1
|
Pittaluga JR, Birnberg-Weiss F, Serafino A, Castro JE, Castillo LA, Martire-Greco D, Barrionuevo P, Fernández GC, Landoni VI. The RNA from Pseudomonas aeruginosa Reduces Neutrophil Responses Favoring Bacterial Survival. J Innate Immun 2024; 16:489-500. [PMID: 39293427 PMCID: PMC11521516 DOI: 10.1159/000541414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024] Open
Abstract
INTRODUCTION Epithelial and endothelial cells modulate innate immune responses in the lung, including the arrival of neutrophils (PMN), which are crucial cells for the antibacterial host defense. Cells are exposed to prokaryotic RNA (pRNA) during bacterial infections and different pRNA may promote or attenuate the inflammatory response on different immune cells. Pseudomonas aeruginosa (PAE) can cause severe pneumonia and has several immune-evading mechanisms. The aim of this study was to determine the effects of the RNA from PAE (RNAPAE) on lung epithelial, endothelial cells, and PMN, and its impact on bacterial elimination. METHODS Purified total RNAPAE was used as a stimulus on a human lung epithelial cell line (Calu-6), human microvascular endothelial cell line HMEC-1 and isolated healthy human PMN. Activation and cytokine secretion were evaluated. In addition, PMN elimination of live ECO or PAE was determined in the presence of RNAPAE. RESULTS We found that RNAPAE either induced a pro-inflammatory response on Calu-6 and HMEC-1 or PMN. Pre-stimulation of PMN with RNAPAE diminished activation and chemotaxis induced by live bacteria. Moreover, we found that RNAPAE reduced phagocytosis of live ECO. Finally, we also found that non-degraded fragments of small RNA (<200 bp) were responsible for the PMN microbicidal attenuation during PAE elimination. CONCLUSION Our results indicated that short fragments of RNAPAE diminished the immune response of PMN favoring bacterial survival.
Collapse
Affiliation(s)
- Jose R Pittaluga
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Federico Birnberg-Weiss
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Agustina Serafino
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Joselyn E Castro
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Luis A Castillo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Daiana Martire-Greco
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Paula Barrionuevo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Gabriela C Fernández
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Verónica I Landoni
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
2
|
García-López JP, Grimaldi A, Chen Z, Meneses C, Bravo-Tello K, Bresciani E, Banderas A, Burgess SM, Hernández PP, Feijoo CG. Ontogenetically distinct neutrophils differ in function and transcriptional profile in zebrafish. Nat Commun 2023; 14:4942. [PMID: 37582932 PMCID: PMC10427629 DOI: 10.1038/s41467-023-40662-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 08/07/2023] [Indexed: 08/17/2023] Open
Abstract
The current view of hematopoiesis considers leukocytes on a continuum with distinct developmental origins, and which exert non-overlapping functions. However, there is less known about the function and phenotype of ontogenetically distinct neutrophil populations. In this work, using a photoconvertible transgenic zebrafish line; Tg(mpx:Dendra2), we selectively label rostral blood island-derived and caudal hematopoietic tissue-derived neutrophils in vivo during steady state or upon injury. By comparing the migratory properties and single-cell expression profiles of both neutrophil populations at steady state we show that rostral neutrophils show higher csf3b expression and migration capacity than caudal neutrophils. Upon injury, both populations share a core transcriptional profile as well as subset-specific transcriptional signatures. Accordingly, both rostral and caudal neutrophils are recruited to the wound independently of their distance to the injury. While rostral neutrophils respond uniformly, caudal neutrophils respond heterogeneously. Collectively, our results reveal that co-existing neutrophils populations with ontogenically distinct origin display functional differences.
Collapse
Affiliation(s)
- Juan P García-López
- Fish Immunology Laboratory, Faculty of Life Science, Andres Bello University, Santiago, Chile
| | - Alexandre Grimaldi
- Stem Cells & Development Unit, Institut Pasteur, 75015, Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Zelin Chen
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Claudio Meneses
- Millennium Nucleus Development of Super Adaptable Plants (MN-SAP), Santiago, 8331150, Chile
- Millennium Institute Center for Genome Regulation (CRG), Santiago, 8331150, Chile
- Departamento de Fruticultura y Enología, Facultad de Agronomía e Ingeniería Forestal, Pontificia Universidad Católica de Chile, Santiago, 7820436, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - Karina Bravo-Tello
- Fish Immunology Laboratory, Faculty of Life Science, Andres Bello University, Santiago, Chile
| | - Erica Bresciani
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Alvaro Banderas
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, 75005, Paris, France
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA.
| | - Pedro P Hernández
- Institut Curie, PSL Research University, INSERM U934/CNRS UMR3215, Development and Homeostasis of Mucosal Tissues Lab, Paris, France.
| | - Carmen G Feijoo
- Fish Immunology Laboratory, Faculty of Life Science, Andres Bello University, Santiago, Chile.
| |
Collapse
|
3
|
Masgrau-Alsina S, Wackerbarth LM, Lim DS, Sperandio M. MST1 controls murine neutrophil homeostasis via the G-CSFR/STAT3 axis. Front Immunol 2022; 13:1038936. [PMID: 36618429 PMCID: PMC9816424 DOI: 10.3389/fimmu.2022.1038936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
The release of neutrophils from the bone marrow into the blood circulation is essential for neutrophil homeostasis and the protection of the organism from invading microorganisms. Granulocyte colony-stimulating factor (G-CSF) plays a pivotal role in this process and guides granulopoiesis as well as the release of bone marrow neutrophils into the blood stream both during homeostasis and in case of infection through activation of the G-CSF receptor/signal transduction and activation of transcription 3 (STAT3) signaling pathway. Here, we investigated the role of the mammalian sterile 20-like kinase 1 (MST1) for neutrophil homeostasis and neutrophil mobilization. We found increased plasma levels of G-CSF in Mst1 -/- mice compared to wild type mice both under homeostatic conditions as well as after stimulation with the proinflammatory cytokine TNF-α. In addition, G-CSF-induced mobilization of neutrophils from the bone marrow into the blood circulation in vivo was markedly reduced in the absence of MST1. Interestingly, this was not accompanied by differences in the number of blood neutrophils. Addressing the underlying molecular mechanism of MST1-regulated neutrophil mobilization, we found reduced STAT3 phosphorylation and impaired upregulation of CXCR2 in Mst1 -/- bone marrow neutrophils compared to wild type cells, while JAK2 phosphorylation was not altered. Taken together, we identify MST1 as a critical modulator of neutrophil homeostasis and neutrophil mobilization from the bone marrow, which adds another important aspect to the complex role of MST1 in regulating innate immunity.
Collapse
Affiliation(s)
- Sergi Masgrau-Alsina
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Lou Martha Wackerbarth
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Dae-sik Lim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians University Munich, Munich, Germany,*Correspondence: Markus Sperandio,
| |
Collapse
|
4
|
Li F, Gao C, Li X, Wang J, Zhao Y, Ke Y, Liu Y, Liu HM, Hu Z, Wei L, Chen ZS. Jiyuan oridonin A induces differentiation of acute myeloid leukemia cells including leukemic stem-like cells. Front Pharmacol 2022; 13:1001552. [PMID: 36133825 PMCID: PMC9484275 DOI: 10.3389/fphar.2022.1001552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive form of hematological neoplasia characterized by failure of myeloid differentiation. AML is a leading cause of death from leukemia. Cytarabine chemotherapy resistance is a major source of refractory/relapsed AML. A major obstacle to the successful treatment of AML results from residual disease maintained by leukemic stem cells (LSCs), which are mostly resistant to conventional chemotherapy. Here, we determined the effect of a natural compound, Jiyuan oridonin A (JOA), on the differentiation blockade in the M2 subtype [particularly t (8;21)] of AML cells, M3 subtype of AML cells (APL cells), and leukemic stem-like cells both in vitro and in vivo. We found that JOA induced cell differentiation and suppressed the colony formation capacity in various AML cell lines (Kasumi-1, KG-1, MUTZ-8, NB4, and HL-60) without eliciting apoptosis. The mechanism of JOA-induced cell differentiation depends on the specificity of cell type. JOA mediated the differentiation of Kasumi-1 cells by activating the hematopoietic cell lineage signaling pathway, while inhibition of c-MYC was involved in the JOA-induced differentiation of NB4 cells. Moreover, JOA was identified to target leukemic stem-like cells by induced cell differentiation in vivo. These findings demonstrated that JOA could inhibit the proliferation of M2 and M3 subtypes of AML cells and leukemic stem-like cells by overcoming the differentiation blockade, which may offer a novel therapeutic strategy for AML to overcome relapse and drug resistance in patients with AML. Our findings highlight the possibility of using compounds like JOA as a promising differentiation-induced agent for the treatment of AML.
Collapse
Affiliation(s)
- Fahui Li
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Congying Gao
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Xueming Li
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Jiangyun Wang
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Yao Zhao
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yu Ke
- School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Ying Liu
- School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Hong-Min Liu
- School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Zhenbo Hu
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Zhenbo Hu, ; Liuya Wei, ; Zhe-Sheng Chen,
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, China
- *Correspondence: Zhenbo Hu, ; Liuya Wei, ; Zhe-Sheng Chen,
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
- *Correspondence: Zhenbo Hu, ; Liuya Wei, ; Zhe-Sheng Chen,
| |
Collapse
|
5
|
Malengier-Devlies B, Metzemaekers M, Wouters C, Proost P, Matthys P. Neutrophil Homeostasis and Emergency Granulopoiesis: The Example of Systemic Juvenile Idiopathic Arthritis. Front Immunol 2021; 12:766620. [PMID: 34966386 PMCID: PMC8710701 DOI: 10.3389/fimmu.2021.766620] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are key pathogen exterminators of the innate immune system endowed with oxidative and non-oxidative defense mechanisms. More recently, a more complex role for neutrophils as decision shaping cells that instruct other leukocytes to fine-tune innate and adaptive immune responses has come into view. Under homeostatic conditions, neutrophils are short-lived cells that are continuously released from the bone marrow. Their development starts with undifferentiated hematopoietic stem cells that pass through different immature subtypes to eventually become fully equipped, mature neutrophils capable of launching fast and robust immune responses. During severe (systemic) inflammation, there is an increased need for neutrophils. The hematopoietic system rapidly adapts to this increased demand by switching from steady-state blood cell production to emergency granulopoiesis. During emergency granulopoiesis, the de novo production of neutrophils by the bone marrow and at extramedullary sites is augmented, while additional mature neutrophils are rapidly released from the marginated pools. Although neutrophils are indispensable for host protection against microorganisms, excessive activation causes tissue damage in neutrophil-rich diseases. Therefore, tight regulation of neutrophil homeostasis is imperative. In this review, we discuss the kinetics of neutrophil ontogenesis in homeostatic conditions and during emergency myelopoiesis and provide an overview of the different molecular players involved in this regulation. We substantiate this review with the example of an autoinflammatory disease, i.e. systemic juvenile idiopathic arthritis.
