregulated FLT3 pathway in MDS cases, whether this observation adds independent prognostic value to the existing prognostic system needs to be further explored in future prospective studies.
Collapse
Affiliation(s)
- Juehua Gao
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA.
| | - Suchitra Swaminathan
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - Navin Pai
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - Zachary Johnson
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - Yi-Hua Chen
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - LoAnn Peterson
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - Charles Goolsby
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| |
Collapse
|