Collapse
Affiliation(s)
- Bert Malengier-Devlies
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mieke Metzemaekers
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Carine Wouters
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.,Division of Pediatric Rheumatology, University Hospitals Leuven, Leuven, Belgium.,European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at University Hospital Leuven, Leuven, Belgium
| | - Paul Proost
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Martin KR, Wong HL, Witko-Sarsat V, Wicks IP. G-CSF - A double edge sword in neutrophil mediated immunity. Semin Immunol 2021; 54:101516. [PMID: 34728120 DOI: 10.1016/j.smim.2021.101516] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/23/2021] [Indexed: 11/15/2022]
Abstract
Neutrophils are vital for the innate immune system's control of pathogens and neutrophil deficiency can render the host susceptible to life-threatening infections. Neutrophil responses must also be tightly regulated because excessive production, recruitment or activation of neutrophils can cause tissue damage in both acute and chronic inflammatory diseases. Granulocyte colony stimulating factor (G-CSF) is a key regulator of neutrophil biology, from production, differentiation, and release of neutrophil precursors in the bone marrow (BM) to modulating the function of mature neutrophils outside of the BM, particularly at sites of inflammation. G-CSF acts by binding to its cognate cell surface receptor on target cells, causing the activation of intracellular signalling pathways mediating the proliferation, differentiation, function, and survival of cells in the neutrophil lineage. Studies in humans and mice demonstrate that G-CSF contributes to protecting the host against infection, but conversely, it can play a deleterious role in inflammatory diseases. As such, neutrophils and the G-CSF pathway may provide novel therapeutic targets. This review will focus on understanding the role G-CSF plays in the balance between effective neutrophil mediated host defence versus neutrophil-mediated inflammation and tissue damage in various inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Katherine R Martin
- WEHI, 1G Royal Parade, Parkville, Victoria, 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Huon L Wong
- WEHI, 1G Royal Parade, Parkville, Victoria, 3052, Australia
| | | | - Ian P Wicks
- WEHI, 1G Royal Parade, Parkville, Victoria, 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia.
| |
Collapse
|
7
|
Castillo LA, Birnberg Weiss F, Rodriguez-Rodrigues N, Pittaluga JR, Martire-Greco D, Milillo MA, Grinstein SF, Camelli MR, Mena Aybar AJ, Landoni VI, Fernández GC. Prokaryotic RNA activates endothelial cells promoting neutrophil transmigration. Immunol Cell Biol 2019; 97:815-825. [PMID: 31264260 DOI: 10.1111/imcb.12282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/27/2019] [Accepted: 06/29/2019] [Indexed: 01/18/2023]
Abstract
Endothelial cell (EC)-neutrophil (PMN) interactions are crucial in the resolution of bacterial infections. Prokaryotic RNA (pRNA) has been reported as a pathogen-associated molecular pattern that is released from bacteria upon death and is able to activate PMN. In this work, we studied the effects of pRNA on EC and investigated whether these effects could modulate EC-PMN interaction. For this purpose, we purified total pRNA from Escherichia coli and used it as a stimulus for Human Umbilical Vein Endothelial Cells (HUVEC). We found that the incubation of pRNA with HUVEC caused the increase of surface intercellular adhesion molecule 1 (ICAM-1 or CD54) expression on HUVEC, and the secretion of IL-8 and von Willebrand factor, characteristics consistent with HUVEC activation, without causing toxic effects. Moreover, pRNA-treated HUVEC also induced PMN adhesion and the conditioned medium obtained from treated-HUVEC was chemotactic for PMN and caused their activation, as determined by CD11b upregulation. As reported previously, the degradation products of pRNA induced similar biological effects. The treatment of HUVEC with endocytosis inhibitors revealed that the entry of pRNA partially relied on a clathrin-dependent mechanism, whereas the effects of degradation products could not be inhibited by any of the inhibitors tested. Using a transwell system, we found that pRNA or degraded pRNA were also able to stimulate HUVEC when recognized from the basolateral side. Our results indicate that pRNA activates EC, resulting in the modulation of EC-PMN interaction by inducing PMN chemotaxis, adhesion and activation. In the context of infection, pRNA sensed by EC and PMN could favor bacterial clearance.
Collapse
Affiliation(s)
- Luis A Castillo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Pacheco de Melo 3081, C1425AUM, Ciudad Autónoma de Buenos Aires, Argentina
| | - Federico Birnberg Weiss
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Pacheco de Melo 3081, C1425AUM, Ciudad Autónoma de Buenos Aires, Argentina
| | - Nahuel Rodriguez-Rodrigues
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Pacheco de Melo 3081, C1425AUM, Ciudad Autónoma de Buenos Aires, Argentina
| | - José R Pittaluga
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Pacheco de Melo 3081, C1425AUM, Ciudad Autónoma de Buenos Aires, Argentina
| | - Daiana Martire-Greco
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Pacheco de Melo 3081, C1425AUM, Ciudad Autónoma de Buenos Aires, Argentina
| | - Maria A Milillo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Pacheco de Melo 3081, C1425AUM, Ciudad Autónoma de Buenos Aires, Argentina
| | - Sebastian F Grinstein
- Servicio de Obstetricia, Hospital Militar Central Cirujano Mayor Dr. Cosme Argerich, Luis María Campos 726, C1426BOR, Ciudad Autónoma de Buenos Aires, Argentina
| | - María R Camelli
- Servicio de Obstetricia, Hospital Militar Central Cirujano Mayor Dr. Cosme Argerich, Luis María Campos 726, C1426BOR, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ana J Mena Aybar
- Servicio de Obstetricia, Hospital Militar Central Cirujano Mayor Dr. Cosme Argerich, Luis María Campos 726, C1426BOR, Ciudad Autónoma de Buenos Aires, Argentina
| | - Verónica I Landoni
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Pacheco de Melo 3081, C1425AUM, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gabriela C Fernández
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Pacheco de Melo 3081, C1425AUM, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
8
|
Castillo LA, Birnberg-Weiss F, Rodriguez-Rodrigues N, Martire-Greco D, Bigi F, Landoni VI, Gomez SA, Fernandez GC. Klebsiella pneumoniae ST258 Negatively Regulates the Oxidative Burst in Human Neutrophils. Front Immunol 2019; 10:929. [PMID: 31105712 PMCID: PMC6497972 DOI: 10.3389/fimmu.2019.00929] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/11/2019] [Indexed: 11/13/2022] Open
Abstract
The epidemic clone of Klebsiella pneumoniae (Kpn), sequence type 258 (ST258), carbapenamase producer (KPC), commonly infects hospitalized patients that are left with scarce therapeutic option since carbapenems are last resort antibiotics for life-threatening bacterial infections. To improve prevention and treatment, we should better understand the biology of Kpn KPC ST258 infections. Our hypothesis was that Kpn KPC ST258 evade the first line of defense of innate immunity, the polymorphonuclear neutrophil (PMN), by decreasing its functional response. Therefore, our aim was to evaluate how the ST258 Kpn clone affects PMN responses, focusing on the respiratory burst, compared to another opportunistic pathogen, Escherichia coli (Eco). We found that Kpn KPC ST258 was unable to trigger bactericidal responses as reactive oxygen species (ROS) generation and NETosis, compared to the high induction observed with Eco, but both bacterial strains were similarly phagocytized and cause increases in cell size and CD11b expression. The absence of ROS induction was also observed with other Kpn ST258 strains negative for KPC. These results reflect certain selectivity in terms of the functions that are triggered in PMN by Kpn, which seems to evade specifically those responses critical for bacterial survival. In this sense, bactericidal mechanisms evasion was associated with a higher survival of Kpn KPC ST258 compared to Eco. To investigate the mechanisms and molecules involved in ROS inhibition, we used bacterial extracts (BE) and found that BE were able to inhibit ROS generation triggered by the well-known ROS inducer, fMLP. A sequence of experiments led us to elucidate that the polysaccharide part of LPS was responsible for this inhibition, whereas lipid A mediated the other responses that were not affected by bacteria, such as cell size increase and CD11b up-regulation. In conclusion, we unraveled a mechanism of immune evasion of Kpn KPC ST258, which may contribute to design more effective strategies for the treatment of these multi-resistant bacterial infections.
Collapse
Affiliation(s)
- Luis A Castillo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)- Consejo Nacional de investigaciones Científicas y Tecnológicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Federico Birnberg-Weiss
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)- Consejo Nacional de investigaciones Científicas y Tecnológicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Nahuel Rodriguez-Rodrigues
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)- Consejo Nacional de investigaciones Científicas y Tecnológicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Daiana Martire-Greco
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)- Consejo Nacional de investigaciones Científicas y Tecnológicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Fabiana Bigi
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Veronica I Landoni
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)- Consejo Nacional de investigaciones Científicas y Tecnológicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Sonia A Gomez
- Servicio de Antimicrobianos, Instituto Nacional de Enfermedades Infecciosas Dr. Carlos G. Malbrán (INEI), Administración Nacional de Laboratorios e Institutos de Salud (ANLIS), Buenos Aires, Argentina
| | - Gabriela C Fernandez
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)- Consejo Nacional de investigaciones Científicas y Tecnológicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
9
|
Mycobacterium tuberculosis Multidrug-Resistant Strain M Induces Low IL-8 and Inhibits TNF- α Secretion by Bronchial Epithelial Cells Altering Neutrophil Effector Functions. Mediators Inflamm 2017; 2017:2810606. [PMID: 28852268 PMCID: PMC5568625 DOI: 10.1155/2017/2810606] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/22/2017] [Accepted: 07/02/2017] [Indexed: 12/17/2022] Open
Abstract
M strain, the most prevalent multidrug-resistant strain of Mycobacterium tuberculosis (Mtb) in Argentina, has mounted mechanisms to evade innate immune response. The role of human bronchial epithelium in Mtb infection remains unknown as well as its crosstalk with neutrophils (PMN). In this work, we evaluate whether M and H37Rv strains invade and replicate within bronchial epithelial cell line Calu-6 and how conditioned media (CM) derived from infected cells alter PMN responses. We demonstrated that M infects and survives within Calu-6 without promoting death. CM from M-infected Calu-6 (M-CM) did not attract PMN in correlation with its low IL-8 content compared to H37Rv-CM. Also, PMN activation and ROS production in response to irradiated H37Rv were impaired after treatment with M-CM due to the lack of TNF-α. Interestingly, M-CM increased H37Rv replication in PMN which would allow the spreading of mycobacteria upon PMN death and sustain IL-8 release. Thus, our results indicate that even at low invasion/replication rate within Calu-6, M induces the secretion of factors altering the crosstalk between these nonphagocytic cells and PMN, representing an evasion mechanism developed by M strain to persist in the host. These data provide new insights on the role of bronchial epithelium upon M infection.
Collapse
|
10
|
Rodriguez-Rodrigues N, Castillo LA, Landoni VI, Martire-Greco D, Milillo MA, Barrionuevo P, Fernández GC. Prokaryotic RNA Associated to Bacterial Viability Induces Polymorphonuclear Neutrophil Activation. Front Cell Infect Microbiol 2017; 7:306. [PMID: 28730145 PMCID: PMC5498479 DOI: 10.3389/fcimb.2017.00306] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/20/2017] [Indexed: 11/13/2022] Open
Abstract
Polymorphonuclear neutrophils (PMN) are the first cellular line of antibacterial host defense. They sense pathogens through recognition of pathogen-associated molecular patterns (PAMPs) by innate pattern recognition receptors, such as Toll-like receptors (TLR). The aim of this study was to investigate whether PMN sense bacterial viability and explore which viability factor could be involved in this phenomenon. For this purpose, different functions were evaluated in isolated human PMN using live Escherichia coli (Ec) and heat-killed Ec (HK-Ec). We found that bacterial viability was indispensable to induce PMN activation, as measured by forward-scatter (FSC) increase, CD11b surface expression, chemotaxis, reactive oxygen species (ROS) generation and neutrophil extracellular trap (NET) formation. As uncapped non-polyadenylated prokaryotic mRNA has been recognized as a PAMP associated to bacterial viability by macrophages and dendritic cells, total prokaryotic RNA (pRNA) from live Ec was purified and used as a stimulus for PMN. pRNA triggered similar responses to those observed with live bacteria. No RNA could be isolated from HK-Ec, explaining the lack of effect of dead bacteria. Moreover, the supernatant of dead bacteria was able to induce PMN activation, and this was associated with the presence of pRNA in this supernatant, which is released in the killing process. The induction of bactericidal functions (ROS and NETosis) by pRNA were abolished when the supernatant of dead bacteria or isolated pRNA were treated with RNAse. Moreover, endocytosis was necessary for pRNA-induced ROS generation and NETosis, and priming was required for the induction of pRNA-induced ROS in whole blood. However, responses related to movement and degranulation (FSC increase, CD11b up-regulation, and chemotaxis) were still triggered when pRNA was digested with RNase, and were not dependent on pRNA endocytosis or PMN priming. In conclusion, our results indicate that PMN sense live bacteria through recognition of pRNA, and this sensing triggers potent bactericidal mechanisms.
Collapse
Affiliation(s)
- Nahuel Rodriguez-Rodrigues
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental CONICET, Academia Nacional de MedicinaBuenos Aires, Argentina
| | - Luis A Castillo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental CONICET, Academia Nacional de MedicinaBuenos Aires, Argentina
| | - Verónica I Landoni
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental CONICET, Academia Nacional de MedicinaBuenos Aires, Argentina
| | - Daiana Martire-Greco
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental CONICET, Academia Nacional de MedicinaBuenos Aires, Argentina
| | - M Ayelén Milillo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental CONICET, Academia Nacional de MedicinaBuenos Aires, Argentina
| | - Paula Barrionuevo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental CONICET, Academia Nacional de MedicinaBuenos Aires, Argentina
| | - Gabriela C Fernández
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental CONICET, Academia Nacional de MedicinaBuenos Aires, Argentina
| |
Collapse
|
11
|
Pedersen CC, Borup R, Fischer-Nielsen A, Mora-Jensen H, Fossum A, Cowland JB, Borregaard N. Changes in Gene Expression during G-CSF-Induced Emergency Granulopoiesis in Humans. THE JOURNAL OF IMMUNOLOGY 2016; 197:1989-99. [PMID: 27481851 DOI: 10.4049/jimmunol.1502690] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/27/2016] [Indexed: 12/30/2022]
Abstract
Emergency granulopoiesis refers to the increased production of neutrophils in bone marrow and their release into circulation induced by severe infection. Several studies point to a critical role for G-CSF as the main mediator of emergency granulopoiesis. However, the consequences of G-CSF stimulation on the transcriptome of neutrophils and their precursors have not yet been investigated in humans. In this work, we examine the changes in mRNA expression induced by administration of G-CSF in vivo, as a model of emergency granulopoiesis in humans. Blood samples were collected from healthy individuals after 5 d of G-CSF administration. Neutrophil precursors were sorted into discrete stages of maturation by flow cytometry, and RNA was subjected to microarray analysis. mRNA levels were compared with previously published expression levels in corresponding populations of neutrophil precursors isolated from bone marrow of untreated, healthy individuals. One thousand one hundred and ten mRNAs were differentially expressed >2-fold throughout terminal granulopoiesis. Major changes were seen in pathways involved in apoptosis, cytokine signaling, and TLR pathways. In addition, G-CSF treatment reduced the levels of four of five measured granule proteins in mature neutrophils, including the proantibacterial protein hCAP-18, which was completely deficient in neutrophils from G-CSF-treated donors. These results indicate that multiple biological processes are altered to satisfy the increased demand for neutrophils during G-CSF-induced emergency granulopoiesis in humans.
Collapse
Affiliation(s)
- Corinna C Pedersen
- Granulocyte Research Laboratory, Department of Hematology, National University Hospital, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Rehannah Borup
- Center for Genomic Medicine, National University Hospital, DK-2100 Copenhagen, Denmark
| | - Anne Fischer-Nielsen
- Department of Clinical Immunology, Cell Therapy Facility, National University Hospital, DK-2100 Copenhagen, Denmark; and
| | - Helena Mora-Jensen
- Granulocyte Research Laboratory, Department of Hematology, National University Hospital, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Anna Fossum
- Biotech and Research Innovation Centre, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Jack B Cowland
- Granulocyte Research Laboratory, Department of Hematology, National University Hospital, University of Copenhagen, DK-2100 Copenhagen, Denmark;
| | - Niels Borregaard
- Granulocyte Research Laboratory, Department of Hematology, National University Hospital, University of Copenhagen, DK-2100 Copenhagen, Denmark;
| |
Collapse
|
12
|
Goldberg GL, Cornish AL, Murphy J, Pang ES, Lim LL, Campbell IK, Scalzo-Inguanti K, Chen X, McMenamin PG, Maraskovsky E, McKenzie BS, Wicks IP. G-CSF and Neutrophils Are Nonredundant Mediators of Murine Experimental Autoimmune Uveoretinitis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:172-84. [DOI: 10.1016/j.ajpath.2015.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 09/11/2015] [Accepted: 09/24/2015] [Indexed: 10/22/2022]
|
13
|
Landoni VI, Chiarella P, Martire-Greco D, Schierloh P, van-Rooijen N, Rearte B, Palermo MS, Isturiz MA, Fernández GC. Tolerance to lipopolysaccharide promotes an enhanced neutrophil extracellular traps formation leading to a more efficient bacterial clearance in mice. Clin Exp Immunol 2012; 168:153-63. [PMID: 22385250 DOI: 10.1111/j.1365-2249.2012.04560.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Tolerance to lipopolysaccharide (LPS) constitutes a stress adaptation, in which a primary contact with LPS results in a minimal response when a second exposure with the same stimulus occurs. However, active important defence mechanisms are mounted during the tolerant state. Our aim was to assess the contribution of polymorphonuclear neutrophils (PMN) in the clearance of bacterial infection in a mouse model of tolerance to LPS. After tolerance was developed, we investigated in vivo different mechanisms of bacterial clearance. The elimination of a locally induced polymicrobial challenge was more efficient in tolerant mice both in the presence or absence of local macrophages. This was related to a higher number of PMN migrating to the infectious site as a result of an increased number of PMN from the marginal pool with higher chemotactic capacity, not because of differences in their phagocytic activity or reactive species production. In vivo, neutrophils extracellular trap (NET) destruction by nuclease treatment abolished the observed increased clearance in tolerant but not in control mice. In line with this finding, in vitro NETs formation was higher in PMN from tolerant animals. These results indicate that the higher chemotactic response from an increased PMN marginal pool and the NETs enhanced forming capacity are the main mechanisms mediating bacterial clearance in tolerant mice. To sum up, far from being a lack of response, tolerance to LPS causes PMN priming effects which favour distant and local anti-infectious responses.
Collapse
Affiliation(s)
- V I Landoni
- Immunology Department of the Instituto de Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lin L, Betsuyaku T, Heimbach L, Li N, Rubenstein D, Shapiro SD, An L, Giudice GJ, Diaz LA, Senior RM, Liu Z. Neutrophil elastase cleaves the murine hemidesmosomal protein BP180/type XVII collagen and generates degradation products that modulate experimental bullous pemphigoid. Matrix Biol 2012; 31:38-44. [PMID: 21979170 PMCID: PMC3261345 DOI: 10.1016/j.matbio.2011.09.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 09/16/2011] [Accepted: 09/19/2011] [Indexed: 12/21/2022]
Abstract
Bullous pemphigoid (BP) is an autoimmune subepidermal blistering disease associated with autoantibodies against the hemidesmosomal proteins BP180 and BP230. In the IgG passive transfer model of BP, blister formation is triggered by anti-BP180 IgG and depends on complement activation, mast cell degranulation, and neutrophil recruitment. Mice lacking neutrophil elastase (NE) do not develop experimental BP. Here, we demonstrated that NE degrades recombinant mouse BP180 within the immunodominant extracellular domain at amino acid positions 506 and 561, generating peptide p561 and peptide p506. Peptide p561 is chemotactic for neutrophils both in vitro and in vivo. Local injection of NE into B6 mice recruits neutrophils to the skin, and neutrophil infiltration is completely blocked by co-injection with the NE inhibitor α1-proteinase inhibitor. More importantly, NE directly cleaves BP180 in mouse and human skin, as well as the native human BP180 trimer molecule. These results demonstrate that (i) NE directly damages the extracellular matrix and (ii) NE degradation of mouse BP180 generates neutrophil chemotactic peptides that amplify disease severity at the early stage of the disease.
Collapse
Affiliation(s)
- Lan Lin
- Dalian University of Technology, Dalian, Liaoning, P.R.C
- Department of Dermatology, University of North Carolina, Chapel Hill, NC, U.S.A
| | - Tomoko Betsuyaku
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | - Lisa Heimbach
- Department of Dermatology, University of North Carolina, Chapel Hill, NC, U.S.A
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, U.S.A
| | - Ning Li
- Department of Dermatology, University of North Carolina, Chapel Hill, NC, U.S.A
| | - David Rubenstein
- Department of Dermatology, University of North Carolina, Chapel Hill, NC, U.S.A
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, U.S.A
| | - Steven D. Shapiro
- Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, U.S.A
| | - Lijia An
- Dalian University of Technology, Dalian, Liaoning, P.R.C
| | - George J. Giudice
- Department of Dermatology, University of Iowa, Iowa City, IA 52246, U.S.A
| | - Luis A. Diaz
- Department of Dermatology, University of North Carolina, Chapel Hill, NC, U.S.A
| | - Robert M. Senior
- Pulmonary and Critical Care Medicine, Washington University, St. Louis, MO, U.S.A
| | - Zhi Liu
- Department of Dermatology, University of North Carolina, Chapel Hill, NC, U.S.A
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, U.S.A
| |
Collapse
|
15
|
Etulain J, Negrotto S, Carestia A, Pozner RG, Romaniuk MA, D'Atri LP, Klement GL, Schattner M. Acidosis downregulates platelet haemostatic functions and promotes neutrophil proinflammatory responses mediated by platelets. Thromb Haemost 2011; 107:99-110. [PMID: 22159527 DOI: 10.1160/th11-06-0443] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 10/19/2011] [Indexed: 01/06/2023]
Abstract
Acidosis is one of the hallmarks of tissue injury such as trauma, infection, inflammation, and tumour growth. Although platelets participate in the pathophysiology of all these processes, the impact of acidosis on platelet biology has not been studied outside of the quality control of laboratory aggregation assays or platelet transfusion optimization. Herein, we evaluate the effect of physiologically relevant changes in extracellular acidosis on the biological function of platelets, placing particular emphasis on haemostatic and secretory functions. Platelet haemostatic responses such as adhesion, spreading, activation of αIIbβ3 integrin, ATP release, aggregation, thromboxane B2 generation, clot retraction and procoagulant activity including phosphatidilserine exposure and microparticle formation, showed a statistically significant inhibition of thrombin-induced changes at pH of 7.0 and 6.5 compared to the physiological pH (7.4). The release of alpha granule content was differentially regulated by acidosis. At low pH, thrombin or collagen-induced secretion of vascular endothelial growth factor and endostatin were dramatically reduced. The release of von Willebrand factor and stromal derived factor-1α followed a similar, albeit less dramatic pattern. In contrast, the induction of CD40L was not changed by low pH, and P-selectin exposure was significantly increased. While the generation of mixed platelet-leukocyte aggregates and the increased chemotaxis of neutrophils mediated by platelets were further augmented under acidic conditions in a P-selectin dependent manner, the increased neutrophil survival was independent of P-selectin expression. In conclusion, our results indicate that extracellular acidosis downregulates most of the haemostatic platelet functions, and promotes those involved in amplifying the neutrophil-mediated inflammatory response.
Collapse
Affiliation(s)
- Julia Etulain
- Thrombosis I Laboratory, National Academy of Medicine, CONICET, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Semple BD, Bye N, Ziebell JM, Morganti-Kossmann MC. Deficiency of the chemokine receptor CXCR2 attenuates neutrophil infiltration and cortical damage following closed head injury. Neurobiol Dis 2010; 40:394-403. [PMID: 20621186 DOI: 10.1016/j.nbd.2010.06.015] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 06/23/2010] [Accepted: 06/26/2010] [Indexed: 12/21/2022] Open
Abstract
The contribution of infiltrated neutrophils to secondary damage following traumatic brain injury remains controversial. Chemokines that regulate neutrophil migration by signaling through the CXCR2 receptor are markedly elevated by brain injury and are associated with the propagation of secondary damage. This study thus investigated the function of CXCR2 in posttraumatic inflammation and secondary degeneration by examining Cxcr2-deficient (Cxcr2(-/-)) mice over 14 days following closed head injury (CHI). We demonstrate a significant attenuation of neutrophil infiltration in Cxcr2(-/-) mice at 12 hours and 7 days after CHI, despite increased levels of CXC neutrophil-attracting chemokines in the lesioned cortex. This coincides with reduced tissue damage, neuronal loss, and cell death in Cxcr2(-/-) mice compared to wild-type controls, with heterozygotes showing intermediate responses. In contrast, blood-brain barrier permeability and functional recovery did not appear to be affected by Cxcr2 deletion. This study highlights the deleterious contribution of neutrophils to posttraumatic neurodegeneration and demonstrates the importance of CXC chemokine signaling in this process. Therefore, CXCR2 antagonistic therapeutics currently in development for other inflammatory conditions may also be of benefit in posttraumatic neuroinflammation.
Collapse
Affiliation(s)
- Bridgette D Semple
- National Trauma Research Institute, The Alfred Hospital, Melbourne, Victoria, Australia.
| | | | | | | |
Collapse
|
17
|
Shiga toxin 1-induced inflammatory response in lipopolysaccharide-sensitized astrocytes is mediated by endogenous tumor necrosis factor alpha. Infect Immun 2009; 78:1193-201. [PMID: 20008539 DOI: 10.1128/iai.00932-09] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hemolytic-uremic syndrome (HUS) is generally caused by Shiga toxin (Stx)-producing Escherichia coli. Endothelial dysfunction mediated by Stx is a central aspect in HUS development. However, inflammatory mediators such as bacterial lipopolysaccharide (LPS) and polymorphonuclear neutrophils (PMN) contribute to HUS pathophysiology by potentiating Stx effects. Acute renal failure is the main feature of HUS, but in severe cases, patients can develop neurological complications, which are usually associated with death. Although the mechanisms of neurological damage remain uncertain, alterations of the blood-brain barrier associated with brain endothelial injury is clear. Astrocytes (ASTs) are the most abundant inflammatory cells of the brain that modulate the normal function of brain endothelium and neurons. The aim of this study was to evaluate the effects of Stx type 1 (Stx1) alone or in combination with LPS in ASTs. Although Stx1 induced a weak inflammatory response, pretreatment with LPS sensitized ASTs to Stx1-mediated effects. Moreover, LPS increased the level of expression of the Stx receptor and its internalization. An early inflammatory response, characterized by the release of tumor necrosis factor alpha (TNF-alpha) and nitric oxide and PMN-chemoattractant activity, was induced by Stx1 in LPS-sensitized ASTs, whereas activation, evidenced by higher levels of glial fibrillary acid protein and cell death, was induced later. Furthermore, increased adhesion and PMN-mediated cytotoxicity were observed after Stx1 treatment in LPS-sensitized ASTs. These effects were dependent on NF-kappaB activation or AST-derived TNF-alpha. Our results suggest that TNF-alpha is a pivotal effector molecule that amplifies Stx1 effects on LPS-sensitized ASTs, contributing to brain inflammation and leading to endothelial and neuronal injury.
Collapse
|
18
|
Moriyama C, Betsuyaku T, Ito Y, Hamamura I, Hata J, Takahashi H, Nasuhara Y, Nishimura M. Aging enhances susceptibility to cigarette smoke-induced inflammation through bronchiolar chemokines. Am J Respir Cell Mol Biol 2009; 42:304-11. [PMID: 19491340 DOI: 10.1165/rcmb.2009-0025oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cigarette smoking and aging are major risk factors for chronic obstructive pulmonary disease. An unsolved question is whether elderly lungs are particularly vulnerable to cigarette smoke (CS) exposure. In this study, we used a mouse model to test the hypothesis that aging increases the susceptibility to CS-induced pulmonary inflammation. We subjected 9-week-old and 69-week-old C57BL/6J mice to CS (whole-body exposure, 90 min/d), and evaluated neutrophil infiltration in the lungs, the levels of keratinocyte-derived chemokine (KC) and macrophage inflammatory protein (MIP)-2 in bronchoalveolar lavage fluid, and mRNA expression in bronchiolar epithelium retrieved by laser capture microdissection. The 69-week-old mice showed a greater number of neutrophils and higher levels of bronchiolar KC and MIP-2 expression than 9-week-old mice after 9 days of CS exposure. Furthermore, single CS exposure induced the rapid up-regulation of KC and MIP-2 in bronchiolar epithelium in both 9-week-old and 69-week-old mice, and the much higher levels in 69-week-old mice were associated with greater nuclear translocation of NF-kappaB. In contrast, no age-related differences were observed in the bronchiolar expression of NF-E2-related factor 2-regulated antioxidant and detoxification genes, heme oxygenase-1, reduced nicotinamide adenine dinucleotide phosphate quinone reductase 1, and glutamate-cysteine ligase, modifier unit, or antioxidant activity in bronchoalveolar lavage fluid, regardless of CS exposure. In summary, aging increases susceptibility to CS-induced inflammation in a mouse model, and robust mRNA up-regulation and nuclear translocation of NF-kappaB in bronchiolar epithelium may be involved.
Collapse
Affiliation(s)
- Chinatsu Moriyama
- First Department of Medicine, Hokkaido University School of Medicine, N-15, W-7, Kita-ku, Sapporo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Zhang Y, Cheng G, Yang K, Fan R, Xu Z, Chen L, Li Q, Yang A, Jin B. A novel function of granulocyte colony‐stimulating factor in mobilization of human hematopoietic progenitor cells. Immunol Cell Biol 2009; 87:428-32. [DOI: 10.1038/icb.2009.9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yun Zhang
- Department of Immunology, The Fourth Military Medical University Xi'an China
| | - Guang Cheng
- Department of Neurosurgery, Xijing Hospital, The Fourth Military Medical University Xi'an China
| | - Kun Yang
- Department of Immunology, The Fourth Military Medical University Xi'an China
| | - Rong Fan
- Department of Physiology, The Fourth Military Medical University Xi'an China
| | - Zhuwei Xu
- Department of Immunology, The Fourth Military Medical University Xi'an China
| | - Lihua Chen
- Department of Immunology, The Fourth Military Medical University Xi'an China
| | - Qi Li
- Department of Immunology, The Fourth Military Medical University Xi'an China
| | - Angang Yang
- Department of Immunology, The Fourth Military Medical University Xi'an China
| | - Boquan Jin
- Department of Immunology, The Fourth Military Medical University Xi'an China
| |
Collapse
|
20
|
UEMURA Y, TAGUCHI T, KUBOTA T, SAITO T, BANDOBASHI K, YOKOYAMA A. Neutrophil function and cytokine-specific signaling in chronic neutrophilic leukemia. Int J Lab Hematol 2009; 31:36-47. [DOI: 10.1111/j.1751-553x.2007.01000.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Jilma B, Hergovich N, Homoncik M, Jilma-Stohlawetz P, Kreuzer C, Eichler HG, Zellner M, Pugin J. Granulocyte colony-stimulating factor (G-CSF) downregulates its receptor (CD114) on neutrophils and induces gelatinase B release in humans. Br J Haematol 2008. [DOI: 10.1111/j.1365-2141.2000.02320.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
22
|
Xu J, Gao XP, Ramchandran R, Zhao YY, Vogel SM, Malik AB. Nonmuscle myosin light-chain kinase mediates neutrophil transmigration in sepsis-induced lung inflammation by activating beta2 integrins. Nat Immunol 2008; 9:880-6. [PMID: 18587400 DOI: 10.1038/ni.1628] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Accepted: 05/29/2008] [Indexed: 11/09/2022]
Abstract
Nonmuscle myosin light-chain kinase (MYLK) mediates increased lung vascular endothelial permeability in lipopolysaccharide-induced lung inflammatory injury, the chief cause of the acute respiratory distress syndrome. In a lung injury model, we demonstrate here that MYLK was also essential for neutrophil transmigration, but that this function was mostly independent of myosin II regulatory light chain, the only known substrate of MYLK. Instead, MYLK in neutrophils was required for the recruitment and activation of the tyrosine kinase Pyk2, which mediated full activation of beta(2) integrins. Our results demonstrate that MYLK-mediated activation of beta(2) integrins through Pyk2 links beta(2) integrin signaling to the actin motile machinery of neutrophils.
Collapse
Affiliation(s)
- Jingsong Xu
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois 60612, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Panopoulos AD, Watowich SS. Granulocyte colony-stimulating factor: molecular mechanisms of action during steady state and 'emergency' hematopoiesis. Cytokine 2008; 42:277-88. [PMID: 18400509 DOI: 10.1016/j.cyto.2008.03.002] [Citation(s) in RCA: 279] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Revised: 02/14/2008] [Accepted: 03/03/2008] [Indexed: 01/13/2023]
Abstract
Neutrophils are phagocytes whose principal function is to maintain anti-bacterial immunity. Neutrophils ingest and kill invading bacteria, releasing cytotoxic, chemotactic and inflammatory mediators at sites of infection. This serves to control the immediate host immune response and attract other cells, such as macrophages and dendritic cells, which are important for establishing long-term adaptive immunity. Neutrophils thus contribute to both the initiation and the maintenance of inflammation at sites of infection. Aberrant neutrophil activity is deleterious; suppressed responses can cause extreme susceptibility to infection while overactivation can lead to excessive inflammation and tissue damage. This review will focus on neutrophil regulation by granulocyte colony-stimulating factor (G-CSF), the principal cytokine controlling neutrophil development and function. The review will emphasize the molecular aspects of G-CSF-driven granulopoiesis in steady state (healthy) conditions and during demand-driven or 'emergency' conditions elicited by infection or clinical administration of G-CSF. Understanding the molecular control of granulopoiesis will aid in the development of new approaches designed to treat disorders of neutrophil production and function.
Collapse
Affiliation(s)
- Athanasia D Panopoulos
- Department of Immunology and Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, PO Box 301402, Unit 902, Houston, TX 77030, USA
| | | |
Collapse
|
24
|
Witowski J, Ksiazek K, Warnecke C, Kuźlan M, Korybalska K, Tayama H, Wiśniewska-Elnur J, Pawlaczyk K, Trómińska J, Breborowicz A, Jörres A. Role of mesothelial cell-derived granulocyte colony-stimulating factor in interleukin-17-induced neutrophil accumulation in the peritoneum. Kidney Int 2007; 71:514-25. [PMID: 17228364 DOI: 10.1038/sj.ki.5002082] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recent studies suggest that peritoneal CD4(+) T lymphocytes may control recruitment of polymorphonuclear leukocytes (PMN) during peritonitis by an interleukin-17 (IL-17)-dependent mechanism. IL-17 and granulocyte colony-stimulating factor (G-CSF) have been proposed to form an axis that regulates PMN transmigration. Here we report on the role of G-CSF released by human peritoneal mesothelial cells (HPMCs) in IL-17A-mediated peritoneal PMN accumulation. In vitro exposure of HPMCs to IL-17A resulted in a time- and dose-dependent release of G-CSF. This effect was related to the induction of G-CSF mRNA and mediated through the nuclear factor-kappaB (NF-kappaB) pathway. The novel observation was that IL-17A-stimulated NF-kappaB activation in HPMCs followed a biphasic profile, with an early induction (45 min), followed by the return to basal levels (90 min), and a delayed induction (3 h). Tumor necrosis factor alpha synergistically amplified IL-17A-induced G-CSF production by enhanced NF-kappaB activation and through stabilization of G-CSF mRNA. Intraperitoneal (i.p.) administration of IL-17A in Balb/c mice resulted in increased local levels of G-CSF and selective PMN accumulation. Administration of anti-G-CSF blocking antibody before IL-17A injection significantly reduced the IL-17A-triggered PMN infiltration. This effect occurred despite increased i.p. levels of PMN-specific chemokines KC and macrophage inflammatory protein-2 seen in animals treated with anti-G-CSF antibody. These data demonstrate that the mesothelium-derived G-CSF plays an important role in IL-17A-induced PMN recruitment into the peritoneum.
Collapse
Affiliation(s)
- J Witowski
- Department of Pathophysiology, University Medical School, Poznan, Poland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gregory AD, Hogue LA, Ferkol TW, Link DC. Regulation of systemic and local neutrophil responses by G-CSF during pulmonary Pseudomonas aeruginosa infection. Blood 2006; 109:3235-43. [PMID: 17185469 PMCID: PMC1852251 DOI: 10.1182/blood-2005-01-015081] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) regulates the production, maturation, and function of neutrophils. Its expression is often induced during infection, resulting in high concentrations of G-CSF in inflammatory exudates and in the blood, suggesting that it may regulate both local and systemic neutrophil responses. Herein, we characterize the neutrophil response in G-CSFR(-/-) mice following intratracheal injection with Pseudomonas aeruginosa-laden agarose beads, modeling the pulmonary infection observed in many patients with cystic fibrosis. G-CSFR(-/-) mice are markedly susceptible to bronchopulmonary P aeruginosa infection, exhibiting decreased survival and bacterial clearance as well as extensive damage to lung tissue. The systemic neutrophil response was mediated primarily by enhanced neutrophil release from the bone marrow rather than increased neutrophil production and was attenuated in G-CSFR(-/-) mice. Despite normal to increased local production of inflammatory chemokines, neutrophil accumulation into the infected lung of G-CSFR(-/-) mice was markedly reduced. Moreover, the percentage of apoptotic neutrophils in the lung was elevated, suggesting that G-CSF signals may play an important role in regulating neutrophil survival at the inflammatory site. Collectively, these data provide new evidence that G-CSF signals play important but specific roles in the regulation of the systemic and local neutrophil response following infection.
Collapse
Affiliation(s)
- Alyssa D Gregory
- Department of Medicine, Division of Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
26
|
Schuster DP, Brody SL, Zhou Z, Bernstein M, Arch R, Link D, Mueckler M. Regulation of lipopolysaccharide-induced increases in neutrophil glucose uptake. Am J Physiol Lung Cell Mol Physiol 2006; 292:L845-51. [PMID: 17122354 DOI: 10.1152/ajplung.00350.2006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The pathogenesis of many lung diseases involves neutrophilic inflammation. Neutrophil functions, in turn, are critically dependent on glucose uptake and glycolysis to supply the necessary energy to meet these functions. In this study, we determined the effects of p38 mitogen-activated protein kinase and hypoxia-inducible factor (HIF)-1, as well as their potential interaction, on the expression of membrane glucose transporters and on glucose uptake in murine neutrophils. Neutrophils were harvested and purified from C57BL/6 mice and stimulated with lipopolysaccharide (LPS) in the presence or absence of specific p38 and HIF-1 inhibitors. Glucose uptake was measured as the rate of [3H]deoxyglucose (DG) uptake. We identified GLUT-1 in mouse neutrophils, but neither GLUT-3 nor GLUT-4 were detected using Western blot analysis, even after LPS stimulation. LPS stimulation did not increase GLUT-1 protein levels but did cause translocation of GLUT-1 from the cell interior to the cell surface, together with a dose-dependent increase in [3H]DG uptake, indicating that glucose uptake is regulated in these cells. LPS also activated both p38 and the HIF-1 pathway. Inhibitors of p38 and HIF-1 blocked GLUT-1 translocation and [3H]DG uptake. These data suggest that LPS-induced increases in neutrophil glucose uptake are mediated by GLUT-1 translocation to the cell surface in response to sequential activation of neutrophil p38 and HIF-1alpha in neutrophils. Given that neutrophil function and glucose metabolism are closely linked, control of the latter may represent a new target to ameliorate the deleterious effects of neutrophils on the lungs.
Collapse
Affiliation(s)
- Daniel P Schuster
- Department of Internal Medicine, Washington University School of Medicine, Univ. Box 8225, St. Louis, MO 63110, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Cataisson C, Pearson AJ, Tsien MZ, Mascia F, Gao JL, Pastore S, Yuspa SH. CXCR2 ligands and G-CSF mediate PKCalpha-induced intraepidermal inflammation. J Clin Invest 2006; 116:2757-66. [PMID: 16964312 PMCID: PMC1560349 DOI: 10.1172/jci27514] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 07/18/2006] [Indexed: 12/11/2022] Open
Abstract
Transgenic mice overexpressing PKCalpha in the epidermis (K5-PKCalpha mice) exhibit an inducible severe intraepidermal neutrophilic inflammation and systemic neutrophilia when PKCalpha is activated by topical 12-O-tetradecanoylphorbol-13-acetate (TPA). This inducible model of cutaneous inflammation was used to define mediators of skin inflammation that may have clinical relevance. Activation of cutaneous PKCalpha increased the production of the chemotactic factors cytokine-induced neutrophil chemoattractant (KC) and macrophage inflammatory protein 2 (MIP-2) in murine plasma. TPA treatment of cultured K5-PKCalpha keratinocytes also released KC and MIP-2 into culture supernatants through an NF-kappaB-dependent pathway. MIP-2 and KC mediated the infiltration of neutrophils into the epidermis, since this was prevented by ablating CXCR2 in K5-PKCalpha mice or administering neutralizing antibodies against KC or MIP-2. The neutrophilia resulted from PKCalpha-mediated upregulation of cutaneous G-CSF released into the plasma independent of CXCR2. These responses could be inhibited by topical treatment with a PKCalpha-selective inhibitor. Inhibiting PKCalpha also reduced the basal and TNF-alpha- or TPA-induced expression of CXCL8 in cultured psoriatic keratinocytes, suggesting that PKCalpha activity may contribute to psoriatic inflammation. Thus, skin can be the source of circulating factors that have both local and systemic consequences, and these factors, their receptors, and possibly PKCalpha could be therapeutic targets for inhibition of cutaneous inflammation.
Collapse
Affiliation(s)
- Christophe Cataisson
- Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, and
Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA.
Istituto Dermopatico dell’Immacolata, Rome, Italy
| | - Andrea J. Pearson
- Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, and
Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA.
Istituto Dermopatico dell’Immacolata, Rome, Italy
| | - Margaret Z. Tsien
- Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, and
Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA.
Istituto Dermopatico dell’Immacolata, Rome, Italy
| | - Francesca Mascia
- Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, and
Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA.
Istituto Dermopatico dell’Immacolata, Rome, Italy
| | - Ji-Liang Gao
- Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, and
Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA.
Istituto Dermopatico dell’Immacolata, Rome, Italy
| | - Saveria Pastore
- Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, and
Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA.
Istituto Dermopatico dell’Immacolata, Rome, Italy
| | - Stuart H. Yuspa
- Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, and
Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA.
Istituto Dermopatico dell’Immacolata, Rome, Italy
| |
Collapse
|
28
|
Panopoulos AD, Zhang L, Snow JW, Jones DM, Smith AM, El Kasmi KC, Liu F, Goldsmith MA, Link DC, Murray PJ, Watowich SS. STAT3 governs distinct pathways in emergency granulopoiesis and mature neutrophils. Blood 2006; 108:3682-90. [PMID: 16888100 PMCID: PMC1895456 DOI: 10.1182/blood-2006-02-003012] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) is essential for the host response to bacterial infection by controlling neutrophil production in the bone marrow. The G-CSF receptor (G-CSFR) activates the Jak/STAT pathway, although little is understood about how these signals regulate basal and stress-induced granulopoiesis. We examined STAT3 function in granulocytes using a bone marrow conditional knockout mouse model. Our results show that STAT3 has a crucial role in emergency granulopoiesis and mature neutrophil function. STAT3-deficient mice have an aberrant response to G-CSF in vivo, characterized by failure to accumulate immature granulocytes and an increased ratio of mature to immature neutrophils in the bone marrow, peripheral blood, and spleen. Acute neutrophil mobilization is impaired in STAT3-deficient mice as judged by their failure to up-regulate circulating neutrophils following short-term G-CSF exposure. STAT3 also controls neutrophil chemotactic responses to natural ligands for CXCR2 and regulates the magnitude of chemoattractant-induced actin polymerization. These functions of STAT3 are independent of its principal target gene Socs3, which encodes a crucial feedback inhibitor of cytokine signaling. Our results demonstrate the existence of distinct STAT3 target pathways in neutrophils required for granulopoiesis and innate immunity.
Collapse
Affiliation(s)
- Athanasia D Panopoulos
- Department of Immunology, The University of Texas M. D. Anderson Cancer Center, PO Box 301402, Unit 902, Houston, TX 77030-1903, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Gombart AF, Krug U, O'Kelly J, An E, Vegesna V, Koeffler HP. Aberrant expression of neutrophil and macrophage-related genes in a murine model for human neutrophil-specific granule deficiency. J Leukoc Biol 2005; 78:1153-65. [PMID: 16204633 DOI: 10.1189/jlb.0504286] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Neutrophil-specific granule deficiency involves inheritance of germline mutations in the CCAAT/enhancer-binding protein epsilon (C/EBPE) gene. Humans and mice lacking active C/EBPepsilon suffer frequent bacterial infections as a result of functionally defective neutrophils and macrophages. We hypothesized that these defects reflected dysregulation of important immune response genes. To test this, gene expression differences of peritoneally derived neutrophils and macrophages from C/EBPepsilon-/- and wild-type mice were determined with DNA microarrays. Of 283 genes, 146 known genes and 21 expressed sequence tags (ESTs) were down-regulated, and 85 known genes and 31 ESTs were up-regulated in the C/EBP-/- mice. These included genes involved in cell adhesion/chemotaxis, cytoskeletal organization, signal transduction, and immune/inflammatory responses. The cytokines CC chemokine ligand 4, CXC chemokine ligand 2, and interleukin (IL)-6, as well as cytokine receptors IL-8RB and granulocyte-colony stimulating factor, were down-regulated. Chromatin immunoprecipitation analysis identified binding of C/EBPepsilon to their promoter regions. Increased expression for lipid metabolism genes apolipoprotein E (APOE), scavenger receptor class B-1, sorting protein-related receptor containing low-density lipoprotein receptor class A repeat 1, and APOC2 in the C/EBPepsilon-/- mice correlated with reduced total cholesterol levels in these mice before and after maintenance on a high-fat diet. Also, C/EBPepsilon-deficient macrophages showed a reduced capacity to accumulate lipids. In summary, dysregulation of numerous, novel C/EBPepsilon target genes impairs innate immune response and possibly other important biological processes mediated by neutrophils and macrophages.
Collapse
Affiliation(s)
- Adrian F Gombart
- Cedars-Sinai Medical Center, Division of Hematology/Oncology, Burns & Allen Research Institute and David Geffen School of Medicine at University of California Los Angeles, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Gómez SA, Fernández GC, Camerano G, Dran G, Rosa FA, Barrionuevo P, Isturiz MA, Palermo MS. Endogenous glucocorticoids modulate neutrophil function in a murine model of haemolytic uraemic syndrome. Clin Exp Immunol 2005; 139:65-73. [PMID: 15606615 PMCID: PMC1809272 DOI: 10.1111/j.1365-2249.2005.02659.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Haemolytic uraemic syndrome (HUS) is caused by Shiga-toxin-producing Escherichia coli (STEC). Although, Shiga toxin type 2 (Stx2) is responsible for the renal pathogenesis observed in patients, the inflammatory response, including cytokines and polymorphonuclear neutrophils (PMN), plays a key role in the development of HUS. Previously, we demonstrated that Stx2 injection generates an anti-inflammatory reaction characterized by endogenous glucocorticoid (GC) secretion, which attenuates HUS severity in mice. Here, we analysed the effects of Stx2 on the pathogenic function of PMN and the potential role of endogenous GC to limit PMN activation during HUS development in a murine model. For this purpose we assessed the functional activity of isolated PMN after in vivo treatment with Stx2 alone or in simultaneous treatment with Ru486 (GC receptor antagonist). We found that Stx2 increased the generation of reactive oxygen intermediates (ROI) under phobol-myristate-acetate (PMA) stimulation and that the simultaneous treatment with Ru486 strengthened this effect. Conversely, both treatments significantly inhibited in vitro phagocytosis. Furthermore, Stx2 augmented in vitro PMN adhesion to fibrinogen (FGN) and bovine serum albumin (BSA) but not to collagen type I (CTI). Stx2 + Ru486 caused enhanced adhesion to BSA and CTI compared to Stx2. Whereas Stx2 significantly increased migration towards N-formyl-methionyl-leucyl-phenylalanine (fMLP), Stx2 + Ru486 treatment enhanced and accelerated this process. The percentage of apoptotic PMN from Stx2-treated mice was higher compared with controls, but equal to Stx2 + Ru486 treated mice. We conclude that Stx2 activates PMN and that the absence of endogenous GC enhances this activation suggesting that endogenous GC can, at least partially, counteract PMN inflammatory functions.
Collapse
Affiliation(s)
- S A Gómez
- Departamento de Inmunología, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Lieber JG, Webb S, Suratt BT, Young SK, Johnson GL, Keller GM, Worthen GS. The in vitro production and characterization of neutrophils from embryonic stem cells. Blood 2003; 103:852-9. [PMID: 14525782 DOI: 10.1182/blood-2003-04-1030] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
An embryonic stem (ES) cell/OP9 coculture system for the effective production of functional neutrophils is described. A 3-step differentiation strategy was developed that uses liquid culture, enabling reliable and abundant production of neutrophils at high purity without the need of sorting for isolation of mature neutrophils. Use of the OP9 stromal cell line significantly enhances the number, percentage, and duration of differentiated neutrophils produced from embryonic stem cells. Effective and sustained differentiation of ES cells into neutrophils provides a useful model system for studying neutrophil differentiation and function and the factors that regulate them. Morphologic and functional evaluation of these ES-derived neutrophils indicates that large numbers of mature neutrophils can be produced from pluripotent ES cells in vitro. Specifically, their morphology, ability to produce superoxides, flux calcium, undergo chemotaxis in response to macrophage inflammatory protein 2 (MIP-2), stain for the granulocyte-specific marker-specific chloroacetate esterase, and contain the neutrophil-specific markers Gr-1 and the mouse neutrophil-specific antigen indicates that they are comparable with purified mouse bone marrow neutrophils. They also express gelatinase and lactoferrin granule proteins. During the differentiation of these ES-derived neutrophils, regional areas of neutrophil production can be identified that have been designated as neutrophil generating regions (NGRs).
Collapse
Affiliation(s)
- Jonathan G Lieber
- National Jewish Medical and Research Center, 1400 Jackson St, Denver, CO 80206, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Adair-Kirk TL, Atkinson JJ, Broekelmann TJ, Doi M, Tryggvason K, Miner JH, Mecham RP, Senior RM. A site on laminin alpha 5, AQARSAASKVKVSMKF, induces inflammatory cell production of matrix metalloproteinase-9 and chemotaxis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:398-406. [PMID: 12817023 DOI: 10.4049/jimmunol.171.1.398] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Several peptide sequences in laminin alpha1, the alpha-chain of laminin (Ln)-1, mediate biological responses in vitro, but Ln-1 is rare in vivo. Since Ln-5 and Ln-10, which contain the alpha3 and alpha5 chains, respectively, are the most prominent laminin heterotrimers in normal adult tissues and few functional domains in other laminin chains have been identified, we are investigating the alpha3 and alpha5 chains for biological activities. Incubation of mouse macrophages with the laminin alpha5 peptide AQARSAASKVKVSMKF resulted in marked increase in matrix metalloproteinase (MMP)-9 mRNA and gelatinolytic activity in the conditioned media, whereas the corresponding alpha3 peptide QQARDAANKVAIPMRF had no effect. AQARSAASKVKVSMKF also induced expression of MMP-14, while MMP-2, MMP-3, MMP-7, MMP-12, and MMP-13 were not induced by this peptide. Deletion analyses indicated that a minimal sequence of ASKVKVSMKF was sufficient for increasing MMP-9 expression. AQARSAASKVKVSMKF was also chemotactic for neutrophils and macrophages in vitro, and induced accumulation of neutrophils and macrophages in lung airspaces in vivo following intranasal instillation into mice. Comparable accumulation occurred in MMP-9-deficient mice, indicating that MMP-9 was not required for AQARSAASKVKVSMKF-induced inflammatory cell emigration in the lung. A scrambled version of the minimal peptide, KAKSFVMVSK, was inactive. These data indicate that laminin alpha5-derived peptides can induce inflammatory cell chemotaxis and metalloproteinase activity.
Collapse
Affiliation(s)
- Tracy L Adair-Kirk
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine and Barnes-Jewish Hospital, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Frasci G. Treatment of breast cancer with chemotherapy in combination with filgrastim: approaches to improving therapeutic outcome. Drugs 2003; 62 Suppl 1:17-31. [PMID: 12479592 DOI: 10.2165/00003495-200262001-00002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Chemotherapy improves disease-free and overall survival in breast cancer, and its benefit is directly related to the percentage of the planned dose that is actually administered. In all current chemotherapeutic regimens, a substantial proportion of patients have reductions and/or delays in dosage due to side effects. In about half such cases, the delays or reductions are related to neutropenia. Overall, approximately 30% of patients have a reduction to less than 85% of the planned dosage. Women aged > or = 50 years are more likely to experience a reduction or delay in dose. Dose-intense regimens (excluding myeloablative high-dose chemotherapy) which increase the dose of chemotherapy or reduce the interval between cycles, or both, are a promising approach now under investigation. The human granulocyte colony-stimulating factor filgrastim reduces the incidence of neutropenia and facilitates adherence to full dose intensity in both standard and dose-intensified regimens. A model based on the first-cycle absolute neutrophil count nadir has been developed and validated to determine which patients should receive filgrastim. A cost benefit associated with the use of filgrastim in patients with breast cancer has been realised. This may lead to a re-evaluation of the current treatment guidelines.
Collapse
|
34
|
Schagat TL, Wofford JA, Greene KE, Wright JR. Surfactant protein A differentially regulates peripheral and inflammatory neutrophil chemotaxis. Am J Physiol Lung Cell Mol Physiol 2003; 284:L140-7. [PMID: 12388367 DOI: 10.1152/ajplung.00125.2002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Surfactant protein A (SP-A), a pulmonary lectin, plays an important role in regulating innate immune cell function. Besides accelerating pathogen clearance by pulmonary phagocytes, SP-A also stimulates alveolar macrophage chemotaxis and directed actin polymerization. We hypothesized that SP-A would also stimulate neutrophil chemotaxis. With the use of a Boyden chamber assay, we found that SP-A (0.5-25 microg/ml) did not stimulate chemotaxis of rat peripheral neutrophils or inflammatory bronchoalveolar lavage (BAL) neutrophils isolated from LPS-treated lungs. However, SP-A affected neutrophil chemotaxis toward the bacterial peptide formyl-met-leu-phe (fMLP). Surprisingly, the effect was different for the two neutrophil populations: SP-A reduced peripheral neutrophil chemotaxis toward fMLP (49 +/- 5% fMLP alone) and enhanced inflammatory BAL neutrophil chemotaxis (277 +/- 48% fMLP alone). This differential effect was not seen for the homologous proteins mannose binding lectin and complement protein 1q but was recapitulated by type IV collagen. SP-A bound both neutrophil populations comparably and did not alter formyl peptide binding. These data support a role for SP-A in regulating neutrophil migration in pulmonary tissue.
Collapse
Affiliation(s)
- Trista L Schagat
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
35
|
Turesin F, Sadr A, Davison JS, Mathison R. The tripeptide FEG ameliorates systemic inflammatory responses to rat intestinal anaphylaxis. BMC PHYSIOLOGY 2002; 2:13. [PMID: 12199907 PMCID: PMC126222 DOI: 10.1186/1472-6793-2-13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2002] [Accepted: 08/19/2002] [Indexed: 11/25/2022]
Abstract
BACKGROUND Food allergies are generally associated with gastrointestinal upset, but in many patients systemic reactions occur. However, the systemic effects of food allergies are poorly understood in experimental animals, which also offer the opportunity to explore the actions of anti-allergic drugs. The tripeptide D-phenylalanine-D-glutamate-Glycine (feG), which potentially alleviates the symptoms of systemic anaphylactic reactions, was tested to determine if it also reduced systemic inflammatory responses provoked by a gastric allergic reaction. RESULTS Optimal inhibition of intestinal anaphylaxis was obtained when 100 microg/kg of feG was given 20 min before the rats were challenged with antigen. The increase in total circulating neutrophils and accumulation of neutrophils in the heart, developing 3 h and 24 h, respectively, after antigen challenge were reduced by both feG and dexamethasone. Both anti-inflammatory agents reduced the increase in vascular permeability induced by antigen in the intestine and the peripheral skin (pinna), albeit with different time courses. Dexamethasone prevented increases in vascular permeability when given 12 h before antigen challenge, whereas feG was effective when given 20 min before ingestion of antigen. The tripeptide prevented the anaphylaxis induced up regulation of specific antibody binding of a cell adhesion molecule related to neutrophil activation, namely CD49d (alpha4 integrin). CONCLUSIONS Aside from showing that intestinal anaphylaxis produces significant systemic inflammatory responses in non-intestinal tissues, our results indicate that the tripeptide feG is a potent inhibitor of extra-gastrointestinal allergic reactions preventing both acute (30 min) and chronic (3 h or greater) inflammatory responses.
Collapse
Affiliation(s)
- Fusun Turesin
- Faculty of Medicine, Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta T2N 4N1, USA
| | - Aida Sadr
- Faculty of Medicine, Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta T2N 4N1, USA
| | - Joseph S Davison
- Faculty of Medicine, Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta T2N 4N1, USA
| | - Ronald Mathison
- Faculty of Medicine, Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta T2N 4N1, USA
| |
Collapse
|
36
|
Panopoulos AD, Bartos D, Zhang L, Watowich SS. Control of myeloid-specific integrin alpha Mbeta 2 (CD11b/CD18) expression by cytokines is regulated by Stat3-dependent activation of PU.1. J Biol Chem 2002; 277:19001-7. [PMID: 11889125 PMCID: PMC2388249 DOI: 10.1074/jbc.m112271200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) plays an essential role in regulating multiple aspects of hematopoiesis. To elucidate the role of G-CSF in controlling hematopoietic cell migration capabilities, we studied inducible expression of the myeloid-specific marker, integrin alpha(M)beta(2) (CD11b/CD18, Mac-1), in the myeloid cell line, 32D. We found that G-CSF stimulates the synthesis and cell surface expression of alpha(M) and beta(2) integrin subunits. Induction of both alpha(M) and beta(2) is dependent on Stat3, a major G-CSF-responsive signaling protein. However, the kinetics of expression suggested the involvement of an intermediate protein regulated by Stat3. Our results demonstrate that Stat3 signaling stimulates the expression of PU.1, a critical regulator of myelopoiesis. Furthermore, we show that PU.1 is an essential intermediate for the inducible expression of alpha(M)beta(2) integrin. Thus, Stat3 promotes alpha(M)beta(2) integrin expression through its activation of PU.1. These findings indicate that G-CSF-dependent Stat3 signals stimulate the changes in cell adhesion and migration capabilities that occur during myeloid cell development. These data also demonstrate a link between Stat3 and PU.1, suggesting that Stat3 may play an instructive role in hematopoiesis.
Collapse
Affiliation(s)
| | | | | | - Stephanie S. Watowich
- ‡ To whom correspondence should be addressed: University of Texas M.D. Anderson Cancer Center, Box 178, 1515 Holcombe Blvd., Houston, TX 77030. Tel.: 713-792-8376; Fax: 713-794-1322; E-mail:
| |
Collapse
|
37
|
Nagase H, Miyamasu M, Yamaguchi M, Imanishi M, Tsuno NH, Matsushima K, Yamamoto K, Morita Y, Hirai K. Cytokine‐mediated regulation of CXCR4 expression in human neutrophils. J Leukoc Biol 2002. [DOI: 10.1189/jlb.71.4.711] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Hiroyuki Nagase
- Department of Respiratory Medicine, University of Tokyo Graduate School of Medicine, Japan
| | - Misato Miyamasu
- Department of Allergy and Rheumatology, University of Tokyo Graduate School of Medicine, Japan
| | - Masao Yamaguchi
- Department of Allergy and Rheumatology, University of Tokyo Graduate School of Medicine, Japan
| | - Masako Imanishi
- Department of Bioregulatory Function, University of Tokyo Graduate School of Medicine, Japan
| | - Nelson H. Tsuno
- Department of Transfusion Medicine, University of Tokyo Graduate School of Medicine, Japan and
| | - Kouji Matsushima
- Department of Molecular Preventive Medicine and CREST, University of Tokyo Graduate School of Medicine, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, University of Tokyo Graduate School of Medicine, Japan
| | - Yutaka Morita
- Department of Respiratory Medicine, University of Tokyo Graduate School of Medicine, Japan
| | - Koichi Hirai
- Department of Bioregulatory Function, University of Tokyo Graduate School of Medicine, Japan
| |
Collapse
|
38
|
Starckx S, Van den Steen PE, Wuyts A, Van Damme J, Opdenakker G. Neutrophil gelatinase B and chemokines in leukocytosis and stem cell mobilization. Leuk Lymphoma 2002; 43:233-41. [PMID: 11999552 DOI: 10.1080/10428190290005982] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Leukocytosis is a physiopathological mechanism primarily to combat infections, whereas stem cell mobilization is induced for therapeutical purposes. Both processes are dependent on the balance between leukocyte and stem cell retention and mobilization. The retention is mediated by the specific architecture of the bone marrow, adhesion molecules and the production of chemokines in the bone marrow, which attract escaped immature cells to the marrow. Mobilization is the effect of the action of "peripheral" chemokines, such as interleukin-8 (IL-8 or CXCL8) and the remodeling of the matrix and basement membranes by matrix enzymes, such as gelatinase B (MMP-9). Recent studies lead to the conclusion that neutrophils, IL-8/CXCL8 and gelatinase B/MMP-9 play control roles in leukocytosis and stem cell mobilization. Neutrophils are the predominant circulating leukocyte type and IL-8/CXCL8 is the major neutrophil chemoattractant in humans. Gelatinase B and no gelatinase A is rapidly released from prestored granules after activation of neutrophils by IL-8/CXCL8. Moreover, neutrophils do not produce TIMP-1 and can chemically activate latent progelatinase B. Activated gelatinase B catalyses the aminoterminal truncation of IL-8/CXCL8 into a tenfold more potent chemokine. This implies that, when IL-8/CXCL8 appears in the circulation, the bone marrow is instructed to release neutrophils and concomitantly stem cells. These studies suggest that IL-8/CXCL8 and gelatinase B/MMP-9 are targets for the modulation of stem cell mobilization.
Collapse
Affiliation(s)
- S Starckx
- Laboratory of Molecular Immunology, Rega Institute, University of Leuven, Belgium
| | | | | | | | | |
Collapse
|
39
|
Sweeney EA, Lortat-Jacob H, Priestley GV, Nakamoto B, Papayannopoulou T. Sulfated polysaccharides increase plasma levels of SDF-1 in monkeys and mice: involvement in mobilization of stem/progenitor cells. Blood 2002; 99:44-51. [PMID: 11756151 DOI: 10.1182/blood.v99.1.44] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It was previously reported that treatment with the sulfated polysaccharide fucoidan or the structurally similar dextran sulfate increased circulating mature white blood cells and hematopoietic progenitor/stem cells (HPCs) in mice and nonhuman primates; however, the mechanism mediating these effects was unclear. It is reported here that plasma concentrations of the highly potent chemoattractant stromal-derived factor 1 (SDF-1) increase rapidly and dramatically after treatment with fucoidan in monkeys and in mice, coinciding with decreased levels in bone marrow. In vitro and in vivo data suggest that the SDF-1 increase is due to its competitive displacement from heparan sulfate proteoglycans that sequester the chemokine on endothelial cell surfaces or extracellular matrix in bone marrow and other tissues. Although moderately increased levels of interleukin-8, MCP1, or MMP9 were also present after fucoidan treatment, studies in gene-ablated mice (GCSFR(-/-), MCP1(-/-), or MMP9(-/-)) and the use of metalloprotease inhibitors do not support their involvement in the concurrent mobilization. Instead, SDF-1 increases, uniquely associated with sulfated glycan-mobilizing treatments and not with several other mobilizing agents tested, are likely responsible. To the authors' knowledge, this is the first published report of disrupting the SDF-1 gradient between bone marrow and peripheral blood through a physiologically relevant mechanism, resulting in mobilization with kinetics similar to other mobilizing CXC chemokines. The study further underscores the importance of the biological roles of carbohydrates.
Collapse
|
40
|
Ito T, Fujihara M, Abe H, Yamaguchi M, Wakamoto S, Takeoka S, Sakai H, Tsuchida E, Ikeda H, Ikebuchi K. Effects of poly(ethyleneglycol)-modified hemoglobin vesicles on N-formyl-methionyl-leucyl-phenylalanine-induced responses of polymorphonuclear neutrophils in vitro. ARTIFICIAL CELLS, BLOOD SUBSTITUTES, AND IMMOBILIZATION BIOTECHNOLOGY 2001; 29:427-37. [PMID: 11795629 DOI: 10.1081/bio-100108548] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
[Poly(ethyleneglycol)]-modified hemoglobin vesicles (PEG-HbV), a type of encapsulated hemoglobin, have been developed as artificial oxygen carriers and it is important to evaluate their blood compatibility. We studied the effects of PEG-HbV on human polymor phonuclear neutrophils (PMNs) in vitro, focusing on the functional responses to N-formyl-methionyl-leucyl-phenylalanine (fMLP) as an agonist. The pretreatment of the PMNs with PEG-HbV up to a concentration of 60 mg/dl Hb did not affect the fMLP-triggered chemotactic activity. In parallel to these results, the fMLP-induced upregulation of CD11b (Mac-1) levels on the PEG-HbV-pretreated PMNs was comparable to that of untreated cells. Furthermore, the pretreatment of the PMNs with the PEG-HbV even at 600 mg/dl Hb did not affect the gelatinase B (Matrix methalloproteinase-9 (MMP-9)) release, suggesting that the fMLP-induced release of secondary and tertiary granules was normal. In addition, the fMLP-triggered superoxide production of the PMNs was unchanged by the pretreatment with the PEG-HbV at 600 mg/dl Hb. Thus, these results suggest that PEG-HbV, at the concentrations studied, have no aberrant effects on the fMLP-triggered functions of human PMNs.
Collapse
Affiliation(s)
- T Ito
- Hokkaido Red Cross Blood Center, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
King AG, Horowitz D, Dillon SB, Levin R, Farese AM, MacVittie TJ, Pelus LM. Rapid mobilization of murine hematopoietic stem cells with enhanced engraftment properties and evaluation of hematopoietic progenitor cell mobilization in rhesus monkeys by a single injection of SB-251353, a specific truncated form of the human CXC chemokine GRObeta. Blood 2001; 97:1534-42. [PMID: 11238087 DOI: 10.1182/blood.v97.6.1534] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
SB-251353 is an N-terminal truncated form of the human CXC chemokine GRObeta. Recombinant SB-251353 was profiled in murine and rhesus monkey peripheral blood stem cell mobilization and transplantation models. SB-251353 rapidly and transiently mobilized hematopoietic stem cells and neutrophils into the peripheral blood after a single subcutaneous injection. Transplantation of equivalent numbers of hematopoietic stem cells mobilized by SB-251353 into lethally irradiated mice resulted in faster neutrophil and platelet recovery than stem cells mobilized by granulocyte colony-stimulating factor (G-CSF). A single injection of SB-251353 in combination with 4 days of G-CSF administration resulted in augmented stem and progenitor cell mobilization 5-fold greater than G-CSF alone. Augmented stem cell mobilization could also be demonstrated in mice when a single injection of SB-251353 was administered with only one-day treatment with G-CSF. In addition, SB-251353, when used as a single agent or in combination with G-CSF, mobilized long-term repopulating stem cells capable of hematopoietic reconstitution of lethally irradiated mice. In rhesus monkeys, a single injection of SB-251353 induced rapid increases in peripheral blood hematopoietic progenitor cells at a 50-fold lower dose than in mice, which indicates a shift in potency. These studies provide evidence that the use of SB-251353 alone or in combination with G-CSF mobilizes hematopoietic stem cells with long-term repopulating ability. In addition, this treatment may (1) reduce the number of apheresis sessions and/or amount of G-CSF required to collect adequate numbers of hematopoietic stem cells for successful peripheral blood cell transplantation and (2) improve hematopoietic recovery after transplantation.
Collapse
Affiliation(s)
- A G King
- Department of Molecular Virology and Host Defense, SmithKline Beecham Pharmaceuticals, Collegeville, PA 19426-0989, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Meuli M, Liu Y, Liggitt D, Kashani-Sabet M, Knauer S, Meuli-Simmen C, Harrison MR, Adzick NS, Heath TD, Debs RJ. Efficient gene expression in skin wound sites following local plasmid injection. J Invest Dermatol 2001; 116:131-5. [PMID: 11168808 DOI: 10.1046/j.1523-1747.2001.00139.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transfection of the skin by local gene delivery, as well as widespread transfection of systemic tissues following intravenous injection of cationic liposome/DNA complexes have been reported. Here, we show that surgically wounded mouse skin can be transfected either by local injection of DNA alone or by intravenous injection of optimized cationic liposome/DNA complexes; however, direct cutaneous injection produces much higher levels of gene expression in the skin, which is targeted to dermal and subdermal layers. High levels of chloramphenicol acetyltransferase activity were present from 3 h to 2 wk following direct injection of a gene expression plasmid into wounded skin and were maintained at detectable levels up to 8 wk after injection. Expression of transferred chloramphenicol acetyltransferase as well as beta-GAL genes was localized to fibroblasts, macrophages, and adipocytes as determined by histochemistry and immunohistochemistry. Further- more, local injection of a human granulocyte- colony-stimulating factor gene expression plasmid produced high levels of the biologically relevant human granulocyte-colony-stimulating factor protein in wounded mouse skin. This efficient and simple method of site-specific gene transfer into wounds may lead to the development of cutaneous gene therapy directed against disorders of abnormal cutaneous wound healing.
Collapse
Affiliation(s)
- M Meuli
- Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Akbarzadeh S, Layton JE. Granulocyte colony-stimulating factor receptor: Structure and function. VITAMINS & HORMONES 2001; 63:159-94. [PMID: 11358114 DOI: 10.1016/s0083-6729(01)63006-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Affiliation(s)
- S Akbarzadeh
- Ludwig Institute for Cancer Research, Melbourne Tumour Biology Branch, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | |
Collapse
|
44
|
Jilma B, Hergovich N, Homoncik M, Jilma-Stohlawetz P, Kreuzer C, Eichler HG, Zellner M, Pugin J. Granulocyte colony-stimulating factor (G-CSF) downregulates its receptor (CD114) on neutrophils and induces gelatinase B release in humans. Br J Haematol 2000; 111:314-20. [PMID: 11091218 DOI: 10.1046/j.1365-2141.2000.02320.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Despite the increasing use of granulocyte colony-stimulating factor (G-CSF) for the mobilization of stem cells and neutrophils, its pharmacodynamic actions are not fully understood. Because of the roles of G-CSF and gelatinase B in leucokinetics, we set out to characterize the interaction of G-CSF with its receptor in humans and its effects on gelatinase B release. G-CSF was infused at bolus doses of 1 microg/kg and 5 microg/kg, and compared to placebo and dexamethasone (1 mg/kg b.i.d), which enhances the plasma levels of endogenous G-CSF. The study was randomized, double-blind, four-way crossover, in eight healthy male volunteers. G-CSF dose-independently induced profound neutropenia (> 95%) within minutes and downregulated its own receptor (CD114) on neutrophils by 75%. The G-CSF/CD114 interaction dose-independently induced degranulation of neutrophils as evidenced by a 300-400% increase in CD11b expression. Degranulation induced up to a 10-fold increase in plasma levels of gelatinase B, an enzyme known to precipitate neutropenia and subsequent neutrophilia in animals. In this study, it was shown that G-CSF downmodulates CD114 expression on the surface of neutrophils in humans and the consequent degranulation enhances gelatinase B release into plasma, which may contribute to mobilization of neutrophils or stem cells.
Collapse
Affiliation(s)
- B Jilma
- Department of Clinical Pharmacology-TARGET, Vienna University School of Medicine, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Abstract
Lymphocyte-specific protein 1, recently renamed leukocyte-specific protein 1 (LSP1), is an F-actin binding protein expressed in lymphocytes, macrophages, and neutrophils in mice and humans. This study examines LSP1-deficient (Lsp1−/−) mice for the development of myeloid and lymphocytic cell populations and their response to the development of peritonitis induced by thioglycollate (TG) and to a T-dependent antigen.Lsp1−/− mice exhibit significantly higher levels of resident macrophages in the peritoneum compared to wild-type (wt) mice, whereas the development of myeloid cells is normal. This increase, which is specific for conventional CD5−macrophages appears to be tissue specific and does not result from differences in adhesion to the peritoneal mesothelium. The level of peritoneal lymphocytes is decreased inLsp1−/− mice without affecting a particular lymphocytic subset. The proportions of precursor and mature lymphocytes in the central and peripheral tissues of Lsp1−/−mice are similar to those of wt mice andLsp1−/−mice mount a normal response to the T-dependent antigen, ovalbumin (OVA). On injection of TG, theLsp1−/−mice exhibit an accelerated kinetics of changes in peritoneal macrophage and neutrophil numbers as compared to wt including increased influx of these cells. LSP1− neutrophils demonstrate an enhanced chemotactic response in vitro to N-formyl methionyl-leucyl-phenylalanine (FMLP) and to the C-X-C chemokine, KC, indicating that their enhanced influx into the peritoneum may be a result of increased motility. Our data demonstrate that LSP1 is a negative regulator of neutrophil chemotaxis.
Collapse
|
46
|
Abstract
Lymphocyte-specific protein 1, recently renamed leukocyte-specific protein 1 (LSP1), is an F-actin binding protein expressed in lymphocytes, macrophages, and neutrophils in mice and humans. This study examines LSP1-deficient (Lsp1−/−) mice for the development of myeloid and lymphocytic cell populations and their response to the development of peritonitis induced by thioglycollate (TG) and to a T-dependent antigen.Lsp1−/− mice exhibit significantly higher levels of resident macrophages in the peritoneum compared to wild-type (wt) mice, whereas the development of myeloid cells is normal. This increase, which is specific for conventional CD5−macrophages appears to be tissue specific and does not result from differences in adhesion to the peritoneal mesothelium. The level of peritoneal lymphocytes is decreased inLsp1−/− mice without affecting a particular lymphocytic subset. The proportions of precursor and mature lymphocytes in the central and peripheral tissues of Lsp1−/−mice are similar to those of wt mice andLsp1−/−mice mount a normal response to the T-dependent antigen, ovalbumin (OVA). On injection of TG, theLsp1−/−mice exhibit an accelerated kinetics of changes in peritoneal macrophage and neutrophil numbers as compared to wt including increased influx of these cells. LSP1− neutrophils demonstrate an enhanced chemotactic response in vitro to N-formyl methionyl-leucyl-phenylalanine (FMLP) and to the C-X-C chemokine, KC, indicating that their enhanced influx into the peritoneum may be a result of increased motility. Our data demonstrate that LSP1 is a negative regulator of neutrophil chemotaxis.
Collapse
|
47
|
Wooten DK, Xie X, Bartos D, Busche RA, Longmore GD, Watowich SS. Cytokine signaling through Stat3 activates integrins, promotes adhesion, and induces growth arrest in the myeloid cell line 32D. J Biol Chem 2000; 275:26566-75. [PMID: 10858439 PMCID: PMC2396147 DOI: 10.1074/jbc.m003495200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Hematopoietic cell development and function is dependent on cytokines and on intercellular interactions with the microenvironment. Although the intracellular signaling pathways stimulated by cytokine receptors are well described, little is known about the mechanisms through which these pathways modulate hematopoietic cell adhesion events in the microenvironment. Here we show that cytokine-activated Stat3 stimulates the expression and function of cell surface adhesion molecules in the myeloid progenitor cell line 32D. We generated an erythropoietin receptor (EpoR) isoform (ER343/401-S3) that activates Stat3 rather than Stat5 by substituting the Stat3 binding/activation sequence motif from gp130 for the sequences surrounding tyrosines 343 and 401 in the receptor cytoplasmic region. Activation of Stat3 leads to homotypic cell aggregation, increased expression of intercellular adhesion molecule 1 (ICAM-1), CD18, and CD11b, and activation of signaling through CD18-containing integrins. Unlike the wild type EpoR, ER343/401-S3 is unable to support long term Epo-dependent proliferation in 32D cells. Instead, Epo-treated ER343/401-S3 cells undergo G(1) arrest and express elevated levels of the cyclin-dependent kinase inhibitor p27(Kip1). Sustained activation of Stat3 in these cells is required for their altered morphology and growth properties since constitutive SOCS3 expression abrogates homotypic cell aggregation, signaling through CD18-containing integrins, G(1) arrest, and accumulation of p27(Kip1). Collectively, our results demonstrate that cytokine-activated Stat3 stimulates the expression and function of cell surface adhesion molecules, indicating that a role for Stat3 is to regulate intercellular contacts in myeloid cells.
Collapse
Affiliation(s)
- David K. Wooten
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Xiaoling Xie
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - David Bartos
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Ruth A. Busche
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Gregory D. Longmore
- Departments of Medicine and Cell Biology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Stephanie S. Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
- To whom correspondence should be addressed: The University of Texas MD Anderson Cancer Center, Box 178, 1515 Holcombe Blvd., Houston, TX 77030. Tel.: 713-792-8376; Fax: 713-794-1322; E-mail:
| |
Collapse
|
48
|
Watari K, Tojo A, Nagamura-Inoue T, Matsuoka M, Irie S, Tani K, Yamada Y, Asano S. Hyperfunction of neutrophils in a patient with BCR/ABL negative chronic myeloid leukemia. Cancer 2000. [DOI: 10.1002/1097-0142(20000801)89:3<551::aid-cncr10>3.0.co;2-e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
49
|
Betsuyaku T, Fukuda Y, Parks WC, Shipley JM, Senior RM. Gelatinase B is required for alveolar bronchiolization after intratracheal bleomycin. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 157:525-35. [PMID: 10934155 PMCID: PMC1850142 DOI: 10.1016/s0002-9440(10)64563-4] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Increased expression of matrix metalloproteinases, particularly gelatinase B (MMP-9), has been described in the lungs in pulmonary fibrosis. Intratracheal bleomycin is often used experimentally to produce lesions resembling human fibrosing alveolitis. To assess the role of gelatinase B in bleomycin-induced fibrosing alveolitis, we instilled bleomycin intratracheally into gelatinase B-deficient mice and gelatinase B+/+ littermates. Twenty-one days after bleomycin the two groups of mice were indistinguishable in terms of pulmonary histology and total lung collagen and elastin. However, the lungs of gelatinase B-deficient mice showed minimal alveolar bronchiolization, whereas bronchiolization was prominent in the lungs of gelatinase B+/+ mice. Gelatinase B was identified immunohistochemically in terminal bronchiolar cells and bronchiolized cells 7 and 14 days after bleomycin in gelatinase B+/+ mice, and whole lung gelatinase B mRNA was increased at the same times. Many bronchiolized cells displayed Clara cell features by electron microscopy. Some bronchiolized cells stained with antibody to helix transcription factor 4, a factor associated with the ciliated cell phenotype. Thus, fibrosing alveolitis develops after intratracheal bleomycin irrespective of gelatinase B. However, gelatinase B is required for alveolar bronchiolization, perhaps by facilitating migration of Clara cells and other bronchiolar cells into the regions of alveolar injury.
Collapse
Affiliation(s)
- T Betsuyaku
- Departments of Medicine, Pediatrics, and Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | |
Collapse
|
50
|
Ward AC, Loeb DM, Soede-Bobok AA, Touw IP, Friedman AD. Regulation of granulopoiesis by transcription factors and cytokine signals. Leukemia 2000; 14:973-90. [PMID: 10865962 DOI: 10.1038/sj.leu.2401808] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The development of mature granulocytes from hematopoietic precursor cells is controlled by a myriad of transcription factors which regulate the expression of essential genes, including those encoding growth factors and their receptors, enzymes, adhesion molecules, and transcription factors themselves. In particular, C/EBPalpha, PU.1, CBF, and c-Myb have emerged as critical players during early granulopoiesis. These transcription factors interact with one another as well as other factors to regulate the expression of a variety of genes important in granulocytic lineage commitment. An important goal remains to understand in greater detail how these various factors act in concert with signals emanating from cytokine receptors to influence the various steps of maturation, from the pluripotent hematopoietic stem cell, to a committed myeloid progenitor, to myeloid precursors, and ultimately to mature granulocytes.
Collapse
Affiliation(s)
- A C Ward
- Institute of Hematology, Erasmus University Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|